WO2011072243A1 - Activateurs d'histone acétyltransférase et utilisation de ceux-ci - Google Patents

Activateurs d'histone acétyltransférase et utilisation de ceux-ci Download PDF

Info

Publication number
WO2011072243A1
WO2011072243A1 PCT/US2010/059925 US2010059925W WO2011072243A1 WO 2011072243 A1 WO2011072243 A1 WO 2011072243A1 US 2010059925 W US2010059925 W US 2010059925W WO 2011072243 A1 WO2011072243 A1 WO 2011072243A1
Authority
WO
WIPO (PCT)
Prior art keywords
formula
compound
disease
body weight
mice
Prior art date
Application number
PCT/US2010/059925
Other languages
English (en)
Inventor
Yen Feng
Mauro Fa
Ottavio Arancio
Shixian Deng
Donald W. Landry
Yitshak Francis
Original Assignee
The Trustees Of Columbia University In The City Of New York
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 The Trustees Of Columbia University In The City Of New York filed Critical The Trustees Of Columbia University In The City Of New York
Priority to JP2012543311A priority Critical patent/JP6093180B2/ja
Priority to EP10836767.3A priority patent/EP2509590B1/fr
Priority to ES10836767T priority patent/ES2764999T3/es
Priority to EP19205945.9A priority patent/EP3632901B1/fr
Publication of WO2011072243A1 publication Critical patent/WO2011072243A1/fr
Priority to US13/493,490 priority patent/US10640457B2/en
Priority to US16/708,190 priority patent/US11034647B2/en
Priority to US17/317,133 priority patent/US20210317074A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/27Esters, e.g. nitroglycerine, selenocyanates of carbamic or thiocarbamic acids, meprobamate, carbachol, neostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/42Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/44Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton with carbon atoms of carboxamide groups and singly-bound oxygen atoms bound to carbon atoms of the same non-condensed six-membered aromatic ring
    • C07C235/58Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton with carbon atoms of carboxamide groups and singly-bound oxygen atoms bound to carbon atoms of the same non-condensed six-membered aromatic ring with carbon atoms of carboxamide groups and singly-bound oxygen atoms, bound in ortho-position to carbon atoms of the same non-condensed six-membered aromatic ring
    • C07C235/64Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton with carbon atoms of carboxamide groups and singly-bound oxygen atoms bound to carbon atoms of the same non-condensed six-membered aromatic ring with carbon atoms of carboxamide groups and singly-bound oxygen atoms, bound in ortho-position to carbon atoms of the same non-condensed six-membered aromatic ring having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/23Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton
    • C07C323/39Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton at least one of the nitrogen atoms being part of any of the groups, X being a hetero atom, Y being any atom
    • C07C323/40Y being a hydrogen or a carbon atom
    • C07C323/42Y being a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/23Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton
    • C07C323/39Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton at least one of the nitrogen atoms being part of any of the groups, X being a hetero atom, Y being any atom
    • C07C323/43Y being a hetero atom
    • C07C323/44X or Y being nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/50Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton
    • C07C323/62Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atom of at least one of the thio groups bound to a carbon atom of a six-membered aromatic ring of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/64Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and sulfur atoms, not being part of thio groups, bound to the same carbon skeleton
    • C07C323/67Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and sulfur atoms, not being part of thio groups, bound to the same carbon skeleton containing sulfur atoms of sulfonamide groups, bound to the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • C07D213/82Amides; Imides in position 3
    • 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
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/26Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D333/38Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals

Definitions

  • Cognitive neurodegenerative disorders are characterized by synaptic dysfunction, cognitive abnormalities, and/or the presence of inclusion bodies throughout the CNS containing, for example, but not limited to native beta-amyloid fragments, native and phosphorylated Tau, native and phosphorylated alpha-synuclein, lipofuscin, cleaved
  • AD Alzheimer's disease
  • amyloid ⁇ -peptides
  • ⁇ 42 amyloid-P-peptide 1-42
  • Histone Acetyltransferases are involved in histone acetylation (leading to gene activation), chromosome decondensation, DNA repair and non-histone substrate modification.
  • the invention is directed to compounds with histone acetyltransferase (HAT) activity, high selectivity, and blood-brain-barrier (BBB) permeability.
  • HAT histone acetyltransferase
  • BBB blood-brain-barrier
  • the compound is a compound of Formula (I),
  • R 1 is H, or CF 3 ;
  • R 2 is H, CI, or CN
  • R 3 is H, O-methyl, O-ethyl, S-ethyl, O-cyclopentyl, OCH 2 CH 2 N(CH 3 ) 2 , or CH 2 CH 2 CH 2 N(CH 3 ) 2 ;
  • R 5 is H, Ci-C 5 -alkyl, OH, OCH 3 , O-ethyl, OCH 2 CH 2 N(CH 3 ) 2 ,
  • the compound is a compound of Formula (II),
  • R is H or CF 3 ;
  • R 8 is O-ethyl or S-methyl
  • R 9 is butyl or OCH 2 CH 2 N(CH 3 ) 2 ;
  • Y is C-Cl, C-CN, C-N0 2 , or N;
  • W is CH or N
  • Z is CH or N, or a pharmaceutically acceptable salt or hydrate thereof.
  • the compound is a compound of Formula (III),
  • R 1U is CF 3
  • R 11 is CN
  • R is O-ethyl, or a pharmaceutically acceptable salt or hydrate thereof.
  • the compound is a compound of Formula (IV),
  • X is S, S(0) 2 , NH, O, or C;
  • Y is -C(O), S(0) 2 , or NH-C(O);
  • R 1 is H, Methyl, Ethyl, n-Propyl, Isopropyl, n-butyl, t-butyl,
  • R 2 is H, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, or (Ci-C 6 alkyl)-
  • R 3 is H, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, CF 3 , CC1 3 , Cl 3 ,
  • R 4 is (Ci-C 6 alkyl)-N(R 5 ) 2 - or Ci-C 6 alkyl;
  • R 5 is independently hydrogen, Ci-C 6 alkyl, or C 3 -C 8 cycloalkyl
  • R 6 is hydrogen, Ci-C 6 alkyl, or C 3 -Cg cycloalkyl, or a pharmaceutically salt or hydrate thereof.
  • the compound is a compound of Formula (V),
  • X is S, S(0) 2 , NH, O, or C;
  • Y is -C(O), S(0) 2 , or NH-C(O);
  • ARl is a 5-membered aromatic ring or a 6-membered aromatic ring
  • AR2 is a 5-membered aromatic ring, a 6-membered aromatic ring or a 6- membered aromatic ring containing 1-2 Nitrogens;
  • R 1 is H, Methyl, Ethyl, n-Propyl, Isopropyl, n-butyl, t-butyl,
  • Ci 5 H 2 8 Ci 5 H 3 o, Ci 5 H 32 , SR 4 , or OR 4 ;
  • R 2 is H, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, or (Ci-C 6 alkyl)-
  • R 3 is H, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, CF 3 , CC1 3 , Cl 3 ,
  • R 4 is (Ci-C 6 alkyl)-N(R 5 ) 2 - or Ci-C 6 alkyl; R is independently hydrogen, Ci-C 6 alkyl, or C3-C 8 cycloalkyl; and
  • R 6 is hydrogen, Ci-C 6 alkyl, or C3-C 8 cycloalkyl, or a pharmaceutically acceptable salt or hydrate thereof.
  • the compound of Formula (I) is YF2 compound:
  • R of compound 3 is H, Methyl
  • the compound of Formula (I) is:
  • the compound of Formula (II) is:
  • the compound of Formula (V) is:
  • An aspect of the invention provides a method for screening compounds of Formula (I), Formula (II), Formula (III), Formula (IV), Formula (V), or Formula (VI) to treat conditions associated with accumulated amyloid-beta peptide deposits.
  • the method comprises (a) administering a HAT Activator compound of Formula (I), Formula (II), Formula (III), Formula (IV), Formula (V), or Formula (VI)to an animal model of amyloid- beta peptide deposit accumulation; and (b) selecting a HAT Activator compound of Formula (I), Formula (II), Formula (III), Formula (IV), Formula (V), or Formula (VI) that can modulate histone acetylation after administration of the HAT Activator compound in an animal model of amyloid-beta peptide deposit accumulation.
  • An aspect of the invention further provides a method for identifying a histone acetyltransferase (HAT) activator compound of Formula (I), Formula (II), Formula (III), Formula (IV), Formula (V), or Formula (VI) to treat conditions associated with accumulated amyloid-beta peptide deposits, wherein the method comprises selecting a HAT Activator compound of Formula (I), Formula (II), Formula (III), Formula (IV), Formula (V), or Formula (VI) having one or more of the following features: (a) the EC 50 of the compound is no more than about 1000 nM; (b) the histone acetylation activity in vitro targets histone protein H2, H3, and/or H4; (c) the compound penetrates the blood brain barrier; (d) or a combination thereof.
  • HAT histone acetyltransferase
  • the compound has a molecular mass less than about 500 Da, has a polar surface area less than about 90 A 2 , has less than 8 hydrogen bonds, or a combination thereof, in order to penetrate the blood brain barrier.
  • An aspect of the invention provides a method for reducing amyloid beta ( ⁇ ) protein deposits in a subject wherein the method comprises administering to the subject an effective amount of a composition comprising a HAT Activator compound of Formula (I), Formula (II), Formula (III), Formula (IV), Formula (V), or Formula (VI), thereby decreasing ⁇ protein deposits in the subject.
  • the subject exhibits abnormally elevated levels of amyloid beta plaques.
  • the subject is afflicted with Alzheimer's disease, Lewy body dementia, inclusion body myositis, or cerebral amyloid angiopathy.
  • the ⁇ protein deposit comprises an ⁇ 40 isomer, an ⁇ 42 isomer, or a combination of isomers.
  • the compound is YF2.
  • the effective amount is at least about 1 mg/kg body weight, at least about 2 mg/kg body weight, at least about 3 mg/kg body weight, at least about 4 mg/kg body weight, at least about 5 mg/kg body weight, at least about 6 mg/kg body weight, at least about 7 mg/kg body weight, at least about 8 mg/kg body weight, at least about 9 mg/kg body weight, at least about 10 mg/kg body weight, at least about 15 mg/kg body weight, at least about 20 mg/kg body weight, at least about 25 mg/kg body weight, at least about 30 mg/kg body weight, at least about 40 mg/kg body weight, at least about 50 mg/kg body weight, at least about 75 mg/kg body weight, or at least about 100 mg/kg body weight.
  • the composition crosses the blood brain barrier.
  • the compound is compound 1, compound 2, compound 3, compound 4, compound 5, compound 6, compound 7, compound 8, compound 9, compound 10, compound 11, compound 12, compound 13, compound 14, compound 15, compound 16, compound 17, compound 18, or compound 19.
  • the compound increases histone acetylation.
  • histone acetylation comprises acetylation of histones H2B, H3, H4, or a combination thereof.
  • histone acetylation comprises acetylation of histone lysine residues H3K4, H3K9, H3K14, H4K5, H4K8, H4K12, H4K16, or a combination thereof.
  • An aspect of the invention further provides a method for treating Alzheimer's Disease in a subject, the method comprising administering to a subject a therapeutic amount of a pharmaceutical composition comprising a HAT activator compound.
  • the compound is YF2.
  • the effective amount is at least about 1 mg/kg body weight, at least about 2 mg/kg body weight, at least about 3 mg/kg body weight, at least about 4 mg/kg body weight, at least about 5 mg/kg body weight, at least about 6 mg/kg body weight, at least about 7 mg/kg body weight, at least about 8 mg/kg body weight, at least about 9 mg/kg body weight, at least about 10 mg/kg body weight, at least about 15 mg/kg body weight, at least about 20 mg/kg body weight, at least about 25 mg/kg body weight, at least about 30 mg/kg body weight, at least about 40 mg/kg body weight, at least about 50 mg/kg body weight, at least about 75 mg/kg body weight, or at least about 100 mg/kg body weight.
  • the composition crosses the blood brain barrier.
  • the compound is compound 1, compound 2, compound 3, compound 4, compound 5, compound 6, compound 7, compound 8, compound 9, compound 10, compound 11, compound 12, compound 13, compound 14, compound 15, compound 16, compound 17, compound 18, or compound 19.
  • the compound increases histone acetylation.
  • histone acetylation comprises acetylation of histones H2B, H3, H4, or a combination thereof.
  • histone acetylation comprises acetylation of histone lysine residues H3K4, H3K9, H3K14, H4K5, H4K8, H4K12, H4K16, or a combination thereof.
  • An aspect of the invention further provides a method for treating Alzheimer's Disease in a subject, the method comprising administering to a subject a therapeutic amount of a pharmaceutical composition comprising a compound of Formula (I), Formula (II), Formula (III), Formula (IV), Formula (V), or Formula (VI).
  • the compound is YF2.
  • the effective amount is at least about 1 mg/kg body weight, at least about 2 mg/kg body weight, at least about 3 mg/kg body weight, at least about 4 mg/kg body weight, at least about 5 mg/kg body weight, at least about 6 mg/kg body weight, at least about 7 mg/kg body weight, at least about 8 mg/kg body weight, at least about 9 mg/kg body weight, at least about 10 mg/kg body weight, at least about 15 mg/kg body weight, at least about 20 mg/kg body weight, at least about 25 mg/kg body weight, at least about 30 mg/kg body weight, at least about 40 mg/kg body weight, at least about 50 mg/kg body weight, at least about 75 mg/kg body weight, or at least about 100 mg/kg body weight.
  • the composition crosses the blood brain barrier.
  • the compound is compound 1, compound 2, compound 3, compound 4, compound 5, compound 6, compound 7, compound 8, compound 9, compound 10, compound 11, compound 12, compound 13, compound 14, compound 15, compound 16, compound 17, compound 18, or compound 19.
  • the compound increases histone acetylation.
  • histone acetylation comprises acetylation of histones H2B, H3, H4, or a combination thereof.
  • histone acetylation comprises acetylation of histone lysine residues H3K4, H3K9, H3K14, H4K5, H4K8, H4K12, H4K16, or a combination thereof.
  • Another aspect of the invention provides a method for increasing memory retention in a subject afflicted with a neurodegenerative disease, the method comprising administering to a subject a therapeutic amount of a pharmaceutical composition comprising a compound of Formula (I), Formula (II), Formula (III), Formula (IV), Formula (V), or Formula (VI).
  • the neurodegenerative disease comprises
  • Adrenoleukodystrophy ALD
  • Alcoholism Alexander's disease, Alper's disease, Alzheimer's disease, Amyotrophic lateral sclerosis (Lou Gehrig's Disease), Ataxia telangiectasia, Batten disease (also known as Spielmeyer-Vogt-Sjogren-Batten disease), Bovine spongiform encephalopathy (BSE), Canavan disease, Cockayne syndrome, Corticobasal degeneration, Creutzfeldt- Jakob disease, Familial fatal insomnia, Frontotemporal lobar degeneration, Huntington's disease, HIV-associated dementia, Kennedy's disease, Krabbe's disease, Lewy body dementia, Neuroborreliosis, Machado-Joseph disease (Spinocerebellar ataxia type 3), Multiple System Atrophy, Multiple sclerosis, Narcolepsy, Niemann Pick disease, Parkinson's disease, Pelizaeus-Merzbacher Disease, Pick's disease, Primary lateral s
  • the compound is YF2.
  • the effective amount is at least about 1 mg/kg body weight, at least about 2 mg/kg body weight, at least about 3 mg/kg body weight, at least about 4 mg/kg body weight, at least about 5 mg/kg body weight, at least about 6 mg/kg body weight, at least about 7 mg/kg body weight, at least about 8 mg/kg body weight, at least about 9 mg/kg body weight, at least about 10 mg/kg body weight, at least about 15 mg/kg body weight, at least about 20 mg/kg body weight, at least about 25 mg/kg body weight, at least about 30 mg/kg body weight, at least about 40 mg/kg body weight, at least about 50 mg/kg body weight, at least about 75 mg/kg body weight, or at least about 100 mg/kg body weight.
  • the composition crosses the blood brain barrier.
  • the compound is compound 1, compound 2, compound 3, compound 4, compound 5, compound 6, compound 7, compound 8, compound 9, compound 10, compound 11, compound 12, compound 13, compound 14, compound 15, compound 16, compound 17, compound 18, or compound 19.
  • the compound increases histone acetylation.
  • histone acetylation comprises acetylation of histones H2B, H3, H4, or a combination thereof.
  • histone acetylation comprises acetylation of histone lysine residues H3K4, H3K9, H3K14, H4K5, H4K8, H4K12, H4K16, or a combination thereof.
  • An aspect of the invention further provides a method for increasing synaptic plasticity in a subject afflicted with a neurodegenerative disease, the method comprising administering to a subject a therapeutic amount of a composition that increases histone acetylation in the subject, wherein the composition comprises a compound of Formula (I), Formula (II), Formula (III), Formula (IV), Formula (V), or Formula (VI).
  • a composition that increases histone acetylation in the subject, wherein the composition comprises a compound of Formula (I), Formula (II), Formula (III), Formula (IV), Formula (V), or Formula (VI).
  • the compound is YF2.
  • the effective amount is at least about 1 mg/kg body weight, at least about 2 mg/kg body weight, at least about 3 mg/kg body weight, at least about 4 mg/kg body weight, at least about 5 mg/kg body weight, at least about 6 mg/kg body weight, at least about 7 mg/kg body weight, at least about 8 mg/kg body weight, at least about 9 mg/kg body weight, at least about 10 mg/kg body weight, at least about 15 mg/kg body weight, at least about 20 mg/kg body weight, at least about 25 mg/kg body weight, at least about 30 mg/kg body weight, at least about 40 mg/kg body weight, at least about 50 mg/kg body weight, at least about 75 mg/kg body weight, or at least about 100 mg/kg body weight.
  • the composition crosses the blood brain barrier.
  • the neurodegenerative disease comprises Adrenoleukodystrophy (ALD), Alcoholism, Alexander's disease, Alper's disease, Alzheimer's disease, Amyotrophic lateral sclerosis (Lou Gehrig's Disease), Ataxia telangiectasia, Batten disease (also known as Spielmeyer-Vogt-Sjogren-Batten disease), Bovine spongiform encephalopathy (BSE), Canavan disease, Cockayne syndrome, Corticobasal degeneration, Creutzfeldt- Jakob disease, Familial fatal insomnia, Frontotemporal lobar degeneration, Huntington's disease, HIV- associated dementia, Kennedy's disease, Krabbe's disease, Lewy body dementia,
  • Neuroborreliosis Machado-Joseph disease (Spinocerebellar ataxia type 3), Multiple System Atrophy, Multiple sclerosis, Narcolepsy, Niemann Pick disease, Parkinson's disease,
  • Pelizaeus-Merzbacher Disease Pick's disease, Primary lateral sclerosis, Prion diseasesm Progressive Supranuclear Palsy, Rett's syndrome, Tau-positive FrontoTemporal dementia, Tau-negative FrontoTemporal dementia, Refsum's disease, Sandhoff disease, Schilder's disease, Subacute combined degeneration of spinal cord secondary to Pernicious Anaemia, Spielmeyer-Vogt-Sjogren-Batten disease (also known as Batten disease), Spinocerebellar ataxia (multiple types with varying characteristics), Spinal muscular atrophy, Steele- Richardson-Olszewski disease, Tabes dorsalis, or Toxic encephalopathy.
  • synaptic plasticity comprises learning, memory, or a combination thereof.
  • synaptic plasticity comprises long term potentiation (LTP).
  • the compound is compound 1, compound 2, compound 3, compound 4, compound 5, compound 6, compound 7, compound 8, compound 9, compound 10, compound 11, compound 12, compound 13, compound 14, compound 15, compound 16, compound 17, compound 18, or compound 19.
  • the compound increases histone acetylation.
  • histone acetylation comprises acetylation of histones H2B, H3, H4, or a combination thereof.
  • histone acetylation comprises acetylation of histone lysine residues H3K4, H3K9, H3K14, H4K5, H4K8, H4K12, H4K16, or a combination thereof.
  • An aspect of the invention further provides a method for treating Alzheimer's Disease in a subject, the method comprising administering to a subject a therapeutic amount of a pharmaceutical composition comprising a compound of Formula (I), Formula (II), Formula (III), Formula (IV), Formula (V), or Formula (VI).
  • the compound is YF2.
  • the effective amount is at least about 1 mg/kg body weight, at least about 2 mg/kg body weight, at least about 3 mg/kg body weight, at least about 4 mg/kg body weight, at least about 5 mg/kg body weight, at least about 6 mg/kg body weight, at least about 7 mg/kg body weight, at least about 8 mg/kg body weight, at least about 9 mg/kg body weight, at least about 10 mg/kg body weight, at least about 15 mg/kg body weight, at least about 20 mg/kg body weight, at least about 25 mg/kg body weight, at least about 30 mg/kg body weight, at least about 40 mg/kg body weight, at least about 50 mg/kg body weight, at least about 75 mg/kg body weight, or at least about 100 mg/kg body weight.
  • the composition crosses the blood brain barrier.
  • the compound is compound 1, compound 2, compound 3, compound 4, compound 5, compound 6, compound 7, compound 8, compound 9, compound 10, compound 11, compound 12, compound 13, compound 14, compound 15, compound 16, compound 17, compound 18, or compound 19.
  • the compound increases histone acetylation.
  • histone acetylation comprises acetylation of histones H2B, H3, H4, or a combination thereof.
  • histone acetylation comprises acetylation of histone lysine residues H3K4, H3K9, H3K14, H4K5, H4K8, H4K12, H4K16, or a combination thereof.
  • An aspect of the invention provides a method for ameliorating symptoms of Parkinson's Disease in a subject, the method comprising administering to a subject a therapeutic amount of a pharmaceutical composition comprising a HAT activator compound.
  • the HAT activator compound can be a compound of Formula (I), Formula (II), Formula (III), Formula (IV), Formula (V), or Formula (VI).
  • the compound is YF2.
  • the effective amount is at least about 1 mg/kg body weight, at least about 2 mg/kg body weight, at least about 3 mg/kg body weight, at least about 4 mg/kg body weight, at least about 5 mg/kg body weight, at least about 6 mg/kg body weight, at least about 7 mg/kg body weight, at least about 8 mg/kg body weight, at least about 9 mg/kg body weight, at least about 10 mg/kg body weight, at least about 15 mg/kg body weight, at least about 20 mg/kg body weight, at least about 25 mg/kg body weight, at least about 30 mg/kg body weight, at least about 40 mg/kg body weight, at least about 50 mg/kg body weight, at least about 75 mg/kg body weight, or at least about 100 mg/kg body weight.
  • the composition crosses the blood brain barrier.
  • the compound is compound 1, compound 2, compound 3, compound 4, compound 5, compound 6, compound 7, compound 8, compound 9, compound 10, compound 11, compound 12, compound 13, compound 14, compound 15, compound 16, compound 17, compound 18, or compound 19.
  • the compound increases histone acetylation.
  • histone acetylation comprises acetylation of histones H2B, H3, H4, or a combination thereof.
  • histone acetylation comprises acetylation of histone lysine residues H3K4, H3K9, H3K14, H4K5, H4K8, H4K12, H4K16, or a combination thereof.
  • the symptoms of Parkinson's Disease comprise tremor, bradykinesia, dyskinesia, rigidity, postural instability, dystonia, akathisia, dementia, impaired gross motor coordination, or a
  • the postural instability comprises impaired imbalance, impaired coordination, or a combination thereof.
  • An aspect of the invention also provides a method for treating cancer in a subject, the method comprising administering to a subject a therapeutic amount of a pharmaceutical composition comprising a compound of Formula (I), Formula (II) or Formula (III).
  • the compound is YF2.
  • the effective amount is at least about 1 mg/kg body weight, at least about 2 mg/kg body weight, at least about 3 mg/kg body weight, at least about 4 mg/kg body weight, at least about 5 mg/kg body weight, at least about 6 mg/kg body weight, at least about 7 mg/kg body weight, at least about 8 mg/kg body weight, at least about 9 mg/kg body weight, at least about 10 mg/kg body weight, at least about 15 mg/kg body weight, at least about 20 mg/kg body weight, at least about 25 mg/kg body weight, at least about 30 mg/kg body weight, at least about 40 mg/kg body weight, at least about 50 mg/kg body weight, at least about 75 mg/kg body weight, or at least about 100 mg/kg body weight.
  • the composition crosses the blood brain barrier.
  • the cancer comprises B cell lymphoma, colon cancer, lung cancer, renal cancer, bladder cancer, T cell lymphoma, myeloma, leukemia, chronic myeloid leukemia, acute myeloid leukemia, chronic lymphocytic leukemia, acute lymphocytic leukemia, hematopoietic neoplasias, thymoma, lymphoma, sarcoma, lung cancer, liver cancer, non-Hodgkins lymphoma, Hodgkins lymphoma, uterine cancer, renal cell carcinoma, hepatoma, adenocarcinoma, breast cancer, pancreatic cancer, liver cancer, prostate cancer, head and neck carcinoma, thyroid carcinoma, soft tissue sarcoma, ovarian cancer, primary or metastatic melanoma, squamous cell carcinoma, basal cell carcinoma, brain cancer, angiosarcoma, hemangiosarcoma,
  • endotheliosarcoma lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, testicular cancer, uterine cancer, cervical cancer, gastrointestinal cancer, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, Waldenstroom's macroglobulinemia, papillary adenocarcinomas,
  • cystadenocarcinoma bronchogenic carcinoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms' tumor, lung carcinoma, epithelial carcinoma, cervical cancer, testicular tumor, glioma, astrocytoma, meduUoblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, retinoblastoma, leukemia, melanoma, neuroblastoma, small cell lung carcinoma, bladder carcinoma, lymphoma, multiple myeloma, or medullary carcinoma.
  • the compound is compound 1, compound 2, compound 3, compound 4, compound 5, compound 6, compound 7, compound 8, compound 9, compound 10, compound 11, compound 12, compound 13, compound 14, compound 15, compound 16, compound 17, compound 18, or compound 19.
  • An aspect of the invention provides a method for treating Huntington's Disease in a subject, the method comprising administering to a subject a therapeutic amount of a pharmaceutical composition comprising a HAT activator compound.
  • the HAT activator compound can be a compound of Formula (I), Formula (II), Formula (III), Formula (IV), Formula (V), or Formula (VI).
  • the compound is YF2.
  • the effective amount is at least about 1 mg/kg body weight, at least about 2 mg/kg body weight, at least about 3 mg/kg body weight, at least about 4 mg/kg body weight, at least about 5 mg/kg body weight, at least about 6 mg/kg body weight, at least about 7 mg/kg body weight, at least about 8 mg/kg body weight, at least about 9 mg/kg body weight, at least about 10 mg/kg body weight, at least about 15 mg/kg body weight, at least about 20 mg/kg body weight, at least about 25 mg/kg body weight, at least about 30 mg/kg body weight, at least about 40 mg/kg body weight, at least about 50 mg/kg body weight, at least about 75 mg/kg body weight, or at least about 100 mg/kg body weight.
  • the composition crosses the blood brain barrier.
  • the compound is compound 1, compound 2, compound 3, compound 4, compound 5, compound 6, compound 7, compound 8, compound 9, compound 10, compound 11, compound 12, compound 13, compound 14, compound 15, compound 16, compound 17, compound 18, or compound 19.
  • the compound increases histone acetylation.
  • histone acetylation comprises acetylation of histones H2B, H3, H4, or a combination thereof.
  • histone acetylation comprises acetylation of histone lysine residues H3K4, H3K9, H3K14, H4K5, H4K8, H4K12, H4K16, or a combination thereof.
  • An aspect of the invention provides for a method of treating a neurodegenerative disease in a subject, the method comprising administering to a subject a therapeutic amount of a pharmaceutical composition comprising a compound of Formula (I), Formula (II), Formula (III), or Formula (V), thereby treating the neurodegenerative disease in the subject.
  • the compound is YF2.
  • the effective amount is at least about 1 mg/kg body weight, at least about 2 mg/kg body weight, at least about 3 mg/kg body weight, at least about 4 mg/kg body weight, at least about 5 mg/kg body weight, at least about 6 mg/kg body weight, at least about 7 mg/kg body weight, at least about 8 mg/kg body weight, at least about 9 mg/kg body weight, at least about 10 mg/kg body weight, at least about 15 mg/kg body weight, at least about 20 mg/kg body weight, at least about 25 mg/kg body weight, at least about 30 mg/kg body weight, at least about 40 mg/kg body weight, at least about 50 mg/kg body weight, at least about 75 mg/kg body weight, or at least about 100 mg/kg body weight.
  • the composition crosses the blood brain barrier.
  • the neurodegenerative disease comprises Adrenoleukodystrophy (ALD), Alcoholism, Alexander's disease, Alper's disease, Alzheimer's disease, Amyotrophic lateral sclerosis (Lou Gehrig's Disease), Ataxia telangiectasia, Batten disease (also known as Spielmeyer-Vogt-Sjogren-Batten disease), Bovine spongiform encephalopathy (BSE), Canavan disease, Cockayne syndrome, Corticobasal degeneration, Creutzfeldt- Jakob disease, Familial fatal insomnia, Frontotemporal lobar degeneration, Huntington's disease, HIV- associated dementia, Kennedy's disease, Krabbe's disease, Lewy body dementia,
  • Neuroborreliosis Machado-Joseph disease (Spinocerebellar ataxia type 3), Multiple System Atrophy, Multiple sclerosis, Narcolepsy, Niemann Pick disease, Parkinson's disease, Pelizaeus-Merzbacher Disease, Pick's disease, Primary lateral sclerosis, Prion diseasesm Progressive Supranuclear Palsy, Rett's syndrome, Tau-positive FrontoTemporal dementia, Tau-negative FrontoTemporal dementia, Refsum's disease, Sandhoff disease, Schilder's disease, Subacute combined degeneration of spinal cord secondary to Pernicious Anaemia, Spielmeyer-Vogt-Sjogren-Batten disease (also known as Batten disease), Spinocerebellar ataxia (multiple types with varying characteristics), Spinal muscular atrophy, Steele- Richardson-Olszewski disease, Tabes dorsalis, or Toxic encephalopathy.
  • synaptic plasticity comprises learning, memory, or a combination thereof.
  • synaptic plasticity comprises long term potentiation (LTP).
  • the compound is compound 1, compound 2, compound 3, compound 4, compound 5, compound 6, compound 7, compound 8, compound 9, compound 10, compound 11, compound 12, compound 13, compound 14, compound 15, compound 16, compound 17, compound 18, or compound 19.
  • the compound increases histone acetylation.
  • histone acetylation comprises acetylation of histones H2B, H3, H4, or a combination thereof.
  • histone acetylation comprises acetylation of histone lysine residues H3K4, H3K9, H3K14, H4K5, H4K8, H4K12, H4K16, or a combination thereof.
  • An aspect of the invention provides for a method of decreasing inclusion bodies in a subject afflicted with a neurodegenerative disorder, the method comprising administering to the subject an effective amount of a composition comprising a HAT Activator compound of Formula (I), Formula (II), Formula (III), or Formula (V), thereby decreasing inclusion bodies in the subject.
  • the inclusion bodies comprise beta-amyloid peptides, native and phosphorylated Tau proteins, native and phosphorylated alpha-synuclein, lipofuscin, cleaved TARDBP (TDB-43), or a combination thereof.
  • the subject exhibits abnormally elevated levels of amyloid beta plaques.
  • the beta-amyloid peptides comprises an ⁇ 40 isomer, an ⁇ 42 isomer, or a combination of isomers.
  • the compound is YF2.
  • the effective amount is at least about 1 mg/kg body weight, at least about 2 mg/kg body weight, at least about 3 mg/kg body weight, at least about 4 mg/kg body weight, at least about 5 mg/kg body weight, at least about 6 mg/kg body weight, at least about 7 mg/kg body weight, at least about 8 mg/kg body weight, at least about 9 mg/kg body weight, at least about 10 mg/kg body weight, at least about 15 mg/kg body weight, at least about 20 mg/kg body weight, at least about 25 mg/kg body weight, at least about 30 mg/kg body weight, at least about 40 mg/kg body weight, at least about 50 mg/kg body weight, at least about 75 mg/kg body weight, or at least about 100 mg/kg body weight.
  • the effective amount is at least about 1 mg/kg body weight, at least about 2 mg/
  • the neurodegenerative disease comprises Adrenoleukodystrophy (ALD), Alcoholism, Alexander's disease, Alper's disease, Alzheimer's disease, Amyotrophic lateral sclerosis (Lou Gehrig's Disease), Ataxia telangiectasia, Batten disease (also known as Spielmeyer-Vogt-Sjogren-Batten disease), Bovine spongiform encephalopathy (BSE), Canavan disease, Cockayne syndrome,
  • ALD Adrenoleukodystrophy
  • Alcoholism Alexander's disease
  • Alper's disease Alzheimer's disease
  • Amyotrophic lateral sclerosis Lou Gehrig's Disease
  • Ataxia telangiectasia Batten disease (also known as Spielmeyer-Vogt-Sjogren-Batten disease), Bovine spongiform encephalopathy (BSE), Canavan disease, Cockayne syndrome
  • BSE Bovine spongiform encephalopathy
  • Frontotemporal lobar degeneration Huntington's disease, HIV-associated dementia,
  • Kennedy's disease Krabbe's disease, Lewy body dementia, Neuroborreliosis, Machado- Joseph disease (Spinocerebellar ataxia type 3), Multiple System Atrophy, Multiple sclerosis, Narcolepsy, Niemann Pick disease, Parkinson's disease, Pelizaeus-Merzbacher Disease, Pick's disease, Primary lateral sclerosis, Prion diseasesm Progressive Supranuclear Palsy, Rett's syndrome, Tau-positive FrontoTemporal dementia, Tau-negative FrontoTemporal dementia, Refsum's disease, Sandhoff disease, Schilder's disease, Subacute combined degeneration of spinal cord secondary to Pernicious Anaemia, Spielmeyer-Vogt-Sjogren- Batten disease (also known as Batten disease), Spinocerebellar ataxia (multiple types with varying characteristics), Spinal muscular atrophy, Steele-Richardson-Olszewski disease, Tabes dorsalis, or Toxic encephalopathy.
  • synaptic plasticity comprises learning, memory, or a combination thereof.
  • synaptic plasticity comprises long term potentiation (LTP).
  • the compound is compound 1, compound 2, compound 3, compound 4, compound 5, compound 6, compound 7, compound 8, compound 9, compound 10, compound 11, compound 12, compound 13, compound 14, compound 15, compound 16, compound 17, compound 18, or compound 19.
  • the compound increases histone acetylation.
  • histone acetylation comprises acetylation of histones H2B, H3, H4, or a combination thereof.
  • histone acetylation comprises acetylation of histone lysine residues H3K4, H3K9, H3K14, H4K5, H4K8, H4K12, H4K16, or a combination thereof.
  • FIG. 1 is photograph of a western blot that shows histone 3 acetylation levels of mice hippocampus.
  • FIG. 2 is a bar graph showing that contextual fear conditioning of ⁇ 42 infused memory deficit mice.
  • FIG. 3 is the chemical structure of lead compound YF2, a HAT
  • FIG. 4 is the chemical structure of compound 6J (CTB), N-(4-chloro-3- trifluoromethyl-phenyl)-2-ethoxy-benzamide. 6J had no solubility and precipitated as soon as it was put in H 2 0.
  • FIG. 5 is the chemical structure of compound MOM (YF1).
  • YF1 was administered 25mg/kg to WT mice (i.p.).
  • the mice liver and hippocampus were extracted lhr after treatment. Hippocampus and liver had no increase in AcH3 levels.
  • FIG. 6 is a photograph of a western blot showing acetylation levels of H3 in the liver and hippocampus.
  • FIG. 7 is a photograph of a western blot showing acetylation levels of H3 in the liver, cortex, and hippocampus of mice that were administered by gavage and i.p. 25mg/kg of the HAT agonist, MOM. Mice were subsequently subjected to fear conditioning treatment to see if the drug is active after induction of learning.
  • FIG. 8 is a photograph of a western blot showing acetylation levels of H3 in the cortex and hippocampus. Mice were administered with MOM via cannula (100 ⁇ g/ ⁇ l per side) or mice were administered YF2 (50 mg/kg, i.p.).
  • FIG. 9 is a photograph of a western blot showing acetylation levels of H3 in the hippocampus. Mice were administered with YF2 (i.p. dissolved in saline) at 5 mg/kg, 10 mg/kg, or 20 mg/kg.
  • FIG. 10 is a bar graph demonstrating cued fear conditioning responses after administration of the HAT activator, YF2, to mice treated with amyloid-beta ( ⁇ or A-beta).
  • FIG. 11 is a bar graph demonstrating contextual fear conditioning responses after administration of the HAT activator, YF2, to mice treated with amyloid-beta ( ⁇ or A- beta).
  • FIG. 12 is a bar graph demonstrating the assessment of sensory threshold in mice treated with vehicle, YF2 alone, ⁇ alone, or YF2 plus ⁇ .
  • FIG. 13 is a graph showing the kinetics of the HAT agonist, YF2, in the blood.
  • YF2 was administered (20 mg/kg. i.p.) to mice and then blood was sampled from tails at different time points.
  • FIG. 14 is a graph showing the results from the radial arm water maze for mice that were administered the HAT agonist, YF2.
  • FIG. 15 is a graph showing the results from platform trials for mice that were administered the HAT activator, YF2.
  • FIG. 16 is a graph showing the speed of mice during the radial arm water maze that were administered the HAT activator, YF2.
  • FIG. 18 is a graph that shows that HDAC inhibition improves contextual FC in 3-4 month-old APP/PS l mice.
  • Baseline APP/PS l and WT littermates treated with TSA or vehicle show no difference in immediate freezing in the training chamber.
  • Contextual FC performed 24 hrs after training shows a reduction of freezing in APP/PSl mice treated with vehicle compared to vehicle-treated WT littermates.
  • Treatment with TSA ameliorates deficit in freezing responses in APP/PSl mice, and has no effect in WT mice.
  • * p ⁇ 0.05 vs. APP/PS 1 -vehicle. (n 13 for all groups).
  • FIG. 19 is a schematic of a CBP/Gal4 hybrid and a reporter plasmid in which luciferase expression is driven by a Gal4 yeast promoter.
  • CBP is CREB binding protein.
  • FIG. 20 are graphs that show the Role of CBP and its HAT region on transcription following PS1 stimulation.
  • FIG. 20B shows basal luciferase activity in Fl 1 cells co-transfected with 2 ⁇ g of Gal4 DNA binding sites upstream of the luciferase gene and constructs containing either WT-CBP or WY-CBP linked to the DNA binding domain of Gal4. No significant difference was found.
  • FIG. 21 shows photographs of western blots showing that endogenous expression of CBP and PCAF is reduced in APP/PSl mice. A decrease in PCAF and CBP levels, but not p300 or GCN5, was observed in ⁇ -infused mice. 4 month old APP/PSl mice displayed reduced CBP levels compared to WT controls.
  • FIG. 23 is a schematic showing that CBP is recruited to act as a bridge between DNA-bound phosphorylated CREB and the basal transcription machinery located at the start site of transcription.
  • CBP acts as a HAT, making the chromatin more accessible and increasing the transcription of memory associated genes.
  • HDACs remove an acetyl group from histones, thus restricting access of the transcriptional machinery to the DNA (figure was modified from [Bl]).
  • FIG. 24 depicts HDAC inhibition improves contextual fear conditioning in mice in which dorsal hippocampi were infused with a preparation containing oligomeric ⁇ 42.
  • FIG. 24A is a schematic representation of cannulas implanted bilaterally into the dorsal hippocampi.
  • FIG. 24B is a photograph of a Tris-Tricine PAGE Western blot depicting the analysis of ⁇ preparation in non-denaturing/non-reducing conditions, showing different bands corresponding to monomers (4.5 kDa), trimers (13.5 kDa) and tetramers (18 kDa).
  • FIG. 24A is a schematic representation of cannulas implanted bilaterally into the dorsal hippocampi.
  • FIG. 24B is a photograph of a Tris-Tricine PAGE Western blot depicting the analysis of ⁇ preparation in non-denaturing/non-reducing conditions, showing different bands corresponding to monomers (4.5 kDa), trimers (13.5
  • FIG. 25 depicts histone acetylation reduction in APP/PS1 mice.
  • FIG. 25A is photograph of a western blot (top) and the normalized results in graphic form (bottom).
  • Western blotting of protein extracts from APP/PS1 and WT mice which were injected with vehicle and TSA 2 hours before training, and euthanized 1 hour after contextual fear conditioning.
  • Vehicle-treated APP/PS1 animals showed a decrease in acetylated H4 levels.
  • TSA-treated APP/PS1 mice showed an enhancement of H4 acetylation, reaching the same levels of acetylation as TSA-treated WT mice. Results were normalized against vehicle-treated WT mice.
  • the data shown in FIG. 25 A and FIG. 25B are presented as a ratio of acetylated-H4 to total H4.
  • FIG. 26 is a synthetic scheme showing the synthesis and HAT activity assessment of the agonist MOM.
  • FIG. 26 A shows that the o-ethoxybenzolic acid was refluxed in SOCl 2 (1.66 g, 10 mmol) overnight. Removal of the organic solvent in vacuum gave the corresponding benzoyl chloride, which was dissolved in 20 mL of CH 2 C1 2 , followed by addition of 4-chloro-3-trifluoromethylaniline (1.96 g, 10 mmol). The resulting solution was put in an ice-H 2 0 bath and added 20 mL of saturated NaHC0 3 aqueous solution dropwise. The mixture was then allowed to react at room temperature for 4 hours.
  • FIG. 26B is a photograph of a western blot showing that endogenous levels of acetylated H3 are increased by MOM (100 ⁇ g in 1 ⁇ infused into dorsal hippocampi through cannulas) in mouse hippocampi which were harvested 2 hrs after the agonist administration.
  • FIG. 27 is a photograph of a western blot showing that histone acetylation is reduced in ⁇ (A-beta) infused mice. Vehicle-treated A-beta infused animals showed a decrease in acetylated H3/4 levels both before and after following associative memory.
  • FIG. 28 is a schematic of histone acetylation and the role of HATs in such.
  • YF2 is an example of a compound that activates HAT.
  • FIG. 29 is a synthetic scheme for the HAT Activator compound, YF2.
  • FIG. 30 is a schematic showing the role of a HDAC inhibitor (HDACi; e.g., TSA) in histone acetylation and deacetylation.
  • HDACi HDAC inhibitor
  • FIG. 31 are graphs showing the effect of YF2 on cancer cell viability in NCI- ADR-RES cells (FIG. 31A; ovarian cancer), U251 cells (FIG. 31B; glioblastoma cells), Hs578T cells (FIG. 31C; breast cancer cells), and CCRF-CEM cell (FIG. 31D; leukemia cells). Values represent an average of 3 wells. Error bars represent standard deviations.
  • FIGS. 32A-B are graphs showing the effect of YF2 on cancer cell proliferation in NCI-ADR-RES cells (FIG. 32A; ovarian cancer) and U251 cells (FIG. 32B; glioblastoma cells).
  • FIGS. 32C-D are graphs showing the effect of YF2 on cancer cell proliferation in Hs578T cells (FIG. 32C; breast cancer cells) and CCRF-CEM cell (FIG. 32D; leukemia cells).
  • FIG. 33 is a table showing the IC 50 measurements for YF2 and Vinblastine (VBL) on cell viability (Cell Titer Glo column) and cell proliferation (Cyquant column) in NCI-ADR-RES cells (ovarian cancer), U251 cells (glioblastoma cells), Hs578T cells (breast cancer cells), CCRF-CEM cells (leukemia cells), ACHN cells (human renal carcinoma cells), and A549 cells (human lung carcinoma cells).
  • VBL Vinblastine
  • FIG. 34 is a graph showing YF2 specificity. YF2 activates CREB Binding Protein (CBP) and the histone acetyltransferase, GCN5.
  • CBP CREB Binding Protein
  • FIG. 35 are graphs showing the effect of YF2 on cancer cell viability in ACHN cells (FIG. 35A; human renal carcinoma cells) and A549 cells (FIG. 35B; human lung carcinoma cells). Values represent an average of 3 wells. Error bars represent standard deviations.
  • FIG. 36 are graphs showing the effect of YF2 on cancer cell proliferation in ACHN cells (FIG. 36A; human renal carcinoma cells) and A549 cells (FIG. 36B; human lung carcinoma cells).
  • FIG. 37 is a synthetic scheme for the HAT Activator compound, 10.
  • FIG. 38 is a synthetic scheme for the HAT Activator compound, 20.
  • FIG. 39 is a synthetic scheme for the HAT Activator compound, 11.
  • FIG. 40 is a synthetic scheme for the HAT Activator compound, 12.
  • FIG. 41 is a synthetic scheme for the HAT Activator compound, 14.
  • FIG. 42 is a synthetic scheme for the HAT Activator compound, 13.
  • FIG. 43 is a synthetic scheme for the HAT Activator compound, 15. The synthetic scheme is continued to a second page at the hatched line.
  • FIG. 44 is a synthetic scheme for the HAT Activator compound, 16.
  • FIG. 45 is a synthetic scheme for the HAT Activator compound, 17.
  • FIG. 46 is a synthetic scheme for the HAT Activator compound, 18.
  • FIG. 47 are graphs showing the effect of Vinblastine on cancer cell viability in NCI-ADR-RES cells (FIG. 47A; ovarian cancer), U251 cells (FIG. 47B; glioblastoma cells), Hs578T cells (FIG. 47C; breast cancer cells), and CCRF-CEM cell (FIG. 47D; leukemia cells). Values represent an average of 3 wells. Error bars represent standard deviations.
  • FIG. 48 are graphs showing the effect of Vinblastine on cancer cell viability in ACHN cells (FIG. 48A; human renal carcinoma cells) and A549 cells (FIG. 48B; human lung carcinoma cells). Values represent an average of 3 wells. Error bars represent standard deviations.
  • FIGS. 49A-B are graphs showing the effect of Vinblastine on cancer cell proliferation in NCI-ADR-RES cells (FIG. 49A; ovarian cancer) and U251 cells (FIG. 49B; glioblastoma cells). Values represent an average of 3 wells. Error bars represent standard deviations.
  • FIGS. 49C-D are graphs showing the effect of Vinblastine on cancer cell proliferation in Hs578T cells (FIG. 49C; breast cancer cells) and CCRF-CEM cell (FIG. 49D; leukemia cells). Values represent an average of 3 wells. Error bars represent standard deviations.
  • FIG. 50 are graphs showing the effect of Vinblastine on cancer cell proliferation in ACHN cells (FIG. 50A; human renal carcinoma cells) and A549 cells (FIG. SOB; human lung carcinoma cells). Values represent an average of 3 wells. Error bars represent standard deviations.
  • FIG. 51 is a synthetic scheme for the HAT Activator compound, 19.
  • FIG. 52 is a graph showing the effect of OA2 (YF2 compound) on HDAC 1 activity.
  • FIG. 53 is a graph showing the effect of OA2 (YF2 compound) on
  • FIG. 54 is a graph showing the effect of OA2 (YF2 compound) on HDAC5FL activity.
  • FIG. 55 is a graph showing the effect of OA2 (YF2 compound) on HDAC7
  • FIG. 56 is a graph showing the effect of OA2 (YF2 compound) on HDAC8 activity.
  • FIG. 57 is a graph showing the effect of OA2 (YF2 compound) on HDAC 10 activity.
  • FIG. 58 is a graph showing the effect of OA2 (YF2 compound) on HDAC 11 activity.
  • FIG. 59 is a graph showing the effect of OA2 (YF2 compound) on Sirtuinl activity.
  • FIG. 60 is a graph showing the effect of OA2 (YF2 compound) on Sirtuin2 activity.
  • FIG. 61 is a graph showing the effect of the HDAC inhibitor, SAHA, on HDAC1 activity.
  • FIG. 62 is a graph showing the effect of the HDAC inhibitor, SAHA, on HDAC3/NCOR2 activity.
  • FIG. 63 is a graph showing the effect of the HDAC inhibitor, SAHA, on HDAC 6 activity.
  • 200nM in a final volume of ⁇ over lmin, 75 min before mice were sacrificed was infused through cannulas onto dorsal hippocampi.
  • FIGS. 66A-B are graphs that show the beneficial effect of YF2 on Ap 42 -induced synaptic and cognitive dysfunction.
  • YF2 ameliorates the LTP deficit in Ap 42 -treated slices (FIG. 66A; the graph represents the average of the last 5min of recording at 60min after the tetanus) P ⁇ 0.05.
  • YF2 ameliorates the contextual fear memory deficit in Ap 42 -infused mice, PO.01 (FIG. 66B).
  • FIG. 66C is a graph that shows the beneficial effect of YF2 on Ap 42 -induced cognitive dysfunction using a water maze/reference memory test.
  • YF2 ameliorates the reference memory deficit in Ap 42 -infused mice, P ⁇ 0.01.
  • FIG. 67 shows graphs depicting the beneficial effect of YF2 against memory defect in APP/PSl mice using fear conditioning or reference memory tests.
  • YF2 ameliorates the reference (FIG. 67A) and contextual fear (FIG. 67B) memory deficit in APP/PSl mice compared to WT littermates (P ⁇ 0.02 for both tests).
  • FIGS. 68A-B show that YF2 rescues the reduction in acetylated H3 levels after infusion of ⁇ 42.
  • FIG. 69 are graphs that show the Latency and speed to reach a visible platform and open field assessment of YF2 in mice infused with ⁇ 42.
  • FIG. 70 shows that histone acetylation levels are reduced in AD patients.
  • Alzheimer's disease patients showed a decrease in acetylation of histone residues important for memory.
  • FIG. 71 is a graph showing pharmacokinetic properties of YF2.
  • the amount of YF2 in the brain is higher than that in the plasma.
  • YF2 is rapidly absorbed in the brain (see Table 14).
  • the elimination half- lives of YF2 in the brain and plasma are ⁇ 40 min.
  • the distribution of YF2 to the brain is fast.
  • YF2 does not induce any adverse effects up to 300mg/kg (i.p.) in acute toxicity experiments.
  • FIG. 72A are dose-response curves for human CBP, p300, PCAF and GCN5 activation by different YF2 concentrations.
  • FIG. 72B is a table that represents the calculated EC 50 values of CBP, p300, PCAF, and GCN5.
  • FIG. 73 is a graph showing NIH cell lines growth inhibition values for 10 ⁇ YF2 treatment.
  • FIG. 74 is a graph showing the effect of OA2 (YF2 compound) on HDAC6 DETAILED DESCRIPTION OF THE INVENTION
  • AD Alzheimer's disease
  • ⁇ -amyloid a peptide that is present in high amounts in the disease
  • LTP long-term potentiation
  • Memory is known to be modulated by epigenetics through regulation of gene expression.
  • Epigenetics is defined as the mechanism that changes gene expression by 'marking' DNA or its associated proteins, through processes such as DNA methylation and histone (H) modification, without changing the DNA sequence itself ⁇ .
  • Modification of histones by, for example, the addition or removal of acetyl or methyl functional groups causes the chromatin structure to open or close, so that the information contained within the DNA is made more or less accessible to transcription factors.
  • deregulation of one of the epigenetic mechanisms might lead to memory disruption. For instance, reduction of histone acetylation causes the chromatin structure to close, so that the information contained within the DNA might be less accessible to transcription factors and memory formation ⁇ 9 .
  • HDACs histone deacetylases
  • HATs share a highly conserved motif containing an acetyl-CoA binding site. HATs can be involved in the pathology of cancer, asthma, neurodegenerative diseases and viral infection. This indicates that specific HAT activators are potential tools for
  • This invention is about the synthesis of a new class of HAT activators, which have good potency, excellent solubility, reasonable pharmacokinetic profiles and good membrane and Blood-brain-Barrier (BBB) permeability, hence can be used as a new medicine with minimum amount of side effect for patients with neurodegenerative diseases and cancers.
  • BBB Blood-brain-Barrier
  • the invention provides for compounds with histone acetyltransferase activity, HAT activation potency, high selectivity, reasonable pharmacokinetics and good
  • BBB blood-brain-barrier
  • the invention provides methods for identifying HAT activators that can acetylate histone proteins thus increasing gene expression in a subject resulting in enhanced memory and cognition.
  • the invention provides for the utilization of HAT agonists as memory enhancers in normal subjects (for example, a subject not afflicted with a neurodegenerative disease). In further embodiments, the invention provides for the utilization of HAT agonists as memory enhancers in aging subjects (for example, a subject who is >55 years old). In further embodiments, the invention provides for the utilization of HAT agonists as memory enhancers for other conditions associated with cognitive decrease/impairment.
  • Non-limiting examples of conditions associated with cognitive decrease/impairment include a variety of syndromes associated with mental retardation and syndromes associated with learning disabilities, Parkinson's disease, Pick's disease, a Lewy body disease, amyotrophic lateral sclerosis, Huntington's disease, Creutzfeld- Jakob disease, Down syndrome, multiple system atrophy, neuronal degeneration with brain iron
  • HAT activators can first be screened or selected based on their possession of certain characteristics, such as having one or more of: an EC 50 no greater than about 100 nM; a histone acetylation activity in vitro; and the ability to penetrate the BBB.
  • the candidate pool of HAT activators to be tested in animal models of neurodegenerative diseases such as, but not limited to, animals that exhibit elevated levels of inclusion bodies, for example ⁇ accumulation animal models (e.g., animal models of AD), or, for example, a mouse model for Huntington's disease, can first be screened or selected based on "medicinal chemistry" strategies. For example, based on the structure analysis of reported HAT activators, a class of structurally related, but nevertheless formally independent scaffolds, can be generated to be deemed as HAT activator candidates. Compounds derived from these scaffolds can first be screened and optimized on
  • HAT activator compound does not necessarily preclude the possibility that the compound may also be able to inhibit other HATs.
  • Eukaryotic DNA is highly organized and packaged into the nucleus.
  • the organization and packaging are achieved through the addition of proteins, including core histones H2A, H2B, H3 and H4, which form a complex structure, the chromatin, together with DNA (see FIG. 28).
  • the modification of core histones is of fundamental importance to conformational changes of the chromatin.
  • the level of acetylation is related to transcription activity, and then the acetylation induces an open chromatin confirmation that allows the transcription machinery access to promoters.
  • Histone deacetylase HDAC
  • histone acetyltransferase HAT
  • Chromatin acetylation correlates with transcriptional activity (euchromatin)
  • deacetylation correlates with gene silencing.
  • acetylation of H3 in area CA1 of the hippocampus an area in the brain that plays an important role in long-tem memory
  • Histone acetylation and deacetylation are increasingly recognized for their contribution to the tight regulation of gene activation and silencing, respectively. Hence, it is not surprising that deregulation of these mechanisms might lead to the disruption of memory- associated gene expression, resulting in a number of syndromes associated with mental retardation.
  • Histones The DNA is firstly wrapped around an octamer complex of histones (H) to form nucleosomal units, giving the appearance of beads on a string [B31]. In turn, these nucleosomal units, fold into a higher-order chromatin fiber [B32].
  • H histones
  • Each histone - octamer complex contains two copies of histones H3 and H4 bordered by two copies of histones 2A and 2B [B32].
  • HI and its avian variant H5 are linker histones that bind the nucleosome and both the entry and exit sites of the DNA, thus locking the DNA into place and allowing the formation of higher order structure.
  • histone Every histone has a globular domain, which mediates histone-histone interactions, and an N-terminal 'tail' extension.
  • the histone cores and in particular their tails, are targets for a considerable number of covalent modifications, such as acetylation, ubiquitination, sumoylation, phosphorylation,
  • Histone modifications associated with active gene transcription such as H3 Lys4 methylation and H3 Lys56 acetylation, were found to lead to gene expression.
  • histone modifications associated with the inactivation of gene transcription such as H3 Lys27 methylation and H2A Lysl 19 ubiquitination were found to cause gene silencing.
  • histone 2B, 3 and 4 because they have been shown to be involved in memory processes [B19, B25].
  • HATs and HDACs Histone modifications and their combinations have been proposed to be involved in gene regulation by modifying the chromatin accessibility and by acting as docking sites for transcription factors and modifying enzymes [B34, B35].
  • One of the most studied histone modifications is the acetylation of the evolutionary-conserved lysine residues on the histone N-termini by histone acetyltransferase (HAT).
  • HAT histone acetyltransferase
  • HDAC histone deacetylation, catalyzed by histone deacetylase
  • the HATs involved in the regulation of gene expression include at least three groups of enzymes [B37] .
  • the general control non-derepressible 5 (Gcn5) is the founding member of the Gcn5 N-acetyltransferases (GNATs).
  • GNATs Gcn5 N-acetyltransferases
  • the GNAT family members include Gcn5, PCAF, Elp3, HATlm Hpa2 and Nutl .
  • the MYST family is named after the founding members of the family: Morf, Ybf2, Sas2 and Tip60 [B37].
  • other proteins including CBP/p300, Tafl and a number of nuclear receptor co-activators have been shown to possess intrinsic HAT activity. However, these proteins do not contain a consensus domain and therefore represent an Orphan class' of HAT enzymes [B37].
  • HDACs form repressor complexes with transcription activators and with other HDACs [B38].
  • Mammalian HDACs can be divided into the classical and the silent information regulator 2 (Sir2)-related protein (sirtruin) families [B39].
  • members of the classical family have another subdivision, which include class I, II and IV, that share sequence similarity and require Zn+ for deacetylase activity.
  • Class I HDACs HDAC 1-3, HDAC8 are related to the yeast gene repressor Rpd3p, and are subunits of at least two distinct co-repressor complexes, the Sin3 complex and the NuRD complex.
  • Class II HDACs are similar to the yeast Hdalp HDAC, they act as gene repressors and have been implicated in various roles in cell differentiation and development.
  • Class IV comprises HDAC11, which has some features of both class I and II HDACs.
  • the sirtruin family includes class III HDACs (SIRTl-7), which are similar to yeast Sir2.
  • Class III HDACs are biochemically and structurally distinct from the classical family and require NAD + as a cofactor. HDACs appear to be involved in gene silencing and heterochromatin formation at centromeres and telomeres (for a review see [B40]).
  • HATs Histone Acetyltransferases
  • HDAC Histone Deacytylases
  • HATs include, but are not limited to GCN5, GCN5L, PCAF, HAT1, ELP3, HPA2, ESA1, SAS2, SAS3, TIP60, HBOl, MOZ, MORF, MOF, SRC1, SRC3, TIF2, GRIP1, ATF-2 [see Lee and Workman (2007) Nat Rev Mol Cell Biol, 8(4):284-95, Marmorstein (2001) J Molec Biol. 311 : 433- 444; and Kimura et al., (2005) J Biochem. 138(6): 647-662, which are each hereby incorporated by reference in their entireties].
  • the HAT activator compound of the invention is directed to GCN5, GCN5L, HAT1, PCAF, or a combination thereof.
  • the HAT activator compound of the invention is directed to proteins that possess intrinsic HAT activity, such as nuclear receptor co-activators (for example, CBP/p300 and Tafl).
  • nuclear receptor co-activators for example, CBP/p300 and Tafl.
  • the acetylation of H2, H3, and/or H4 histones is increased.
  • the HAT Activator compound is YF2, depicted in FIG. 3.
  • HAT activator No HAT activator is currently in drug trials, however several HDAC inhibitors are currently in clinical trials. Some of these HDAC inhibitors (HDACi) have shown therapeutic efficacy in preclinical trials. Without being bound by theory, HAT activators can be a useful drug candidate with a role similar to HDACi. However, previously available HAT activators had little solubility and membrane permeability, making them unsuitable as drugs.
  • HDACi HDACi
  • 4SC- 202 Necomed, Germany
  • AR-42 Arno therapeutics, Parsippany, NJ
  • Belinostat TopicoTarget, Rockaway, NJ
  • Entinostat Bayer Schering
  • HDAC inhibitors which include Vorinostat, Depsipeptide, and MGCD0103.
  • HDAC inhibitors in clinical use or development which include hydroxamic acid compounds (e.g., Vorinostat, Trichostatin A, LAQ824,
  • Panobinostat, Belinostat, and ITF2357 cyclic tetrapeptide compounds (e.g., Depsipeptide), benzamide compounds (e.g., Entinostat and MGCD0103), and short-chain aliphatic acid compounds (e.g., valproic acid, phenyl butyrate, and pivanex).
  • cyclic tetrapeptide compounds e.g., Depsipeptide
  • benzamide compounds e.g., Entinostat and MGCD0103
  • short-chain aliphatic acid compounds e.g., valproic acid, phenyl butyrate, and pivanex
  • HDACi are or were being developed for neurological diseases, such as an HDACi from Merck (Whitehouse Station, NJ) that is being used for the treatment of neurodegenerative diseases; and HDACi from TopoTarget (Rockaway, NJ) that was being used for the treatment of Huntington's disease, now discontinued; isovaleramide NPS-1776 (NPS Pharmaceutical, Bedminster, New Jersey) that was being used for bipolar disorder, epilepsy, and migraines, now discontinued; and a histone acetyltransferase inhibitor for cancer from TopoTarget A/S (Kobenhavn, Denmark), which was discontinued in the preclinical stage.
  • HAT activator compound of the invention YF2
  • FIG. 3 can be used as adjuvant therapy in several cancers, psychiatric and neurodegenerative diseases and may improve efficacy and safety of treatment for these disorders.
  • the HAT activator compound, YF2 has a moiety which was not mentioned in the above- referenced patent applications that significantly improves the solubility and membrane and Blood-brain-Barrier (BBB) permeability. See Abel and Zukin (2008) Current Opinion in Pharmacology 8:57-64; and Lee and Workman (2007) Nat Rev Mol Cell Biol 8:284-295.
  • a HAT activator compound can be used to treat a cancer in a subject in need thereof.
  • cancers include B cell lymphoma, colon cancer, lung cancer, renal cancer, bladder cancer, T cell lymphoma, myeloma, leukemia, chronic myeloid leukemia, acute myeloid leukemia, chronic lymphocytic leukemia, acute lymphocytic leukemia, hematopoietic neoplasias, thymoma, lymphoma, sarcoma, lung cancer, liver cancer, non-Hodgkins lymphoma, Hodgkins lymphoma, uterine cancer, renal cell carcinoma, hepatoma, adenocarcinoma, breast cancer, pancreatic cancer, liver cancer, prostate cancer, head and neck carcinoma, thyroid carcinoma, soft tissue sarcoma, ovarian cancer, primary or metastatic melanoma, squamous cell carcinoma, basal cell carcinoma,
  • endotheliosarcoma lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, testicular cancer, uterine cancer, cervical cancer, gastrointestinal cancer, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, Waldenstroom's macroglobulinemia, papillary adenocarcinomas,
  • cystadenocarcinoma bronchogenic carcinoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms' tumor, lung carcinoma, epithelial carcinoma, cervical cancer, testicular tumor, glioma, astrocytoma, meduUoblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, retinoblastoma, leukemia, melanoma, neuroblastoma, small cell lung carcinoma, bladder carcinoma, lymphoma, multiple myeloma, and medullary carcinoma.
  • a HAT activator compound can be used to treat a neurodegenerative disease in a subject in need thereof.
  • a neurodegenerative disease in a subject in need thereof.
  • neurodegenerative diseases include Adrenoleukodystrophy (ALD), Alcoholism, Alexander's disease, Alper's disease, Alzheimer's disease, Amyotrophic lateral sclerosis (Lou Gehrig's Disease), Ataxia telangiectasia, Batten disease (also known as Spielmeyer-Vogt-Sjogren- Batten disease), Bovine spongiform encephalopathy (BSE), Canavan disease, Cockayne syndrome, Corticobasal degeneration, Creutzfeldt- Jakob disease, Familial fatal insomnia, Frontotemporal lobar degeneration, Huntington's disease, HIV-associated dementia,
  • Kennedy's disease Krabbe's disease, Lewy body dementia, Neuroborreliosis, Machado- Joseph disease (Spinocerebellar ataxia type 3), Multiple System Atrophy, Multiple sclerosis, Narcolepsy, Niemann Pick disease, Parkinson's disease, Pelizaeus-Merzbacher Disease, Pick's disease, Primary lateral sclerosis, Prion diseasesm Progressive Supranuclear Palsy, Refsum's disease, Rett's syndrome, Tau-positive FrontoTemporal dementia, Tau-negative FrontoTemporal dementia, Sandhoff disease, Schilder's disease, Subacute combined degeneration of spinal cord secondary to Pernicious Anaemia, Spielmeyer-Vogt-Sjogren- Batten disease (also known as Batten disease), Spinocerebellar ataxia (multiple types with varying characteristics), Spinal muscular atrophy, Steele-Richardson-Olszewski disease, Tabes dorsalis, and Toxic encephalopathy. [00132
  • FIG. 23 A schematic representation of the processes involved in gene transcription and memory is shown in FIG. 23.
  • CBP functions as a co-activator that facilitates interactions with the basal transcription machinery and through its HAT activity catalyzes acetylation of the histones, causing a loss in chromosomal repression and increase in the transcription of memory associated genes.
  • mutations in HAT domain CBP were found to cause LTM impairment. For instance, Korzus et a ⁇ . demonstrated that inducible dominant-negative CBP mice, with no CBP HAT activity, exhibited normal short-tern memory while LTM was impaired [B8]. Moreover, the impaired LTM was rescued by suppression of the transgene expression and by HDAC inhibitor administration [B8].
  • HDAC inhibition may be beneficial in certain neurodegenerative disorders, such as Huntington's disease, spinal muscular atrophy, amyotrophic lateral sclerosis, ischemia and Rubinstein-Taybi syndrome [B19, B25, B41, B42].
  • HDAC inhibition may provide a therapeutic avenue for memory impairment in neurodegenerative diseases characterized by cognitive disorders such as AD.
  • APP(K670N:M671L) transgenic animals showed a decrease in CREB phosphorylation at the downstream level of the ⁇ -7-nicotinic-receptor (a7-nAChR)/ERK/MAPK cascade [B44, B45].
  • Perfusion of hippocampal slices with a preparation containing oligomeric ⁇ 42 revealed a reduction of the tetanus-induced increase in CREB phosphorylation, providing a clue to the mechanisms underlying the ⁇ -mediated changes in LTP [B12]. Consistent with these results ⁇ blocked the glutamate-induced increase in CREB phosphorylation in rat hippocampal cultures [Bl 1].
  • rolipram a selective PDE IV inhibitor which increases cAMP levels and therefore CREB phosphorylation, ameliorates deficits in both LTP and contextual learning in the APP/PS1 double-transgenic mouse [B46].
  • This protective effect is possibly due to CREB activation that might lead to CBP recruitment and thus histone acetylation.
  • PS cDKO conditional double knockout mice showed impaired memory and LTP that were amplified with age.
  • PS cDKO mice showed a reduction in CBP levels and in CRE-dependent gene expression in the cerebral cortex, which is likely to contribute to subsequent neuronal degeneration [B15].
  • Alzheimer's Disease An example of a Neurodegenerative Disease
  • LTM long-term memory
  • B4 synthesis of new proteins
  • B5 structural changes of the synapse
  • B6 epigenetics
  • N-terminal tails of histone proteins are known to undergo posttranslational modifications, such as histone acetylation, ubiquitination, sumoylation, phosphorylation, citrullination, ADP-ribosylation, and methylation that can dictate the transitions between transcriptionally active or
  • AD Alzheimer's disease
  • LTP long-term-potentiation
  • NOS2 NO-synthase 2
  • APP mutated amyloid precursor protein
  • AD Alzheimer's disease
  • ⁇ -amyloid
  • LTP ⁇ A3 A8 ⁇
  • is the proteolytic product of a larger precursor protein, the amyloid precursor protein (APP), which in its mutant form has been found to be implicated in familial AD (FAD) ⁇ .
  • PS1 presenilin 1
  • PS2 presenilin 2
  • AD is characterized neuropathologically by neuronal loss, extracellular senile plaques (SPs) and intracellular neurofibrillary tangles (NFTs).
  • SPs extracellular senile plaques
  • NFTs intracellular neurofibrillary tangles
  • SPs are chiefly comprised of ⁇ aggregates.
  • the major component of NFTs is the microtubule binding protein tau.
  • AD is characterized by cognitive dysfunction and begins as a synaptic disorder that involves progressively larger areas of the brain over time [SI].
  • SI synaptic disorder
  • An emerging view of the processes involved in synaptic impairment shows that the subtlety and variability of the earliest amnesic symptoms, occurring in the absence of any other clinical signs of brain injury, can be due to discrete changes in the function of a single synapse, produced at least in part, by A ⁇ [S5, S7, S10, Sl l].
  • AD Alzheimer's disease
  • CRE binding protein transcription factor CREB
  • CBP coactivator CREB binding protein
  • LTP long-term potentiation
  • NO is a central molecule in cellular biochemical processes.
  • the gas has been established as an important messenger molecule in various steps of brain physiology, from development to synaptic plasticity and learning and memory.
  • NO has been found to have a protective effect on ⁇ -induced damage of the nervous system [S38-S40].
  • the invention provides methods for identifying an agent or compound for the treatment of neurodegenerative diseases (such as AD, Huntington's Disease, Parkinson's Disease, other ⁇ -accumulation related neurodegenerative disorders or diseases characterized by elevated levels of inclusion bodies) that comprise selecting the agent or compound on the basis of having one or more characteristics that make the compound optimized for treating CNS diseases.
  • neurodegenerative diseases such as AD, Huntington's Disease, Parkinson's Disease, other ⁇ -accumulation related neurodegenerative disorders or diseases characterized by elevated levels of inclusion bodies
  • the characteristics can comprise: an ECso no greater than about 100 nM; histone acetylation activity in vitro; the ability to penetrate the BBB; or a combination thereof.
  • the HAT Activator compound is YF2, depicted in FIG. 3.
  • the invention provides methods for identifying or designing agents or compounds for the treatment of neurodegenerative diseases, treatment of conditions associated with, but not limited to, elevated inclusion bodies, (e.g., conditions associated with ⁇ , alpha-synuclein, lipofuscin, cleaved TARDBP-TDP-43, and/or Tau protein accumulation), where computer aided-medicinal chemistry methods are used to identify and/or design agents or compounds tailored to satisfy one or more of the
  • the invention generally provides methods for identifying compounds which can be used for treating a neurodegenerative disease in a subject.
  • the invention provides methods for identifying compounds which can be used for treating subjects that exhibit abnormally elevated amyloid beta plaques, or elevated Tau protein levels, or elevated alpha- synuclein levels, or inclusions, or lipofuscin level or inclusions, or cleaved TARDBP-TDP-43 level or inclusion, or accumulation of cleaved TARDBP / TDP-43 inclusions.
  • the invention provides methods for identifying compounds which can be used for the treatment of Alzheimer's disease, Lewy body dementia, inclusion body myositis, cerebral amyloid angiopathy, Huntington's Disease, Parkinson's Disease, and cancer.
  • the methods can comprise the identification of test compounds or agents (e.g., peptides (such as antibodies or fragments thereof), small molecules, nucleic acids (such as siRNA or antisense RNA), or other agents) that can bind to a HAT polypeptide molecule and/or activate or enhance the biological activity of a HAT polypeptide or its expression.
  • the compound is a HAT activator (for example a HAT activator compound having Formula (I), (II), (III), (IV), (V), or (VI).
  • the HAT Activator compound is YF2, depicted in FIG. 3.
  • modulate refers to a change in the activity or expression of a protein molecule. For example, modulation can cause an increase or a decrease in protein activity, binding characteristics, or any other biological, functional, or immunological properties of a secretase protein molecule.
  • a HAT activator compound can be a peptide fragment of a HAT protein that binds to a histone acetyltransferase protein.
  • the HAT activator molecule can encompass any portion of at least about 8 consecutive amino acids of SEQ ID NO: 1, 3, or 5.
  • the fragment can comprise at least about 10 amino acids, a least about 20 amino acids, at least about 30 amino acids, at least about 40 amino acids, a least about 50 amino acids, at least about 60 amino acids, or at least about 75 amino acids of SEQ ID NO: 1 , 3, or 5.
  • the peptide fragment is directed to a HAT protein, such as GCN5, GCN5L, HATl , or PCAF.
  • HATl The polypeptide sequence of a HAT protein, human HATl , is depicted in SEQ ID NO: 1.
  • the nucleotide sequence of human HATl is shown in SEQ ID NO: 2.
  • Sequence information related to HATl is accessible in public databases by GenBank Accession numbers NM 003642 (for mRNA) and NP 003633 (for protein).
  • HATl is also known as KATl (K(lysine) acetyltransferase 1).
  • the protein encoded by this gene is a type B histone acetyltransferase (HAT) that is involved in the rapid acetylation of newly synthesized cytoplasmic histones, which are in turn imported into the nucleus for de novo deposition onto nascent DNA chains.
  • Histone acetylation, particularly of histone H4 plays an important role in replication-dependent chromatin assembly.
  • SEQ ID NO: 1 is the human wild type amino acid sequence corresponding to the HAT protein, the HATl enzyme (residues 1-419):
  • SEQ ID NO: 2 is the human wild type nucleotide sequence corresponding to HAT protein, the HATl enzyme (residues 1-1682), wherein the underscored ATG denotes the beginning of the open reading frame:
  • polypeptide sequence of a HAT protein is depicted in SEQ ID NO: 3.
  • the nucleotide sequence of human PCAF is shown in SEQ ID NO: 4.
  • Sequence information related to PCAF is accessible in public databases by GenBank Accession numbers NM 003884 (for mRNA) and NP 003875 (for protein).
  • PCAF is also known as KAT2B (K(lysine) acetyltransferase 2B).
  • CBP and p300 are large nuclear proteins that bind to many sequence-specific factors involved in cell growth and/or differentiation, including c- jun and the adenoviral oncoprotein El A. The protein encoded by this gene associates with p300/CBP.
  • SEQ ID NO: 3 is the human wild type amino acid sequence corresponding to the HAT protein, the PCAF enzyme (residues 1-832):
  • SEQ ID NO: 4 is the human wild type nucleotide sequence corresponding to HAT protein, the PCAF enzyme (residues 1-4824), wherein the underscored ATG denotes the beginning of the open reading frame: 1 gcggaaaaga ggccgtgggg ggcctcccag cgctggcaga caccgtgagg ctggcagccg
  • the polypeptide sequence of a HAT protein, human GCN5L is depicted in SEQ ID NO: 5.
  • the nucleotide sequence of human GCN5L is shown in SEQ ID NO: 6.
  • Sequence information related to GCN5L is accessible in public databases by GenBank Accession numbers NM_021078 (for mRNA) and NP_066564.2 (for protein).
  • GCN5L is also known as KAT2A (K(lysine) acetyltransferase 2A).
  • KAT2A, or GCN5 is a histone acetyltransferase (HAT) that functions primarily as a transcriptional activator.
  • SEQ ID NO: 5 is the human wild type amino acid sequence corresponding to the HAT protein, the GCN5L enzyme (residues 1-837):
  • HAT peptide fragments can be obtained commercially or synthesized via liquid phase or solid phase synthesis methods (Atherton et al, (1989) Solid Phase Peptide Synthesis: a Practical Approach. IRL Press, Oxford, England).
  • the HAT peptide fragments can be isolated from a natural source, genetically engineered, or chemically prepared. These methods are well known in the art.
  • a HAT Activator compound can also be a protein, such as an antibody
  • An antibody fragment can be a form of an antibody other than the full-length form and includes portions or components that exist within full-length antibodies, in addition to antibody fragments that have been engineered.
  • Antibody fragments can include, but are not limited to, single chain Fv (scFv), diabodies, Fv, and (Fab') 2 , triabodies, Fc, Fab, CDR1, CDR2, CDR3, combinations of CDR's, variable regions, tetrabodies, bifunctional hybrid antibodies, framework regions, constant regions, and the like (see, Maynard et al., (2000) Ann. Rev.
  • Antibodies can be obtained commercially, custom generated, or synthesized against an antigen of interest according to methods established in the art (Janeway et al, (2001) Immunobiology, 5th ed., Garland Publishing).
  • RNA encoding a HAT protein can effectively modulate the expression of a HAT gene (e.g., GCN5, GCN5L, PCAF, or HAT1) from which the RNA is transcribed.
  • Inhibitors are selected from the group comprising: siRNA, interfering RNA or RNAi; dsRNA; RNA Polymerase III transcribed DNAs; ribozymes; and antisense nucleic acid, which can be RNA, DNA, or artificial nucleic acid.
  • Antisense oligonucleotides act to directly block the translation of mRNA by binding to targeted mRNA and preventing protein translation.
  • antisense oligonucleotides of at least about 15 bases and complementary to unique regions of the DNA sequence encoding a HAT polypeptide can be synthesized, e.g., by conventional phosphodiester techniques (Dallas et al, (2006) Med. Sci. ow*.12(4):RA67-74; Kalota et al., (2006) Handb. Exp. Pharmacol.
  • siR A comprises a double stranded structure containing from about 15 to about 50 base pairs, for example from about 21 to about 25 base pairs, and having a nucleotide sequence identical or nearly identical to an expressed target gene or RNA within the cell.
  • Antisense nucleotide sequences include, but are not limited to: morpho linos, 2'-0-methyl polynucleotides, DNA, RNA and the like.
  • RNA polymerase III transcribed DNAs contain promoters, such as the U6 promoter. These DNAs can be transcribed to produce small hairpin RNAs in the cell that can function as siRNA or linear RNAs that can function as antisense RNA.
  • the HAT activator compound can contain ribonucleotides,
  • deoxyribonucleotides synthetic nucleotides, or any suitable combination such that the target RNA and/or gene is inhibited.
  • these forms of nucleic acid can be single, double, triple, or quadruple stranded, (see for example Bass (2001) Nature, 411, 428 429; Elbashir et al, (2001) Nature, 411, 494 498; and PCT Publication Nos. WO 00/44895, WO 01/36646, WO 99/32619, WO 00/01846, WO 01/29058, WO 99/07409, WO 00/44914).
  • a HAT Activator compound can be a small molecule that binds to a histone acetyltransferase enzyme, such as GCN5, GCN5L, PCAF, or HAT1, and disrupts its function.
  • Small molecules are a diverse group of synthetic and natural substances generally having low molecular weights. They can be isolated from natural sources (for example, plants, fungi, microbes and the like), are obtained commercially and/or available as libraries or collections, or synthesized.
  • Candidate small molecules that interact with a HAT protein can be identified via in silico screening or high-through-put (HTP) screening of combinatorial libraries.
  • Identification and screening antagonists can be further facilitated by determining structural features of the protein, e.g., using X-ray crystallography, neutron diffraction, nuclear magnetic resonance spectrometry, and other techniques for structure determination. These techniques provide for the rational design or identification of antagonists, in addition to protein agonists.
  • the invention provides methods for screening and identifying compounds useful for treating a neurodegenerative disease in a subject.
  • the invention provides methods for identifying compounds which can be used for treating subjects that exhibit, for example, abnormally elevated amyloid beta plaques, or elevated Tau protein levels, or elevated alpha- synuclein levels, or inclusions, or lipofuscin level or inclusions, or cleaved TARDBP-TDP-43 level or inclusion, or accumulation of cleaved TARDBP / TDP-43 inclusions, or a
  • the method comprises selecting a HAT Activator compound that comprises one or both of the following features: (a) the EC50 of the compound is no more than about 1000 nM; (b) the compound penetrates the blood brain barrier; (c) the compound enhances histone acetylation (for example acetylates histone protein H3 or H4), or a combination thereof.
  • the compound for example the HAT Activator, has an EC50 of at least about 0.1 nM, at least about 1 nM, at least about 5 nM, at least about 10 nM, at least about 25 nM, at least about 50 nM, at least about 100 nM, at least about 200 nM, at least about 300 nM, at least about 400 nM, at least about 500 nM, at least about 600 nM, at least about 700 nM, at least about 800 nM, or at least about 900 nM.
  • the HAT Activator compound can have a molecular mass less than about 500 Da in order to penetrate the blood brain barrier.
  • the HAT Activator compound can have a polar surface area less than about 90 A 2 and should have 8 or fewer hydrogen bonds in order to penetrate the blood brain barrier.
  • the screening and identifying of the compound can comprise in silico screening, molecular docking, in vivo screening, in vitro screening, or a combination thereof.
  • Test compounds such as HAT Activator compounds
  • HAT Activator compounds can be screened from large libraries of synthetic or natural compounds (see Wang et al, (2007) Curr Med Chem, 14(2): 133-55; Mannhold (2006) Curr Top Med Chem, 6 (10): 1031-47; and Hensen (2006) Curr Med Chem 13(4) :361-76).
  • Numerous means are currently used for random and directed synthesis of saccharide, peptide, and nucleic acid based compounds.
  • Synthetic compound libraries are commercially available from Maybridge Chemical Co. (Trevillet, Cornwall, UK), Comgenex (Princeton, N. J.), Brandon Associates (Merrimack, N.H.), and Microsource (New Milford, Conn.).
  • a rare chemical library is available from Aldrich (Milwaukee, Wis.).
  • libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available from e.g. Pan Laboratories (Bothell, Wash.) or MycoSearch (N.C.), or are readily producible.
  • natural and synthetically produced libraries and compounds are readily modified through conventional chemical, physical, and biochemical means (Blondelle et al., (1996) Tib Tech 14:60).
  • Libraries of interest in the invention include peptide libraries, randomized oligonucleotide libraries, synthetic organic combinatorial libraries, and the like.
  • Degenerate peptide libraries can be readily prepared in solution, in immobilized form as bacterial flagella peptide display libraries or as phage display libraries.
  • Peptide ligands can be selected from combinatorial libraries of peptides containing at least one amino acid.
  • Libraries can be synthesized of peptoids and non-peptide synthetic moieties. Such libraries can further be synthesized which contain non-peptide synthetic moieties, which are less subject to enzymatic degradation compared to their naturally-occurring counterparts.
  • Libraries are also meant to include for example but are not limited to peptide-on-plasmid libraries, polysome libraries, aptamer libraries, synthetic peptide libraries, synthetic small molecule libraries, neurotransmitter libraries, and chemical libraries.
  • the libraries can also comprise cyclic carbon or heterocyclic structure and/or aromatic or polyaromatic structures substituted with one or more of the functional groups described herein.
  • a combinatorial library of small organic compounds is a collection of closely related analogs that differ from each other in one or more points of diversity and are synthesized by organic techniques using multi-step processes.
  • Combinatorial libraries include a vast number of small organic compounds.
  • One type of combinatorial library is prepared by means of parallel synthesis methods to produce a compound array.
  • a compound array can be a collection of compounds identifiable by their spatial addresses in Cartesian coordinates and arranged such that each compound has a common molecular core and one or more variable structural diversity elements. The compounds in such a compound array are produced in parallel in separate reaction vessels, with each compound identified and tracked by its spatial address. Examples of parallel synthesis mixtures and parallel synthesis methods are provided in U.S.
  • phage display libraries are described in Scott et al, (1990) Science 249:386-390; Devlin et al, (1990) Science, 249:404-406; Christian, et al, (1992) J. Mol. Biol. 227:711-718; Lenstra, (1992) J. Immunol. Meth. 152: 149-157; Kay et al, (1993) Gene 128:59-65; and PCT Publication No. WO 94/18318.
  • non-peptide libraries such as a benzodiazepine library (see e.g., Bunin et al, (1994) Proc. Natl. Acad. Sci. USA 91 :4708-4712), can be screened.
  • Peptoid libraries such as that described by Simon et al, (1992) Proc. Natl. Acad. Sci. USA 89:9367-9371, can also be used.
  • Another example of a library that can be used, in which the amide functionalities in peptides have been permethylated to generate a chemically transformed combinatorial library, is described by Ostresh et al. (1994), Proc. Natl. Acad. Sci. USA 91 : 11138-11142.
  • the three dimensional geometric structure of an active site for example that of a HAT polypeptide can be determined by known methods in the art, such as X-ray
  • a compound that binds to a HAT protein can be identified via: (1) providing an electronic library of test compounds; (2) providing atomic coordinates listed in PDB Entry No.
  • YGH or 2RC4 for at least 20 amino acid residues for the acetyltransferase active site of the HAT protein (the HAT domain), wherein the coordinates have a root mean square deviation therefrom, with respect to at least 50% of Ca atoms, of not greater than about 2 A, in a computer readable format; (3) converting the atomic coordinates into electrical signals readable by a computer processor to generate a three dimensional model of the HAT protein; (4) performing a data processing method, wherein electronic test compounds from the library are docked onto the three dimensional model of the HAT protein; and determining which test compound fits into the active site of the three dimensional model of the HAT protein, thereby identifying which compound would bind to a HAT protein.
  • the method can further comprise: synthesizing or obtaining the compound determined to dock to the active site of the HAT protein; contacting the HAT protein with the compound under a condition suitable for binding; and determining whether the compound modulates HAT protein expression or mRNA expression, or HAT protein activity using a diagnostic assay.
  • One method for preparing mimics of a HAT protein involves the steps of: (i) polymerization of functional monomers around a known substrate (the template) that exhibits a desired activity; (ii) removal of the template molecule; and then (iii) polymerization of a second class of monomers in, the void left by the template, to provide a new molecule which exhibits one or more desired properties which are similar to that of the template.
  • Other binding molecules such as polysaccharides, nucleosides, drugs,
  • nucleoproteins, lipoproteins, carbohydrates, glycoproteins, steroids, lipids, and other biologically active materials can also be prepared. This method is useful for designing various biological mimics that are more stable than their natural counterparts, because they are prepared by the free radical polymerization of functional monomers, resulting in a compound with a nonbiodegradable backbone. Other methods for designing such molecules include , e.g., drug design based on structure activity relationships, which require the synthesis and evaluation of a number of compounds and molecular modeling.
  • the invention also provides in vivo and in vitro methods for identifying a compound that binds to a HAT protein.
  • the method comprises: (a) obtaining a tissue and/or cells that express a HAT protein (such as GCN5, GCN5L, PCAF, or HATl); (b) contacting the tissue and/or cell with a ligand source for an effective period of time; (c) measuring a secondary messenger response, wherein the response is indicative of a ligand binding to a HAT protein; (d) isolating the ligand from the ligand source; and (e) identifying the structure of the ligand that binds a HAT protein, thereby identifying which compound would bind to a HAT protein.
  • a HAT protein such as GCN5, GCN5L, PCAF, or HATl
  • ligand source can be any compound library described herein, or a library of neurotransmitters that can be used to screen for compounds that would act as an agonist of a HAT protein (such as GCN5, GCN5L, PCAF, or HATl).
  • Screening compound libraries listed herein [also see U.S. Patent Application Publication No. 2005/0009163, which is hereby incorporated by reference in its entirety], in combination with in vivo animal studies and functional assays can be used to identify HAT Activator compounds that can be used to treat subjects afflicted with abnormal ⁇ deposits, such as AD or to treat cancer.
  • a HAT Activator compound can be a compound that increases the activity and/or expression of a HAT molecule (e.g., GCN5, GCN5L, PCAF, or HATl) in vivo and/or in vitro.
  • HAT Activator compounds can be compounds that exert their effect on the activity of a HAT protein via the expression, via post-translational modifications, or by other means.
  • a HAT Activator compound can increase HAT protein or mRNA expression, or acetyltransferase activity by at least about 10%, at least about 20%, at least about 30%), at least about 40%>, at least about 50%>, at least about 60%>, at least about 70%>, at least about 75%, at least about 80%>, at least about 85%, at least about 90%>, at least about 95%, at least about 97%, at least about 99%, or 100%.
  • Test compounds or agents which bind to a HAT molecule can be identified by various assays.
  • the assay can be a binding assay comprising direct or indirect measurement of the binding of a test compound or a known HAT ligand to the active site of a HAT protein.
  • the assay can also be an activity assay comprising direct or indirect measurement of the activity of a HAT molecule.
  • the assay can also be an expression assay comprising direct or indirect measurement of the expression of a HAT mRNA or protein.
  • the various screening assays can be combined with an in vivo assay comprising measuring the effect of the test compound on cognitive and synaptic function in an animal model for neurodegenerative disorders, such as, but not imited to, AD or
  • the diagnostic assay of the screening methods of the invention can also involve monitoring the expression of a HAT molecule.
  • inhibitors of the expression of a HAT molecule can be identified via contacting a HAT -positive cell or tissue with a test compound and determining the expression of a HAT protein or HAT mRNA in the cell.
  • the protein or mRNA expression level of a HAT molecule in the presence of the test compound is compared to the protein or mRNA expression level of a HAT protein in the absence of the test compound.
  • the test compound can then be identified as an inhibitor of expression of a HAT protein (such as GCN5, GCN5L, PCAF, or HAT1) based on this comparison.
  • Acivators of the expression of a HAT molecule can also be identified via contacting a HAT- positive cell or tissue with a test compound and determining the expression of a HAT protein or HAT mRNA in the cell.
  • the protein or mRNA expression level of a HAT molecule in the presence of the test compound is compared to the protein or mRNA expression level of a HAT protein in the absence of the test compound.
  • the test compound can then be identified as an activator of expression of a HAT protein (such as GCN5, GCN5L, PCAF, or HAT1) based on this comparison.
  • the compound when expression of HAT protein or mRNA is statistically or significantly more in the presence of the test compound than in its absence, the compound is identified as an activator of the expression of a HAT protein or mRNA.
  • the test compound can also be said to be a HAT Activator compound (such as an agonist).
  • the expression level of a HAT protein or mRNA in cells can be determined by methods described herein.
  • BIA Bimolecular Interaction Analysis
  • the invention provides for compounds that bind to a HAT activator protein, such as GCN5, GCN5L, PCAF, or HAT1. These compounds can be identified by the screening methods and assays described herein, and enhance the activity or expression of HAT activator proteins.
  • the invention encompasses a compound of Formula (I):
  • R 1 is H, or CF 3 ;
  • R 2 is H, CI, or CN
  • R 3 is H, O-methyl, O-ethyl, S-ethyl, O-cyclopentyl, OCH 2 CH 2 N(CH 3 ) 2 , or
  • R 5 is H, Ci-C 5 -alkyl, OH, OCH 3 , O-ethyl, OCH 2 CH 2 N(CH 3 ) 2 ,
  • R 6 is H, O-methyl, O-ethyl, OCH 2 CH 2 N(CH 3 )2;
  • R 1 is CF3
  • R 2 is H, CI, or CN
  • R 3 is H, 0-(Ci-C 2 )-alkyl, 0-(C 3 -C 6 )-cycloalkyl, 0-(Ci-C 2 )-alkyl-C0 2 -(Ci- C 2 )-alkyl, or S-(Ci-C 2 )-alkyl;
  • R 4 is H
  • R 5 is H, (Ci-C 6 )-alkyl, OH, OCH 2 CH 2 N(CH 3 ) 2 , S-(Ci-C 2 )-alkyl, or
  • R 6 is H, or OCH 2 CH 2 N(CH 3 ) 2 ;
  • X S0 2 NH, CONH, NHCO, or NHCONH, or a pharmaceutically acceptable salt or hydrate thereof.
  • the compound of Formula (I) is:
  • the compound of Formula (I) is N-(2-aminoethyl)-2-aminoethyl
  • R 1 is CF 3 ;
  • R 2 is CI
  • R 3 is H, 0-(Ci-C 2 )-alkyl, 0-(C 3 -C 6 )-cycloalkyl, or 0-(Ci-C 2 )-alkyl-C0 2
  • R 4 is H
  • R 5 is H, (C C 6 )-alkyl, OH, OCH 2 CH 2 N(CH 3 ) 2 , S-(C C 2 )-alkyl, or
  • R 6 is H, or OCH 2 CH 2 N(CH 3 ) 2 ;
  • X is S0 2 NH, CONH, NHCO, or NHCONH, or a pharmaceutically acceptable salt or hydrate thereof.
  • the compound of Formula (I) is:
  • R 1 is CF3
  • R 2 is H, or CN
  • R 3 is S-(Ci-C 2 )-alkyl, or OCH 2 CH 2 N(CH 3 )2;
  • R 4 is H
  • R 5 is H, or (Ci-C 4 )-alkyl
  • R 6 is H
  • X is CONH, or a pharmaceutically acceptable salt or hydrate thereof.
  • the compound of Formula (I) is:
  • Ri is CF 3 ;
  • R 2 is CI
  • R 3 is H, 0-(Ci-C 2 )-alkyl, OCH 2 CH 2 N(CH 3 ) 2 , or CH 2 CH 2 CH 2 N(CH 3 ) 2 ;
  • R4 is C(0)NH-(3-CF 3 , 4-Cl-phenyl);
  • R 5 is H, 0-(Ci-C 2 )-alkyl, OCH 2 CH 2 N(CH 3 ) 2 , or CH 2 CH 2 CH 2 N(CH 3 ) 2 ;
  • R 6 is H, or 0-(Ci-C 2 )-alkyl;
  • X CONH, or a pharmaceutically acceptable salt or hydrate thereof.
  • the compound of Formula (I) is:
  • R 1 is CF3
  • R 2 is CI
  • R 3 is 0-(Ci-C 2 )-alkyl
  • R 4 is H
  • R 5 is OCH 2 CH 2 N(CH ) 2 ;
  • X is CONH, or a pharmaceutically acceptable salt or hydrate thereof
  • R 1 is CF3
  • R 2 is CI
  • R 3 is H, 0-(Ci-C 2 )-alkyl
  • R 4 is H
  • R 5 is SCH 2 CH 2 N(CH 3 ) 2 ;
  • R 6 is H
  • X S0 2 NH, or a pharmaceutically acceptable salt or hydrate thereof
  • the invention encompasses HAT Activator compounds of Formula (II), wherein:
  • R' is H or CF 3 ;
  • R 8 is O-ethyl or S-methyl
  • R 9 is butyl or OCH 2 CH 2 N(CH 3 ) 2 ;
  • Y is C-Cl, C-CN, C-N0 2 , or N;
  • W is CH or N
  • Z is CH or N, or a pharmaceutically acceptable salt or hydrate thereof
  • the compound of Formula (II) is:
  • R 10 is CF 3 ;
  • R 11 is CN
  • R 12 is O-ethyl, or a pharmaceutically acceptable salt or hydrate thereof.
  • the invention encompasses HAT Activator compounds of Formula (IV), wherein:
  • X is S, S(0) 2 , NH, O, or C;
  • Y is -C(O), S(0) 2 , or NH-C(O);
  • R 1 is H, Methyl, Ethyl, n-Propyl, Isopropyl, n-butyl, t-butyl, C 8 Hi 8 ,Ci 5 H 26 ,
  • R 2 is H, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, or (Ci-C 6 alkyl)-
  • R 3 is H, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, CF 3 , CC1 3 , Cl 3 , F, CI, I, N0 2 , or CN;
  • R 4 is (Ci-C 6 alkyl)-N(R 5 ) 2 - or Ci-C 6 alkyl;
  • R 5 is independently hydrogen, Ci-C 6 alkyl, or C 3 -C 8 cycloalkyl; and R 6 is hydrogen, Ci-C 6 alkyl, or C 3 -C 8 cycloalkyl, or a pharmaceutically acceptable salt or hydrate thereof.
  • the compound is YF2:
  • the HAT Activator compound, YF2 can be synthesized according to the scheme depicted in FIG. 29.
  • Other analogs of HAT Activator compounds having Formula I can be similarly synthesized.
  • the HAT Activator compound, 10 can be synthesized according to the scheme depicted in FIG. 37.
  • the HAT Activator compound, 11 can be synthesized according to the scheme depicted in FIG. 39.
  • the HAT Activator compound, 12 can be synthesized according to the scheme depicted in FIG. 40.
  • the HAT Activator compound, 13, can be synthesized according to the scheme depicted in FIG. 42.
  • the HAT Activator compound, 14 can be synthesized according to the scheme depicted in FIG. 41.
  • the HAT Activator compound, 15, can be synthesized according to the scheme depicted in FIG. 43.
  • the HAT Activator compound, 16 can be synthesized according to the scheme depicted in FIG. 44.
  • the HAT Activator compound, 17, can be synthesized according to the scheme depicted in FIG. 45.
  • the HAT Activator compound, 18, can be synthesized according to the scheme depicted in FIG. 46.
  • the HAT Activator compound, 19, can be synthesized according to the scheme depicted in FIG. 51.
  • the HAT Activator compound, 20 can be synthesized according to the scheme depicted in FIG. 38.
  • the invention encompasses HAT Activator compounds of
  • X is S, S(0) 2 , NH, O, or C;
  • Y is -C(O), S(0) 2 , or NH-C(O);
  • ARl is a 5-membered aromatic ring or a 6-membered aromatic ring containing 1-2 Nitrogens
  • AR2 is a 5-membered aromatic ring, a 6-membered aromatic ring or a 6-membered aromatic ring containing 1-2 Nitrogens;
  • R 1 is H, Methyl, Ethyl, n-Propyl, Isopropyl, n-butyl, t-butyl, CisH 28 , C15H30, Ci 5 H 32 , SR 4 , or OR 4 ;
  • R 2 is H, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, or (Ci-C 6 alkyl)-C0 2 R 6 ;
  • R 3 is H, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, CF 3 , CC1 3 , Cl 3 , F, CI, I, N0 2 , or CN;
  • R 4 is (Ci-C 6 alkyl)-N(R 5 ) 2 - or C C 6 alkyl;
  • R 5 is independently hydrogen, Ci-C 6 alkyl, or C 3 -Cg cycloalkyl; and R 6 is hydrogen, Ci-C 6 alkyl, or C3-C8 cycloalkyl, or a pharmaceutically acceptable salt or hydrate thereof.
  • the compound of Formula (V) is:
  • the compounds of the invention can be generally synthesized according to the scheme based on the diagram depicted in FIG. 29.
  • a pharmaceutically acceptable salt of a compound of Formula (I) is an acid addition salt, for example a hydrochloride, sulfate, or phosphate salt.
  • a pharmaceutically acceptable salt of a compound of Formula (I) is a base addition salt, for example a sodium, potassium, calcium, or ammonium salt.
  • the base addition salt is a tetrafluoroboro salt.
  • a pharmaceutically acceptable salt of a compound of Formula (II) is an acid addition salt, for example a hydrochloride, sulfate, or phosphate salt.
  • a pharmaceutically acceptable salt of a compound of Formula (II) is an acid addition salt, for example a hydrochloride, sulfate, or phosphate salt.
  • a pharmaceutically acceptable salt of a compound of Formula (II) is an acid addition salt, for example a hydrochloride, sulfate, or phosphate salt.
  • a pharmaceutically acceptable salt of a compound of Formula (II)
  • a pharmaceutically acceptable salt of a compound of Formula (II) is a base addition salt, for example a sodium, potassium, calcium, or ammonium salt.
  • the base addition salt is a tetrafluoroboro salt.
  • a pharmaceutically acceptable salt of a compound of Formula (III) is an acid addition salt, for example a hydrochloride, sulfate, or phosphate salt.
  • a pharmaceutically acceptable salt of a compound of Formula (III) is a base addition salt, for example a sodium, potassium, calcium, or ammonium salt.
  • the base addition salt is a tetrafluoroboro salt.
  • a pharmaceutically acceptable salt of a compound of Formula (IV) is an acid addition salt, for example a hydrochloride, sulfate, or phosphate salt.
  • a pharmaceutically acceptable salt of a compound of Formula (IV) is a base addition salt, for example a sodium, potassium, calcium, or ammonium salt.
  • the base addition salt is a tetrafluoroboro salt.
  • a pharmaceutically acceptable salt of a compound of Formula (V) is an acid addition salt, for example a hydrochloride, sulfate, or phosphate salt.
  • a pharmaceutically acceptable salt of a compound of Formula (IV) is an acid addition salt, for example a hydrochloride, sulfate, or phosphate salt.
  • a pharmaceutically acceptable salt of a compound of Formula (IV) is an acid addition salt, for example a hydrochloride, sulfate, or phosphate salt.
  • a pharmaceutically acceptable salt of a compound of Formula (IV) is an acid addition
  • pharmaceutically acceptable salt of a compound of Formula (V) is a base addition salt, for example a sodium, potassium, calcium, or ammonium salt.
  • the base addition salt is a tetrafluoroboro salt.
  • the invention provides methods for reducing inclusion bodies (e.g., amyloid beta ( ⁇ ) protein deposits, native and phosphorylated Tau proteins, native and
  • a neurodegenerative disease e.g., a AD, Huntington's Disease, or Parkinson's Disease
  • a neurodegenerative disease e.g., a AD, Huntington's Disease, or Parkinson's Disease
  • the invention also provides methods for treating a neurodegenerative disease in a subject by administering any one of the HAT Activator compounds having Formula (I), Formula (II), Formula (III), Formula (IV), or Formula (V).
  • the invention further provides methods for treating cancer in a subject by administering any one of the HAT Activator compounds having Formula (I), Formula (II), Formula (III), Formula (IV), or Formula (V).
  • the compound administered to a subject is any one of compounds 1-9.
  • the HAT Activator compound is YF2, depicted in FIG. 3.
  • the compound administered is a HAT Activator compound of Formula (VI):
  • R 1 is H, Methyl, Ethyl, n-Propyl, Isopropyl, n-butyl, t-butyl, C 8 Hi 8 ,Ci 5 H 2 6, CisH 28 , Ci 5 H 30 , Ci 5 H 32 , S, O, SR 4 , or OR 4 ;
  • R 2 is H, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, or -C(0)R 6 ;
  • R 3 is H, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, CF 3 , CC1 3 , Cl 3 , F, CI, I, N0 2 , or CN;
  • R 4 is -NR 5 ;
  • R 5 is hydrogen, Ci-C 6 alkyl, or C 3 -Cg cycloalkyl
  • R 6 is hydrogen, C C 6 alkyl, or C 3 -C 8 cycloalkyl, C(0)R 7 ;
  • R 7 is hydrogen, Ci-C 6 alkyl, or C 3 -Cg cycloalkyl, or a pharmaceutically acceptable salt or hydrate thereof.
  • a pharmaceutically acceptable salt of a compound of Formula (VI) is an acid addition salt, for example a hydrochloride, sulfate, or phosphate salt.
  • a pharmaceutically acceptable salt of a compound of Formula (VI) is a base addition salt, for example a sodium, potassium, calcium, or ammonium salt.
  • the base addition salt is a tetrafluoroboro salt.
  • HAT activator compounds having Formula (I), Formula (II), Formula (III), Formula (IV), Formula (V), or Formula (VI) are first screened for their ability to satisfy one or more of the following characteristics: an EC 50 no greater than about 100 nM; a histone acetylation activity in vitro; the ability to penetrate the BBB; or a combination thereof.
  • the method comprises administering to the subject an effective amount of a composition comprising a HAT Activator compound.
  • the subject exhibits abnormally elevated amyloid beta plaques, or elevated Tau protein levels, or accumulations of alpha-synuclein, or accumulations of lipofuscin, or accumulation of cleaved TARDBP (TDB-43) levels, or a combination thereof.
  • the ⁇ protein deposit comprises an ⁇ 40 isomer, an ⁇ 42 isomer, or a combination thereof.
  • the subject is afflicted with Alzheimer's disease, Lewy body dementia, inclusion body myositis, Huntington's Disease, Parkinson's Disease, or cerebral amyloid angiopathy. In further embodiments, the subject is afflicted with cancer.
  • the dosage administered can be a therapeutically effective amount of the composition sufficient to result in amelioration of symptoms of a neurogenerative disease such as, but not limited to reducing inclusion bodies (e.g., amyloid beta ( ⁇ ) protein deposits, native and phosphorylated Tau proteins, native and phosphorylated alpha-synuclein, lipofuscin, cleaved TARDBP (TDB-43), or a combination thereof), or reducing memory loss in a subject.
  • reducing inclusion bodies e.g., amyloid beta ( ⁇ ) protein deposits, native and phosphorylated Tau proteins, native and phosphorylated alpha-synuclein, lipofuscin, cleaved TARDBP (TDB-43), or a combination thereof
  • reducing inclusion bodies e.g., amyloid beta ( ⁇ ) protein deposits, native and phosphorylated Tau proteins, native and phosphorylated alpha-synuclein, lipofuscin, cleaved TARDBP (TDB-43
  • observing at least, about a 25% reduction, at least about a 30% reduction, at least about a 40% reduction, at least about a 50% reduction, at least about a 60% reduction, at least about a 70%> reduction, at least about a 80%> reduction, at least about a 85% reduction, at least about a 90%> reduction, at least about a 95% reduction, at least about a 97% reduction, at least about a 98% reduction, or a 100% reduction in inclusion bodies or memory loss in a subject is indicative of amelioration of symptoms of a neurogenerative disease (for example, including, but not limited to, AD, Huntington's Disease, Parkinson's Disease).
  • This efficacy in reducing inclusion occurrence can be, for example, a meaure of ameliorating symptoms of a neurogenerative disease.
  • the therapeutically effective amount is at least about 0.1 mg/kg body weight, at least about 0.25 mg/kg body weight, at least about 0.5 mg/kg body weight, at least about 0.75 mg/kg body weight, at least about 1 mg/kg body weight, at least about 2 mg/kg body weight, at least about 3 mg/kg body weight, at least about 4 mg/kg body weight, at least about 5 mg/kg body weight, at least about 6 mg/kg body weight, at least about 7 mg/kg body weight, at least about 8 mg/kg body weight, at least about 9 mg/kg body weight, at least about 10 mg/kg body weight, at least about 15 mg/kg body weight, at least about 20 mg/kg body weight, at least about 25 mg/kg body weight, at least about 30 mg/kg body weight, at least about 40 mg/kg body weight, at least about 50 mg/kg body weight, at least about 75 mg/kg body weight, at least about 100 mg/kg body weight, at least about 200 mg/kg body weight, at least about 250 mg/
  • a HAT activator compound can be administered to the subject one time (e.g., as a single injection or deposition).
  • a HAT activator compound of the invention can be administered once or twice daily to a subject in need thereof for a period of from about 2 to about 28 days, or from about 7 to about 10 days, or from about 7 to about 15 days. It can also be administered once or twice daily to a subject for a period of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 times per year, or a combination thereof.
  • the dosage administered can vary depending upon known factors such as the pharmacodynamic characteristics of the active ingredient and its mode and route of administration; time of administration of active ingredient; age, sex, health and weight of the recipient; nature and extent of symptoms; kind of concurrent treatment, frequency of treatment and the effect desired; and rate of excretion.
  • Toxicity and therapeutic efficacy of therapeutic compositions of the present invention can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD 50 /ED 50 .
  • Therapeutic agents that exhibit large therapeutic indices are useful.
  • Therapeutic compositions that exhibit some toxic side effects can be used.
  • a therapeutically effective dose of a HAT activator compound can depend upon a number of factors known to those of ordinary skill in the art.
  • the dose(s) of a HAT activator compound for example a compound of Formula (I), Formula (II), Formula (III), Formula (IV), Formula (V), or Formula (VI), can vary, for example, depending upon the identity, size, and condition of the subject or sample being treated, further depending upon the route by which the composition is to be administered, if applicable, and the effect which the practitioner desires the HAT activator compound to have upon a HAT protein or a protein exhibiting intrinsic HAT activity. These amounts can be readily determined by a skilled artisan.
  • HAT activator compounds of the invention can be incorporated into
  • compositions suitable for administration can comprise a HAT activator compound (e.g., a compound of HAT activator compounds having Formula (I), Formula (II), Formula (III), Formula (IV), Formula (V), or Formula (VI), or compound YF2 depicted in FIG. 3) and a pharmaceutically acceptable carrier.
  • a HAT activator compound e.g., a compound of HAT activator compounds having Formula (I), Formula (II), Formula (III), Formula (IV), Formula (V), or Formula (VI), or compound YF2 depicted in FIG.
  • the compositions can be administered alone or in combination with at least one other agent, such as a stabilizing compound, which can be administered in any sterile, biocompatible pharmaceutical carrier including, but not limited to, saline, buffered saline, dextrose, and water.
  • the compositions can be administered to a patient alone, or in combination with other agents, drugs or hormones.
  • a pharmaceutically acceptable carrier can comprise any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Any conventional media or agent that is compatible with the active compound can be used. Supplementary active compounds can also be incorporated into the compositions.
  • any of the therapeutic applications described herein can be applied to any subject in need of such therapy, including, for example, a mammal such as a dog, a cat, a cow, a horse, a rabbit, a monkey, a pig, a sheep, a goat, or a human.
  • a mammal such as a dog, a cat, a cow, a horse, a rabbit, a monkey, a pig, a sheep, a goat, or a human.
  • a pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration.
  • routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor EMTM (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS).
  • the composition must be sterile and should be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, a pharmaceutically acceptable polyol like glycerol, propylene glycol, liquid polyetheylene glycol, and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, and thimerosal. In many cases, it can be useful to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the HAT Activator compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated herein, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated herein.
  • examples of useful preparation methods are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed.
  • compositions can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch
  • a lubricant such as magnesium stearate or sterotes
  • Systemic administration can also be by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
  • Beta-amyloid activates the mitogen- activated protein kinase cascade via hippocampal alpha7 nicotinic acetylcholine receptors: In vitro and in vivo mechanisms related to Alzheimer's disease. J Neurosci, 2001. 21(12): p. 4125-33.
  • Nitric oxide signaling contributes to late- phase LTP and CREB phosphorylation in the hippocampus. J Neurosci, 1999. 19(23): p. 10250-61.
  • Venturini, G., et al., Beta-amyloid inhibits NOS activity by subtracting NADPH availability. Faseb J, 2002. 16(14): p. 1970-2.
  • NOS2 NO synthase 2
  • Amyloid beta-peptide induces nitric oxide production in rat hippocampus: association with cholinergic dysfunction and amelioration by inducible nitric oxide synthase inhibitors. Faseb J, 2001. 15(8): p. 1407-9.
  • Example 1 A HAT Activator compound.
  • YF2 a Histone Acetyltransferase (HAT) Activator of the invention (FIG. 3)
  • HAT Histone Acetyltransferase
  • FIG. 3 a Histone Acetyltransferase Activator of the invention
  • the western blot showed that it not only crosses the BBB, but also increases histone 3 acetylation levels of the hippocampus (FIG. 1).
  • the title compound ameliorates the contextual fear memory deficit in ⁇ 42 - infused mice (FIG. 2).
  • ⁇ 42 is a protein that is produced in high amount in AD and is responsible for the impairment of synaptic functions and memory.
  • MOM has medium solubility (DMSO 10% in H 2 0). MOM was administered 25mg/kg to WT mice (i.p.). The mice liver and hippocampus were extracted lhr after treatment. The liver showed a very slight increase of AcH3, indicating that the drug has either very little efficacy OR very little membrane permeability (FIG. 6). The hippocampus had no increase in AcH3 levels, indicating the drug is either ineffective OR does not cross the blood brain barrier (BBB) (FIG. 6). Although MOM failed to increase AcH3 levels in the hippocampus and liver, the experiment was repeated with a new administration (gavage and i.p. 25mg/kg).
  • BBB blood brain barrier
  • mice fear conditioning treatment of the mice was subsequently carried out to see if the drug is active after induction of learning.
  • the mouse cortex was also extracted. Hippocampus, cortex, and liver samples again showed no increase of AcH3 levels, indicating the drug is either ineffective OR does not cross the BBB OR does not cross the cell membrane (FIG. 7).
  • a new compound YF2 (FIG. 3), was synthesized.
  • the preparation of YF2 was without a column and 2 phases were visible: clear and oily.
  • YF2 (50 mg/kg, i.p.) was subsequently administered to mice. Two and four hrs after its administration, the mice were sacrificed and hippocampi were extracted. Interestingly, YF2 was able to cross the BBB, penetrate the cells and increase AcH3 (lane 1 vs. lanes 9, 10) (FIG. 8). Given that the compound was not 100% clean and needed to be further purified/verified, we synthesized more YF2 and purified it. Purity was verified through Nuclear Magnetic Resonance (NMR).
  • mice were administered with YF2 (i.p. dissolved in saline) at 5, 10, 20 mg/Kg. Hippocampus extraction was made at 3 different time points (0.5, 1 and 2 hrs post treatment). We then ran a western blotting for AcH3. Except for the lhr-lOmg/kg administration of YF2, YF2 dramatically increased AcH3 levels (FIG. 10), indicating that YF2 crosses the BBB and the cell membrane.
  • Contextual and cued fear conditioning was performed to assess whether the compound is capable of ameliorating amyloid-beta ( ⁇ ) induced memory defect.
  • is a peptide which is elevated in Alzheimer's disease.
  • the hippocampus plays a key role in contextual memory and in Alzheimer's Disease. This type of cognitive test is much faster than other behavioral tasks that require multiple days of training and testing [Ql , Q2].
  • Our conditioning chamber was in a sound-attenuating box. A clear Plexiglas window allowed the experimenter to film the mouse performance with a camera placed on a tripod and connected to the Freezeframe software (MED Ass. Inc.).
  • the conditioning chamber had a 36-bar insulated shock grid floor.
  • the floor was removable, and after each experimental subject, we cleaned it with 75% ethanol and then with water. Only one animal at a time was present in the experimentation room.
  • mice were placed in the conditioning chamber for 2 min before the onset of a discrete tone (CS) (a sound that lasted 30 sec at 2800 Hz and 85 dB). In the last 2 sec of the CS, mice were given a foot shock (US) of 0.8 mA for 2 sec through the bars of the floor. After the CS/US pairing, the mice were left in the conditioning chamber for another 30 sec and were then placed back in their home cages. Freezing behavior, defined as the absence of all movement except for that necessitated by breathing, was scored using the Freezeview software.
  • CS discrete tone
  • US foot shock
  • mice were placed in a novel context (triangular cage with smooth flat floor) for 2 min (pre-CS test), after which they were exposed to the CS for 3 min (CS test), and freezing was measured. Sensory perception of the shock was determined through threshold assessment.
  • a sequence of single foot shocks was delivered to animals placed on the same electrified grid used for fear conditioning. Initially, a 0.1 mV shock was delivered for 1 sec, and the animal behavior was evaluated for flinching, jumping, and vocalization.
  • the shock intensity was increased by 0.1 mV to 0.7 mV and then returned to 0 mV in 0.1 mV increments at 30 sec intervals. Threshold to vocalization, flinching, and then jumping was quantified for each animal by averaging the shock intensity at which each animal manifests a behavioral response to the foot shock.
  • YF2 was i.p. administered to mice (one group of mice was administered with 20 mg/kg, 2hrs before the electric shock, whereas another group was administered with 5 mg/kg, 30 minutes before the electric shock). YF2 at both doses was capable of dramatically increasing the freezing time demonstrating that the compound rescues the defect in contextual memory.
  • the compound alone at the highest concentration (20 mg/kg) did not affect contextual memory (FIG. 10) indicating that the compound per se is not toxic with respect to memory.
  • Cued memory was not changed in the different groups indicating that YF2 does not affect amygdala function (FIG. 11).
  • FIG. 12 no difference was observed among different groups of mice in different sets of experiments in which we assessed sensory threshold in the presence of vehicle, YF2 alone, ⁇ alone, or YF2 plus ⁇ ( FIG. 12).
  • Alzheimer mouse model following rolipram treatment J. Clin. Invest., 2004. 114: p. 1624-1634.
  • the task is a hybrid of the Morris Water Maze (MWM) and the radial arm land maze. This task is altered in ⁇ -infused mice.
  • the motivation for the animals is the immersion in water.
  • the mouse needed to swim in 6 alleys (arms) radiating from a central area until it found a hidden (submerged) platform at the end of one of the arms, based on visual cues placed in the room.
  • the goal arm was kept constant for all trials, with a different start arm on successive trials, such that the learning criterion was reached in 2 days.
  • the first day of the protocol was a training day.
  • Mice were trained to identify the platform location by alternating between a visible and a hidden platform in a goal arm.
  • the final 3 trials on day 1 and all 15 trials on day 2 used a hidden escape platform to force mice to use spatial cues to identify the location of the goal arm.
  • the mouse was guided gently through the water by placing a hand behind it to direct it towards the platform. The mouse rested on the platform for 15 sec. After completing the trial, the mouse was removed from the pool, gently towel dried and placed back into its cage under a heat lamp. The goal platform location was different for each mouse.
  • mice from cohort 1 After all the mice in the first cohort have had a trial to locate a visible platform, the platform was switched from visible to hidden. After each mouse from cohort 1 completed six alternating trials between visible and hidden platforms, the mice was left to rest under a heating source, and mice from the second cohort were tested in the same way. After completing the six alternating trials, mice from cohort 2 returned to their cages to rest.
  • mice from the first cohort completed trials 7-12 again using the alternating visible-hidden platform location.
  • mice from the second cohort completed trials 7-12.
  • all mice had performed 3 hidden platform trials.
  • the same procedure was repeated as on day 1 for all 15 trials using only the hidden platform.
  • averages for each mouse were calculated using blocks of 3 trials.
  • vehicle-treated mice exhibit ⁇ 1 error over three trials near the end of the second day.
  • mice failed to learn, making 3-4 errors throughout the training session, with no improvement over trials.
  • Treatment with YF2 [(i.p., 5 mg/kg, 30 min prior to the 1 st trial (for the 1 st group of tests) and 30 min prior to the 7 th trial (for the 2 nd group of tests)] rescued the ⁇ -induced memory impairment.
  • CBP functions as a co-activator that facilitates interactions with the basal transcription machinery by working as an
  • HAT acetyltransferase
  • HDACs were found to remove an acetyl group from histones, thus restricting access of the transcriptional machinery to the DNA.
  • HDAC inhibitors have been shown to enhance LTP and contextual fear memory, a form of associative memory in which animals must associate a neutral stimulus with an aversive one[A25].
  • HDAC inhibitors could affect neuronal function through a variety of mechanisms including epigenetic and non-epigenetic changes [A27]. Whether cognitive deficits following ⁇ elevation may be induced by epigenetic modification on histone acetylation (via chromatin remodeling) has not been determined. [00266] WT-PS1 stimulates the transcriptional activity ability of CBP whereas its AD M146L mutant did not produce such an effect [A20] indicating that CBP and its HAT activity in AD may be involved. In addition, a CBP mutant lacking HAT activity is not capable of responding to WT-PS1 in terms of increased transcription activating ability. Hence, CBP and its HAT region appear to be essential for enhancing transcription in vitro following PS 1 stimulation. The inventors find that histone acetylation level of APP/PS 1 mice is different than in WT mice, thus identifying AD as a disease of epigenetic etiology.
  • epigenetics and histone acetylation might play a fundamental role in AD.
  • inhibition of HDAC or conversely, activation of a HAT can effectively counteract the disease progression.
  • AD is thought to begin as a synaptic disorder that progressively leads to greater neuronal dysfunction, leading to memory loss [A28].
  • TSA HDAC inhibitor
  • TSA treated APP/PS 1 slices was far greater than in vehicle-treated APP/PS 1 slices (FIG. 17). But, TSA did not change the amplitude of LTP in hippocampal slices of WT mice compared to WT slices treated with vehicle alone. Moreover, TSA perfusion did not affect baseline transmission in APP/PSl and WT slices that received no tetanus.
  • mice Following experiments on acute effect of TSA in synaptic dysfunction, we determined if TSA is beneficial against impairment of contextual FC in APP/PSl mice[Ai7] .
  • 4 month-old mice were divided into 4 groups: APP/PSl with TSA, APP/PSl with vehicle, WT with TSA and WT with vehicle.
  • TSA and vehicle control solution were administered i.p. at a concentration of 2 ⁇ g/g body weight.
  • mice were trained to associate neutral stimuli with an aversive one. They were placed in a novel context (FC box), exposed to a white noise cue paired with a mild foot shock, and injected with TSA 2hrs before training.
  • the APP and PS1 transgenes could affect neuronal function through different mechanisms [A30, A31], including direct effects by ⁇ .
  • the trafficking and signaling properties of full-length APP and its cleavage products are likely different, which could impact aspects of synaptic function differently.
  • ⁇ per se is responsible for the deficits observed in our studies on Tg mice. Since it has already been described that natural oligomers of human ⁇ , in the absence of monomers and fibrils, markedly inhibit LTP in vivo [A6], we will apply 200 nM oligomeric ⁇ 42 concurrently with TSA (1.65 ⁇ ) for 30 minutes to WT slices prior to inducing LTP.
  • oligomeric ⁇ 42 should inhibit LTP and fear memory, and demonstrate that TSA reestablishes normal LTP and contextual fear memory following ⁇ 42 treatment. TSA alone should not have any effect.
  • CBP and its HAT region appear to be essential for enhancing transcription in vitro following PS1 stimulation. Additionally, we decided to measure endogenous CBP levels in APP/PS1 mice.
  • Controls will be performed using a latent inhibition training paradigm to exclude that changes in CBP HAT activity are due to novel context alone or the electric shock instead of the association between them [A25].
  • animals will be pre-exposed to a novel context prior to receiving the electric shock so that the animal will form a spatial memory that blocks the formation of an associative contextual fear memory.
  • HDAC activity using a new fluorimetric assay on hippocampi using the experimental paradigm as in the HAT assay. Without being bound by theory, these experiments will establish if CBP and/or HDACs are altered following overexpression of the APP and PS1 transgenes.
  • HDAC inhibition might rescue the histone 4 acetylation levels defect observed in APP/PSl mice was also tested.
  • Injection of TSA (2 ⁇ g/g body weight; i.p) 2 hours prior to contextual fear conditioning enhanced H4 acetylation of APP/PS 1 mice (FIG. 22).
  • AD is likely to be a disease with an epigenetic motif and HDAC inhibitors can elevate decreased levels of histone 4 in an AD mouse model.
  • CA1 fEPSPs will be recorded by placing stimulating and the recording electrodes in CA1 stratum radiatum. Following BST assessment, a 15min baseline will be recorded every min at an intensity that evokes a response -35% of the maximum evoked response.
  • LTP will be induced using ⁇ - burst stimulation (4 pulses at 100Hz, with the bursts repeated at 5Hz and each tetanus including 3 ten-burst trains separated by 15 sec).
  • mice will be placed in the conditioning chamber for 2min before the onset of a discrete tone (CS) (a 30s, 85dB sound at 2800Hz), as described [A17] .
  • CS discrete tone
  • US 0.60mA foot shock
  • mice will be left in the conditioning chamber for 30s and will then be placed back in their home cages. Freezing behavior will be scored using the Freezeview software (MED Ass).
  • MED Ass Freezeview software
  • freezing will be measured for 5min in the chamber in which the mice will be trained 24hr after training.
  • mice will be placed in a novel context for 2min (pre-CS test), after which they will be exposed to the CS for 3min (CS test), and freezing will be measured.
  • CBP levels will be measured with western blot using specific CBP antibodies.
  • the nuclear fraction will be contained in the pellet obtained from homogenated tissue, centrifuged at 7,700xg for lmin.
  • CBP HA T activity will be measured by immunoprecipitation from the lysis of hippocampal extracts using CBP antibodies. After isolation, HAT activity will be assessed using indirect enzyme-linked immunosorbent assay kit to detect acetyl residues according to the manufacturer's instruction (Upstate).
  • HDAC activity assay I will use a fluorimetric kit from Biovision (CA), according to the manufacturer instruction.
  • CA Biovision
  • Histone acetylation assay Western blot will be performed from snap-frozen in liquid nitrogen hippocampi. Nuclear proteins will be acid-extracted and separated onto a denaturing, 7%— 12% acrylamide gel followed by electrob lotting onto nitrocellulose.
  • Acetylated histones (H3, H2A and H2B) will be detected using antibodies purchased from Upstate and the Amersham ECF Kit accordingly to the manufacturer protocol.
  • Nicotinamide an HDAC III inhibitor
  • Nicotinamide was found to restore cognition in the triple transgenic mouse model of AD via a non-epigenetic mechanism involving reduction of cytosolic Thr231-phosphotau [B49].
  • histones H2B and 3 which are also known to play a key role in transcription and memory [B19, B25, B26], abnormally acetylated during memory processes in AD?
  • histone acetylation affected following ⁇ elevation is histone acetylation affected following ⁇ elevation?
  • the inventors will also discuss in this Example whether chromatin changes at the level of histone acetylation occur following elevation of ⁇ during memory processes. Without being bound by theory, the results obtained will provide a new type of mechanism for ⁇ -induced impairment of memory and synaptic function.
  • the therapies for AD include augmentation of the cholinergic system by usage of acetylcholinesterase inhibitors, or blockage of glutamate neurotoxicity through NMD A antagonists. These agents have a limited efficacy. Major efforts are underway to inhibit tangle formation, to combat inflammation and oxidative damage, and to decrease ⁇ load in the brain either by the use of agents that inhibit ⁇ and ⁇ secretases or increase a secretase, by the use of drugs that inhibit ⁇ oligomerization, or by the use of treatments such as immunization with ⁇ that appear to augment the removal of ⁇ from the brain.
  • HDAC inhibition ameliorates deficits in hippocampal long-term
  • HDAC inhibition is capable of rescuing the defect in LTP in the APP/PSl animal model of ⁇ deposition.
  • HDAC inhibition rescues the defect in associative memory in APP/PSl mice. Similar to humans affected by AD [B51], APP/PSl mice exhibit a deficit in associative memory [B46]. In rodents, associative learning can be assessed by contextual fear
  • HDAC inhibition rescues the defect in associative memory by ⁇ elevation.
  • the APP and PS1 transgenes could affect neuronal function through different mechanisms [B23, B24], including direct effects by ⁇ . Full-length APP and its cleavage products could differently impact H4 acetylation. To separate ⁇ effects from other effects of APP and PS1 overexpression, we next determined whether ⁇ per se is responsible for the deficit in H4 acetylation observed in our studies on transgenic mice. Since it has already been described that natural oligomers of human ⁇ , markedly inhibit memory in vivo [B54, B55], we applied a preparation containing oligomeric ⁇ 42 (200 nM in a volume of 1 ⁇ , bilaterally, slowly over 1 min) into dorsal hippocampi (FIG.
  • APP/PSl mice display a reduced endogenous level of histone 4 acetylation in response to a learning task.
  • HDAC inhibitors are known to acetylate other molecules besides histones [B49]
  • our next goal was to determine whether the effect of TSA on the defect in fear memory of APP/PSl mice was linked, at least in part, to chromatin changes at the level of histone acetylation.
  • Acetylation of H4 was shown to play a key role in transcription and memory [B19].
  • Histone acetylation induces chromosomal changes resulting in loss of chromosomal repression (see FIG. 23).
  • mice were divided into 4 groups: APP/PSl with NaB, APP/PSl with vehicle, WT with NaB and WT with vehicle.
  • Controls will be also performed using a latent inhibition training paradigm to exclude that HDAC inhibitors act through an effect on novel context alone or the electric shock instead of the association between them [B26].
  • animals will be pre-exposed to a novel context prior to receiving the electric shock so that the animal will form a spatial memory that blocks the formation of an associative contextual fear memory [B26].
  • mice will perform visible platform testing to exclude the possibility that visual, motor and motivational deficits affect the outcome of the experiments.
  • 3-4 and 7 month-old APP/PS1 and WT littermates will be injected with TSA or NaB, 2 hrs prior to performing the task.
  • Vehicle will be used in control experiments.
  • Similar experiments assessing reference memory will be also performed in Ap42-infused mice (for these experiments mice will be divided into the following groups: Ap42-infused animals + TSA or NaB or vehicle, vehicle-infused animals + TSA or NaB or vehicle).
  • oligomeric ⁇ 42 will inhibits LTP and fear memory, and demonstrate that TSA ameliorates LTP and contextual fear memory following ⁇ 42 treatment. TSA alone should not have any effect. Findings from young mice or ⁇ -infused animals should be confirmed in 7 month old transgenics. If not, as an alternative strategy, we will try longer treatments with the HDAC inhibitors, starting 1 or 4 weeks before testing. Finally, without being bound by theory, findings with the reference memory task should be in agreement with those obtained with fear conditioning. If so, these results should demonstrate that inhibition of histone deacetylase is beneficial against cognitive loss in general in AD-like animal models. If not, this will be equally interesting as it will demonstrate a dissociation between different types of memory and usage of HDAC inhibitors.
  • TSA and other HDAC inhibitors represent a new approach to AD treatment that appears to make the synapse more robust and resistant to the effects of ⁇ .
  • HDAC inhibitors it has been criticized that inhibition of HDACs might alter gene expression globally and thus affect memory processes in a nonspecific manner.
  • Vecsey et al [B53] showed that TSA does not globally alter gene expression but instead increases the expression of specific genes during memory consolidation. They were able to show that HDAC inhibitors, including TSA, enhance memory and synaptic plasticity mainly by the activation of key genes that are dependent on CREB transcriptional activation [B53].
  • TSA may be capable of stopping memory degradation in the presence of ⁇ accumulation as well as improving brain functions that have already deteriorated, as in the case of the 3-month-old APP/PS1 mouse.
  • HDAC inhibitors could be capable of reestablishing neural networks in the AD brain. This indicates that using small molecules to target HDACs in AD patients could facilitate access to long-term memories.
  • HDAC inhibitors with minimized side-effects are currently being developed by the pharmaceutical industry. It remains to be seen if these newer inhibitors can readily enter the brain and if they are as effective as TSA.
  • HDAC inhibitors could affect neuronal function through a variety of
  • HDACs class I/II may increase the acetylation of non-histone substrates that, in turn, can contribute to the amplification of cellular processes associated with memory.
  • Green et al. [B49] showed that inhibition of class III NAD+-dependent HDACs using vitamin B3 restored cognitive deficits in the triple transgenic AD mice, via a mechanism involving the reduction of Thr231-phosphotau in the cytoplasm.
  • Hippocampi from 3-4 month old APP/PS1 mice exhibit, after fear conditioning training, approximately a 50% reduction in acetylated histone 4 (H4) levels, an acetylation that was shown to be important in memory formation [B19].
  • H4 acetylated histone 4
  • brain tissue will be dissected and the two hippocampi from each mouse pooled and flash frozen for later homogenization and analysis. Additional controls will use cerebellum.
  • Example 6 Findings in Example 6 will be confirmed with the structurally dissimilar NaB.
  • NaB 1.2 g/Kg
  • controls will be performed on vehicle -infused mice and using a latent inhibition training paradigm, as well as cerebellum.
  • oligomeric ⁇ 42 will affect H4 acetylation levels and oligomeric ⁇ 42 will produce the same effects on acetylation levels of histones 2B and 3 as in transgenic mice. If not, we will try more prolonged applications of 200 nM oligomeric ⁇ 42 through Alzet osmotic mini-pumps (1 day, 1 week, 1 month). Problems related to the use of a synthetic preparation containing ⁇ 42 are greatly alleviated by the use of transgenic animals which produce natural forms of ⁇ . Studies on H2B and 3 will provide a more complete picture of the type of epigenetic changes occurring at the level of histone acetylation. Finally, research in older mice will help understanding whether HDAC inhibitors might be used at older disease stages. Taken all together, the studies described herein will help establishing epigenetic changes as events occurring following ⁇ elevation.
  • PTMs histone posttranslational modifications
  • H3 Lys4 methylation and H3 Lys56 acetylation were found to lead to gene expression.
  • histone modifications associated with the inactivation of gene transcription such as H3 Lys27 methylation and H2A Lysl 19 ubiquitination were found to cause gene silencing.
  • PTMs that are modified as a consequence of chronic neuronal exposure to oligomeric forms of ⁇ 42 in mice. As described (see FIG.
  • oligomeric ⁇ 42 will be infused into dorsal hippocampi of WT mice.
  • the hippocampi of ⁇ 42- ⁇ 8 ⁇ mice will be removed and compared to those of vehicle-infused mice.
  • mice will be sacrificed at 1 month (prior to plaque formation), 2 months (as plaques start to form), 3-4 months (at early stages of plaque formation), and 7 months (at late stages of plaque formation) of age to test when this histone PTMs occur.
  • epigenetic changes such as reduced histone acetylation, are likely to play an important role in the ⁇ -induced damage of synaptic function and memory associated with AD.
  • CBP HAT activity is essential for enhancing transcription in vitro following PSl stimulation. Furthermore, CBP levels in 3-4 month old APP/PSl mice were found to be lower than in WT mice. In future experiments, we will extend this observation to older transgenic mice and following hippocampal ⁇ infusion. In addition, we will determine if CBP HAT activity is affected both in young and older APP/PSl mice as well as after ⁇ infusion.
  • a HAT agonist such as N-(4-chloro-3-trifluoromethyl-phenyl)-2-ethoxy- benzamide (CTB or also referred to as compound 6 J) [B61] was synthesized.
  • CTB N-(4-chloro-3-trifluoromethyl-phenyl)-2-ethoxy- benzamide
  • MOM benzamide HAT agonist
  • MOM (10 ⁇ ) or vehicle will be applied for 30 min prior to inducing LTP with the ⁇ -burst. WT littermate slices treated with MOM or vehicle will be used as controls.
  • MOM re-establishes normal reference memory.
  • cannulas will be implanted into the dorsal hippocampi of APP/PS 1 mice and WT littermates to deliver it directly into the hippocampi.
  • We will infuse 100 ⁇ g in 1 ⁇ , slowly over 1 min. The infusion will occur 2 hrs prior to applying the foot shock for fear conditioning.
  • we will measure the amount of freezing at 24 hrs to assess contextual fear memory followed by cued fear memory at 48 hrs. For the water maze experiments, in turn, we will measure the number of incorrect arm entries.
  • HAT agonists might ameliorate the defect in LTP, memory and histone acetylation following overexpression of APP and PS1 transgenes.
  • the protocol will also be carried out for testing the effect of the HAT activator, CTB.
  • HDAC activity through a new fluorimetric assay on hippocampi using the experimental paradigm as in the HAT assay (basal, 1 min, 5 min, 20 min and 1 hour after foot shock).
  • HAT assay basic, 1 min, 5 min, 20 min and 1 hour after foot shock.
  • HATs are involved in the reduction of histone acetylation. These include GNATs family, MYST family, p300, and ACTR/SRC-1. We will measure levels and activity of these HATs.
  • Electrophysiological studies We will cut 400 ⁇ hippocampal slices from C57B16 mice and maintain them in an interface chamber at 29° C for 90 min prior to recording, as previously reported [Bl 1].
  • the bath solution will consist of 124.0 mM NaCl, 4.4 mM KC1, 1.0 mM Na 2 HP04, 25.0 mM NaHC0 3 , 2.0 mM CaCl 2 , 2.0 mM MgS0 4 , and 10.0 mM glucose, continuously bubbled with 95% 0 2 and 5% C0 2 .
  • a stimulating electrode we will use a bipolar tungsten electrode, placed at the level of the Schaeffer collateral fibers.
  • Contextual fear learning a type of memory for which hippocampal function is indispensable, will be evaluated 24 hrs after training by measuring freezing for 5 min in the chamber in which the mice will be trained.
  • Cued fear learning a type of memory that depends upon amygdala function, will be evaluated 24 hrs after contextual testing by placing mice in a novel context for 2 min (pre-CS test), after which they will be exposed to the CS for 3min (CS test), and freezing will be measured.
  • conditioning is tested if the experimental procedure does not affect the sensory threshold of the animals.
  • mice will be checked with the open-field test.
  • the open field will be an arena made of white acrylic with internal dimensions of 72 X 72 X 33 cm (An area measuring 36 X 36 cm in the centre of the open field will be defined as the 'central zone').
  • Mice will be placed in the center of a standard open field and their behavior monitored for 1 hr and scored for proportion of time in the center compartment vs. periphery, and number of entries into the center compartment. Mice will be returned for a second hour block after 24 hr. No difference in exploratory behavior as demonstrated by a similar percentage of time spent in the center compartment and the number of entries into the center compartment should be observed if the manipulation does not affect exploratory capabilities of the mice.
  • the first day of the protocol will be a training day. Mice will be trained to identify the platform location by alternating between a visible and a hidden platform in a goal arm. The final 3 trials on day 1 and all 15 trials on day 2 will use a hidden escape platform to force mice to use spatial cues to identify the location of the goal arm. To avoid learning limitations imposed by exhausting practice and to avoid fatigue that may result from consecutive trials, spaced practice training will be established by running the mice in cohorts of 4 and alternating different cohorts through the 15 training trials over 3 hours testing periods each day. On day 1 , a visible platform will be placed in a goal location.
  • Mouse 1 of cohort 1 will be gently placed in the pool near the perimeter of the wall of the first start arm (specified on a score sheet) and facing the center of the pool. The number of incorrect arm entries (entries in arms with no platform) will be counted. If the animal enters the incorrect arm it is gently pulled back to the start arm. Each trial will last up to 1 minute. Failure to select an arm after 15 seconds will be counted as an error and the mouse will be returned to the start arm. After 1 minute, if the platform has not been located, the mouse will be guided gently through the water by placing a hand behind it to direct it towards the platform. The mouse will rest on the platform for 15 seconds. After completing the trial, the mouse will be removed from the pool, gently towel dried and placed back into its cage under a heat lamp.
  • mice from the first cohort After all the mice in the first cohort have had a trial to locate a visible platform, the platform will be switched from visible to hidden. After each mouse from cohort 1 completes six alternating trials between visible and hidden platforms, the mice will be left to rest under a heating source, and mice from the second cohort will be tested in the same way. After completing the six alternating trials, mice from cohort 2 will return to their cages to rest. Next, mice from the first cohort will complete trials 7-12 again using the alternating visible -hidden platform location. During resting time for mice from the first cohort, mice from the second cohort will complete trials 7-12. At this point, all mice will have to perform 3 hidden platform trials.
  • mice will be implanted with a 26-gauge guide cannula into the dorsal part of the hippocampi
  • mice will be injected 20 min prior to performing each session and the probe trial, whereas for fear conditioning mice will receive a single injection 20 min before the training. Mice will be handled once a day for 3 days before behavioral experiments. During infusion animals will be handled gently to minimize stress.
  • Oligomeric ⁇ 42 will be prepared from commercially available synthetic peptides (American Peptides Co), as described [B63, B64]. Briefly, the lyophilized peptide will be resuspended in cold 1,1,1, 3,3, 3-hexafluoro-2-propanol (HFIP, Sigma) and aliquoted in polypropylene vials. After 24 hrs the HFIP solution will be allowed to evaporate in a fume hood until a thin film of peptide is formed on the bottom of the vials. Peptide films will be dried under gentle vacuum and stored in sealed vials at -20°C.
  • HFIP 1,1,1, 3,3, 3-hexafluoro-2-propanol
  • Oligomeric ⁇ 42 will be obtained by incubating an aliquot of monomeric ⁇ /DMSO solution in sterile PBS at 4°C overnight.
  • the quality of these ⁇ preparations will be routinely controlled using Western blot analysis in which ⁇ samples will be resolved by Tris-Tricine PAGE under non-denaturing/non- reducing conditions, and then transferred on nitrocellulose membrane. Subsequent Western blotting will be carried out after membrane incubation with the anti-human ⁇ monoclonal antibody 6E10 (Signet Lab). The immunostaining will be revealed by horseradish peroxidase chemi-luminescence.
  • Histone acetylation assay Western blot will be performed from snap-frozen in liquid nitrogen hippocampi and cerebella. Tissue will be homogenized in lysis buffer (62.5 mM Tris-HCl pH 6.8, 3% SDS, 1 mM DTT) and incubated at 4 °C for 10 min, then sonicated before centrifugation at 2,000 rpm for 5 min. Whole cell extracts will be electrophoresed on 10-20% gradient PAGE gel (Invitrogen) and then immunob lotted. Antibodies will be used at a 1 : 1,000 concentration for immunoblotting. All anti-histone antibodies will be purchased from Millipore. ⁇ - ⁇ -Tubulin antibody will be purchased from Promega.
  • Immunoblot data will be quantified by measuring the band intensity using imaging software (NIH ImageJ). For quantitative immunoblot analysis, equal amounts of proteins will be loaded into each lane. To confirm equal loading, blots will be reprobed with corresponding pan-antibodies or antibodies for house-keeping proteins such as ⁇ - ⁇ -Tubulin. For quantification, we always use a signal in the linear range.
  • Mass Spectrometry Characterization of modifications on immunoprecipitated histones will be carried out by mass spectrometry. Immunoprecipitated histones will be purified by reverse-phase HPLC in the PSR or by SDS-PAGE, then subjected to enzymatic digestion. Resulting peptides will be analyzed by LC-MS/MS on a Waters Qtof mass spectrometer equipped with a Dionex nanflow LC. The standard digestion protocol using trypsin is not feasible due to the number of Lys residues in the N-terminal portion of histones, resulting in peptides too small to be analyzed.
  • CBP levels will be measured with western blot using specific CBP antibodies.
  • the nuclear fraction will be contained in the pellet obtained from homogenated tissue, centrifuged at 7,700xg for lmin.
  • CBP HAT activity will be measured by immunoprecipitation from the lysis of hippocampal extracts using CBP antibodies. After isolation, HAT activity will be assessed using indirect enzyme-linked immunosorbent assay kit to detect acetyl residues according to the manufacturer's instruction (Upstate).
  • HP AC activity will be measured using a fluorimetric kit from Biovision (CA), according to the manufacturer instruction.
  • HDACs Histone deacetylases
  • Beta-amyloid activates the mitogen-activated protein kinase cascade via hippocampal alpha7 nicotinic acetylcholine receptors: In vitro and in vivo mechanisms related to Alzheimer's disease. J Neurosci, 2001. 21(12): p. 4125-33.
  • Nicotinamide restores cognition in Alzheimer's disease transgenic mice via a mechanism involving sirtuin inhibition and selective reduction of Thr231-phosphotau. J Neurosci, 2008. 28(45): p. 11500-10.
  • beta-Amyloid infusion results in delayed and age-dependent learning deficits without role of inflammation or beta-amyloid deposits. Proc Natl Acad Sci U S A, 2006. 103(23): p. 8852-7.
  • Puzzo, D., et al. Picomolar Amyloid- ⁇ beta ⁇ Positively Modulates Synaptic Plasticity and Memory in Hippocampus. J. Neurosci., 2008. 28(53): p. 14537-14545. B65. Garcia, B.A., et al., Chemical derivatization of histories for facilitated analysis by mass spectrometry. Nat Protoc, 2007. 2(4): p. 933-8.
  • Example 7 Cell Viability Assays for ACHN, U251, NCI-ADR-RES, A549, Hs578T,
  • VBL Vinblastine
  • Cells and culture medium All cell lines were purchased from the ATCC and were expanded and archived under liquid nitrogen at CD AS as low passage aliquots. Cells were maintained and passaged in recommended and optimal culture medium (ACHN:
  • EMEM 2 mM L-Gln, 10% FBS
  • A549 Ham's F12, 10% FBS
  • U251 RPMI 1640,2 mM L- Gln, 10% FBS
  • Hs578T DMEM, 4 mM L-Gln, 1 U/mL of Bovine Insulin, 10% FBS;
  • CCRF-CEM ATCC RPMI, 2 mM L-Gln, 10% FBS; NCI-ADR-RES: RPMI 1640, 2 mM L-Gln, 10% FBS). All experiments were carried out with cells which had undergone less than 20 passages. Optimal seed densities were determined for all cell lines. All cells were plated at 1500 cells per well except CCRF-CEM which was plated at 6000 cells per well.
  • Drugs YF2 was supplied as a 80 mM stock solution in 100% DMSO.
  • Vinblastine was purchased from Sigma (Catalogue Number V-1377) and resuspended at 1 X 10 "2 M in 100% DMSO. All dilutions for both drugs were carried out in culture medium containing 0.2% DMSO such that the final solvent concentration never exceeded 0.1 %.
  • Drug treatment YF2 tested at 10 concentrations (0.03, 0.1, 0.25, 0.5, 1,2.5, 5, 15,40 and 80 ⁇ ) in triplicate wells. Vinblastine was used as a reference control and tested at 10 concentrations in a half-log series (0, 0.001, 0.003, 0.01, 0.03, 0.1, 0.3,1,3, and 10 ⁇ ). Cells were resuspended in medium at the appropriate concentration and 180 ⁇ (1500 or 6000 cells) was added to each well following which 20 ⁇ of drug at lOx of the final concentration was added to attain the desired drug concentration in every well. The drug treatment plates were incubated at 37°C for 72 hours, following which cell viability was assayed by the Cell Titer Glo or Cyquant method as described below.
  • Cell Titer Glo Assay Following the 72 hour drug treatment period, the assay plates were centrifuged, and 100 ⁇ , of the medium was aspirated and replaced with 100 J.1L of Cell Titer Glo reagent (Promega) according to the manufacturer's recommended protocol. The reagent was mixed with the cells and the luminescence measured using a Perkin Elmer Envision instrument. The average luminescence signal obtained from wells containing untreated cells which had been incubated for the entire length of the assay period was used to set the 100% viability value. The percent proliferation was calculated as (Test signal)/(Avg. plate background signal) x 100. The % viability was graphed against drug concentration to calculate an IC50 for each drug.
  • Cyquant Assay Following the 72 hour drug treatment period the assay plates were centrifuged, the medium discarded, and frozen overnight. The plates were assayed using the CyquantTM reagent (Invitrogen) according to the manufacturer's recommended protocol. The average fluorescence signal obtained from wells containing untreated cells which had been incubated for the entire length of the assay period was used to set the 100% proliferation value. The percent proliferation was calculated as (Test signal)/(Avg. plate background signal) x 100. The % proliferation was graphed against drug concentration to calculate an IC50 for each drug.
  • Example 8 YF2 Increases Histone Acetylation by HAT Activation, Not HDAC
  • HDAC inhibition causes an increase in histone acetylation.
  • the inventors examined whether histone acetylation occurred via HDAC inhibition.
  • HDAC Class 2a Substrate 1 (BPS number 50040)
  • Assay Conditions A series of dilution of the test compounds were prepared with 10% DMSO in assay buffer and 5 ⁇ 1 of the dilution was added to a 50 ⁇ 1 reaction so that the final concentration of DMSO is 1% in all of reactions. All of the enzymatic reactions were conducted in duplicate at 37°C for 30 minutes except of HDACl 1 at room temperature for 3 hours.
  • the 50 ⁇ 1 reaction mixture contains HDAC assay buffer, 5 ⁇ g BSA, an HDAC substrate, an HDAC enzyme and a test compound. After enzymatic reactions, 50 ⁇ 1 of HDAC Developer was added to each well and the plate was incubated at room temperature for an additional 20 minutes. Fluorescence intensity was measured at an excitation of 360 nm and an emission of 460 nm using a Tecan Infinite Ml 000 microplate reader.
  • Slope slope factor or Hill coefficient.
  • the IC 50 value was determined by the concentration causing a half-maximal percent activity.
  • FIG. 52 corresponds to the results shown in Table 4.
  • FIG. 53 corresponds to the results shown in Table 5.
  • FIG. 54 corresponds to the results shown in Table 6.
  • FIG. 55 corresponds to the results shown in Table 7.
  • FIG. 56 corresponds to the results shown in Table 8.
  • FIG. 57 corresponds to the results shown in Table 9.
  • FIG. 58 corresponds to the results shown in Table 10.
  • FIG. 59 corresponds to the results shown in Table 11.
  • FIG. 60 corresponds to the results shown in Table 12.
  • FIG. 74 corresponds to the results shown in Table 13. [00405] Results of Effect of SAHA on HDAC inhibition
  • SAHA is an HDAC inhibitor (HDACi). It serves as a positive control for HDACs.
  • FIGS. 61-63 show the inhibitory effect of SAHA on the HDACs HDACl, HDAC3/NCOR2, and HDAC 6.
  • SAHA also inhibited HDAC5FL, HDAC7, HDAC 8, HDAC 10, Sirtuin 1, and Sirtuin 2 (see Table 1).
  • AD Alzheimer's disease
  • ⁇ -amyloid a peptide that is produced in elevated amounts in the disease
  • LTP long-term potentiation
  • memory is modulated by epigenetics through regulation of gene expression, deregulation of one of the epigenetic mechanisms such as histone (H) acetylation, might lead to memory disruption. Reduction of histone acetylation causes the chromatin structure to close, so that the information contained within the DNA might be less amenable to transcription factors and memory formation' 9] .
  • HDAC inhibitors The main strategy that is currently used to up-regulate histone acetylation involves HDAC inhibitors.
  • the pleiotropic effect of nonspecific HDAC inhibition may hamper their therapeutic potential [10 ⁇ 13] .
  • CBP and PCAF hippocampal levels of two HATs, CBP and PCAF, are reduced following ⁇ elevation.
  • HATs histone acetylation
  • YF2 histone acetylation
  • BACKGROUND AND SIGNIFICANCE The post-translational acetylation status of chromatin is governed by the competing activities of two classes of enzymes, HATs and HDACs.
  • HDAC inhibitors have been shown to enhance LTP and contextual fear memory, a form of associative memory in which animals must associate a neutral stimulus with an aversive one' P17] .
  • memory and LTP deficits of CBP +/ mice were reversed by HDAC inhibition ⁇ 15 .
  • the potential of inhibiting HDACs to counteract neurodegenerative disorders has been widely explored' 4] . For instance, in a set of experiments, Tsai et al.
  • HDAC inhibitors induced sprouting of dendrites, increased number of synapses, and reinstated learning and access to long-term memories in the CK-p25 Tg mouse model of neurodegeneration' 15 ' l6 Moreover, in recent studies we have shown that the HDAC inhibitor TSA ameliorates LTP and contextual fear conditioning (FC) in the double Tg
  • HATs can be divided in two main groups, the nuclear HATs and cytoplasmic HATs [18] .
  • Nuclear A-type HATs can be grouped into at least 4 different families based on sequence conservation within the HAT domain: Gcn5 and p300/CBP associated factor (PCAF), MYST (MOZ, Ybf2/ Sas3, Sas2 and Tip60), p300 and CBP (named for the two human paralogs p300 and CBP) and Rttl09. While the Gcn5/PCAF and MYST families have homologs from yeast to man, p300/CBP is metazoan-specific, and Rttl09 is fungal- specific. Cytoplasmic B-type HATs, such as HAT1, are involved in histone deposition [P22] . Marmorstein and Roth (2001, Curr Opin in Genet and Develop., 11 : 155-161) list in Table 1 the HAT families and their transcriptional-related functions, the reference which is incorporated by reference in its entirety.
  • PCAF Gcn5 and p300/CBP associated factor
  • MYST MOZ,
  • HATs are highly conserved in mammals' 1 ⁇ . Of all these HATs, three were shown to be involved in memory: CBP, p300 [19 ' 20] , and PCAF [21] . Interestingly, both CBP and PCAF levels are reduced by ⁇ elevation.
  • HAT activators are a viable approach to enhance histone acetylation.
  • Two scaffolds for HAT activators have been identified. The first one includes CTPB and its derivative CTB [22 ' 23] . The second one includes only one compound, nemorosone [24] .
  • CTPB/CTB were found to be insoluble and membrane-impermeable' 22 ' 23 Moreover, CTPB has unfavorable characteristics to be used in CNS diseases (MW equal to 553,29, clogP equal to 12.70) and the clogP of CTB is 5.13. Nemorosone has a MW of 502 and a clogP of 8.42.
  • CNS diseases including AD.
  • clogP equal to 12.70
  • Nemorosone has a MW of 502 and a clogP of 8.42.
  • YF2 (MW 430.13, clogP 5.15, clogBB 0.17) has increased solubility, membrane permeability and blood-brain barrier permeability, is safe in acute toxicity tests.
  • YF2 rescued the reduction in hippocampal levels of H3 acetylation following ⁇ elevation, enhanced enzymatic activity of PCAF, CBP and Gcn5 as well as p300, had excellent selectivity for PCAF, CBP and Gcn5 over HDACs, and was capable of rescuing deficits in fear and reference memory induced by ⁇ exposure.
  • HAT activators bearing different moieties at different positions of one of the two aromatic rings of YF2.
  • HAT activators bearing different moieties at different positions of one of the two aromatic rings of YF2.
  • we will substitute a dimethylamino group which is likely to be alkylated by microsomes and protect an aromatic ring from oxidative reactions.
  • HAT binding site i) by evaluating the importance of the amide group between the two aromatic rings, ii) by constraining the molecular structure, iii) by modifying the distance between two aromatic rings that currently form YF2, iv) by replacing one of the two aromatic rings with an eterocyclic ring, and v) by increasing the hindrance of the substituents of one of the two aromatic rings.
  • HAT activators with a) HAT specificity and potency, b) great CNS penetration, and c) safety.
  • [00415] 2 To identify compounds with high affinity and good selectivity for selective HAT activators that target specifically CBP and PCAF. Following assessment of HAT activator potency (EC 50 ), we will check selectivity with respect to CBP and PCAF. For HAT activators found to show good potency ( ⁇ lOOnM), selectivity (at least 50-fold vs p300, Gcn5, MYST families and HDACs) and solubility, we will use a functional assay to determine if they increase hippocampal H3 and H4 acetylation in adult mice.
  • HAT activator potency EC 50
  • selectivity at least 50-fold vs p300, Gcn5, MYST families and HDACs
  • solubility we will use a functional assay to determine if they increase hippocampal H3 and H4 acetylation in adult mice.
  • HAT activators have good pharmacokinetic (PK) profile and are safe. Selected HAT activators will be screened against unfavorable PK properties including bioavailability, brain uptake, and BBB penetration. Compounds passing these tests will be evaluated for rudimentary ADMET characteristics including tests of acute and chronic toxicity.
  • HAT activators that rescue LTP in APP/PS1 mice.
  • We will determine if selected HAT activators ameliorate LTP defect in APP/PS 1 slices using electrophysiological techniques' 2 ⁇ .
  • 5) Further screen HAT activators to examine if they ameliorate cognitive abnormalities in APP/PSl mice.
  • HAT activators screened in slices can protect APP/PS l mice against impairments of contextual and reference memory using behavioral assays' 25 -'.
  • AD therapies have limited efficacy. Major efforts are underway to inhibit tangle formation, to combat inflammation and oxidative damage, and to decrease ⁇ load in the brain' 26-28] .
  • APP, ⁇ , and the secretases in normal physiological function' 9"3 ⁇ might present a problem in providing effective and safe approaches to AD therapy.
  • Developing agents that interact with ⁇ targets that lead to neuronal dysfunction is another approach that is currently tested by many laboratories.
  • HAT activators represent a new class of compounds that might effectively counteract the disease progression.
  • Lambert, M.P., et al., Diffusible, nonfibrillar ligands derived from Abetal-42 are potent central nervous system neurotoxins. Proc Natl Acad Sci U S A, 1998. 95(1 1): p. 6448-53. Rakyan, V.K., et al., The marks, mechanisms and memory of epigenetic states in mammals. Biochem J, 2001. 356(Pt 1): p. 1-10.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Emergency Medicine (AREA)
  • Epidemiology (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Heterocyclic Compounds Containing Sulfur Atoms (AREA)
  • Pyridine Compounds (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

L'invention divulgue un procédé pour cribler des composés qui se lient à et modulent une protéine d'histone acétyltransférase (HAT). L'invention divulgue en outre des procédés pour traiter des désordres neurodégénératifs, des conditions associées à des dépôts accumulés de peptides bêta-amyloïdes, des niveaux de protéines Tau, et/ou des accumulations d'alpha-synucléine ainsi que le cancer en administrant à un sujet un composé d'activation de HAT.
PCT/US2010/059925 2009-12-10 2010-12-10 Activateurs d'histone acétyltransférase et utilisation de ceux-ci WO2011072243A1 (fr)

Priority Applications (7)

Application Number Priority Date Filing Date Title
JP2012543311A JP6093180B2 (ja) 2009-12-10 2010-12-10 ヒストンアセチルトランスフェラーゼ活性化剤及びその使用
EP10836767.3A EP2509590B1 (fr) 2009-12-10 2010-12-10 Activateurs d'histone acétyltransférase et utilisation de ceux-ci
ES10836767T ES2764999T3 (es) 2009-12-10 2010-12-10 Activadores de histona acetiltransferasa y usos de los mismos
EP19205945.9A EP3632901B1 (fr) 2009-12-10 2010-12-10 Activateurs de l'histone acétyltransférase et leurs utilisations
US13/493,490 US10640457B2 (en) 2009-12-10 2012-06-11 Histone acetyltransferase activators and uses thereof
US16/708,190 US11034647B2 (en) 2009-12-10 2019-12-09 Histone acetyltransferase activators and uses thereof
US17/317,133 US20210317074A1 (en) 2009-12-10 2021-05-11 Histone acetyltransferase activators and uses thereof

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
US28528709P 2009-12-10 2009-12-10
US61/285,287 2009-12-10
US31776510P 2010-03-26 2010-03-26
US61/317,765 2010-03-26
US35496410P 2010-06-15 2010-06-15
US35511010P 2010-06-15 2010-06-15
US61/354,964 2010-06-15
US61/355,110 2010-06-15
US36300910P 2010-07-09 2010-07-09
US61/363,009 2010-07-09

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/493,490 Continuation-In-Part US10640457B2 (en) 2009-12-10 2012-06-11 Histone acetyltransferase activators and uses thereof

Publications (1)

Publication Number Publication Date
WO2011072243A1 true WO2011072243A1 (fr) 2011-06-16

Family

ID=44145939

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/059925 WO2011072243A1 (fr) 2009-12-10 2010-12-10 Activateurs d'histone acétyltransférase et utilisation de ceux-ci

Country Status (4)

Country Link
EP (2) EP2509590B1 (fr)
JP (4) JP6093180B2 (fr)
ES (2) ES2907862T3 (fr)
WO (1) WO2011072243A1 (fr)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012088420A1 (fr) 2010-12-22 2012-06-28 The Trustees Of Columbia University In The City Of New York Modulateurs de l'histone acétyltransférase et leurs utilisations
WO2012171008A1 (fr) 2011-06-10 2012-12-13 The Trustees Of Columbia University In The City Of New York Utilisations d'activateurs d'histone acétyl tansférase
WO2013116663A1 (fr) 2012-02-01 2013-08-08 The Trustees Of Columbia University In The City Of New York Nouveaux inhibiteurs de protéases à cystéine et leurs utilisations
JP2013208060A (ja) * 2012-03-30 2013-10-10 Nissin Foods Holdings Co Ltd 薬物代謝酵素の活性誘導方法及び薬物代謝酵素の活性が誘導されたヒト培養細胞
WO2014145485A2 (fr) 2013-03-15 2014-09-18 The Trustees Of Columbia University In The City Of New York Modulateurs de map kinase et utilisations de ceux-ci
WO2015009930A2 (fr) 2013-07-17 2015-01-22 The Trustees Of Columbia University In The City Of New York Nouveaux inhibiteurs de la phosphodiestérase et utilisations de ceux-ci
WO2018017858A1 (fr) 2016-07-20 2018-01-25 The Trustees Of Columbia University In The City Of New York Activateurs d'histone acétyltransférase et compositions et utilisations associées
EP3398937A1 (fr) 2014-03-31 2018-11-07 The Trustees Of Columbia University In The City Of New York Activateurs de l'histone acétyltransférase et leurs utilisations
US10640457B2 (en) 2009-12-10 2020-05-05 The Trustees Of Columbia University In The City Of New York Histone acetyltransferase activators and uses thereof
EP3920900A4 (fr) * 2019-02-08 2022-11-16 The Trustees of Columbia University in the City of New York Modulateurs d'histone acétyltransférase et compositions et utilisations associées
EP3920897A4 (fr) * 2019-02-08 2022-11-16 The Trustees of Columbia University in the City of New York Régulateurs d'histone acétyltransférase (hat) et utilisations associées

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3254120A (en) 1961-12-05 1966-05-31 Pharmazeutische Fabrik Montavit Gmbh N-(tertiaryaminoalkoxy-benzoyl) anilides
WO1991005058A1 (fr) 1989-10-05 1991-04-18 Glenn Kawasaki Synthese et isolation sans cellule de nouveaux genes et de nouveaux polypeptides
WO1993020242A1 (fr) 1992-03-30 1993-10-14 The Scripps Research Institute Bibliotheques chimiques combinatoires codees
WO1994018318A1 (fr) 1993-02-01 1994-08-18 The University Of North Carolina At Chapel Hill Reactifs d'affinite entierement synthetiques
WO1995018972A1 (fr) 1994-01-05 1995-07-13 Arqule, Inc. Production modulaire systematique de molecules a base d'aminimide et d'oxazolone ayant des proprietes choisies
WO1996022529A1 (fr) 1995-01-20 1996-07-25 Arqule, Inc. Procede de production de differents composes chimiques constituant un reseau spatial
WO2004053140A2 (fr) 2002-12-12 2004-06-24 Jawaharlal Nehru Centre For Advanced Scientific Research Modulateurs (inhibiteurs/activateurs) d'histone acetyltransferases
US20050009163A1 (en) 2003-01-10 2005-01-13 Liang Tong Methods of using crystal structure of carboxyltransferase domain of acetyl-CoA carboxylase, modulators thereof, and computer methods
US20050227915A1 (en) * 2001-05-02 2005-10-13 Steffan Joan S Methods and reagents for treating neurodegenerative diseases and motor deficit disorders
US20060167107A1 (en) 2002-12-12 2006-07-27 Kundu Tapas K Modulators (inhibitors/ activators) of histone acetyltransferases
US20070265296A1 (en) * 2005-11-28 2007-11-15 Dalton James T Nuclear receptor binding agents
US20090111863A1 (en) 2007-10-31 2009-04-30 Esposito Luke A Compounds, Compositions and Methods for the Treatment of Beta-Amyloid Diseases and Synucleinopathies
US20090264414A1 (en) * 2003-02-07 2009-10-22 High Point Pharmaceuticals, Llc Amide Derivatives and Pharmaceutical Use Thereof

Family Cites Families (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE1303920C2 (de) * 1966-01-15 1975-06-26 Farbwerke Hoechst AG vormals Meister Lucius & Biüning, 6000 Frankfurt Gamma-resorcylsaeure-anilide und verfahren zu ihrer herstellung
DE1642224B2 (de) * 1967-04-28 1976-04-29 Basf Ag, 6700 Ludwigshafen Verwendung von substituierten benzoesaeureaniliden zur bekaempfung von pilzen aus der klasse der basidiomyceten
US4088770A (en) * 1971-05-04 1978-05-09 Eli Lilly And Company Substituted 2-anilinobenzoxazoles used as immunosuppressive agents
JPS5842812B2 (ja) * 1976-05-14 1983-09-22 三共株式会社 木材防腐剤
JPS5459325A (en) * 1977-10-14 1979-05-12 Nippon Nohyaku Co Ltd Agent for controlling attached aquatic life
US4218438A (en) * 1979-02-14 1980-08-19 Eli Lilly And Company Anticoccidial combinations comprising nicarbazin and the polyether antibiotics
JPS57139053A (en) * 1981-02-24 1982-08-27 Showa Denko Kk Preparation of benzanilide compound
US5565325A (en) 1992-10-30 1996-10-15 Bristol-Myers Squibb Company Iterative methods for screening peptide libraries
US5484926A (en) * 1993-10-07 1996-01-16 Agouron Pharmaceuticals, Inc. HIV protease inhibitors
DK41193D0 (da) * 1993-04-07 1993-04-07 Neurosearch As Ionkanalaabnere
JP3160882B2 (ja) * 1996-02-02 2001-04-25 日本製紙株式会社 感熱記録シート
TW589189B (en) 1997-08-04 2004-06-01 Scras Kit containing at least one double-stranded RNA combined with at least one anti-viral agent for therapeutic use in the treatment of a viral disease, notably of viral hepatitis
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
AU3665199A (en) * 1998-04-29 1999-11-16 Vertex Pharmaceuticals Incorporated Inhibitors of impdh enzyme
GB9827152D0 (en) 1998-07-03 1999-02-03 Devgen Nv Characterisation of gene function using double stranded rna inhibition
EP2314700A1 (fr) 1999-01-28 2011-04-27 Medical College of Georgia Research Institute, Inc Composition et méthode destinées à l'attenuation in vivo et in vitro de l'expression génique utilisant de l'ARN double brin
DE19956568A1 (de) 1999-01-30 2000-08-17 Roland Kreutzer Verfahren und Medikament zur Hemmung der Expression eines vorgegebenen Gens
CA2386270A1 (fr) 1999-10-15 2001-04-26 University Of Massachusetts Genes de voies d'interference d'arn en tant qu'outils d'interference genetique ciblee
GB9927444D0 (en) 1999-11-19 2000-01-19 Cancer Res Campaign Tech Inhibiting gene expression
JP2001151742A (ja) * 1999-11-26 2001-06-05 Mitsui Chemicals Inc アニリド誘導体及びそれを含有する抗不整脈剤
US7429593B2 (en) * 2001-09-14 2008-09-30 Shionogi & Co., Ltd. Utilities of amide compounds
AU2003289222A1 (en) * 2002-12-06 2004-06-30 Toray Industries, Inc. Benzomorpholine derivatives
CA2532313A1 (fr) * 2003-07-16 2005-01-27 Institute Of Medicinal Molecular Design. Inc. Medicament pour le traitement de pigmentation de la peau
US8338638B2 (en) * 2006-08-25 2012-12-25 Unichem Laboratories Ltd. Antimicrobial derivatives of anacardic acid and process for preparing the same
KR20100039429A (ko) * 2007-08-02 2010-04-15 에프. 호프만-라 로슈 아게 Cns 질환의 치료를 위한 벤즈아미드 유도체의 용도
US9034387B2 (en) * 2007-10-03 2015-05-19 Jawaharlal Nehru Centre For Advanced Scientific Research Intrinsically fluorescent carbon nanospheres and a process thereof
TW200922468A (en) * 2007-10-11 2009-06-01 Sumitomo Chemical Co α, β-unsaturated imine compound and use thereof for pest control
JP2016081709A (ja) * 2014-10-16 2016-05-16 Tdk株式会社 リチウムイオン二次電池用負極活物質、およびそれを含む負極並びにリチウムイオン二次電池

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3254120A (en) 1961-12-05 1966-05-31 Pharmazeutische Fabrik Montavit Gmbh N-(tertiaryaminoalkoxy-benzoyl) anilides
WO1991005058A1 (fr) 1989-10-05 1991-04-18 Glenn Kawasaki Synthese et isolation sans cellule de nouveaux genes et de nouveaux polypeptides
WO1993020242A1 (fr) 1992-03-30 1993-10-14 The Scripps Research Institute Bibliotheques chimiques combinatoires codees
WO1994018318A1 (fr) 1993-02-01 1994-08-18 The University Of North Carolina At Chapel Hill Reactifs d'affinite entierement synthetiques
WO1995018972A1 (fr) 1994-01-05 1995-07-13 Arqule, Inc. Production modulaire systematique de molecules a base d'aminimide et d'oxazolone ayant des proprietes choisies
US5712171A (en) 1995-01-20 1998-01-27 Arqule, Inc. Method of generating a plurality of chemical compounds in a spatially arranged array
WO1996022529A1 (fr) 1995-01-20 1996-07-25 Arqule, Inc. Procede de production de differents composes chimiques constituant un reseau spatial
US20050227915A1 (en) * 2001-05-02 2005-10-13 Steffan Joan S Methods and reagents for treating neurodegenerative diseases and motor deficit disorders
WO2004053140A2 (fr) 2002-12-12 2004-06-24 Jawaharlal Nehru Centre For Advanced Scientific Research Modulateurs (inhibiteurs/activateurs) d'histone acetyltransferases
US20060167107A1 (en) 2002-12-12 2006-07-27 Kundu Tapas K Modulators (inhibitors/ activators) of histone acetyltransferases
US20090076155A1 (en) 2002-12-12 2009-03-19 Tapas Kumar Kundu Modulators (inhibitors/activators) of histone acetyltransferases
US20050009163A1 (en) 2003-01-10 2005-01-13 Liang Tong Methods of using crystal structure of carboxyltransferase domain of acetyl-CoA carboxylase, modulators thereof, and computer methods
US20090264414A1 (en) * 2003-02-07 2009-10-22 High Point Pharmaceuticals, Llc Amide Derivatives and Pharmaceutical Use Thereof
US20070265296A1 (en) * 2005-11-28 2007-11-15 Dalton James T Nuclear receptor binding agents
US20090111863A1 (en) 2007-10-31 2009-04-30 Esposito Luke A Compounds, Compositions and Methods for the Treatment of Beta-Amyloid Diseases and Synucleinopathies

Non-Patent Citations (28)

* Cited by examiner, † Cited by third party
Title
BERGE ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, no. 1, January 1977 (1977-01-01), pages 1 - 19, XP002675560, DOI: doi:10.1002/jps.2600660104
BRENNER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 89, 1992, pages 5381 - 5383
BUNIN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 91, 1994, pages 4708 - 4712
CHRISTIAN ET AL., J. MOL. BIOL., vol. 227, 1992, pages 711 - 718
DEVLIN ET AL., SCIENCE, vol. 249, 1990, pages 404 - 406
ERB ET AL., PROC. NATL. ACAD. SCI. USA, vol. 91, 1994, pages 11422 - 11426
FODOR ET AL., SCIENCE, vol. 251, 1991, pages 767 - 773
FORD, W. T.,: "ACS Symposium Series", 1986, AMERICAN CHEMICAL SOCIETY, pages: 186 - 230
GALLOP ET AL., J. MEDICINAL CHEMISTRY, vol. 37, no. 9, 1994, pages 1233 - 1251
HOUGHTEN ET AL., BIOTECHNIQUES, vol. 13, 1992, pages 412
HOUGHTEN ET AL., NATURE, vol. 354, 1991, pages 84 - 86
JAYAWICKREME ET AL., PROC. NATL. ACAD. SCI. USA, vol. 91, 1994, pages 1614 - 1618
KAY ET AL., GENE, vol. 128, 1993, pages 59 - 65
KIMURA ET AL., J BIOCHEM., vol. 138, no. 6, 2005, pages 647 - 662
LAM ET AL., NATURE, vol. 354, 1991, pages 82 - 84
LANE; CHABNER, J CLIN ONCOL., vol. 27, no. 32, 2009, pages 5459 - 68
LENSTRA, J. IMMUNOL. METH., vol. 152, 1992, pages 149 - 157
MANTELINGU ET AL.: "Activation of p300 Histone Acetyltransferase by Small Molecules Altering Enzyme Structure: Probed by Surface-Enhanced Raman Spectroscopy.", J. PHYS. CHEM. B, vol. 111, 2007, pages 4527 - 4534, XP055075158, DOI: doi:10.1021/jp067655s *
MARMORSTEIN, J MOLEC BIOL., vol. 311, 2001, pages 433 - 444
MATTHEAKIS ET AL., PROC. NATL. ACAD. SCI. USA, vol. 91, 1994, pages 9022 - 9026
MEDYNSKI, BIOTECHNOLOGY, vol. 12, 1994, pages 709 - 710
NAT REV MOL CELL BIOL., vol. 8, no. 4, 2007, pages 284 - 95
OHLMEYER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 10922 - 10926
OSTRESH ET AL., PROC. NATL. ACAD. SCI. USA, vol. 91, 1994, pages 11138 - 11142
SALMON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 11708 - 11712
SCOTT ET AL., SCIENCE, vol. 249, 1990, pages 386 - 390
See also references of EP2509590A4
SIMON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 89, 1992, pages 9367 - 9371

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10640457B2 (en) 2009-12-10 2020-05-05 The Trustees Of Columbia University In The City Of New York Histone acetyltransferase activators and uses thereof
US11034647B2 (en) 2009-12-10 2021-06-15 The Trustees Of Columbia University In The City Of New York Histone acetyltransferase activators and uses thereof
CN106432137B (zh) * 2010-12-22 2020-11-06 纽约市哥伦比亚大学理事会 组蛋白乙酰转移酶调节剂和其用途
CN103347392A (zh) * 2010-12-22 2013-10-09 纽约市哥伦比亚大学理事会 组蛋白乙酰转移酶调节剂和其用途
US9969677B2 (en) 2010-12-22 2018-05-15 The Trustees Of Columbia University In The City Of New York Histone acetyltransferase modulators and uses thereof
EP2654428A1 (fr) * 2010-12-22 2013-10-30 The Trustees of Columbia University in the City of New York Modulateurs de l'histone acétyltransférase et leurs utilisations
WO2012088420A1 (fr) 2010-12-22 2012-06-28 The Trustees Of Columbia University In The City Of New York Modulateurs de l'histone acétyltransférase et leurs utilisations
EP2654428A4 (fr) * 2010-12-22 2014-08-27 Univ Columbia Modulateurs de l'histone acétyltransférase et leurs utilisations
JP2017193578A (ja) * 2010-12-22 2017-10-26 ザ トラスティーズ オブ コロンビア ユニヴァーシティ イン ザ シティ オブ ニューヨーク ヒストンアセチル基転移酵素モジュレーターおよびその使用
CN106432137A (zh) * 2010-12-22 2017-02-22 纽约市哥伦比亚大学理事会 组蛋白乙酰转移酶调节剂和其用途
EP3323813A1 (fr) 2010-12-22 2018-05-23 The Trustees of Columbia University in the City of New York Modulateurs de l'histone acétyltransférase et leurs utilisations
EP2717688A1 (fr) * 2011-06-10 2014-04-16 The Trustees of Columbia University in the City of New York Utilisations d'activateurs d'histone acétyl tansférase
WO2012171008A1 (fr) 2011-06-10 2012-12-13 The Trustees Of Columbia University In The City Of New York Utilisations d'activateurs d'histone acétyl tansférase
US9809532B2 (en) 2011-06-10 2017-11-07 The Trustees Of Columbia University In The City Of New York Uses of histone acetyltransferase activators
EP2717688A4 (fr) * 2011-06-10 2014-11-19 Univ Columbia Utilisations d'activateurs d'histone acétyl tansférase
US10647709B2 (en) 2012-02-01 2020-05-12 The Trustees Of Columbia University In The City Of New York Cysteine protease inhibitors and uses thereof
WO2013116663A1 (fr) 2012-02-01 2013-08-08 The Trustees Of Columbia University In The City Of New York Nouveaux inhibiteurs de protéases à cystéine et leurs utilisations
JP2013208060A (ja) * 2012-03-30 2013-10-10 Nissin Foods Holdings Co Ltd 薬物代謝酵素の活性誘導方法及び薬物代謝酵素の活性が誘導されたヒト培養細胞
US11149020B2 (en) 2013-03-15 2021-10-19 The Trustees Of Columbia University In The City Of New York MAP kinase modulators and uses thereof
EP3521284A1 (fr) 2013-03-15 2019-08-07 The Trustees of Columbia University in the City of New York Composés de pyrazine comme modulateurs de map kinase et leurs utilisations
US10428047B2 (en) 2013-03-15 2019-10-01 The Trustees Of Columbia University In The City Of New York MAP kinase modulators and uses thereof
US11566016B2 (en) 2013-03-15 2023-01-31 The Trustees Of Columbia University In The City Of New York MAP kinase modulators and uses thereof
WO2014145485A2 (fr) 2013-03-15 2014-09-18 The Trustees Of Columbia University In The City Of New York Modulateurs de map kinase et utilisations de ceux-ci
US9783525B2 (en) 2013-03-15 2017-10-10 The Trustees Of Columbia University In The City Of New York Map kinase modulators and uses thereof
WO2015009930A2 (fr) 2013-07-17 2015-01-22 The Trustees Of Columbia University In The City Of New York Nouveaux inhibiteurs de la phosphodiestérase et utilisations de ceux-ci
US11191768B2 (en) 2014-03-31 2021-12-07 The Trustees Of Columbia University In The City Of New York Histone acetyltransferase activators and uses thereof
EP3398937A1 (fr) 2014-03-31 2018-11-07 The Trustees Of Columbia University In The City Of New York Activateurs de l'histone acétyltransférase et leurs utilisations
US10653648B2 (en) 2016-07-20 2020-05-19 The Trustees Of Columbia University In The City Of New York Histone acetyltransferase activators and compositions and uses thereof
US20230165813A1 (en) * 2016-07-20 2023-06-01 The Trustees Of Columbia University In The City Of New York Histone acetyltransferase activators and compositions and uses thereof
US11406608B2 (en) 2016-07-20 2022-08-09 The Trustees Of Columbia University In The City Of New York Histone acetyltransferase activators and compositions and uses thereof
WO2018017858A1 (fr) 2016-07-20 2018-01-25 The Trustees Of Columbia University In The City Of New York Activateurs d'histone acétyltransférase et compositions et utilisations associées
EP3920900A4 (fr) * 2019-02-08 2022-11-16 The Trustees of Columbia University in the City of New York Modulateurs d'histone acétyltransférase et compositions et utilisations associées
EP3920897A4 (fr) * 2019-02-08 2022-11-16 The Trustees of Columbia University in the City of New York Régulateurs d'histone acétyltransférase (hat) et utilisations associées

Also Published As

Publication number Publication date
ES2764999T3 (es) 2020-06-05
EP2509590A4 (fr) 2013-05-29
EP3632901B1 (fr) 2022-02-02
ES2907862T3 (es) 2022-04-26
EP2509590B1 (fr) 2019-10-30
JP2013513618A (ja) 2013-04-22
EP3632901A1 (fr) 2020-04-08
JP2016196459A (ja) 2016-11-24
JP2018027960A (ja) 2018-02-22
JP6093180B2 (ja) 2017-03-08
EP2509590A1 (fr) 2012-10-17
JP2020059733A (ja) 2020-04-16

Similar Documents

Publication Publication Date Title
US11034647B2 (en) Histone acetyltransferase activators and uses thereof
EP2509590B1 (fr) Activateurs d'histone acétyltransférase et utilisation de ceux-ci
JP6629797B2 (ja) ヒストンアセチル基転移酵素モジュレーターおよびその使用
JP2019527226A (ja) ヒストンアセチルトランスフェラーゼ活性化剤及び組成物並びにそれらの使用
US20180050982A1 (en) Uses of histone acetyltransferase activators
Zhao et al. Class I histone deacetylase inhibition by tianeptinaline modulates neuroplasticity and enhances memory
D’Mello Histone deacetylases as targets for the treatment of human neurodegenerative diseases
Henry et al. Quantitative measurement of histone tail acetylation reveals stage-specific regulation and response to environmental changes during Drosophila development
Ganai Histone Deacetylase Inhibitors-Epidrugs for Neurological Disorders
Basso et al. Designing Novel Dual Transglutaminase 2/Histone Deacetylase Inhibitors Effective in Halting Neuronal Death
Labrie Histone Deacetylase Inhibitors: A Novel Therapeutic Approach for Cognitive Disorders

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10836767

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2012543311

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2010836767

Country of ref document: EP