CA2584485C - Stilbenes and chalcones for the prevention and treatment of cardiovascular diseases - Google Patents

Stilbenes and chalcones for the prevention and treatment of cardiovascular diseases Download PDF

Info

Publication number
CA2584485C
CA2584485C CA2584485A CA2584485A CA2584485C CA 2584485 C CA2584485 C CA 2584485C CA 2584485 A CA2584485 A CA 2584485A CA 2584485 A CA2584485 A CA 2584485A CA 2584485 C CA2584485 C CA 2584485C
Authority
CA
Canada
Prior art keywords
hydroxyl
hydroxy
heteroaryl
aryl
formula
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CA2584485A
Other languages
French (fr)
Other versions
CA2584485A1 (en
Inventor
Fabrizio S. Chiacchia
Henrik C. Hansen
David Mccaffrey
Joseph E. L. Tucker
Norman C. W. Wong
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Resverlogix Corp
Original Assignee
Resverlogix Corp
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 Resverlogix Corp filed Critical Resverlogix Corp
Publication of CA2584485A1 publication Critical patent/CA2584485A1/en
Application granted granted Critical
Publication of CA2584485C publication Critical patent/CA2584485C/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • 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
    • C07D213/62Oxygen or sulfur atoms
    • C07D213/63One oxygen atom
    • C07D213/64One oxygen atom attached in position 2 or 6
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/05Phenols
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/05Phenols
    • A61K31/055Phenols the aromatic ring being substituted by halogen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/166Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the carbon of a carboxamide group directly attached to the aromatic ring, e.g. procainamide, procarbazine, metoclopramide, labetalol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/18Sulfonamides
    • 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
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/216Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acids having aromatic rings, e.g. benactizyne, clofibrate
    • 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
    • A61K31/265Esters, e.g. nitroglycerine, selenocyanates of carbonic, thiocarbonic, or thiocarboxylic acids, e.g. thioacetic acid, xanthogenic acid, trithiocarbonic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • A61K31/3533,4-Dihydrobenzopyrans, e.g. chroman, catechin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/357Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having two or more oxygen atoms in the same ring, e.g. crown ethers, guanadrel
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/382Heterocyclic compounds having sulfur as a ring hetero atom having six-membered rings, e.g. thioxanthenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4375Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having nitrogen as a ring heteroatom, e.g. quinolizines, naphthyridines, berberine, vincamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4412Non condensed pyridines; Hydrogenated derivatives thereof having oxo groups directly attached to the heterocyclic ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4418Non condensed pyridines; Hydrogenated derivatives thereof having a carbocyclic group directly attached to the heterocyclic ring, e.g. cyproheptadine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4433Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with oxygen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/465Nicotine; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/536Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines ortho- or peri-condensed with carbocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C205/00Compounds containing nitro groups bound to a carbon skeleton
    • C07C205/13Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by hydroxy groups
    • C07C205/20Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by hydroxy groups having nitro groups and hydroxy groups bound to carbon atoms of six-membered aromatic rings
    • C07C205/21Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by hydroxy groups having nitro groups and hydroxy groups bound to carbon atoms of six-membered aromatic rings having nitro groups and hydroxy groups bound to carbon atoms of the same non-condensed six-membered aromatic ring
    • C07C205/22Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by hydroxy groups having nitro groups and hydroxy groups bound to carbon atoms of six-membered aromatic rings having nitro groups and hydroxy groups bound to carbon atoms of the same non-condensed six-membered aromatic ring having one nitro groups bound to the ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C311/00Amides of sulfonic acids, i.e. compounds having singly-bound oxygen atoms of sulfo groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/22Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound oxygen atoms
    • C07C311/29Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound oxygen atoms having the sulfur atom of at least one of the sulfonamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C317/00Sulfones; Sulfoxides
    • C07C317/16Sulfones; Sulfoxides having sulfone or sulfoxide groups and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C317/22Sulfones; Sulfoxides having sulfone or sulfoxide groups and singly-bound oxygen atoms bound to the same carbon skeleton with sulfone or sulfoxide groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C327/00Thiocarboxylic acids
    • C07C327/20Esters of monothiocarboxylic acids
    • C07C327/28Esters of monothiocarboxylic acids having sulfur atoms of esterified thiocarboxyl groups bound to carbon atoms of hydrocarbon radicals substituted by singly-bound oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C37/00Preparation of compounds having hydroxy or O-metal groups bound to a carbon atom of a six-membered aromatic ring
    • C07C37/001Preparation of compounds having hydroxy or O-metal groups bound to a carbon atom of a six-membered aromatic ring by modification in a side chain
    • C07C37/003Preparation of compounds having hydroxy or O-metal groups bound to a carbon atom of a six-membered aromatic ring by modification in a side chain by hydrogenation of an unsaturated part
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C37/00Preparation of compounds having hydroxy or O-metal groups bound to a carbon atom of a six-membered aromatic ring
    • C07C37/01Preparation of compounds having hydroxy or O-metal groups bound to a carbon atom of a six-membered aromatic ring by replacing functional groups bound to a six-membered aromatic ring by hydroxy groups, e.g. by hydrolysis
    • C07C37/055Preparation of compounds having hydroxy or O-metal groups bound to a carbon atom of a six-membered aromatic ring by replacing functional groups bound to a six-membered aromatic ring by hydroxy groups, e.g. by hydrolysis the substituted group being bound to oxygen, e.g. ether group
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C37/00Preparation of compounds having hydroxy or O-metal groups bound to a carbon atom of a six-membered aromatic ring
    • C07C37/11Preparation of compounds having hydroxy or O-metal groups bound to a carbon atom of a six-membered aromatic ring by reactions increasing the number of carbon atoms
    • C07C37/18Preparation of compounds having hydroxy or O-metal groups bound to a carbon atom of a six-membered aromatic ring by reactions increasing the number of carbon atoms by condensation involving halogen atoms of halogenated compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C39/00Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a six-membered aromatic ring
    • C07C39/12Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a six-membered aromatic ring polycyclic with no unsaturation outside the aromatic rings
    • C07C39/14Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a six-membered aromatic ring polycyclic with no unsaturation outside the aromatic rings with at least one hydroxy group on a condensed ring system containing two rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C39/00Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a six-membered aromatic ring
    • C07C39/12Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a six-membered aromatic ring polycyclic with no unsaturation outside the aromatic rings
    • C07C39/15Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a six-membered aromatic ring polycyclic with no unsaturation outside the aromatic rings with all hydroxy groups on non-condensed rings, e.g. phenylphenol
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C39/00Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a six-membered aromatic ring
    • C07C39/205Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a six-membered aromatic ring polycyclic, containing only six-membered aromatic rings as cyclic parts with unsaturation outside the rings
    • C07C39/21Compounds having at least one hydroxy or O-metal group bound to a carbon atom of a six-membered aromatic ring polycyclic, containing only six-membered aromatic rings as cyclic parts with unsaturation outside the rings with at least one hydroxy group on a non-condensed ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C43/00Ethers; Compounds having groups, groups or groups
    • C07C43/02Ethers
    • C07C43/20Ethers having an ether-oxygen atom bound to a carbon atom of a six-membered aromatic ring
    • C07C43/23Ethers having an ether-oxygen atom bound to a carbon atom of a six-membered aromatic ring containing hydroxy or O-metal groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C46/00Preparation of quinones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/76Ketones containing a keto group bound to a six-membered aromatic ring
    • C07C49/82Ketones containing a keto group bound to a six-membered aromatic ring containing hydroxy groups
    • C07C49/83Ketones containing a keto group bound to a six-membered aromatic ring containing hydroxy groups polycyclic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/76Ketones containing a keto group bound to a six-membered aromatic ring
    • C07C49/82Ketones containing a keto group bound to a six-membered aromatic ring containing hydroxy groups
    • C07C49/835Ketones containing a keto group bound to a six-membered aromatic ring containing hydroxy groups having unsaturation outside an aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C50/00Quinones
    • C07C50/26Quinones containing groups having oxygen atoms singly bound to carbon atoms
    • C07C50/32Quinones containing groups having oxygen atoms singly bound to carbon atoms the quinoid structure being part of a condensed ring system having two rings
    • 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
    • C07D213/62Oxygen or sulfur atoms
    • C07D213/63One oxygen atom
    • C07D213/65One oxygen atom attached in position 3 or 5
    • 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
    • C07D213/79Acids; Esters
    • C07D213/80Acids; Esters in position 3
    • 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
    • C07D213/89Heterocyclic 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 with hetero atoms directly attached to the ring nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms 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
    • C07D215/20Oxygen atoms
    • C07D215/22Oxygen atoms attached in position 2 or 4
    • C07D215/233Oxygen atoms attached in position 2 or 4 only one oxygen atom which is attached in position 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/12Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with radicals, substituted by hetero atoms, attached to carbon atoms of the nitrogen-containing ring
    • C07D217/14Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with radicals, substituted by hetero atoms, attached to carbon atoms of the nitrogen-containing ring other than aralkyl radicals
    • C07D217/16Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with radicals, substituted by hetero atoms, attached to carbon atoms of the nitrogen-containing ring other than aralkyl radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/22Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems 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 carbon atoms of the nitrogen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/22Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems 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 carbon atoms of the nitrogen-containing ring
    • C07D217/24Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D265/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one oxygen atom as the only ring hetero atoms
    • C07D265/041,3-Oxazines; Hydrogenated 1,3-oxazines
    • C07D265/121,3-Oxazines; Hydrogenated 1,3-oxazines condensed with carbocyclic rings or ring systems
    • C07D265/141,3-Oxazines; Hydrogenated 1,3-oxazines condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • C07D265/201,3-Oxazines; Hydrogenated 1,3-oxazines condensed with carbocyclic rings or ring systems condensed with one six-membered ring with hetero atoms directly attached in position 4
    • C07D265/22Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/22Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4
    • C07D311/26Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with aromatic rings attached in position 2 or 3
    • C07D311/28Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with aromatic rings attached in position 2 or 3 with aromatic rings attached in position 2 only
    • C07D311/30Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with aromatic rings attached in position 2 or 3 with aromatic rings attached in position 2 only not hydrogenated in the hetero ring, e.g. flavones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/22Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4
    • C07D311/26Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with aromatic rings attached in position 2 or 3
    • C07D311/28Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with aromatic rings attached in position 2 or 3 with aromatic rings attached in position 2 only
    • C07D311/322,3-Dihydro derivatives, e.g. flavanones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/22Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4
    • C07D311/26Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with aromatic rings attached in position 2 or 3
    • C07D311/34Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with aromatic rings attached in position 2 or 3 with aromatic rings attached in position 3 only
    • C07D311/36Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with aromatic rings attached in position 2 or 3 with aromatic rings attached in position 3 only not hydrogenated in the hetero ring, e.g. isoflavones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/58Benzo[b]pyrans, not hydrogenated in the carbocyclic ring other than with oxygen or sulphur atoms in position 2 or 4
    • C07D311/60Benzo[b]pyrans, not hydrogenated in the carbocyclic ring other than with oxygen or sulphur atoms in position 2 or 4 with aryl radicals attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D319/00Heterocyclic compounds containing six-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D319/101,4-Dioxanes; Hydrogenated 1,4-dioxanes
    • C07D319/141,4-Dioxanes; Hydrogenated 1,4-dioxanes condensed with carbocyclic rings or ring systems
    • C07D319/161,4-Dioxanes; Hydrogenated 1,4-dioxanes condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • C07D319/201,4-Dioxanes; Hydrogenated 1,4-dioxanes condensed with carbocyclic rings or ring systems condensed with one six-membered ring with substituents attached to the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D335/00Heterocyclic compounds containing six-membered rings having one sulfur atom as the only ring hetero atom
    • C07D335/04Heterocyclic compounds containing six-membered rings having one sulfur atom as the only ring hetero atom condensed with carbocyclic rings or ring systems
    • C07D335/06Benzothiopyrans; Hydrogenated benzothiopyrans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/052Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being six-membered

Abstract

The present disclosure provides non-naturally occurring polyphenol compounds that upregulate the expression of Apolipoprotein A-I (ApoA-I). The disclosed compositions and methods can be used for treatment and prevention of cardiovascular disease and related disease states, including cholesterol or lipid related disorders, such as, e.g., atherosclerosis.

Description

tv/

OF CARDIOVASCULAR DISEASES
_ Technical Field [002] The present disclosure relates to polyphenol compounds, which = are useful for regulating the expression of apolipoprotein A-I (ApoA-I), and their use for treatment and prevention of cardiovascular disease and related disease states, including cholesterol or lipid related disorders, such as, e.g., atherosclerosis.
BACKGROUND
[003] Epidemiologic data demonstrate an inverse relationship between circulating levels of high density lipoprotein cholesterol (HDL-C) and the incidence of clinically significant atherosclerosis. Each 1 mg/d1 increment in the HDL-C

serum level is associated with a 2-3% decrement in cardiovascular risk; a 1%
reduction in LDL-C reduces coronary heart disease (CHD) risk by 2%. Gordon et al., Am. J. Med. 62(5):707-14 (1997). Experimental evidence further supports the protective effect of HDL against cardiovascular disease. For example, in subjects with low HDL-C, administration of gemfibrozil results in a 6% increase in the HDL-C level and a corresponding 22% reduction of the CHD risk Rubins et al., N.
Engl.

J. Med. 341(6):410-8 (1999). Observations in genetic disorders associated with low HDL due to reduced ApoA-I expression, also indicate the link between elevated risk of CHD and low HDL-C.
[004] HDL appears to exert its antiatherogenic effect by mediating reverse cholesterol transport (RCT), in which cholesterol is recruited from peripheral tissues and transported to the liver. In addition, HDL also exerts anti-inflammatory, andtioxidant effects and promotes fibrinolysis. HDL paticles protect against oxidation of LDL, an important initial step in promoting cholseteol uptake by arterial macrophages. HDL exists in two main forms, one containing both apolipoprotein A-I (ApoA-I) and apolipoprotein A-II (ApoA-II), and the other containing ApoA-I without ApoA-II. Schultz et al., Nature 365(6448):762-4 (1993).
The cardioprotective effect of HDL is mostly, but not exclusively, attributable to ApoA-I.
[005] Clinical and experimental data suggest that the production of ApoA-I is a critical determinant of circulating HDL. For example, persons with familial hyperalphalipoproteinemia (elevated ApoA-I) appear to be protected from atherosclerosis, while those deficient in ApoA-I (hypoalphalipoproteinemia) show accelerated cardiovascular disease. In addition, various experimental manipulations to increase production of ApoA-I are associated with reduced atherogenicity. For example, human ApoA-I is protective in transgenic animal models (Shah et al., Circulation 97(8):780-5 (1998); Rubin et al., Nature 353(6341):265-7 (1991)), and treatment with ApoA-IMilano prevents atherosclerotic lesions and leads to regression of atherosclerotic plaques in human patients (Nissen et al., JAMA 290(17):2292-300 (2003)). Further lines of research supporting an antiatherogenic role of ApoA-I, include: enhancement of reverse cholesterol transport, attenuation of oxidative stress, increased peroxonase activity, enhanced anticoagulant activity, and anti-inflammatory activity.
Accordingly, ApoA-I is an attractive target for therapeutic intervention.
[006] Currently available therapeutic agents that increase the plasma concentration of ApoA-I, for example, recombinant ApoA-I or peptides that mimic ApoA-I, have potential drawbacks related to manufacturing and reproducibility, e.g., stability during storage, delivery of an active product, and in vivo half-life.
Therefore, small molecule compounds that upregulate the production of endogenous ApoA-I, such as, e.g., transcriptional upregulators of ApoA-I
expression, are very attractive as new therapeutic agents for cardiovascular disease.
[007] One class of compounds that are thought to contribute to the prevention of various diseases, including cancer and cardiovascular diseases, is polyphenols. Polyphenols are common constituents of the human diet, present in most food and beverages of plant origin, and are the most abundant dietary antioxidants. However, polyphenols protective properties are often minimized due to poor bioavailability, lack of clinical significance, and deleterious effects at high concentrations. For example, the most abundant and available source of resveratrol for consumers, red wine, cannot be consumed in therapeutically efficacious quantities on a daily basis due to the numerous well documented deleterious effects of excessive alcohol consumption. The actions of resveratrol may be better or safer in the absence of alcohol.
[008] Several human clinical studies, involving foods or beverages, have yet to demonstrate an unequivocal benefit on primary clinical endpoints, such as oxidative stress, lipemia and inflammation. For example, out of 12 recent intervention studies with differing polyphenol sources; 6 showed no effect on lipid parameters and the other 6 showed an improvement in the lipid parameters.
Manach, Curr. Opin. Lipidol. 16(1):77-84 (2005). Such contradictory data has limited the potential use of polyphenols, despite their many beneficial properties.
[009] The use of naturally occurring polyphenols as a potential therapy has also been impeded by the inability to achieve efficacious levels of bioavailability. The bioavailability of polyphenols in humans range from 1% to 26% and has a large inter-individual variability as well as variability between different polyphenols. Polyphenols differ in how they are absorbed, metabolized, and excreted. For example, polyphenol flavonoids, such as quercetin, have been reported to have less than 1% intestinal absorption following oral administration.
Gugler et al., Eur. J. Clin. Pharm. 9:223 (1975). In addition, metabolites are known to negatively influence the biological activity of the parent compounds.

Such metabolites often differ from the parent compound in terms of toxicity, efficacy and length of residence in the plasma. Another limiting factor may be polyphenols' poor solubility in water which limits the routes of administration.
These and other factors have made it difficult to determine appropriate dosages of the naturally occurring polyphenols, naringenin or resveratrol, for use in humans.
[010] Thus, there exists a need for synthetic polyphenols to be developed as therapeutic agents for the treatment and prevention of cardiovascular and related diseases, particularly, cholesterol or lipid related disorders, such as, e.g., atherosclerosis. It is therefore one of the objects of the present invention to provide compounds that upregulate the expression of ApoA-I, while having more favorable pharmacological properties than naturally occurring polyphenols.

SUMMARY
[011] The methods of invention include administering to a mammal (e.g., a human) in need thereof a therapeutically effective amount of a stilbene compound of Formula I:
(R4)p (R5)p (ROI) (R7)P
W=W W==W
(R3)pµ / _______________________ X z _____ W
.µ"V
(R2)p (R1)p (R10)p (R9) Formula I
wherein:
X is selected from C, CRii, CR11R13, CO, CS, 0, S, SO, SO2, N and NRii, wherein R11 may be the same or different than R13;
Y is selected from C, CR12, CR12R14, CO, CS, 0, S, SO, SO2, N and NR12, wherein R12 may be the same or different than R14;
R1, R2, R3, Ra, R5, R6, R7, Rg, Rg, R10, R11, R12, R13, and R1.4 are each independently selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone, or two adjacent substituents selected from R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, and R14 are connected in a 5 or 6-membered ring to form a bicyclic aryl or bicyclic heteroaryl;
each W is independently selected from C and N, wherein if W is N, then p is 0 and if .W is C, then p is 1;
Z is selected from a single bond, a double bond and a triple bond;

and pharmaceutically acceptable salts and hydrates thereof;
wherein if R3 and R9 are each hydroxyl, then at least one of R11 and R12 is not alkyl;
wherein if Z is a double bond, then R2, R4 and R9 are not each hydroxyl;
and wherein if Formula 1 has the structure:

OH

then at least one of R7 and R9 is not alkyl.
[012] Methods of invention also include administering to a mammal (e.g., a human) in need thereof a therapeutically effective amount of a chalcone compound of Formula IV:
(R7)p \w/
(R4)8 (RA' w w(Rs)p (R9)p 1z2 I
(Rio)p ZY
(Ri)p Formula IV
wherein:
X1 is selected from C, CRit CR11R13, CO, CS, 0, S, SO, SO2, N and NR11, wherein R11 may be the same or different than R13;
X2 is selected from C, CR17, CR17R18, CO, CS, 0, S, SO, SO2, N and NR17, wherein R17 may be the same or different than R18;

Y is selected from C, CR12, CR12R14, CO, CS, S, SO, SO2, N and NR12, wherein R12 may be the same or different than R14, R1, R2, R3, Ra, R5, R6, R7, R8, R9, Rio, R11, R12, R13, R14, R17 and R18 are each independently selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone, or two adjacent substituents selected from Ri, R2, R3, R4, R5, R6, R7, R8, R9, Rio, R11, R12, R13, R14, R17 and R18 are connected in a 5 or 6-membered ring to form a bicyclic aryl or bicyclic heteroaryl;
each W is independently selected from C and N, wherein if W is N, then p is 0 and if W is C, then p is 1;
Zi and Z2 are each independently selected from a single bond, a double bond, and a triple bond;
and pharmaceutically acceptable salts and hydrates thereof.
[013] In certain embodiments, the methods and compositions of the invention are useful for treatment of diseases characterized by reduced ApoA-I

and/or HDL. The compounds and compositions of the invention can be used to increase expression of ApoA-I. Increasing expression of ApoA-I refers to transcriptionally modulating the expression of the ApoA-I gene thereby affecting the level of the ApoA-I protein expressed, i.e., synthesized and secreted, by the cell. An increase in ApoA-I protein expression leads to an increase in blood levels of HDL. Thus, the methods and compounds of the invention may further be used to reduce plasma cholesterol levels. Accordingly, the methods and compositions of the invention can be used for treatment and prevention of cardiovascular disease and related disease states, particularly, cholesterol or lipid related disorders, such as, atherosclerosis, dyslipidemias, dyslipoproteinemias, hypertension, coronary artery disease, cerebrovascular disease, and the like.
[014] In one aspect, the invention provides a method for prevention of arteriosclerosis lesion development in a mammal, including the development of new arteriosclerotic lesions. In another aspect, the present invention provides a method regressing arteriosclerosis lesions.
[015] The methods and compounds of the invention may further be used to lower blood levels of LDL and triglycerides and/or to increase free radical scavenging. In addition, these methods and compositions may also be used to inhibit HMG-CoA reductase, inhibit ACAT, and/or increase ABCA-I activity.
[016] In a further aspect, the invention provides methods and compositions for effecting an increase of HDL in a mammal comprising, wherein the compound is a stilbene or a chalcone, each derivatized with covalently bonded niacin. In certain embodiments, the covalent bond comprises a reverse ester linkage.
DETAILED DESCRIPTION
Definitions [017] The term "aldehyde" or "formyl" as used herein refers to the radical -CHO.
[018] The term "alkenyl" as used herein refers to an unsaturated straight or branched hydrocarbon having at least one carbon-carbon double bond, such as a straight or branched group of 2-22, 2-8, or 2-6 carbon atoms, referred to herein as (C2_C22)alkenyl, (C2_C8)alkenyl, and (C2_C6)alkenyl, respectively.
Exemplary alkenyl groups include, but are not limited to, vinyl, allyl, butenyl, pentenyl, hexenyl, butadienyl, pentad ienyl, hexadienyl, 2-ethylhexenyl, 2-propy1-2-butenyl, 4-(2-methyl-3-butene)-pentenyl, etc.
[019] The term "alkoxy" as used herein refers to an alkyl group attached to an oxygen (-0-alkyl-). "Alkoxy" groups also include an alkenyl group attached to an oxygen ("alkenoxy") or an alkynyl group attached to an oxygen ("alkynoxy") groups. Exemplary alkoxy groups include, but are not limited to, groups with an alkyl, alkenyl or alkynyl group of 1-22, 1-8, or 1-6 carbon atoms, referred to herein as (C1_C22)alkoxy, (C1_C8)alkoxy, and (C1_C8)alkoxy, respectively. Exemplary alkoxy groups include, but are not limited to methoxy, ethoxy, etc.
[020] The term "alkyl" as used herein refers to a saturated straight or branched hydrocarbon, such as a straight or branched group of 1-22, 1-8, or 1-carbon atoms, referred to herein as (C1_C22)alkyl, (C1_C8)alkyl, and (C1_C8)alkyl, respectively. Exemplary alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, 2-methyl-1-propyl, 2-methyl-2-propyl, 2-methyl-1-butyl, 3-methyl-1-butyl, 2-methyl-3-butyl, 2,2-dimethy1-1-propyl, 2-methyl-1-pentyl, 3-methy1-pentyl, 4-methyl-1-pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethy1-1-butyl, 3,3-dimethy1-1-butyl, 2-ethyl-1-butyl, butyl, isobutyl, t-butyl, pentyl, isopentyl, neopentyl, hexyl, heptyl, octyl, etc.
[021] The term "alkynyl" as used herein refers to an unsaturated straight or branched hydrocarbon having at least one carbon-carbon triple bond, such as a straight or branched group of 2-22, 2-8, or 2-6 carbon atoms, referred to herein as (C2_C22)alkynyl, (C2_C8)alkynyl, and (C2_C6)alkynyl, respectively.
Exemplary alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, pentynyl, hexynyl, methylpropynyl, 4-methyl-1-butynyl, 4-propy1-2-pentynyl, and 4-butyl-hexynyl, etc.
[022] The term "amide" as used herein refers to a radical of the form -RaC(0)N(Rb)-, -RaC(0)N(Rb)Rc_, or -C(0)NRbRc, wherein Rb and Rc are each independently selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone.
The amide can be attached to another group through the carbon, the nitrogen, Rb, Rc, or Ra. The amide also may be cyclic, for example Rb and Rc, Ra and Rb, or Ra and Rc may be joined to form a 3- to 12-membered ring, such as a 3- to 10-membered ring or a 5- to 6-membered ring. The term "amide" encompasses groups such as sulfonamide, urea, carbamate, carbamic acid, and cyclic versions thereof. The term "amide" also encompasses an amide group attached to a carboxy group, e.g., ¨amide-000H or salts such as ¨amide-COONa, etc, an amino group attached to a carboxy group, e.g., ¨amino-COOH or salts such as ¨
amino-COONa, etc.
[023] The term "amine" or "amino" as used herein refers to a radical of the form -NRdRe, -N(Rd)Re.., or -ReN(Rd)Rf_ where Rd, Re, and Rf are independently selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone.
The amino can be attached to the parent molecular group through the nitrogen, Rd, Re or Rf. The amino also may be cyclic, for example any two of Ra, Rb, and Rc may be joined together or with the N to form a 3-to 12-membered ring, e.g., morpholino or piperidinyl. The term amino also includes the corresponding quaternary ammonium salt of any amino group, e.g., ¨[N(Rd)(Re)(Rf)].
Exemplary amino groups include aminoalkyl groups, wherein at least one of Rd, Re, or Rf is an alkyl group.
[024] The term "aryl" as used herein refers to a mono-, bi-, or other multi-carbocyclic, aromatic ring system. The aryl group can optionally be fused to one or more rings selected from aryls, cycloalkyls, and heterocyclyls. The aryl groups of this invention can be substituted with groups selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone. Exemplary aryl groups include, but are not limited to, phenyl, tolyl, anthracenyl, fluorenyl, indenyl, azulenyl, and naphthyl, as well as benzo-fused carbocyclic moieties such as 5,6,7,8-tetrahydronaphthyl. Exemplary aryl groups also include, but are not limited to a monocyclic aromatic ring system, wherein the ring comprises 6 carbon atoms, referred to herein as "(C6)aryl."
[025] The term "arylalkyl" as used herein refers to an aryl group having at least one alkyl substituent, e.g. -aryl-alkyl-. Exemplary arylalkyl groups include, but are not limited to, arylalkyls having a monocyclic aromatic ring system, wherein the ring comprises 6 carbon atoms, referred to herein as "(C6)arylalkyl."
[026] The term "aryloxy" as used herein refers to an aryl group attached to an oxygen atom. Exemplary aryloxy groups include, but are not limited to, aryloxys having a monocyclic aromatic ring system, wherein the ring comprises carbon atoms, referred to herein as "(C6)aryloxy."
[027] The term "arylthio" as used herein refers to an aryl group attached to an sulfur atom. Exemplary arylthio groups include, but are not limited to, arylthios having a monocyclic aromatic ring system, wherein the ring comprises 6 carbon atoms, referred to herein as "(C6)arylthio."
[028] The term "arylsulfonyl" as used herein refers to an aryl group attached to a sulfonyl group, e.g., ¨S(0)2-aryl--. Exemplary arylsulfonyl groups include, but are not limited to, arylsulfonyls having a monocyclic aromatic ring system, wherein the ring comprises 6 carbon atoms, referred to herein as "(C6)arylsulfonyl."
[029] The term "benzyl" as used herein refers to the group -CH2-phenyl.
[030] The term "bicyclic aryl" as used herein refers to an aryl group fused to another aromatic or non-aromatic carbocylic or heterocyclic ring. Exemplary bicyclic aryl groups include, but are not limited to, naphthyl or partly reduced forms thereof, such as di-, tetra-, or hexahydronaphthyl.
[031] The term "bicyclic heteroaryl" as used herein refers to a heteroaryl group fused to another aromatic or non-aromatic carbocylic or heterocyclic ring.
Exemplary bicyclic heteroaryls include, but are not limited to, 5,6 or 6,6-fused systems wherein one or both rings contain heteroatoms. The term "bicyclic heteroaryl" also encompasses reduced or partly reduced forms of fused aromatic system wherein one or both rings contain ring heteroatoms. The ring system may contain up to three heteroatoms, independently selected from oxygen, nitrogen, or sulfur. The bicyclic system may be optionally substituted with one or more groups selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone. Exemplary bicyclic heteroaryl's include, but are not limited to, quinazolinyl, benzothiophenyl, benzoxazolyl, benzimidazolyl, benzothiazolyl, benzofuranyl, indolyl, quinolinyl, isoquinolinyl, phthalazinyl, benzotriazolyl, benzopyridinyl, and benzofuranyl.
[032] The term "carbamate" as used herein refers to a radical of the form -Rg0C(0)N(Rh), -Rg0C(0)N(Rh)Ri_, or -0C(0)NRhRi, wherein Rg, Rh and Ri are each independently selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone. Exemplary carbamates include, but are not limited to, arylcarbamates or heteroaryl carbamates, e.g. wherein at least one of Rg, Rh and Ri are independently selected from aryl or heteroaryl, such as pyridine, pyridazine, pyrimidine, and pyrazine.
[033] The term "carbonyl" as used herein refers to the radical -C(0)-.
[034] The term "carboxy" as used herein refers to the radical -COOH or its corresponding salts, e.g. ¨COONa, etc. The term carboxy also includes "carboxycarbonyl," e.g. a carboxy group attached to a carbonyl group, e.g., ¨C(0)-COOH or salts such as -C(0)-COONa, etc..
[035] The term "cyano" as used herein refers to the radical -CN.
[036] The term "cycloalkoxy" as used herein refers to a cycloalkyl group attached to an oxygen.
[037] The term "cycloalkyl" as used herein refers to a monovalent saturated or unsaturated cyclic, bicyclic, or bridged bicyclic hydrocarbon group of 3-12 carbons, or 3-8 carbons, referred to herein as "(C3-C8)cycloalkyl,"
derived from a cycloalkane. Exemplary cycloalkyl groups include, but are not limited to, cyclohexanes, cyclohexenes, cyclopentanes, and cyclopentenes. Cycloalkyl groups may be substituted with alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone.
Cycloalkyl groups can be fused to other cycloalkyl, aryl, or heterocyclyl groups.
[038] The term "dicarboxylic acid" as used herein refers to a group containing at least two carboxylic acid groups such as saturated and unsaturated hydrocarbon dicarboxylic acids and salts thereof. Exemplary dicarboxylic acids include alkyl dicarboxylic acids. Dicarboxylic acids may be substituted with alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone. Dicarboxylic acids include, but are not limited to succinic acid, glutaric acid, adipic acid, suberic acid, sebacic acid, azelaic acid, maleic acid, phthalic acid, aspartic acid, glutamic acid, malonic acid, fumaric acid, (+)/(-)-malic acid, (+)/(-) tartaric acid, isophthalic acid, and terephthalic acid. Dicarboxylic acids further include carboxylic acid derivatives thereof, such as anhydrides, imides, hydrazides, etc., for example, succinic anhydride, succinimide, etc.
[039] The term "ester" refers to a radical having the structure -C(0)0-, -RkC(0)0-Rj_1 or -RkC(0)0-, where 0 is not bound to hydrogen, and Ri and Rk can independently be selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, cycloalkyl, ether, formyl, haloalkyl, halogen, heteroaryl, heterocyclyl, ketone, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid and thioketone. Rk can be a hydrogen, but Ri cannot be hydrogen. The ester may be cyclic, for example the carbon atom and R, the oxygen atom and Rk, or R. and Rk may be joined to form a 3- to 12-membered ring. Exemplary esters include, but are not limited to, alkyl esters wherein at least one of Rj or Rk is alkyl, such as ¨alkyl-C(0)-0¨, ¨C(0)-0-alkyl¨, ¨alkyl¨C(0)-0-alkyl¨, etc. Exemplary esters also include aryl or heteoraryl esters, e.g. wherein at least one of Rj or Rk is a heteroaryl group such as pyridine, pyridazine, pyrmidine and pyrazine, such as a nicotinate ester. Exemplary esters also include reverse esters having the structure -RkC(0)0-, where the oxygen is bound to the parent molecular group.
Exemplary reverse esters include succinate, D-argininate, L-argininate, L-lysinate and D-lysinate. Esters also include carboxylic acid anhydrides and acid halides.
[040] The term "ether" refers to a radical having the structure -RIO-Rm_, where R1 and Rm can independently be alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heterocyclyl, or ether. The ether can be attached to the parent molecular group through R1 or Rm. Exemplary ethers include, but are not limited to, alkoxyalkyl and alkoxyaryl groups. Ethers also includes polyethers, e.g., where one or both of R1 and Rm are ethers.
[041] The terms "halo" or "halogen" or "Hal" as used herein refer to F, Cl, Br, or I.
[042] The term "haloalkyl" as used herein refers to an alkyl group substituted with one or more halogen atoms. "Haloalkyls" also encompass alkenyl or alkynyl groups substituted with one or more halogen atoms.
[043] The term "heteroaryl" as used herein refers to a mono-, bi-, or multi-cyclic, aromatic ring system containing one or more heteroatoms, for example 1 to 3 heteroatoms, such as nitrogen, oxygen, and sulfur. Heteroaryls can be substituted with one or more substituents including alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone.
Heteroaryls can also be fused to non-aromatic rings. Illustrative examples of heteroaryl groups include, but are not limited to, pyridinyl, pyridazinyl, pyrimidyl, pyrazyl, triazinyl, pyrrolyl, pyrazolyl, imidazolyl, (1,2,3,)- and (1,2,4)-triazolyl, pyrazinyl, pyrimidilyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, furyl, phenyl, isoxazolyl, and oxazolyl. Exemplary heteroaryl groups include, but are not limited to, a monocyclic aromatic ring, wherein the ring comprises 2 to 5 carbon atoms and 1 to 3 heteroatoms, referred to herein as "(C2-05)heteroaryl."
[044] The terms "heterocycle," "heterocyclyl," or "heterocyclic" as used herein refer to a saturated or unsaturated 3-, 4-, 5-, 6- or 7-membered ring containing one, two, or three heteroatoms independently selected from nitrogen, oxygen, and sulfur. Heterocycles can be aromatic (heteroaryls) or non-aromatic.
Heterocycles can be substituted with one or more substituents including alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone.
[045] Heterocycles also include bicyclic, tricyclic, and tetracyclic groups in which any of the above heterocyclic rings is fused to one or two rings independently selected from aryls, cycloalkyls, and heterocycles. Exemplary heterocycles include acrid inyl, benzimidazolyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, biotinyl, cinnolinyl, dihydrofuryl, dihydroindolyl, dihydropyranyl, dihydrothienyl, dithiazolyl, furyl, homopiperidinyl, imidazolidinyl, imidazolinyl, imidazolyl, indolyl, isoquinolyl, isothiazolidinyl, isothiazolyl, isoxazolidinyl, isoxazolyl, morpholinyl, oxadiazolyl, oxazolidinyl, oxazolyl, piperazinyl, piperidinyl, pyranyl, pyrazolidinyl, pyrazinyl, pyrazolyl, pyrazolinyl, pyridazinyl, pyridyl, pyrimidinyl, pyrimidyl, pyrrolidinyl, pyrrolidin-2-onyl, pyrrolinyl, pyrrolyl, quinolinyl, quinoxaloyl, tetrahydrofuryl, tetrahydroisoquinolyl, tetrahydropyranyl, tetrahydroquinolyl, tetrazolyl, thiadiazolyl, thiazolidinyl, thiazolyl, thienyl, thiomorpholinyl, thiopyranyl, and triazolyl.
[046] The terms "hydroxy" and "hydroxyl" as used herein refers to the radical -OH.
[047] The term "hydroxyalkyl" as used herein refers to a hydroxy radical attached to an alkyl group.
[048] The term "hydroxyaryl" as used herein refers to a hydroxy radical attached to an aryl group.
[049] The term "ketone" as used herein refers to a radical having the structure -C(0)-Rn (such as acetyl, -C(0)CH3) or -Rn_C(0)-Ro_. The ketone can be attached to another group through Rn or Ro. Rn or Ro can be alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl or aryl, or Rn or Ro can be joined to form a 3- to 12-membered ring.
[050] The term "monoester" as used herein refers to an analogue of a dicarboxylic acid wherein one of the carboxylic acids is functionalized as an ester and the other carboxylic acid is a free carboxylic acid or salt of a carboxylic acid.
Examples of monoesters include, but are not limited to, to monoesters of succinic acid, glutaric acid, adipic acid, suberic acid, sebacic acid, azelaic acid, oxalic and maleic acid.
[051] The term "nitro" as used herein refers to the radical -NO2.
[052] The term "perfluoroalkoxy" as used herein refers to an alkoxy group in which all of the hydrogen atoms have been replaced by fluorine atoms.
[053] The term "perfluoroalkyl" as used herein refers to an alkyl group in which all of the hydrogen atoms have been replaced by fluorine atoms.
Exemplary perfluroalkyl groups include, but are not limited to, C1-5 perfluoroalkyl, such as trifluorom ethyl, etc.
[054] The term "perfluorocycloalkyl" as used herein refers to a cycloalkyl group in which all of the hydrogen atoms have been replaced by fluorine atoms.
[055] The term "phenyl" as used herein refers to a 6-membered carbocyclic aromatic ring. The phenyl group can also be fused to a cyclohexane or cyclopentane ring. Phenyl can be substituted with one or more substituents including alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone.
[056] The term "phosphate" as used herein refers to a radical having the structure -0P(0)02-, ¨Rx0P(0)02-, ¨0P(0)02Ry-, or ¨Rx0P(0)02Ry-, wherein Rx and Ry can be alkyl, alkenyl, alkynyl, alkoxy, amide, amino, aryl, aryloxy, carboxy, cyano, cycloalkyl, ester, ether, halogen, heterocyclyl, hydrogen, hydroxy, ketone, nitro, sulfonate, sulfonyl, and thio.
[057] The term "sulfide" as used herein refers to the radical having the structure RS-, where Rz can be alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cycloalkyl, ester, ether, formyl, haloalkyl, heteroaryl, heterocyclyl, and ketone. The term "alkylsulfide" as used herein refers to an alkyl group attached to a sulfur atom.
[058] The term "sulfinyl" as used herein refers to a radical having the structure -S(0)0-, -RS(0)O-, -RpS(0)0Rq-, or -S(0)0Rq-, wherein Rp and Rs can be alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, phosphate, sulfide, sulfonyl, sulfonic acid, sulfonamide and thioketone.
Exemplary sulfinyl groups include, but are not limited to, alkylsulfinyls wherein at least one of Rp or Rq is alkyl, alkenyl or alkynyl.
[059] The term "sulfonamide" as used herein refers to a radical having the structure ¨(Rr)-N-S(0)2-Rs¨ or ¨Rt(Rr)-N-S(0)2-Rs, where Rt, Rr, and Rs can be, for example, hydrogen, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, and heterocyclyl.
Exemplary sulfonamides include alkylsulfonamides (e.g., where Rs is alkyl), arylsulfonamides (e.g., where Rsis aryl), cycloalkyl sulfonamides (e.g., where Rs is cycloalkyl), and heterocyclyl sulfonamides (e.g., where Rs is heterocyclyl), etc.
[060] The term "sulfonate" as used herein refers to the radical -0S03-.
Sulfonate includes salts such as -0S03Na, -0S03K, etc. and the acid -0S03H
[061] The term "sulfonic acid" refers to the radical -S03H-- and its corresponding salts, e.g. -S03K--, -SO3Na--.
[062] The term "sulfonyl" as used herein refers to a radical having the structure RuS02-, where Ru can be alkyl, alkenyl, alkynyl, amino, amide, aryl, cycloalkyl, and heterocyclyl, e.g., alkylsulfonyl. The term "alkylsulfonyl" as used herein refers to an alkyl group attached to a sulfonyl group. "Alkylsulfonyl"
groups can optionally contain alkenyl or alkynyl groups.
[063] The term "thioketone" refers to a radical having the structure -Rv_C(S)-Rw_. The ketone can be attached to another group through Rv or Rw.
Rv or Rw can be alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl or aryl, or Rv or Rw can be joined to form a 3-to 12-membered ring.
[064] "Alkyl," "alkenyl," and "alkynyl" groups, collectively referred to as "saturated and unsaturated hydrocarbons," can be substituted with or interrupted by at least one group selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide, thioketone, and N.
[065] As used herein, a "suitable substituent" refers to a group that does not nullify the synthetic or pharmaceutical utility of the compounds of the invention or the intermediates useful for preparing them. Examples of suitable substituents include, but are not limited to: C1-22, C1-8, and C1-6 alkyl, alkenyl or alkynyl; C1-6 aryl, C2-6 heteroaryl; C3-7 cycloalkyl; C1-22, C1-8, and C1-6 alkoxy; C6 aryloxy; -CN;

-OH; oxo; halo, carboxy; amino, such as -NH(C1-22, C1_8, or C1_6 alkyl), -N((C1-22, C1_8, and C1_6 alky1)2, -NH((C6)ary1), or -N((C6)ary1)2; formyl; ketones, such as -CO(C1_22, C1_8, and C1-6 alkyl), --00((C6 aryl) esters, such as -0O2(C1-22, C1-8, and C1_6 alkyl) and -CO2 (C6 aryl). One of skill in art can readily choose a suitable substituent based on the stability and pharmacological and synthetic activity of the compound of the invention.
[066] The term "pharmaceutically acceptable carrier" as used herein refers to any and all solvents, dispersion media, coatings, isotonic and absorption delaying agents, and the like, that are compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. The compositions may also contain other active compounds providing supplemental, additional, or enhanced therapeutic functions.
[067] The term "pharmaceutically acceptable composition" as used herein refers to a composition comprising at least one compound as disclosed herein formulated together with one or more pharmaceutically acceptable carriers.
[068] The term "pharmaceutically acceptable prodrugs" as used herein represents those prodrugs of the compounds of the present invention that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, commensurate with a reasonable benefit/ risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention. A discussion is provided in Higuchi et al., "Pro-drugs as Novel Delivery Systems," ACS Symposium Series, Vol. 14, and in Roche, ed.

; C:' = Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987.
[069] The term "pharmaceutically acceptable salt(s)" refers to salts of acidic or basic groups that may be present in compounds used in the present compositions. Compounds included in the present compositions that are basic in nature are capable of forming a wide variety of salts with various inorganic and organic acids. The acids=that may be used to prepare pharmaceutically acceptable acid addition salts of such basic compounds are those that form non-toxic acid addition salts, i.e., salts containing pharmacologicaHy acceptabie anions, including but not limited to sulfate, citrate, matate, acetate, oxalate, chloride, bromide, iodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate and.
pamoate (i.e., 1,1'-methylene-bis-(2-hydroxy-3-naphthoate)) salts. Compounds included in the present compositions that include an amino moiety may form pharmaceutically acceptable salts with various amino acids, in addition to the acids mentioned above. Compounds included in the present compositions, that are acidic in nature are capable of forming base salts.with various pharmacologically acceptable cations. Examples of such salts include alkali metal or alkaline earth metal salts and, particularly, calcium, magnesium, sodium, =
lithium, zinc, potassium, and iron salts.
[070] The compounds of the disclosure may contain one or more chiral centers and/or double bonds and, therefore, exist as stereoisomers, such as geometric isomers, enantiomers or diastereomers. The term "stereoisomers"
when used herein consist of all geometric isomers, enantiomers or diastereomers.
These compounds may be designated by the symbols "R" or "S," depending on the configuration of substituents around the stereogenic carbon atom. The present invention encompasses various stereoisomers of these compounds and mixtures thereof. Stereoisomers include enantiomers and diastereomers. Mixtures of enantiomers or diastereomers may be designated "( )" in nomenclature, but the skilled artisan will recognize that a structure may denote a chiral center implicitly.
[071] Individual stereoisomers of compounds of the present invention can be prepared synthetically from commercially available starting materials that contain asymmetric or stereogenic centers, or by preparation of racemic mixtures followed by resolution methods well known to those of ordinary skill in the art.
These methods of resolution are exemplified by (1) attachment of a mixture of enantiomers to a chiral auxiliary, separation of the resulting mixture of diastereomers by recrystallization or chromatography and liberation of the optically pure product from the auxiliary, (2) salt formation employing an optically active resolving agent, or (3) direct separation of the mixture of optical enantiomers on chiral chromatographic columns. Stereoisomeric mixtures can also be resolved into their component stereoisomers by well known methods, such as chiral-phase gas chromatography, chiral-phase high performance liquid chromatography, crystallizing the compound as a chiral salt complex, or crystallizing the compound in a chiral solvent. Stereoisomers can also be obtained from stereomerically-pure intermediates, reagents, and catalysts by well known asymmetric synthetic methods.
[072] Geometric isomers can also exist in the compounds of the present invention. The present invention encompasses the various geometric isomers and mixtures thereof resulting from the arrangement of substituents around a carbon-carbon double bond or arrangement of substituents around a carbocyclic ring. Substituents around a carbon-carbon double bond are designated as being in the "Z' or "E' configuration wherein the terms "Z" and "E' are used in accordance with IUPAC standards. Unless otherwise specified, structures depicting double bonds encompass both the E and Z isomers. For example, the structure below represents a genus of alkenes in which the double bond is either an "E-double bond" or a "Z-double bond."
(R4)p (R6)p (R7)p (R3)p (R8)p (R2)p (R1)p (R10) (R9) [073] Substituents around a carbon-carbon double bond alternatively can be referred to as "cis" or "trans," where "cis" represents substituents on the same side of the double bond and "trans" represents substituents on opposite sides of the double bond. The arrangement of substituents around a carbocyclic ring are designated as "cis" or "trans." The term "cis" represents substituents on the same side of the plane of the ring and the term "trans" represents substituents on opposite sides of the plane of the ring. Mixtures of compounds wherein the substituents are disposed on both the same and opposite sides of plane of the ring are designated "cis/trans."

Embodiments of the Invention [074] Disclosed herein are methods for increasing expression of ApoA-I
in a mammal comprising administering a therapeutically effective amount of a compound of Formula I:
(R4)p (Rop (Rop (R7)p w.w (R3)p-µ X z Y i/W-(R8)p W-W W-W
(R2)p (R1)p (Rio) (R9) Formula I
wherein:
X is selected from C, CRii, CR11R13, CO, CS, 0, S, SO, SO2, N and NR11, wherein R11 may be the same or different than R13;
Y is selected from C, CR12, CR12R14, CO, CS, 0, S, SO, SO2, N and NR12, wherein R12 may be the same or different than R14;
R1, R2, R3, R4, R5, Rs, R7, R8, R9, R10, R11, R12, R13, and R14 are each independently selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone, or two adjacent substituents selected from R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, and R14 are connected in a 5 or 6-membered ring to form a bicyclic aryl or bicyclic heteroaryl;
each W is independently selected from C and N, wherein if W is N, then p is 0 and if W is C, then p is 1;
Z is selected from a single bond, a double bond and a triple bond;

and pharmaceutically acceptable salts and hydrates thereof;
wherein if R3 and R8 are each hydroxyl, then at least one of R11 and R12 is not alkyl;
wherein if Z is a double bond, then R2, R4 and R8 are not each hydroxyl;
and wherein if Formula 1 has the structure:

________________________________ = OH
then at least one of R7 and R9 is not alkyl.
[075] An alternative embodiment provides stilbene compounds of Formula 1:

\ / \ /
w¨w R11 R12 w¨w /I \
R3¨W\\ z ______________ \ ___ N-R8 w¨w R13 R14 W¨W
\ \

or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, and R14 are independently selected from the group consisting of (Ci_C22)alkyl, (C2L22)alkenyl, (C2_C22)alkynyl, aryl, heteroaryl, alkoxy, aryloxy, benzyl, phenyl, carbonyl, thioketone, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, amides, carbamates, halogen, bromide [Br], iodide [I], fluoride [F], chloride [Cl], CF3, CCI3, sulfonic acid [-S03H], phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA glucuronic) acid conjugates], monoesters, dicaboxylic acid, #STR55#, #STR66#, #STR77#, #STR88#, #STR99#, #STR100#, wherein W can be C or N;
wherein when W is an nitrogen atom, the nitrogen atom will only bind to three covalent bonds due to available valence electrons.
[076] The structures below demonstrate a nitrogen arrangement of one embodiment of the compounds of Formula 1:

N_W R11 R12 w-w \

\\ / I I \

R2 R1 R10 R9 wherein the same applies to any W;
or NW R11 R12 w-w I I
R3 -W/ )/ X Y-K \
\ W-R8 / I I \ //

R2 R1 R10 R9 wherein the same applies to any W:
wherein X can be CH, CH2, CR11, CR13, CHR11, CHR13, CR11R13, CO, CS, 0, S, SO, SO2, NH, NR11 with the proviso that X and Y do not exceed the number of valence electrons available as per definitions of X and Y above Y can be CH, CH2, CR12, CR14, CHR12, CHR14, CR11R14, CO, CS, 0, S, SO, SO2, NH, NR11 with the proviso that X and Y do not exceed the number of valence electrons available as per definitions of X and Y above Z can be a single, double bond or triple bond, with the proviso that X and Y
do not exceed the number of valence electrons available as per definitions of X
and Y above;
wherein 1,R15 R 5 1 1H I 2 0=S=0 0=S=0 0=S=0 #STR55# is or =
o0 #STR66# is ...L.-, =

0 R15 HO R15 0,, OH 0 0 0NH2 #STR77# is ; ; ;
R

0,NH

or ;

R15, 0 #STR88# is or =

R15 R15 C) (D
N-H sN-R16 NH NH
#STR99# is=
or #STR100# is II or .
R15 and R16 are substituents independently selected from the group consisting of (C1_C22)alkyl, (C2_C22)alkenyl, (C2_C22)alkynyl, aryl, heteroaryl, alkoxy, aryloxy, benzyl, phenyl, carbonyl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, amides, carbamates, halogen, bromide [Br], iodide [I], fluoride [F], chloride [Cl], CF3, 0013, sulfonic acid (--S03H], phosphate, or a derivative thereof, wherein said derivative is optionally substituted and optionally branched, and may have one or more of the C atoms replaced by S, N or 0;
wherein the compounds of Formula 1 have at least one proviso selected from the following R7 is a hydroxyl;
at least one W is a N;
at least one of R1-R12 is #STR77#, #STR88# or #STR99#;
R8 is #STR66#;
at least one of R1-R12 is #STR66#;
one of R1-R12 is a manoester;
one of R1-R12 is dicarboxylic acid;
one of R1-R12 is succinic acid;
that R8 and R3 are hydroxyls; and that R8 and R3 are #STR55#.
[077] Another embodiment provides for compounds of Formula 1 wherein:
R8 is a hydroxyl and at least one W is a N;
R8 is a hydroxyl and at least one of R1-R7 and R9-R12 is #STR66#;
R8 is a hydroxyl and at least one of R1-R7 and R9-R12 is #STR77#, #STR88# or #STR99#; and R8 is #STR66# and at least one W is a N.
[078] Non-limiting examples of compounds of Formula 1 include:

\ / \
R11 R12 w-1 W=W
/ I 1 ____ \
R3 _W\

\ 1( I ,/,w¨R8 vv vli\ ---__ R13 R14 yv¨

v\
/ , \
(I) R2 R1 R10 R9 R4 R5 R6 R7 =
\ / \ /
W=--W R11 R12 w¨w / I I ____ \
R3 ¨Wµ X=Y W¨R8 \\ //
W¨W W¨W
/ \ / \
(II) R2 R1 R10 R9 \ / \ /
W=W W=W
/\
R3 ¨Wsµ . __ X=Y W¨R8 \\ .
W¨W W¨W
/ \ / \
(III) R2 R1 R10 R9 =
, \ / \ /
W=W R11 R12 w¨w . / \
R3 ¨W\\ R8 /7¨.
r ¨W R13 14 W¨W
/ \ / \
(IV) R2 R1 R10 R9 =

I I I
R4".., .,"=%.,..,-"\.....,../..-'\'` ./W---, R9 W \ W
II I .

R1 R14 R10 =
I
(V) R2 \ / \ / .
W=W W=W
/ . \ .
\
R3 ¨W\ = /W¨R8 W¨W
VV¨V/
/ \ / \
(VI) R2 R1 R10 R9 , w./*R8 I I I I
R4WNõ.,N...,...=-=-=,wW.õ..

I I I I I

I
(VII) R2 . i7 R6õ, ,....W,õ ,R8 w w I I
N R9 ,...Wõ,.

II I
W W
R3 µN
/ === \ o I
(VIII) R2 R7 =

I I I I

R3-- -W -R1 \-.
I
(IX) R2 'I' R6õ, *W..õ .,,R8 I I

=õ..wõ i0õ,...icõ,"k.wWõ, R3* .w'' -.1:21 I
(X) R2 6 R6 _..-W
\ -..-- -=w.---R8 FI W
I I
R4,,, ...õW S..

R12 is.......--,k. ......W,,.
W i I I I

R3...-' \w---- '1,t1 I
(XI) R2 R6õ, ,..;,W,,, R8 ../s...=-="====.R9 W ,=W R14 R10 I
(XII) R2 Fr R6,%._ I W
// II

......w...... ..zz,76,....<<;,%.w..,,W.

W ,,W R14 R10 / \ -..=-= "R1 I
(XIII) R2 R5R6,,.... *W.,.. R8 W W
I I
W 0 ........--=,... .õWõ.

I I I I

R3' W R1 I
(XIV) R2 R6 .--**R8 W

(XV) R2 (XVI) R2 R6*W=%,R8 (XVII) R2 W W

w R14 R12 R10 R3./ v.:1%* s=-= R1 (XVIII) R2 R6\ ..-;:w===.,R8 R3 '====R1 R14 (XIX) R2 R6\ *W=-..R8 (XX) R2 R3 "=-=R1 (XXI) R2 Fr 0 R6 W R8 R3wR1 (XXII) R2 R6,,, W,, ..õR8 I II
R4 W ......õ,õ=<:...,,,. .....m.......

II//

W ,...W
R3./ `=w-. "====R1 R10 I
(XXIII) R2 = R6,.µ *W.. ...,R8 W W
Fr5 S
II
R4 ..........õ..,....... _w.......
1/µ1WN W R9 II I I
vv..,..W.,.., R14 R10 I
(XXIV) R2 R6\w;:=W,. .-,R8 W

I I
W
''''-µi\r"====,.R9 ..õ...w,, R12 R10 R3 W '- R1 I
(XXV) R2 =H =H
S

11101 R6..,. õ::,.. W.,, ..õR8 75 R5 ,OH
W W
I II R4, ,, W
IR4., I I I R3-,.R.1 R3--, ==== ==='R 1 I R10\ \...,.. R6 W
I R2 w õ..W w po." -- `.....' ..., \
...., w R7 OH . I
(XXVI ) R8 or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, and R14 are independently selected from the group consisting of (Ci_C22)alkyl, (C2_C22)alkenyl, (C2_C22)alkynyl, aryl, heteroaryl, alkoxy, aryloxy, benzyl, phenyl, carbonyl, thioketone, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, amides, carbamates, halogen, bromide [Br], iodide [I], fluoride [F], chloride [Cl], CF3, CCI3, sulfonic acid [-S03H], phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA glucuronic) acid conjugates], monoesters, dicaboxylic acid, #STR55#, #STR66#, #STR77#, #STR88#, #STR99#, #STR100#, wherein W can be C or N;
wherein when W is an nitrogen atom, the nitrogen atom will only bind to three covalent bonds due to available valence electrons.
[079] The structures below demonstrate a nitrogen arrangement of one embodiment of the compounds of Formula 1:

/Di N-vv IL W-W
\W -R8 R3 -V% vv/ I I /A/
/\ R13 R14 \

wherein the same applies to any W;
or \ /
N=W R11 R12 w¨w I I
R3-11( >¨R8 W¨W R13 R14 W¨W
\ \

wherein the same applies to any W:
wherein X can be CH, CH2, CR11, CR13, CHR11, CHR13, CR11R13, CO, CS, 0, S, SO, SO2, NH, NR11 with the proviso that X and Y do not exceed the number of valence electrons available as per definitions of X and Y above Y can be CH, CH2, CR12, CR14, CHR12, CHR14, CR11R14, CO, CS, 0, -S, SO, 502, NH, NR11 with the proviso that X and Y do not exceed the number of valence electrons available as per definitions of X and Y above Z can be a single, double bond or triple bond, with the proviso that X and Y
do not exceed the number of valence electrons available as per definitions of X
and Y above wherein 1,R15 0, R 5 2 0=S=0 0=S=0 0=S=0 #STR55# is or =
IN

#5TR66# is =

0R15 H0R15 00H 00 0NH2 0õ1\1H
#si-R77# is or ON...

;

R15, 04o #STR88# is or ;

R15, R15 o 04 N-H sN-R16 NH NH
#STR99# is or =
#STR100# is.
or R15 and R16 are substituents independently selected from the group consisting of (C1_C22)alkyl, (C2_C22)alkenyl, (C2_C22)alkynyl, aryl, heteroaryl, alkoxy, aryloxy, benzyl, phenyl, carbonyl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, amides, carbamates, halogen, bromide [Br], iodide [I], fluoride [F], chloride [Cl], CF3, CCI3, sulfonic acid (--S03H], phosphate, or a derivative thereof, wherein said derivative is optionally substituted and optionally branched, and may have one or more of the C atoms replaced by S, N or 0;
wherein the compounds of Formula 1 have at least one proviso selected from the following R7 is a hydroxyl;
at least one W is a N;
at least one of R1-R12 is #STR77#, #STR88# or #STR9911;
R8 is #STR66#;
at least one of R1-R12 is #STR66#;

one of R1-R12 is a manoester;
one of R1-R12 is dicarboxylic acid;
one of R1-R12 is succinic acid;
that R8 and R3 are hydroxyls; and that R8 and R3 are #STR55#.
[080] Another alternative embodiment provides for compounds of Formula 1 wherein:
R8 is a hydroxyl and at least one W is a N;
R8 is a hydroxyl and at least one of R1-R7 and R9-R12 is #STR66#;
R8 is a hydroxyl and at least one of R1-R7 and R9-R12 is #STR77#, #STR88# or #STR99#; and R8 is #STR66# and at least one W is a N.
[081] Another alternative embodiment provides stilbene compounds of Formula 2:

W W

W x W W R9 or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11 and R12 are independently selected from the group consisting of (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, Bromide [Br], Iodide [I], methoxy [OCH3], ethoxy [OCH2CH3], fluoride [F], chloride [Cl], CF3, CCI3, phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA glucuronic) acid conjugates], #STR66#, #STR100#, #STR77#, #STR99#, #STR88#, wherein W can be C or N;
wherein when W is an nitrogen atom, the nitrogen atom will only bind to three covalent bonds due to available valence electrons.
[082] The structures below demonstrate a nitrogen arrangement of one embodiment of the compounds of Formula 2:
R5 R11R6 W*W R8 X W, R9 *INR12 R10 wherein the same applies to any W; or R11, W
x w .=====

wherein the same applies to any W:
wherein #STR66# is wherein X can be a single, double bond or triple bond, with the proviso that if a triple bond R11 and R12 are not present;
wherein #STR100# means ;
ovo #STR77# means ^"^ ;
HNõr0,R14 #STR88# means , or =
y0(R13 OR13 0 R14 #STR99# means ^-1^ , , or ;
R13 is pyridine, pyridazine, pyrimidine, pyrazine; and R14 is (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl or a derivative thereof, wherein said derivative is optionally substituted and optionally branched, and may have one or more of the C atoms replaced by S, N or 0;
with the proviso that;
a) at least one W is a N and R8 is a hydroxyl; or b) at least one W is a N and R8 is a hydroxyl, and optionally;

c) at least one of R1-12 is #STR77#, or #STR88#, or #STR99#, and/or;
d) at least one of R1-R12 is #STR66#.
[083] Another alternative embodiment provides compounds comprising the general stilbene structure of Formula 2:
R6\ *vv R8 W, X VY IA( R9 R3 \A/- R1 or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11 and R12 are independently selected from the group consisting of (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, Bromide [Br], Iodide [I], methoxy [OCF13], ethoxy [OCH2CH3], fluoride [F], chloride [Cl], CF3, CCI3, phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA glucuronic) acid conjugates], #STR66#, #STR100#, #STR77#, #STR99#, #STR88#, wherein W can be C or N;
wherein when W is an nitrogen atom, the nitrogen atom will only bind to three covalent bonds due to available valence electrons.
[084] The structures below demonstrate a nitrogen arrangement of one embodiment of the compounds of Formula 2:

*VV R8 R4õ, WI X W, R9 wherein the same applies to any W; or wherein the same applies to any W:
wherein #STR66# is wherein X can be a single, double bond or triple bond, with the proviso that if a triple bond R11 and R12 are not present;
wherein #STR100# means ;

#STR77# means y HN 0, R14 #STR88# menas , or =
0yR13 Os.R:13 0yR14 #STR99# means " , , or ;
R13 is pyridine, pyridazine, pyrimidine, pyrazine; and R14 is (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl or a derivative thereof, wherein said derivative is optionally substituted and optionally branched, and may have one or more of the C atoms replaced by S, N or 0;
with the proviso that;
a) at least one of R1-12 is selected from #STR77# or #STR88# or #STR99#, and R8 is a hydroxyl; or b) at least one of R1-12 is selected from #STR77# or #STR88# or #STR99#, and R8 is a hydroxyl, and optionally;
c) at least one W is a N, and/or;
d) at least one of R1-R12 is #STR66#.
[085] Another alternative embodiment provides compounds comprising the general stilbene structure of Formula 2:

R5 R11 W õ,...
= I
X W, R9 or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11 and R12 are independently selected from the group consisting of (Ci_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, Bromide [Br], Iodide [I], methoxy [OCH3], ethoxy [OCH2CH3], fluoride [F], chloride [CI], CF3, CCI3, phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA glucuronic) acid conjugates], #STR66#, #STR100#, #STR77#, #STR99#, #STR88#, wherein W can be C or N;
wherein when W is an nitrogen atom, the nitrogen atom will only bind to three covalent bonds due to available valence electrons.
[086] The structures below demonstrate a nitrogen arrangement of one embodiment of the compounds of Formula 2:

R6 <,,=WR8 wherein the same applies to any W; or R11R6W*W R 8 \ ?vv R9 wherein the same applies to any W:
wherein #STR66# is wherein X can be a single, double bond or triple bond, with the proviso that if a triple bond R11 and R12 are not present;
wherein #STR100# means ;

#STR77# means "I" =
=
HN 0, y R14 #STR88# means 0 , or 0=
0yR14 0yR13 0===R14 #STR99# means , or =
R13 is pyridine, pyridazine, pyrimidine, pyrazine; and R14 is (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl or a derivative thereof, wherein said derivative is optionally substituted and optionally branched, and may have one or more of the C atoms replaced by S, N or 0;
with the proviso that;
a) R8 is a hydroxyl or #STR66# and if R8 is a hydroxyl then at least one of (R1-R7 and R9-R12) is #STR66#; or b) R8 is a hydroxyl or #STR66# and if R8 is a hydroxyl then at least one of (R1-R7 and R9-R12) is #STR66# and optionally;
c) at least one W is a N, and/or;
d) at least one of R1-12 is #STR77#, or #STR88#, or #STR99#.
[087] Another alternative embodiment provides compounds comprising the general stilbenes structure of Formula 2:

R5 R1'16W*vv wI IW
R4 \ w \ R9 or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11 and R12 are independently selected from the group consisting of (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, Bromide [Br], Iodide [I], methoxy [OCH3], ethoxy [OCH2CH3], fluoride [F], chloride [Cl], CF3, CCI3, phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA glucuronic) acid conjugates], #STR66#, #STR100#, #STR77#, #STR99#, #STR88#, wherein W can be C or N;
wherein when W is an nitrogen atom, the nitrogen atom will only bind to three covalent bonds due to available valence electrons.
[088] The structures below demonstrate a nitrogen arrangement of one embodiment of the compounds of Formula 2:

R5 R11W\A/ R8 IW
R4\ \ R9 W, X W
I I

=R3 W" R1 wherein the same applies to any W; or wherein the same applies to any W:
wherein #STR66# is wherein X can be a single, double bond or triple bond, with the proviso that if a triple bond R11 and R12 are not present;
wherein #STR100# means ;
#STR77# means HNõO, - R14 y), #STR88# menas 0 , or 0=

1 0==='R14 #STR99# means ^-4^ , , or ;
R13 is pyridine, pyridazine, pyrimidine, pyrazine; and R14 is (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl or a derivative thereof, wherein said derivative is optionally substituted and optionally branched, and may have one or more of the C atoms replaced by S, N or 0;
with the proviso that:
a) R8 is a hydroxyl or #STR66#, and b) at least one W is a N, and c) at least one of R1-12 is #STR77#, or #STR88#, or #STR99#, and d) at least one of R1-R12 is #STR66#.
[089] Non-limiting examples of compounds of Formula 2 include:

R5 R11 W* \/R8 W W
R4\

(XXVII) R2 R6\w*WR8 R4\
W ==== W
R3w*W \ R1 R10 (XXVIII) R2 i R6\ w R3/-wW R1 R10 (XXIX) R2 R4 R5 Re R7 VV VV
¨ W=W
( __________________________________________ Re VV=W W¨W
(XXX) R2 R1 Rlo R9 R6\ w*W R8 R4N., R9 R3w%W \ R1 R10 (XXXI ) R2 I I I
RLk., R3/-w*W R1 R10 (XXXII) R2 or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11 and R12 are independently selected from the group consisting of (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, Bromide [Br], Iodide [I], methoxy [OCH3), ethoxy [OCH2CH3], fluoride [F], chloride [Cl], CF3, CCI3, phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA glucuronic) acid conjugates], #STR66#, #STR100#, #STR77#, #STR99#, #STR88#, wherein W can be C or N;
wherein when W is an nitrogen atom, the nitrogen atom will only bind to three covalent bonds due to available valence electrons.
[090] The structures below demonstrate a nitrogen arrangement of one embodiment of the compounds of Formula 2:
R6 R8 =
R5 R11 W*w\/
R4"N, R9 WI X W, I *W,Ns R12 R10 R2 wherein the same applies to any W; or R4N., w R9 R2 wherein the same applies to any W:
wherein #STR66# is wherein X can be a single, double bond or triple bond, with the proviso that if a triple bond R11 and R12 are not present;
wherein #STR100# means ;

#STR77# means ;
HN 0, yR14 R14 #STR88# means 0 , or =

#STR99# means ^'4^ , , or R13 is pyridine, pyridazine, pyrimidine, pyrazine; and R14 is (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl or a derivative thereof, wherein said derivative is optionally substituted and optionally branched, and may have one or more of the C atoms replaced by S, N or 0;
wherein the compounds of Formula 2 have one of the following groups of provisos:
1) with the proviso that:
a) R8 is a hydroxyl; or b) R8 is a hydroxyl, and optionally;
c) at least one W is a N, and/or;
d) at least one of R1-12is #STR77#, or #STR88#, or #STR99#, and/or;
e) at least one of R1-R12 is #STR66#.

2) with the proviso that;
a) at least one W is a N and R8 is a hydroxyl; or b) at least one W is a N and R8 is a hydroxyl, and optionally;
c) at least one of R1-12 is #STR77#, or #STR88#, or #STR99#, and/or;
d) at least one of R1-R12 is #STR66#.
3) with the proviso that:
a) at least one of R1-12 is selected from #STR77# or #STR88# or #STR99#, and R8 is a hydroxyl; or b) at least one of R1-12 is selected from #STR77# or #STR88# or #STR99#, and R8 is a hydroxyl, and optionally;
c) at least one W is a N, and/or;
d) at least one of R1-R12 is #STR66#.
4) with the proviso that:
a) R8 is a hydroxyl or #STR66# and if R8 is a hydroxyl at least one of (R1-R7 and R8-R12) is #STR66#; or b) R8 is a hydroxyl or #STR66# and if R8 is a hydroxyl at least one of (R1-R7 and R9-R12) is #STR66# and optionally;
c) at least one W is a N, and/or;
d) at least one of R1-12 is #STR77#, or #STR88#, or #STR99#.
5) with the proviso that;
a) R8 is a hydroxyl or #STR66#, and b) at least one W is a N, and c) at least one of R1-12 is #STR77#, or #STR88#, or #STR99#, and d) at least one of R1-R12 is #STR66#.
[091] Another alternative embodiment provides stilbene compounds of Formula 3:

R6,õ R8 X W, R9 and a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11 and R12 are independently selected from the group consisting of (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, Bromide [Br], Iodide [I], methoxy [OCH3], ethoxy [OCH2CH3], fluoride [F], chloride [Cl], CF3, CCI3, phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA glucuronic) acid conjugates], wherein #STR66# is wherein X can be a single, double bond or triple bond, with the proviso that if a triple bond R11 and R12 are not present;
W can be C or N;

with the proviso that a) R3 and R8 are hydroxyls and at least one W is a N and optionally:
b) at least one of R1-R12 is #STR66#; and/or c) R8 is #STR66#
[092] Another alternative embodiment provides compounds comprising of the general stilbenes structure of Formula 3:

R6 lei R8 R6 Am R8 111. R9 and a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11 and R12 are independently selected from the group consisting of (Ci_C22)alkyl, (C2.C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, Bromide [Br], Iodide [I], methoxy [OCH3], ethoxy [OCH2CH3], fluoride [F], chloride [Cl], CF3, CCI3, phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA glucuronic) acid conjugates], wherein #STR66# is wherein X can be a single, double bond or triple bond, with the proviso that if a triple bond R11 and R12 are not present;
with the proviso that a) R3 and R8 are hydroxyls, and optionally:
b) at least one of R1-R12 is #STR66#: and/or c) R8 is #STR66#.
[093] Non-limiting examples of stilbene compounds of Formula 3 include:

W W
R 4\ ./* .,=====

(XXXIII) R2 R6 wW R8 W
R4\

(XXXIV) R2 R3/-w*W R1 R10 (XXXV) R2 W¨W W=W
R3 ( ________________________________________ Re W=W W¨W
(X000/1) R2 Ri Rio R9 and a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11 and R12 are independently selected from the group consisting of (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, Bromide [Br], Iodide [I], methoxy [OCH3], ethoxy [OCH2CH3], fluoride [F], chloride [Cl], CF3, CCI3, phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA glucuronic) acid conjugates], wherein v.

#STR66# means wherein X can be a single, double bond or triple bond, with the proviso that if a triple bond R11 and R12 are not present;
W can be C or N;
with the 'proviso that a) R3 and R8 are hydroxyls and at least one W is a N and optionally:
b) at least one of R1-R12 is #STR66#; and/or c) R8 is #STR66#
[094] Another alternative embodiment provides compounds comprising of the general stilbene structure of Formula 3:

which can be further subdivided into the following structures: .

R6ei R8 (XXXVI I ) R2 R6 ei R8 (=Nil') R2 (XXXIX) R2 =R60 R8 (XL) R2 (XLI) R2 W¨N
R3 ___________________________________ ) ___ R8 W=W W¨W
\R, (XLII) R2 R Rio (XLIII) R2 R1 Rio R9 and a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11 and R12 are independently selected from the group consisting of (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, Bromide [Br], Iodide [I], methoxy [OCH3], ethoxy [OCH2CI-13], fluoride [F], chloride [CI], CF3, CCI3, phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA glucuronic) acid conjugates], wherein #STR66# means wherein X can be a single, double bond or triple bond, with the proviso that if a triple bond R11 and R12 are not present;
with the proviso that a) R3 and R8 are hydroxyls, and optionally:
b) at least one of R1-R12 is #STR66#; and/or c) R8 is #STR66#.
[095] Disclosed herein are methods for increasing expression of ApoA-I
in a mammal comprising administering a therapeutically effective amount of a compound of Formula IV:
(R7)p (R4)p W W
(I33)P W,WT5)1' X2W W(I'tg)p Z

(Rio)p (ROp W X1 Z1 (FlOp Formula IV
wherein:
X1 is selected from C, CRii, CR11R13, CO, CS, 0, S, SO, SO2, N and NRii, wherein R11 may be the same or different than R13;

X2 is selected from C, CR17, CR17R18, CO, CS, 0, S, SO, SO2, N and NI:Z17, wherein R17 may be the same or different than R18;
Y is selected from C, CR12, CR12R14, CO, CS, S, SO, SO2, N and NR.12, wherein R12 may be the same or different than R14;
R1, R2, R3, 1:24, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, R17 and R18 are each independently selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone, or two adjacent substituents selected from R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, R17 and R18 are connected in a 5 or 6-membered ring to form a bicyclic aryl or bicyclic heteroaryl;
each W is independently selected from C and N, wherein if W is N, then p is 0 and if W is C, then p is 1;
Z1 and Z2 are each independently selected from a single bond, a double bond, and a triple bond;
and pharmaceutically acceptable salts and hydrates thereof.
[096] An alternative embodiment provides chalcone compounds of Formula 4:
R4R6\ ;:-W /R8 I
w ==='. w R13--x"-v R9 I Z

I R13/ \R11 or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, and R14 are independently selected from the group consisting of (Ci_C22)alkyl, (C2.C22)alkenyl, (C1_C22)alkynyl, aryl, heteroaryl, alkoxy, aryloxy, benzyl, phenyl, carbonyl, thioketone, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, amides, carbamates, halogen, bromide [Br], iodide [I], fluoride [F], chloride [Cl], CF3, CCI3, sulfonic acid [-S03H], phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA glucuronic) acid conjugates], monoesters, dicaboxylic acid, #STR55#, #STR66#, #STR77#, #STR88#, #STR99#, #STR100#, wherein W can be C or N;
wherein when W is an nitrogen atom, the nitrogen atom will only bind to three covalent bonds due to available valence electrons.
[097] The structures below demonstrate a nitrogen arrangement of one embodiment of the compounds of Formula 4:

R6\R8 R3\ I
w R13--x R9 I Z
/Y.c-R12 R10 I R131 \R11 wherein the same applies to any W;
or R3 z I
w R13¨x µf%/ R9 I Z
R2 W /X \Z R14 I R13/ \R11 ./Y.c¨R12 R10 wherein the same applies to any W:
wherein X can be CH, CH2, CR11, CR13, CHR11, CHR13, CR11R13, CO, CS, 0, S, SO, SO2, NH, NR11 with the proviso that X and Y do not exceed the number of valence electrons available as per definitions of X and Y above Y can be CH, CH2, CR12, CR14, CHR12, CHR14, CR11R14, CO, CS, 0, S, SO, SO2, NH, NR11 with the proviso that X and Y do not exceed the number of valence electrons available as per definitions of X and Y above Z can be a single, double bond or triple bond, with the proviso that X and Y
do not exceed the number of valence electrons available as per definitions of X
and Y above wherein ,R15 0, ,R15 I I 1 2 S
N S 0=S=0 0 =S=0 0=S=0 #STR55# is or #STR66# is =

H0,...R15 0 #STR77# is ;

or ;

R15, Oo #STR88# is or R15µ11-1-1 N-R16 NH NH
#STR99# is or ;
#STR100# isII .
or R15 and R16 are substituents independently selected from the group consisting of (C1_C22)alkyl, (C2_C22)alkenyl, (C2_C22)alkynyl, aryl, heteroaryl, alkoxy, aryloxy, benzyl, phenyl, carbonyl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, amides, carbamates, halogen, bromide [Br], iodide [I], fluoride [F], chloride [Cl], CF3, CCI3, sulfonic acid [-S03H], phosphate, or a derivative thereof, wherein said derivative is optionally substituted and optionally branched, and may have one or more of the C atoms replaced by S, N or 0;
wherein Formula 4 compounds have at least one proviso selected from the following:
R7 is a hydroxyl;
at least one W is a N;
at least one of R1-R10 is #STR77#, #STR88# or #STR99#;
at least one of R1-R10 is #STR66#;
R7 is #STR66#;

one of R1-R10 is a monoester;
one of R1-R10 is succinic acid;
R7 is #STR55#;
R7 and R2 are #STR55#;
R7 and R2 are hydroxyls; and one of R1-R10 is a dicarboxylic acid.
[098] Another alternative embodiment provides for compounds of Formula IV wherein:
R7 is a hydroxyl and at least one W is a N;
R7 is a hydroxyl and at least one of R1-R6 and R8-R10 is #STR66#, R7 is a hydroxyl and at least one of R1-R6 and R8-R10 is #STR77#, #STR88# or #S1R99#; and R7 is #STR66# and at least one W is a N.
[099] Non-limiting examples of compounds of Formula 4 include the E
and Z isomers of the following structures:
R6\ *W / R8 II
R3wWw/R13¨j(wWR9 \NIT1R12 R10 (XLIV) R13/ x \R11 R6\ -j=

I I
,R5 R3\ -;,.--W\w,R13¨x/'',:' ¨ R9 /W

I
is\ R14 R13' R11 (XLV) R1 W w w=-="" w R13---x R9 R2WWXY\R14 (XLVI) R1 7' Wõ, /R5 1.õ.õ..=
X

/W*

(XLVII) R1 0 R5 R6, ,..WõR8 µ4(/
R12\ 114 I I
(XLVIII) R1 0 0 R10 or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, and R14 are independently selected from the group consisting of (C1_C22)alkyl, (C2_C22)alkenyl, (C1_C22)alkynyl, aryl, heteroaryl, alkoxy, aryloxy, benzyl, phenyl, carbonyl, thioketone, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, amides, carbamates, halogen, bromide [Br], iodide [I], fluoride [F], chloride [Cl], CF3, CCI3, sulfonic acid [-S03H], phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA glucuronic) acid conjugates], monoesters, dicaboxylic acid, #STR55#, #STR66#, #STR77#, #STR88#, #STR99#, #STR100#, wherein W can be C or N;
wherein when W is an nitrogen atom, the nitrogen atom will only bind to three covalent bonds due to available valence electrons. The structures below demonstrate a nitrogen arrangement of one embodiment of the compounds of Formula 4:

I I

wI I Z
:*WIC.ZY'R14R12 R10 I \
R1 R13 ' R11 wherein the same applies to any W;
or w w R13--x R9 I I I Z

I R13/ \R11 wherein the same applies to any W:
wherein X can be CH, CE-12, CR11, CR13, CHR11, CHR13, CR11R13, CO, CS, 0, S, SO, SO2, NH, NR11 with the proviso that X and Y do not exceed the number of valence electrons available as per definitions of X and Y above Y can be CH, CH2, CR12, CR14, CHR12, CHR14, CR11R14, CO, CS, 0, S, SO, SO2, NH, NR11 with the proviso that X and Y do not exceed the number of valence electrons available as per definitions of X and Y above Z can be a single, double bond or triple bond, with the proviso that X and Y
do not exceed the number of valence electrons available as per definitions of X
and Y above wherein ,R15 0, R1 5 I I 2 S
NS
0=S=0 0=S=0 0=S7---0 #STR55# is or =

#STR66# is =

= 0 R15 HO R15 OOH

#STR77# is ;

or ^^^ ;

R15, Oo #STR88# is or "'L =

R15, R15 () O

#STR99# is or ;
#STR100# isII .
or R15 and R16 are substituents independently selected from the group consisting of (C1_C22)alkyl, (C2_C22)alkenyl, (C2_C22)alkynyl, aryl, heteroaryl, alkoxy, aryloxy, benzyl, phenyl, carbonyl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, amides, carbamates, halogen, bromide [Br], iodide [I], fluoride [9, chloride [Cl], CF3, CCI3, sulfonic acid [-S03H], phosphate, or a derivative thereof, wherein said derivative is optionally substituted and optionally branched, and may have one or more of the C atoms replaced by S, N or 0;
wherein Formula 4 compounds have at least one proviso selected from the following:
R7 is a hydroxyl;
at least one W is a N;
at least one of R1-R10 is #STR77#, #STR88# or #STR99#;
at least one of R1-R10 is #STR66#;
R7 is #STR66#;
one of R1-R10 is a monoester;
one of R1-R10 is succinic acid;
R7 is #STR55#;
R7 and R2 are #STR55#;
R7 and R2 are hydroxyls; and one of R1-R10 is a dicarboxylic acid.
[0100] Another alternative embodiment provides for compounds of Formula 4 wherein:
R7 is a hydroxyl and at least one W is a N;
R7 is a hydroxyl and at least one of R1-R6 and R8-R10 is #STR66#;
R7 is a hydroxyl and at least one of R1-R6 and R8-R10 is #STR77#, #STR88# or #STR99#; and R7 is #STR66# and at least one W is a N.
[0101] Another alternative embodiment provides chalcone compounds of Formula 5:

x I

R2 z Y R14 or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R8, R9, and R10 are independently selected from the group consisting of (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, Bromide [Br], Iodide [I], methoxy [OCH3], ethoxy [OCH2CH3], fluoride [F], chloride [Cl], CF3, CCI3, phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA
glucuronic) acid conjugates], #STR66#, #STR77#, #STR88#, #STR99#, #STR100#
wherein W can be C or N;
wherein when W is an nitrogen atom, the nitrogen atom will only bind to three covalent bonds due to available valence electrons.
[0102] The structures below demonstrate a nitrogen arrangement of one embodiment of the compounds of Formula 5:

x I

R2 W z Y R14 R1 wherein the same applies to any W; or R5w x R2 z Y R14 R1 wherein the same applies to any W:
wherein #STR66# is wherein X can be single or double bond;
Y can be single or double bond;
Z can be 0, C, N, S, CR13, or NR13;
wherein R13 is #STR77#, #STR88#, #STR99#, or #STR100#, wherein #STR77# means ;
HNy0,R12 .yo, #STR88# means , or =

Q)...õ,R11 0R11 0,y,R12 #STR99# means , , or #STR100# means ;
R11 is pyridine, pyridazine, pyrimidine, pyrazine; and R12 is (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl or a derivative thereof, wherein said derivative is optionally substituted and optionally branched, and may have one or more of the C atoms replaced by S, N or 0;
with the proviso that;
a) R7 is a hydroxyl; OR
b) R7 is a hydroxyl, and optionally;
c) at least one W is a N, and/or;
d) at least one of R1-10 is #STR77#, or #STR88#, or #STR99#, and/or;
e) at least one of R1-R10 is #STR66#.
[0103] Another alternative embodiment provides compounds comprising the general chalcone structure of Formula 5:

XI

R2 z Y R14 or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R8, R9, and R10 are independently selected from the group consisting of (Ci_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, Bromide [Br], Iodide [I], methoxy [OCH3], ethoxy [OCH2CH3], fluoride [F], chloride [Cl], CF3, CCI3, phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA
glucuronic) acid conjugates], #STR66#, #STR77#, #STR88#, #STR99#, #STR100#
wherein W can be C or N;
wherein when W is an nitrogen atom, the nitrogen atom will only bind to three covalent bonds due to available valence electrons.
[0104] The structures below demonstrate a nitrogen arrangement of one embodiment of the compounds of Formula 5:

x I

R2 z Y R14 R1 wherein the same applies to any W; or x I

R2 z Y R14 R1 wherein the same applies to any W:
wherein #STR66# is wherein X can be single or double bond;
Y can be single or double bond;
Z can be 0, C, N, S, CR13, or NR13;
wherein R13 is #STR77#, #STR88#, #STR99#, or #STR100#, wherein #STR77# means ;
HNy0,R12 #STR88# means , or =
0yR11 0,...):õR: 1 0yR12 #STR99# means "'L , , Or #STR100# means R11 is pyridine, pyridazine, pyrimidine, pyrazine; and R12 is (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl or a derivative thereof, wherein said derivative is optionally substituted and optionally branched, and may have one or more of the C atoms replaced by S, N or 0;

with the proviso that;
a) at least one W is a N and R7 is a hydroxyl; or b) at least one W is a N and R7 is a hydroxyl, and optionally;
C) at least one of R1-10 is #STR77#, or #STR88#, or #STR99#, and/or;
d) at least one of R1-R10 is #STR66#.
[0105] Another alternative embodiment provides compounds comprising of the chalcone structure of Formula 5:
R5.",w*w`===,/ R7 R3, R1 0 x I

R2 \Ar z Y R14 or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R8, R9, and R10 are independently selected from the group consisting of (Ci.C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, Bromide [Br], Iodide methoxy [OCH3], ethoxy [OCH2CH3], fluoride [F], chloride [CI], CF3, CCI3, phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA
glucuronic) acid conjugates], #STR66#, #STR77#, #STR88#, #STR99#, #STR100#
wherein W can be C or N;
wherein when W is an nitrogen atom, the nitrogen atom will only bind to three covalent bonds due to available valence electrons. The structures below demonstrate a nitrogen arrangement of one embodiment of the compounds of Formula 5:
Fi24 x R2 z R14 Si wherein the same applies to any W; or R5 \.w*WR7 x I

R2 z Y R14 R1 wherein the same applies to any W:
wherein #STR66# is wherein X can be single or double bond;
Y can be single or double bond;
Z can be 0, C, N, S, CR13, or NR13;
wherein R13 is #STR77#, #STR88#, #STR99#, or #STR100#, wherein 0,0 #STR77# means ".^^ ;
HN 0, yR12R12 #STR88# means , or =

#STR99# means , , or #STR100# means ;
R11 is pyridine, pyridazine, pyrimidine, pyrazine; and R12 is (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl or a derivative thereof, wherein said derivative is optionally substituted and optionally branched, and may have one or more of the C atoms replaced by S, N or 0;
with the proviso that;
a) at least one of R1-10 is selected from #STR77# or #STR88# or #STR99#, and R7 is a hydroxyl;
b) at least one of R1-10 is selected from #STR77# or #STR88# or #S1R99#, and R7 is a hydroxyl, and optionally;
c) at least one W is a N, and/or;
d) at least one of R1-R10 is #STR66#.
[0106] Another alternative embodiment provides compounds comprising the chalcone structure of Formula 5:

x I

R2 z Y R14 or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R8, R9, and R10 are independently selected from the group consisting of (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, Bromide [Br], Iodide [I], methoxy [OCH3], ethoxy [OCH2CH3], fluoride [F], chloride [Cl], CF3, CCI3, phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA
glucuronic) acid conjugates], #STR66#, #STR77#, #STR88#, #STR99#, #STR100#
wherein W can be C or N;
wherein when W is an nitrogen atom, the nitrogen atom will only bind to three covalent bonds due to available valence electrons.
[0107] The structures below demonstrate a nitrogen arrangement of one embodiment of the compounds of Formula 5:
R5\wi=Ws=-=õ/R7 x R2 z Y R14 R1 wherein the same applies to any W; or R5\wWR7 R3N,w ====,, x I

R2 z Y R14 R1 wherein the same applies to any W:
wherein #STR66# is wherein X can be single or double bond;
Y can be single or double bond;
Z can be 0, C, N, S, CR13, or NR13;
wherein R13 is #STR77#, #STR88#, #STR99#, or #STR100#, wherein 0,0 #STR77# means ;
HNy0,R12 #STR88# means , or =

Oy R11 0R11 0yR12 0R12 ,or #STR99# means , .

#STR100# means ;
R11 is pyridine, pyridazine, pyrimidine, pyrazine; and R12 is (C1_C22)alkyl, (C2_c22)alkenyl, aryl, heteroaryl or a derivative thereof, wherein said derivative is optionally substituted and optionally branched, and may have one or more of the C atoms replaced by S, N or 0;
with the proviso that;
a) R7 is a hydroxyl or #STR66# and if R7 is a hydroxyl at least one of (R1-R6 and R8-R10) is #STR66#; or b) R7 is a hydroxyl or #STR66# and if R7 is a hydroxyl at least one of (R1-R6 and R8-R10) is #STR66# and optionally;
c) at least one W is a N, and/or;
d) at least one of R1-10 is #STR77#, or #STR88#, or #STR99#.
[0108] Another alternative embodiment provides compounds comprising the chalcone structure of Formula 5:
R5\ w*WR7 R3,RiO

x I

R2 \Ar z Y R14 or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R8, R9, and R10 are independently selected from the group consisting of (Ci_C22)alkyl, (C2L22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, Bromide [Br], Iodide [I], methoxy [OCH3], ethoxy [OCH2CH3l, fluoride [F], chloride [Cl], CF3, CCI3, phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA
glucuronic) acid conjugates], #STR66#, #STR77#, #STR88#, #STR99#, #STR100#
wherein W can be C or N;
wherein when W is an nitrogen atom, the nitrogen atom will only bind to three covalent bonds due to available valence electrons. The structures below demonstrate a nitrogen arrangement of one embodiment of the compounds of Formula 5:

x I

R2 z Y R14 R1 wherein the same applies to any W; or R5\.1,11W\R7 x I

R2 z Y R14 R1 wherein the same applies to any W:
wherein #STR66# is wherein X can be single or double bond;
Y can be single or double bond;
Z can be 0, C, N, S, CR13, or NR13;
wherein R13 is #STR77#, #STR88#, #STR99#, or #STR100#, wherein 0,0 #STR77# means ;
HNy0,R12 =y0,R12 #STR88# means , or 0 =
0R11 Of 1 0R12 0R12 #STR99# means ^'L , , or #STR100# means ;
R11 is pyridine, pyridazine, pyrimidine, pyrazine; and R12 is (Ci_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl or a derivative thereof, wherein said derivative is optionally substituted and optionally branched, and may have one or more of the C atoms replaced by S, N or 0;

with the proviso that;
a) R7 is a hydroxyl or #STR66#, and b) at least one W is a N, and C) at least one of R1-10 is #STR77#, or #STR88#, or #STR99#, and d) at least one of R1-R10 is #STR66#.
[0109] Non-limiting examples of chalcone compounds of Formula 5 include:
R5\w*WR7 (XLIX) R1 R15 (L) R1 R15 W

(LI) R1 R15 R5õNw.....W,,.N...R7 (LII) R1 0 R5 w*w R7 W

(LIII) R1 0 I
W

(LIV) R1 0 R5õõ
R3õõ

(LV) R1 0 R3.õõ
W' R8 (LVI) R1 0 It4 R3w*W
I I

=

(LVII) R1 0 (LVIII) R1 0 0 R9 R5õ.
W

w R9 (LIX) R1 \ IN*

(LX) R1 R5w*WR7 Irt (LXI) R1 R15 I I
R3.,... *W,.......,A10 ,,.......-......... ,,W,, I I
-=""====`,,. ..--W..... R9 (LXII) R1 = Fr I I I

W"

õ,õ.."....... R9 I I
(LXIII) R1 R15 .

I
2/\/,, R2/":"...'; .õ. R9 (LXIV) R1 fit4 R3õ, R10 ,,,,...-...., ..W,,.
i I
R2,./'''.=.: ,.,-W., õ...."-\. R9 I //=

(LXV) R1 Fr W
I
R3,,...w*W.,,R10 ..---,,wW,...

I I
,===.:1,.... ..., ,,.W,... N,.,. R9 I //.
o (LXVI) R1 0 R5\w=WR7 (LXVII) R1 R5\ w*WR7 RiO

I

R2 z_Sµ\ R14 I dr \'0 (LXVIII) R1 R5\w*WR7 Fi24 R3õ. *W,R10 (LXIX) R1 or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R4, R5, R6, R8, R9, R10, R14, and R15 are independently selected from the group consisting of (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl [OH], acetyl, hydroxyalkyl, aminoalkyl, Bromide [Br], Iodide [I], methoxy [OCH3], ethoxy [OCH2CH3], fluoride [F], chloride [Cl], CF3, CCI3, phosphate, 0-sulfate [the sulfate conjugate], 0-glucoronidate [the glucoronic (AKA glucuronic) acid conjugates], #STR66#, #STR77#, #STR88#, #STR99#, #STR100#
wherein can be C or N;

wherein when W is an nitrogen atom, the nitrogen atom will only bind to three covalent bonds due to available valence electrons.
[0110] The structures below demonstrate a nitrogen arrangement of one embodiment of the compounds of Formula 5:

R2 wherein the same applies to any W; or x I

R2 z Y R14 R1 wherein the same applies to any W:
wherein #STR66# is wherein X can be single or double bond;
Y can be single or double bond;
Z can be 0, C, N, S, CR13, or NR13;
wherein R13 is #STR77#, #STR88#, #STR99#, or #STR100#, wherein 0,0 #STR77# means HN 0, y R12 #STR88# means , or =
0yR110 R12 0yR12 #STR99# means ^'L , , or #STR100# means ;
R11 is pyridine, pyridazine, pyrimidine, pyrazine; and R12 is (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl or a derivative thereof, wherein said derivative is optionally substituted and optionally branched, and may have one or more of the C atoms replaced by S, N or 0;
wherein the compounds of Formula 5 have at least one of the following groups of provisos:
1) with the proviso that;
a) R7 is a hydroxyl; or b) R7 is a hydroxyl, and optionally;
C) at least one W is a N, and/or;
d) at least one of R1-10 is #STR77#, or #STR88#, or #STR99#, and/or;
e) at least one of R1-R10 is #STR66#.
2) with the proviso that;

a) at least one W is a N and R7 is a hydroxyl; or b) at least one W is a N and R7 is a hydroxyl, and optionally;
c) at least one of R1-10 is #STR77#, or #STR88#, or #STR99#, and/or;
d) at least one of R1-R10 is #STR66#.
3) with the proviso that;
a) at least one of R1-10 is selected from #STR77# or #STR88# or #STR99#, and R7 is a hydroxyl; or b) at least one of R1-10 is selected from #STR77# or #STR88# or #STR99#, and R7 is a hydroxyl, and optionally;
c) at least one W is a N, and(or;
d) at least one of R1-R10 is #STR66#.
4) with the proviso that;
a) R7 is a hydroxyl or #STR66# and if R7 is a hydroxyl at least one of (R1-R6 and R8-R10) is #STR66#; or b) R7 is a hydroxyl or #STR66# and if R7 is a hydroxyl at least one of (R1-R6 and R8-R10) is #STR66# and optionally;
c) at least one W is a N, and/or;
d) at least one of R1-10 is #STR77#, or #STR88#, or #STR99#, and/or;
5) with the proviso that;
a) R7 is a hydroxyl or #STR66#, and b) at least one W is a N, and c) at least one of R1-10 is #STR77#, or #STR88#, or #STR99#, and d) at least one of R1-R10 is #STR66#.
[0111] The following is a list of specific exemplary embodiments encompassed by the invention:
1. A method for increasing expression of ApoA-I in a mammal comprising administering a therapeutically effective amount of a compound of Formula I:
(R4)p (R9)p (R6)p (R7)p / \
W=W W=W
(R3)p-µ __ ) X z Y
W-W W-W
/ \ / \
(R2)p (R1)p (R10) (R9) Formula I
wherein:
X is selected from C, CR11, CR11R13, CO, CS, 0, S, SO, SO2, N and NR11, wherein R11 may be the same or different than R13, Y is selected from C, CR12, CR12R14, CO, CS, 0, S, SO, SO2, N and NR12, wherein R12 may be the same or different than R14;
R1, R2, R3, R4, R5, R6, R7, Rg, Rg, R10, R11, R12, R13, and R14 are each independently selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone, or two adjacent substituents selected from R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, and R14 are connected in a 5 or 6-membered ring to form a bicyclic aryl or bicyclic heteroaryl;

each W is independently selected from C and N, wherein if W is N, then p is 0 and if W is C, then p is 1;
Z is selected from a single bond, a double bond and a triple bond;
and pharmaceutically acceptable salts and hydrates thereof;
wherein if R3 and R8 are each hydroxyl, then at least one of R11 and R12 is not alkyl;
wherein if Z is a double bond, then R2, R4 and R8 are not each hydroxyl;
and wherein if Formula 1 has the structure:

" 9 then at least one of R7 and R9 is not alkyl.
2. The method of embodiment 1, wherein at least one W is N.
3. The method of embodiment 1, wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, trichloromethyl, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
4. The method of embodiment 3, wherein at least one W is N.

5. The method of embodiment 1, wherein at least one of R1, R2, R3, R4, R5, R6, R7, Rg, R9, R10, R11, R12, R13 and R14 is selected from 0-sulfate and glucoronidate.
6. The method of embodiment 5, wherein at least one W is N.
7. The method of embodiment 1, wherein at least one of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13 and R14 is selected from succinate, D-argininate, L-argininate, L-lysinate and D-lysinate.
8. The method of embodiment 7, wherein at least one W is N.
9. The method of embodiment 1, wherein at least one of R1, R2, R3, R4, R5, R6, R7, Rg, R9, R10, R11, R12, R13 and R14 is selected from amide, amino, carbamate, carboxy, ester, ether, formyl, and ketone.
10. The method of embodiment 9, wherein at least one W is N.
11. The method of embodiment 9, wherein at least one of R1, R2, R3, R4, R5, R6, R7, Rg, R9, R10, R11, R12, R13 and R14 is a nicotinate ester.
12. The method of embodiment 11, wherein at least one W is N.
13. The method of embodiment 1, wherein Z is a single bond.
14. The method of embodiment 13 wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, trichloromethyl, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
15. The method of embodiment 13, wherein at least one W is N.
16. The method of embodiment 14, wherein at least one W is N.

17. The method of embodiment 13, wherein Xis CRi Ri 3 and Y is CRi2R14.
18. The method of embodiment 17, wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, trichloromethyl, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
19. The method of embodiment 17, wherein at least one W is N.
20. The method of embodiment 18, wherein at least one W is N.
21. The method of embodiment 13, wherein X is NRii and Y is NR12.
22. The method of embodiment 21, wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, trichloromethyl, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
23. The method of embodiment 21, wherein at least one W is N.
24. The method of embodiment 22, wherein at least one W is N.
25. The method of embodiment 1, wherein Z is a double bond.
26. The method of embodiment 25, wherein the double bond is an E-double bond.

27. The method of embodiment 25, wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, trichloromethyl, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
28. The method of embodiment 25, wherein at least one W is N.
29. The method of embodiment 27, wherein at least one W is N.
30. The method of embodiment 25, wherein X is N and Y is N.
31. The method of embodiment 30, wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, trichloromethyl, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
32. The method of embodiment 30, wherein at least one W is N.
33. the method of embodiment 31, wherein at least one W is N.
34. The method of embodiment 25, wherein X is CRii and Y is CR12.
35. The method of embodiment 34, wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, trichloromethyl, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
36. The method of embodiment 34, wherein at least one W is N.
37. The method of embodiment 35, wherein at least one W is N.
38. The method of embodiment 1, wherein Z is a triple bond.
39. The method of embodiment 38, wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, trichloromethyl, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
40. The method of embodiment 38, wherein at least one W is N.
41. The method of embodiment 39, wherein at least one W is N.
42. The method of embodiment 1, wherein Formula I has the structure:
(R4)p (R6)p (R7)p _N
(R3)p3 X Y
(R8)p (R2)p (R1)p (R10) (R9)p 43. The method of embodiment 42, wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
44. The method of embodiment 42, wherein at least one W is N.
45. The method of embodiment 43, wherein at least one W is N.
46. The method of embodiment 1, wherein Formula I has the structure:
(R4)p (Rop (R7)p (R3)p (ROI) (R2)p (R1)p (R10)p (R9)p 47. The method of embodiment 46, wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
48. The method of embodiment 46, wherein at least one W is N.
49. The method of embodiment 47, wherein at least one W is N.
50. The method of embodiment 1, wherein:
X is selected from CRii, CR11R13, N and NRii, wherein R11 may be the same or different than R13; and Y is selected from CR12, CR12R14, N and NR12, wherein R12 may be the same or different than R14.
51. The method of embodiment 50 wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
52. The method of embodiment 50, wherein at least one W is N.
53. The method of embodiment 51, wherein at least one W is N
54. The method of embodiment 1, wherein X is selected from N and NRii, and Y is selected from N and NR12.
55. The method of embodiment 54 wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
56. The method of embodiment 54, wherein at least one W is N.
57. The method of embodiment 55, wherein at least one W is N.
58. The method of embodiment 1, wherein the therapeutically effective amount of the compound of Formula I is sufficient to establish a concentration ranging from about 0.001 pM to about 100 pM in the mammal.
59. The method of embodiment 2, wherein the concentration ranges from about 1 pM to about 20 pM.
60. The method of embodiment 1, wherein the therapeutically effective amount of the compound of Formula I is administered with a pharmaceutically acceptable carrier in a pharmaceutically acceptable composition.

61. A method for increasing expression of ApoA-I in a mammal comprising administering a therapeutically effective amount of a compound of Formula II:

\ \ /
W =W
W =W ( \

\I21 \Rg Formula II
wherein:
X is selected from CH, CH2, CRii, CR13, CHRii, CHR13, CR11R13, CO, CS, 0, S, SO, SO2, NH and NRii, wherein R11 may be the same or different than R13;
Y is selected from CH, CH2, CHR12, CHR14, CR12, CR14, CR12R14, CO, CS, 0, S, SO, SO2, NH and NR12, wherein R12 may be the same or different than R14;
R1, R2, R3, R4, R5, R6, R7, Rg, Rg, R10, R11, R12, R13,and R14 are each independently selected from (C1_C22)alkyl, (C2_C22)alkenyl, (C2_C22)alkynyl, aryl, heteroaryl, alkoxy, aryloxy, benzyl, phenyl, carbonyl, thioketone, hydrogen, hydroxyl, acetyl, hydroxyalkyl, aminoalkyl, amide, carbamate, halogen, CF3, CCI3, sulfonic acid, phosphate, 0-sulfate, 0-glucoronidate, monoester, dicarboxylic acid, J, K, L, M, P and Q;
each W is independently selected from C and N;
Z is selected from a single bond, a double bond, and a triple bond;

,R15 0=S=0 J is selected from NH

0=S=0 and =

'N

K is =

0R15 Ha,,,R15 0<.._,õ=0H 0 o L is selected from , , , ==,.." 0 NR16 "======'. and ;

M is selected from and ;

NH N'R16 C)NH NH
Pis selected from and ;
Q is selected from II and ;
R15 and R16 are each independently selected from (C1_C22)alkyl, (C2-C22)alkenyl, (C2_C22)alkynyl, aryl, heteroaryl, alkoxy, aryloxy, benzyl, phenyl, carbonyl, hydrogen, hydroxyl, acetyl, hydroxyalkyl, aminoalkyl, amide, carbamate, halogen, CF3, CCI3, sulfonic acid and phosphate;
and pharmaceutically acceptable salts thereof;
subject to at least one proviso selected from:
1) R8 iS hydroxyl;
2) at least one W is N;
3) at least one of R1-R12 is selected from L, M and P;
4) at least one of R1-R12 is selected from K;
5) R8 IS K;

6) one of R1-R12 is a monoester;
7) one of R1-R12 is a dicarboxylic acid;
8) one of R1-R12 is succinic acid;
9) R8 and R3 are each hydroxyl;
10) R8 is selected from J; and 11) R8 and R3 are each selected from J.
62. The method of embodiment 61, wherein if proviso 1 is selected, then at least one of provisos 3-8, 10 and 11 is selected;
if proviso 2 is selected, then at least one of provisos 3-8, 10 and 11 is selected; and if proviso 9 is selected and Z is a double bond, then at least one of R11 and R12 is not alkyl.
63. The method of embodiment 61, wherein at least one proviso is selected from provisos 1, 2, 4, 5, and 9.
64. The method of embodiment 61, wherein R8 is hydroxyl and at least one W is N.
65. The method of embodiment 61, wherein R8 is hydroxyl and at least one of R1-R7 and R9-R12 is K.
66. The method of embodiment 61, wherein R8 is hydroxyl and at least one of R1-R7 and R9-R12 is selected from L, M and P.
67. The method of embodiment 61, wherein R8 is K and at least one W
is N.
68. The method of embodiment 61, wherein Z is a single bond.
69. The method of embodiment 61, wherein Z is a double bond.

70. The method of embodiment 68, wherein X is CR11R13, and Y is CRi2R14, 71. The method of embodiment 69, wherein X is CRii, and Y is CR12.
72. The method of embodiment 68, wherein X is NRii, and Y is NR12.
73. The method of embodiment 68, wherein X is NRii, and Y is CRi2Ria=
74. The method of embodiment 68, wherein X is 0, and Y is CR12R14.
75. The method of embodiment 68, wherein X is S, and Y is CR12R14.
76. The method of embodiment 68, wherein X is SO, and Y is CR12R14.
77. The method of embodiment 68, wherein X is SO2, and Y is CR12R14.
78. The method of embodiment 68, wherein X is 0, and Y is NR12.
79. The method of embodiment 68, wherein X is S, and Y is NR12.
80. The method of embodiment 68, wherein X is SO, and Y is NR12.
81. The method of embodiment 68, wherein X is SO2, and Y is NR12.
82. The method of embodiment 68, wherein X is CO, and Y is CR12R14.
83. The method of embodiment 68, wherein X is CO, and Y is NR12.
84. The method of embodiment 68, wherein Xis CO, and Y is O.
85. The method of embodiment 68, wherein X is CO, and Y is S.
86. =The method of embodiment 68, wherein X is CO, and Y is SO.
87. The method of embodiment 68, wherein X is CO, and Y is SO2.
88. The method of embodiment 68, wherein X is SO, and Y is NR12.
89. The method of embodiment 68, wherein X is SO, and Y is CR12R14.
90. The method of embodiment 61, wherein Formula ll has the structure:

OH OH

\ \
W = W W = W
R3-W\ X ,W-R8 wherein the double bond is selected from an E-double bond and a Z-double bond.
91. The method of embodiment 90, wherein at least one W is N.
92. The method of embodiment 61, wherein Formula II has the structure:

\
N=W W=W
R3_ W\\ W -R8 W - W

93. A method for increasing expression of ApoA-I in a mammal comprising administering a therapeutically effective amount of a compound selected from Formulae IIA and IIB:

\ \
W = W W =W
R3 -W = \W -R
/i 8 W-W W-W

Formula IIA

***-..vli.->w"..vv N._ µ/Nr R9 I I

..%'W* R1 Formula IIB
wherein:
R1, R2, R3, R4, R5, Rs, R7, R8, Rg, and R10 are each independently selected from (Ci_C22)alkyl, (C2_C22)alkenyl, (C2_C22)alkynyl, aryl, heteroaryl, alkoxy, aryloxy, benzyl, phenyl, carbonyl, thioketone, hydrogen, hydroxyl, acetyl, hydroxyalkyl, aminoalkyl, amide, carbamate, halogen, CF3, CCI3, sulfonic acid, phosphate, 0-sulfate, 0-glucoronidate, monoester, dicarboxylic acid, J, K, L, M, P
and Q;
each W is independently selected from C and N;

,R15 (Do S 0=S=0 0=S=0 J is selected from NH

0=S=0 and =
'====

K is HO R15 0,, OH 0 0 L is selected from 0,¨NH2 0 NH 0 N

, and ;

C:0\o M is selected from and N--11 N'R16 NH NH
P is selected from and ;
Q is selected from "L` and W;
R15 and R16 are each independently selected from (C1_C22)alkyl, (C2-C22)alkenyl, (C2_C22)alkynyl, aryl, heteroaryl, alkoxy, aryloxy, benzyl, phenyl, carbonyl, hydrogen, hydroxyl, acetyl, hydroxyalkyl, aminoalkyl, amide, carbamate, halogen, CF3, CCI3, sulfonic acid and phosphate;
and pharmaceutically acceptable salts thereof;
subject to at least one proviso selected from:
1) R8 is hydroxyl;
2) at least one W is N;
3) at least one of R1-R10 is selected from L, M and P;
4) at least one of R1-R10 is selected from K;
5) R8 is K;
6) one of R1-R10 is a monoester;
7) one of R1-R10 is a dicarboxylic acid;
8) one of R1-R10 is succinic acid;
9) R8 and R3 are each hydroxyl;
10) R8 is selected from J; and 11) R8 and R3 are each selected from J.

94. A method for increasing expression of ApoA-I in a mammal comprising administering a therapeutically effective amount of a compound of Formula III:

W=W R11 R12 W = W
//
\W -- lig z W-W

Formula III
wherein:
R1, R2, R3, Ra, R5, R6, R7, Rg, R10, R11, and R12 are each independently selected from (Ci_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl, acetyl, hydroxyalkyl, aminoalkyl, bromide, iodide, methoxy, ethoxy, fluoride, chloride, CF3, CCI3, phosphate, 0-sulfate, 0-glucoronidate, K, D, E, F and G;
Rg is selected from hydroxyl and K;
each W is independently selected from C and N;
Z is selected from a single bond, a double bond, and a triple bond, provided that when Z is a triple bond, R11 and R12 are not present;
wherein:

K is =

D is 2c E is selected from and =
0 Ri6 01R16 0.yRi5 F is selected from , and 01:16 G is =
R16 is selected from pyridine, pyridazine, pyrimidine, and pyrazine;
R16 is selected from (C1_C22)alkyl, (C2_C22)alkenyl, aryl and heteroaryl;
and pharmaceutically acceptable salts thereof;
subject to at least one proviso selected from:
1) at least one W is N;
2) at least one of R1-7 and R9-12 is selected from D, E and F; and 3) at least one of R1-7 and R9-12 is selected from K.
95. The method of embodiment 94, wherein R8 is hydroxyl and at least one of R1_7 and R9-12 is selected from K.
96. The method of embodiment 95, subject to at least one proviso selected from 1) at least one W is N; and 2) at least one of R1_7 and R9-12 is selected from D, E and F.
97. The method of embodiment 94, wherein at least one W is N, at least one of R1-7 and R9_12 is selected from D, E, F, and K.

98. The method of embodiment 94, wherein R8 is hydroxyl and at least one W is N.
99. The method of embodiment 98, wherein at least one of R1-7 and R9_ 12 is selected from D, E, F, and K.
100. The method of embodiment 94, wherein at least one W is N; and if R8 is hydroxyl, then at least one of R1_7 and R9-12 is K.
101. The method of embodiment 94, wherein E is:
,R

102. The method of embodiment 94, wherein E is:
103. The method of embodiment 94, wherein F is selected from:
oyFR15 oRis and .
104. The method of embodiment 94, wherein F is selected from:
oiR16 , and 105. The method of embodiment 94, wherein Z is a single bond.
106. The method of embodiment 94, wherein Z is a double bond.
107. The method of embodiment 94, wherein Z is a triple bond.
108. The method of embodiment 94, wherein Formula Ill has the structure:

\
W -N R11 R12 W = W

- z VV-VV VV-VV

109. A method for increasing expression of ApoA-I in a mammal comprising administering a therapeutically effective amount of a compound selected from Formulae IIIA and IIIB:

ft5 R6\ w*WR8 i 0 1 R15 r 1 R4\ \ R9 R3 \ R1 R3/\wµ/V \ R1 Formula IIIA Formula IIIB
wherein:
R1, R2, R3, R4, R5, R6, R7, Rs, and R10 are each independently selected from (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl, acetyl, hydroxyalkyl, aminoalkyl, bromide, iodide, methoxy, ethoxy, fluoride, chloride, CF3, CCI3, phosphate, 0-sulfate, 0-glucoronidate, K, D, E, F and G;
R8 is selected from hydroxyl and K;
each W is independently selected from C and N;
wherein:

K is =

D is ;
HN
=
E is selected from and oyRi, 0 R15 0R16 F is selected from , and oR16 =
G is =
R15 is selected from pyridine, pyridazine, pyrimidine, and pyrazine;
R16 is selected from (Ci_C22)alkyl, (C2_C22)alkenyl, aryl and heteroaryl;
and pharmaceutically acceptable salts thereof;
subject to at least one proviso selected from:
1) at least one W is N;
2) at least one of R1-7 and R9-12 is selected from D, E and F; and 3) at least one of R1-7 and R9-12 is selected from K.
110. A method for increasing expression of ApoA-I in a mammal comprising administering a therapeutically effective amount of a compound of Formula III:

\ \
W=W R11 R12 W=W
VI --R3 W\ V z R 8 VV VV
R2 Ri R10 R9 Formula III
wherein R1, R2, R4, R5, R6, R7, R9, R10, R11, and R12 are each independently selected from (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl, acetyl, hydroxyalkyl, aminoalkyl, bromide, iodide, methoxy, ethoxy, fluoride, chloride, CF3, CCI3, phosphate, 0-sulfate, and 0-glucoronidate;
R3 and R8 are hydroxyl;
each W is independently selected from C and N; wherein at least one W is N; and Z is selected from a single bond, a double bond, and a triple bond, provided that when Z is a triple bond, R11 and R12 are not present.
111. The method of embodiment 110, wherein Z is a single bond.
112. The method of embodiment 110, wherein Z is a double bond.
113. The method of embodiment 110, wherein Z is a triple bond.
114. The method of embodiment 110, wherein at least one of R1, R2, R4, R5, R6, R7, R9, R10, R11, and R12 is:
IN
115. The method of embodiment 114, wherein Z is a single bond.
116. The method of embodiment 114, wherein Z is a double bond.
117. The method of embodiment 114, wherein Z is a triple bond.
118. A method for increasing expression of ApoA-I in a mammal comprising administering a therapeutically effective amount of a compound of Formula III:

W=W R11 R12 W=W
/
R W¨R
3¨ W% W Z W / 8 W¨W W¨W

Formula Ill wherein:
R1, R2, Ra, R5, R6, R7, R9, R10, R11, and R12 are each independently selected from (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl, acetyl, hydroxyalkyl, aminoalkyl, bromide, iodide, methoxy, ethoxy, fluoride, chloride, CF3, CCI3, phosphate, 0-sulfate, and 0-glucoronidate;
R3 is hydroxyl;

=
R5 is each W is independently selected from C and N, wherein at least one W is N; and Z is selected from a single bond, a double bond, and a triple bond, provided that when Z is a triple bond, R11 and R12 are not present.
119. The method of embodiment 118, wherein Z is a single bond.
120. The method of embodiment 118, wherein Z is a double bond.
121. The method of embodiment 118, wherein Z is a triple bond.
122. The method of embodiment 110, wherein Formula III has the structure:

R3 \

R2 R1 R10 Rg 123. The method of embodiment 122, wherein Z is a single bond.
124. The method of embodiment 122, wherein Z is a double bond.
125. The method of embodiment 122, wherein Z is a triple bond.
126. The method of embodiment 114, wherein Formula Ill has the structure:
= R4 R8 R7 R3 \

R2 R1 R10 Rg 127. The method of embodiment 126, wherein Z is a single bond.
128. The method of embodiment 126, wherein Z is a double bond.
129. The method of embodiment 126, wherein Z is a triple bond.
130. The method of embodiment 118, wherein Formula Ill has the structure:

R3 \
131. The method of embodiment 130, wherein Z is a single bond.
132. The method of embodiment 130, wherein Z is a double bond.
133. The method of embodiment 130, wherein Z is a triple bond.
134. A method of treating or preventing cardiovascular, cholesterol or lipid related disorders comprising administering a therapeutically effective amount of a compound of Formula I:
(R4) (R5)p (R6)p (ROp W=W W
(R3)/3-µ X z Y /V-(R8)p W -W W-W
(R2)p (R1)p (R10) (R9) Formula I
wherein:
X is selected from C, CRii, CR11R13, CO, CS, 0, S, SO, SO2, N and NRii, wherein R11 may be the same or different than R13, Y is selected from C, CR12, CR12R14, CO, CS, 0, S, SO, SO2, N and NR12, wherein R12 may be the same or different than R14;
R1, R2, R3, R4, R5, Rs, R7, R8, R9, R10, R11, R12, R13, and R14 are each independently selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone, or two adjacent substituents selected from R1, R2, R3, R4, R59 R6, R72 R8, R9, R10, R11, R12, R13, and R14 are connected in a 5 or 6-membered ring to form a bicyclic aryl or bicyclic heteroaryl;
each W is independently selected from C and N, wherein if W is N, then p is 0 and if W is C, then p is 1;
Z is selected from a single bond, a double bond and a triple bond;
and pharmaceutically acceptable salts and hydrates thereof;

wherein if R3 and R9 are each hydroxyl, then at least one of R11 and R12 is not alkyl;
wherein if Z is a double bond, then R2 and R4 and R9 are not simultaneously hydroxyl; and wherein if Formula 1 has the structure:

OH

then at least one of R7 and R9 is not alkyl.
135. The method of embodiment 134, wherein the cardiovascular, cholesterol or lipid related disorder is selected from acute coronary syndrome, angina pectoris, arteriosclerosis, atherosclerosis, carotid atherosclerosis, cerebrovascular disease, cerebral infarction, congestive heart failure, congenital heart disease, coronary heart disease, coronary artery disease, coronary plaque stabilization, dyslipidemias, dyslipoproteinemias, endothelium dysfunctions, familial hypercholeasterolemia, familial combined hyperlipidemia, hypertension, hyperlipidemia, intermittent claudication, ischemia, ischemia reperfusion injury, ischemic heart diseases, multi-infarct dementia, myocardial infarction, peripheral vascular disease, restenosis, renal artery atherosclerosis, rheumatic heart disease, stroke, thrombotic disorder, transitory ischemic attacks, and lipoprotein abnormalities associated with Alzheimer's disease, obesity, diabetes mellitus, syndrome X
and impotence.
136. The method of embodiment 135, wherein treating or preventing a cholesterol disorder comprises decreasing blood cholesterol levels.
137. The method of embodiment 135, wherein treating or preventing a cholesterol disorder comprises increasing blood ApoA-I levels.
138. The method of embodiment 135, wherein the therapeutically effective amount of the compound of Formula I is administered with a pharmaceutically acceptable carrier in a pharmaceutically acceptable composition.
139. The method of embodiment 135, wherein the therapeutically effective amount of the compound of Formula I is sufficient to establish a concentration ranging from about 0.001 pM to about 100 pM in the mammal.
140. The method of embodiment 136, wherein the concentration ranges from about 1 pM to about 20 pM.
141. A method for increasing expression of ApoA-I in a mammal comprising administering a therapeutically effective amount of a compound of Formula IV:
(R7)p (R4)p (134 W
(114 1z2 I
(R10)P

(ROp Formula IV
wherein:
X1 is selected from C, CRii, CR11R13, CO, CS, 0, S, SO, SO2, N and NRii, wherein R11 may be the same or different than R13;

X2 is selected from C, CR17, CR17R18, CO, CS, 0, S, SO, SO2, N and NR17, wherein R17 may be the same or different than R18;
Y is selected from C, CR12, CR12R14, CO, CS, S, SO, SO2, N and NR12, wherein R12 may be the same or different than R14;
R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R121 R13, R14, R17 and R18 are each independently selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, = halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone, or two adjacent substituents selected from R1, R2, R3, R4, R5, Rs, R7, R8, Rs, R10, R11, R12, R13, R14, R17 and R18 are connected in a 5 or 6-membered ring to form a bicyclic aryl or bicyclic heteroaryl;
each W is independently selected from C and N, wherein if W is N, then p = is 0 and if W is C, then p is 1;
Z1 and Z2 are each independently selected from a single bond, a double bond, and a triple bond;
and pharmaceutically acceptable salts and hydrates thereof.
142. The method of embodiment 6, wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, trichloromethyl, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
143. The method of embodiment 6, wherein at least one W is N.
144. The method of embodiment 142, wherein at least one W is N.
145. The method of embodiment 6, wherein at least one of R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, R17 and Rig is selected from 0-sulfate and 0-glucoronidate.
146. The method of embodiment 6, wherein at least one of R1, R2, R3, R4, R5, R6, R7, Rg, R9, R10, R11, R12, R13, R14, R17 and Rig is selected from succinate, D-argininate, L-argininate, L-lysinate and D-lysinate.
147. The method of embodiment 6, wherein at least one of R1, R2, R3, R4, R5, R6, R7, Rg, R9, R10, R11, R12, R13, R14, R17 and Rig is selected from amide, amino, carbamate, carboxy, ester, ether, formyl, and ketone.
148. The method of embodiment 147, wherein at least one of R1, R2, R3, R4, R5, R6, R7, Rg, R9, R10, R11, R12, R13, R14 and R17 is a nicotinate ester.
149. The method of embodiment 6, wherein Z1 is a double bond, X is selected from C, CRii and N, and Y is selected from C, CR17, and N.
150. The method of embodiment 149, wherein Rg is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, trichloromethyl, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
151. The method of embodiment 149, wherein at least one W is N.
152. The method of embodiment 150, wherein at least one W is N.
153. The method of embodiment 6, wherein Z2 is a double bond, X is selected from C, CRii and N, and Y is selected from C, CR17, and N.
154. The method of embodiment 153, wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, trichloromethyl, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
155. The method of embodiment 153, wherein at least one W is N.
156. The method of embodiment 154, wherein at least one W is N.
157. The method of embodiment 153, wherein Z1 is a single bond and X1 is CO.
158. The method of embodiment 157, wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, trichloromethyl, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
159. The method of embodiment 157, wherein at least one W is N.
160. The method of embodiment 158, wherein at least one W is N.
161. The method of embodiment 157, wherein Y is CR14.
162. The method of embodiment 161, wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, trichloromethyl, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
163. The method of embodiment 161, wherein at least one W is N.
164. The method of embodiment 162, wherein at least one W is N.
165. The method of embodiment 149, wherein Z2 is a double bond.
166. The method of embodiment 165, wherein at least one W is N.
167. The method of embodiment 6, wherein Formula IV has the structure:
(R7)p (R8)p (R8)p (R4)P
(R3)P
W W X2 (ROp (Rio)p (R2)p X1 Zi (ROp 168. The method of embodiment 167, wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, trichloromethyl, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
169. The method of embodiment 167, wherein at least one W is N.
170. The method of embodiment 168, wherein at least one W is N.
171. The method of embodiment 167, wherein Z1 is a single bond, Z2 is a double bond, and X1 is CO.
172. The method of embodiment 171, wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, trichloromethyl, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
173. The method of embodiment 171, wherein at least one W is N.
174. The method of embodiment 172, wherein at least one W is N.
175. The method of embodiment 171, wherein Y is CR14.
176. The method of embodiment 175, wherein R8 is hydroxyl, and R7 and R9 are independently selected from arylalkyl, carboxy, cyano, cycloalkyl, ester, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, hydroxyalkyl, hydroxyaryl, ketone, perfluoroalkyl, perfluorocyclolkyl, perfluoralkoxy, trichloromethyl, 0-sulfate, and 0-glucoronidate, subject to the proviso that R7 and R9 are not both simultaneously hydrogen.
177. The method of embodiment 175, wherein at least one W is N.
178. The method of embodiment 176, wherein at least one W is N.
179. The method of embodiment 6, wherein the therapeutically effective amount of the compound of Formula I is sufficient to establish a concentration ranging from about 0.001 pM to about 100 pM in the mammal.
180. The method of embodiment 180, wherein the concentration ranges from about 1 pM to about 20 pM.
181. The method of embodiment 6, wherein the therapeutically effective amount of the compound of Formula IV is administered with a pharmaceutically acceptable carrier in a pharmaceutically acceptable composition.
182. A method for increasing expression of ApoA-I in a mammal comprising administering a therapeutically effective amount of a compound of Formula IV:

\

I Z

Formula V
wherein:
X is selected from CH, CH2, CR11, CR13, CHRii, CHR13, CR11R13, CO, CS, 0, S, SO, SO2, NH and NR-H, wherein R11 may be the same or different than R13, Y is selected from CH, CH2, CR12, CR14, CHR12, CHR14, CR12R14, CO, CS, 0, S, SO, SO2, NH and NR12, wherein R12 may be the same or different than R14;
R1, R2, R3, R4, R5, R6, R7, Rg, Rs, R10, R11, R12, R13,and R14 are each independently selected from (Ci_C22)alkyl, (C2.C22)alkenyl, (C1_C22)alkynyl, aryl, heteroaryl, alkoxy, aryloxy, benzyl, phenyl, carbonyl, thioketone, hydrogen, hydroxyl, acetyl, hydroxyalkyl, aminoalkyl, amide, carbamate, halogen, CF3, 00I3, sulfonic acid, phosphate, 0-sulfate, 0-glucoronidate, monoester, dicarboxylic acid, J, K, L, M, P and Q;
each W is independently selected from C and N;
Z is selected from a single bond, a double bond, and a triple bond;

s'R15 0-.. R15 I I
0=S=0 0=S=0 J is selected from ..1, ..1, .-J--and NH

0=S=0 ..,-....L. =
I
N
I
-z----......,,. =,' K is ....-.. ;

=:,,,,,.,,, 15 FIORi5 0....<_õ,OH 0 o Lis selected from ........-...... , ........--.. ---------.
..-...,.
, I I
0 NH2 0 NH C) ,N, ..."...--...--. , õ,.......,s,", and w ;

"0 (:)\

M is selected from and --/-- ;
_./
Ri5 \ R1, R15 0.--R15 N--1-1 N- I:) "R16 C).------\NH NH
P is selected from ..--...L .--,...),,, .../... and .....4,-. =
, , , o s ..---....,..
Q is selected from 1 and Ri6 and R16 are each independently selected from (Ci_C22)alkyl, (C2-C22)alkenyl, (C1_C22)alkynyl, aryl, heteroaryl, alkoxy, aryloxy, benzyl, phenyl, carbonyl, hydrogen, hydroxyl, acetyl, hydroxyalkyl, aminoalkyl, amide, carbamate, halogen, CF3, CCI3, sulfonic acid and phosphate;
and pharmaceutically acceptable salts thereof;
subject to at least one proviso selected from:
a. R8 is hydroxyl;
b. at least one W is N;
c. at least one of R1-R12 is selected from L, M and P;
d. at least one of R1-R12 is selected from K;
e. R8 is K;
f. one of R1-R12 is a monoester;
g. one of R1-R12 is a dicarboxylic acid;
h. one of R1-R12 is succinic acid;
i. R8 and R3 are each hydroxyl;
j. R8 is selected from J; and k. R8 and R3 are each selected from J.
183. The method of embodiment 182, wherein the proviso is selected from provisos 1, 2,4, 5, and 9.
184. The method of embodiment 182, wherein R8 is hydroxyl and at least one W is N.
185. The method of embodiment 182, wherein R8 is hydroxyl and at least one of R1-R7 and R9-R12 is K.
186. The method of embodiment 182, wherein R8 is hydroxyl and at least one of R1-R7 and R9-R12 is selected from L, M and P.
187. The method of embodiment 182, wherein Z is a single bond.
188. The method of embodiment 182, wherein Formula V is selected from Formulae VA-VE:

R6 w R8 Re W
III I II
R3 \ w "I w /R5 21V R3 X W R9 \ w,/135 xw/W
I l II I I II I Rg R
wx R10 W)IY Rig I

Formula VA Formula VB
ir III i4 R3W\ jWWR5 X\ / w/W W Re II I I I
R2.õ- W Ra w RI' IT
w.,....... ,..--....I .)( Ri 0 X
R2 W Rg I I I
Ri R i 0 0 Rio Formula VC Formula VD

R6 \ A \ R6 W W
II
R3,..% ..." ,.., /Rs .......=,..% ....,,W%, II Z I
,/,'.=====\I . /...Y R10 I
Ri .
Formula VE
189. A method for increasing expression of ApoA-I in a mammal comprising administering a therapeutically effective amount of a compound of Formula VI:

Re\ RE, I II Rg vv ,, R10 R2 W X z1 R14 Formula VI
wherein:
R1, R2, R3, Ra, R5, R6, R7, R9, R10, and R14 are each independently selected from (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl, acetyl, hydroxyalkyl, aminoalkyl, bromide, iodide, methoxy, ethoxy, fluoride, chloride, CF3, CCI3, phosphate, 0-sulfate, 0-glucoronidate, K, D, E, F and G;
R8 is selected from hydroxyl or K;
each W is independently selected from C and N;
X is selected from 0, C, N, S, CR13 and NR13;
Z1 and Z2 are each independently is selected from a single bond and a double bond;
R13 is selected from D, E, F and G;
wherein:
o?-,o K is =

D is ;

HN)(R Ri5 015' E is selected from and O R
y 15 0 Ri5 0yrµ16 F is selected from , and is =

=
G is R15 is selected from pyridine, pyridazine, pyrimidine, and pyrazine;
R16 is selected from (C1_C22)alkyl, (C2_C22)alkenyl, aryl and heteroaryl;
and pharmaceutically acceptable salts thereof;
subject to at least one proviso selected from:
1) at least one W is N;
2) at least one of R1_7 and R9-12 is selected from D, E and F; and 3) at least one of R1-7 and R9-12 is selected from K.
190. The method of embodiment 189, wherein R8 is hydroxyl and at least one of R1_7 and R9_10 is selected from K.
191. The method of embodiment 190, subject to at least one proviso selected from:
1) at least one W is N; and 2) at least one of R1_7 and R9_10 is selected from D, E and F.
192. The method of embodiment 189, wherein at least one W is N, at least one of R16 and R8_10 is selected from D, E and F, and at least one of R1.7 and R9-10 is selected from K.
193. The method of embodiment 189, wherein X is 0, and Z1 and Z2 are each single bonds.
194. The method of embodiment 189, wherein Xis 0, Z1 is a single bond and Z2 is a double bond.
195. The method of embodiment 189, wherein Xis N, Z1 is a double bond and Z2 is a single bond.
196. The method of embodiment 189, wherein X is S, and Z1 and Z2 are each single bonds.
197. The method of embodiment 189, wherein X is S, Z1 is a double bond and Z2 is a single bond.
198. The method of embodiment 189, wherein Formula VI is selected from Formulae VIA to VIJ :

Re_ µ/V R6 W R8 R = w y )W W

Ii , ..õW.y N...
/..%

Rio R( "W X Zi R14 R2 W R14 R10 Ri R1 R17 Formula VIA Formula VIB

I I
R6W 1 , W.õõ..r....... R8 . I II 74 II
R3 õ Wõ., _....... R5 ..,74.õ.\.õ IN ....,W.
R3, ..,,W,õ..........õ R5 ........jk., = W \AP./ II
R9 W..- R9 T. 1 1 ,,,1õ.... ......W R10 )% /W \r Rio R2 W ' R14 R2 W R14 I I
Ri R17 R1 R17 =
Formula VIC Formula VID

I

I R6: Wy R8 R6, ......Wõ.........õ, R9 ....w.....;. W ''.. II R4 I II I
R3, ....,W.......y...õ R5 ..........L., .....W R3,.w WR5 ..........14,,.: .....Wõ...
W '... II W Rg W R9 Il ........Lõ ....,,W It10 ,/L,N. ,.. IW I it i R2 W -Ir.-- R14 R2 W Y'..'.....R14 I I
Ri 0 R1 0 Formula VIE
1.....w,.... ....,..........., Rlo ...)......Form_ula VIF .

I I
R6tr.r....W.õõ...r.e.,.. R8 R6 Wõ.....e....., R8 .

õ

W
I II I R5 .......1., I I
R3, R3 W.õ../ `..õ ......W,,, .

II

I II
,...L ,...W.õ,r,,.... R10 I I I

Formula VIG
w.............. .....w....... if....õ,..6.... W Y R98 Formula VIH
...*WRI12710 . R4 ' I R5 R3, .....,,Wõ / .... ==-=,... ..,,Wõ....
IT = I w R
I
R2 W N R14 .
I I
R1 R17 .
Formula VIJ
199. A method for increasing expression of ApoA-I in a mammal comprising administering a therapeutically effective amount of a compound selected from Formulae VIK-VIR:

I I

... ...::...=
R4 ...w.f.* y I I
R3, ...,Wsõ...r....., R6 R31 R8 (L.. ....w......n.
w- 1 w -R9 W W 1,9 I I
)IV
N \ R10 .........L .....W R10 W y R2 W y R14 R2 N
I I
Ri 0 R1 0 Formula VIK Formula VIL

R6, ......W..õµ.....õ. R8 I
R4 W 1 1 R6, ..,VV.,. R8 ... ...õ,W
R3 0 `,..., ...õW.,...

W R
w....'W '.... R9 1 wi 1 ...õ.L. .õI vvI
Rio- µl R10 R2 W R14 R2 W' Y
I I

Formula VIM Formula VIN
R7 R7 , I I
R6... ..õ,W.........".... R8 R6 õ ..."...._ ,... R8 II 74 1 i R3 õ ,...,W, R5 .......,..k...... ....,W,.... R3 W,,,..., R5 .../ -''''''. IN /V1L\ R9 II I II I
,,.w.õ.s.,......õ. Rio ...4,1k, WõN.,,.. R10 R2 W IR\ 14 R2 W
k R14 I , \ 0 I i Formula VIO Formula VIP

R6 W Ra R6 W R8 Ii R3, R5R5 W w I I
R10 ,.N 3 R10 R2W R= 14 R2 W
I % I A
Formula VIQ Formula VIR
wherein:
R1, R2, R3, R4, R5, R6, R7, R9, R10, R14, and R17 are each independently selected from (C1_C22)alkyl, (C2_C22)alkenyl, aryl, heteroaryl, hydrogen, hydroxyl, acetyl, hydroxyalkyl, aminoalkyl, bromide, iodide, methoxy, ethoxy, fluoride, chloride, CF3, CCI3, phosphate, 0-sulfate, 0-glucoronidate, K, D, E, F and G;
R7 is selected from hydroxyl or K;
each W is independently selected from C and N;
X is selected from 0, C, N, S, CR13 and NR13;
R13 is selected from D, E, F and G;
wherein:

K is =
0,0 D is ;

HN0.R15 E is selected from and O R y 15 R15 OyR16 F is selected from , and oiRis G is "I" ;
R15 is selected from pyridine, pyridazine, pyrimidine, and pyrazine;
R16 is selected from (C1_C22)alkyl, (C2_C22)alkenyl, aryl and heteroaryl;
and pharmaceutically acceptable salts thereof;
subject to at least one proviso selected from:
1) at least one W is N;
2) at least one of R1_7 and R9_12 is selected from D, E and F; and 3) at least one of R1_7 and R9_12 is selected from K.
200. A method of treating or preventing cardiovascular, cholesterol or lipid related disorders comprising administering a therapeutically effective amount of a compound of Formula IV:
(R7)p (Re)p (R4)p X2 (144 10)P
(ROI,W X1 Z1 (R1)p Formula IV
wherein:

X1 is selected from C, CR11, CR11R13, CO, CS, 0, S, SO, SO2, N and NRii, wherein R11 may be the same or different than R13;
X2 is selected from C, CR17, CR17R18, CO, CS, 0, S, SO, SO2, N and NR17, wherein R17 may be the same or different than R18;
Y is selected from C, CR12, CR12R14, CO, CS, S, SO, SO2, N and NR12, wherein R12 may be the same or different than R14;
R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, R17 and R18 are each independently selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone, or two adjacent substituents selected from R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, R17 and R18 are connected in a 5 or 6-membered ring to form a bicyclic aryl or bicyclic heteroaryl;
each W is independently selected from C and N, wherein if W is N, then p is 0 and if W is C, then p is 1;
Z1 and Z2 are each independently selected from a single bond, a double bond, and a triple bond;
and pharmaceutically acceptable salts and hydrates thereof.
201. The method of embodiment 200, wherein the cardiovascular, cholesterol or lipid related disorder is selected from acute coronary syndrome, angina pectoris, arteriosclerosis, atherosclerosis, carotid atherosclerosis, cerebrovascular disease, cerebral infarction, congestive heart failure, congenital heart disease, coronary heart disease, coronary artery disease, coronary plaque stabilization, dyslipidemias, dyslipoproteinemias, endothelium dysfunctions, familial hypercholeasterolemia, familial combined hyperlipidemia, hypertension, hyperlipidemia, intermittent claudication, ischemia, ischemia reperfusion injury, ischemic heart diseases, multi-infarct dementia, myocardial infarction, peripheral vascular disease, restenosis, renal artery atherosclerosis, rheumatic heart disease, stroke, thrombotic disorder, transitory ischemic attacks, and lipoprotein abnormalities associated with Alzheimer's disease, obesity, diabetes mellitus, syndrome X
and impotence.
202. The method of embodiment 200, wherein treating or preventing a cholesterol disorder comprises decreasing blood cholesterol levels.
203. The method of embodiment 200, wherein treating a or preventing cholesterol disorder comprises increasing blood ApoA-I levels.
204. The method of embodiment 200, wherein the therapeutically effective amount of the compound of Formula IV is administered with a pharmaceutically acceptable carrier in a pharmaceutically acceptable composition.
205. The method of embodiment 200, wherein the therapeutically effective amount of the compound of Formula I is sufficient to establish a concentration ranging from about 0.001 pM to about 100 pM in the mammal.
206. The method of embodiment 205, wherein the concentration ranges from about 1 pM to about 20 pM.
Pharmaceutical Formulations and Methods of Treatment [0112] The present disclosure also provides pharmaceutical compositions comprising compounds as disclosed herein formulated together with one or more pharmaceutically acceptable carriers. These formulations include those suitable for oral, rectal, topical, buccal and parenteral (e.g. subcutaneous, intramuscular, intradermal, or intravenous) administration, although the most suitable form of administration in any given case will depend on the degree and severity of the condition being treated and on the nature of the particular compound being used.
[0113] Formulations suitable for oral administration may be presented in discrete units, such as capsules, cachets, lozenges, or tablets, each containing a predetermined amount of the compound as powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water or water-in-oil emulsion. As indicated, such formulations may be prepared by any suitable method of pharmacy which includes the step of bringing into association the active compound and the carrier or excipient (which may constitute one or more accessory ingredients). The carrier must be acceptable in the sense of being compatible with the other ingredients of the formulation and must not be deleterious to the recipient. The carrier may be a solid or a liquid, or both, and may be formulated with the compound as a unit-dose formulation, for example, a tablet, which may contain from 0.05% to 95% by weight of the active compound.
Other pharmacologically active substances may also be present including other compounds. The formulations of the invention may be prepared by any of the well known techniques of pharmacy consisting essentially of admixing the components.
[0114] For solid compositions, conventional nontoxic solid carriers include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talc, cellulose, glucose, sucrose, magnesium carbonate, and the like. Liquid pharmacologically administrable compositions can, for example, be prepared by dissolving, dispersing, etc., an active compound as =

described herein and optional pharmaceutical adjuvants in an excipient, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to thereby form a solution or suspension. In general, suitable formulations may be prepared by uniformly and intimately admixing the active compound with a liquid or finely divided solid carrier, or both, and then, if necessary, shaping the product.
For example, a tablet may be prepared by compressing or molding a powder or granules of the compound, optionally with one or more assessory ingredients.
Compressed tablets may be prepared by compressing, in a suitable machine, the compound in a free-flowing form, such as a powder or granules optionally mixed with a binder, lubricant, inert diluent and/or surface active/dispersing agent(s).
Molded tablets may be made by molding, in a suitable machine, the powdered compound moistened with an inert liquid diluent.
[0115] Formulations suitable for buccal (sub-lingual) administration include lozenges comprising a compound in a flavored base, usually sucrose and acacia or tragacanth, and pastilles comprising the compound in an inert base such as gelatin and glycerin or sucrose and acacia.
[0116] Formulations of the present invention suitable for parenteral administration comprise sterile aqueous preparations of the compounds, which are approximately isotonic with the blood of the intended recipient. These preparations are administered intravenously, although administration may also be effected by means of subcutaneous, intramuscular, or intradermal injection. Such preparations may conveniently be prepared by admixing the compound with water and rendering the resulting solution sterile and isotonic with the blood. Injectable compositions according to the invention may contain from 0.1 to 5% w/w of the active compound.

[0117] Formulations suitable for rectal administration are presented as unit-dose suppositories. These may be prepared by admixing the compound with one or more conventional solid carriers, for example, cocoa butter, and then shaping the resulting mixture.
[0118] Formulations suitable for topical application to the skin may take the form of an ointment, cream, lotion, paste, gel, spray, aerosol, or oil.
Carriers and excipients which may be used include Vaseline, lanoline, polyethylene glycols, alcohols, and combinations of two or more thereof. The active compound is generally present at a concentration of from about 0.1% to about 15% w/w of the composition, for example, from about 0.5 to about 2%.
[0119] The amount of active compound administered may be dependent on the subject being treated, the subject's weight, the manner of administration and the judgment of the prescribing physician. For example, a dosing schedule may involve the daily or semi-daily administration of the encapsulated compound at a perceived dosage of about 1 pg to about 1000 mg. In another embodiment, intermittent administration, such as on a monthly or yearly basis, of a dose of the encapsulated compound may be employed.Encapsulation facilitates access to the site of action and allows the administration of the active ingredients simultaneously, in theory producing a synergistic effect. In accordance with standard dosing regimens, physicians will readily determine optimum dosages and will be able to readily modify administration to achieve such dosages.
[0120] A therapeutically effective amount of a compound or composition disclosed herein can be measured by the therapeutic effectiveness of the compound. Compounds of the invention may be administered in a dose of about 1 pg/kg to about 200 mg/kg daily; such as from about 1 pg/kg to about 150 mg/kg, from about 1 mg/kg to about 200 mg/kg, from about 1 pg/kg to about 100 mg/kg, from about 1 pg/kg to about 1 mg/kg, from about 50 pg/kg to about 200 mg/kg, from about 10 pg/kg to about 1 mg/kg, from about 10 pg/kg to about 100 pg/kg, from about 100 pg to about 10 mg/kg, and from about 500 pg/kg to about 50 mg/kg.
The dosages, however, may be varied depending upon the requirements of the patient, the severity of the condition being treated, and the compound being used.
In one embodiment, the therapeutically effective amount of a disclosed compound is sufficient to establish a maximal plasma concentration ranging from about 0.001 pM to about 100 pM , e.g., from about 1 pM to about 20 pM. Preliminary doses as, for example, determined according to animal tests, and the scaling of dosages for human administration is performed according to art-accepted practices.
[0121] Toxicity and therapeutic efficacy can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (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 LD50/ED50. Compositions that exhibit large therapeutic indices are preferable.
[0122] The therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the therapeutic which achieves a half-maximal inhibition of symptoms) as determined in cell culture assays or animal models. Levels in plasma may be measured, for example, by high performance liquid chromatography. The effects of any particular dosage can be monitored by a suitable bioassay. Examples of dosages are: about 0.1 X IC50, about 0.5 x IC50, about 1 x IC50, about 5 x IC50, 10 x IC50, about 50x IC50, and about 100 x IC50.
[0123] Data obtained from the cell culture assays or animal studies can be used in formulating a range of dosage for use in humans. Therapeutically effective dosages achieved in one animal model may be converted for use in another animal, including humans, using conversion factors known in the art (see, e.g., Freireich et al., Cancer Chemother. Reports 50(4):219-244 (1966) and Table 1 for equivalent Surface Area Dosage Factors).
Table 1 To: Mouse Rat Monkey Dog Human From. (20 g) (150 g) (3.5 kg) (8 kg) (60 kg) Mouse 1 1/2 1/4 1/6 1/12 Rat 2 1 1/2 1/4 1/7 Monkey 4 2 1 3/5 1/3 Dog 6 4 3/5 1 1/2 Human 12 7 3 2 1 [0124] The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. Generally, a therapeutically effective amount may vary with the subject's age, condition, and sex, as well as the severity of the medical condition in the subject. The dosage may be determined by a physician and adjusted, as necessary, to suit observed effects of the treatment.
[0125] In one embodiment, a compound as disclosed herein, or a pharmaceutically acceptable salt or hydrate thereof, is administered in combination with another therapeutic agent. The other therapeutic agent can provide additive or synergistic value relative to the administration of a stilbene or chalcone compound alone. The therapeutic agent can be, for example, a statin; a PPAR agonist, e.g., a thiazolidinedione or fibrate; a bile-acid-binding-resin; a niacin; a RXR
agonist; an anti-obesity drug; a hormone; a tyrophostine; a sulfonylurea-based drug; a biguanide; an alpha-glucosidase inhibitor; apolipoprotein E; a cardiovascular drug;
an HDL-raising drug; an HDL enhancer; or a regulator of the apolipoprotein A-IV
and/or apolipoprotein genes.
[0126] In one embodiment, a method of treating or preventing cardiovascular, cholesterol or lipid related disorders comprises administering a therapeutically effective amount of a disclosed compound. The disclosed compound may be administered as a pharmaceutically acceptable composition, comprising a disclosed compound and a pharmaceutically acceptable carrier. Another embodiment provides methods for the prevention of a cardiovascular, cholesterol or lipid related disorder, comprising administering to a mammal a therapeutically effective amount of a presently disclosed compound or composition.
[0127] Exemplary cardiovascular, cholesterol or lipid related disorders include, but are not limited to.acute coronary syndrome, angina pectoris, arteriosclerosis, atherosclerosis, carotid atherosclerosis, cerebrovascular disease, cerebral infarction, congestive heart failure, congenital heart disease, coronary heart disease, coronary artery disease, coronary plaque stabilization, dyslipidemias, dyslipoproteinemias, endothelium dysfunctions, familial hypercholeasterolemia, familial combined hyperlipidemia, hypertension, hyperlipidemia, intermittent claudication, ischemia, ischemia reperfusion injury, ischemic heart diseases, multi-infarct dementia, myocardial infarction, peripheral vascular disease, restenosis, renal artery atherosclerosis, rheumatic heart disease, stroke, thrombotic disorder, transitory ischemic attacks, and lipoprotein abnormalities associated with Alzheimer's disease, obesity, diabetes mellitus, syndrome X and impotence.
[0128] As used herein, the term "cardiovascular disease" refers to diseases and disorders of the heart and circulatory system. These diseases are often associated with dyslipoproteinemias and/or dyslipidemias. "Endothelium dysfunction(s)" include, but are not limited to, dysfunctions affecting blood vessel elasticity; peripheral vascular disease; coronary heart disease; myocardial infarcation; cerebral infarction and restenosis. "Syndrome X" or "Metabolic Syndrome(s)" include, but are not limited to hypertension and dyslipidemia/dyslipoproteinemia.
[0129] In one embodiment, "treatment" or "treating" refers to an amelioration of a disease or disorder, or at least one discernible symptom thereof. In another embodiment, "treatment" or "treating" refers to an amelioration of at least one measurable physical parameter, not necessarily discernible by the patient. In yet another embodiment, "treatment" or "treating" refers to inhibiting the progression of a disease or disorder, either physically, e.g., stabilization of a discernible symptom, physiologically, e.g., stabilization of a physical parameter, or both. In yet another embodiment, "treatment" or "treating" refers to delaying the onset of a disease or disorder. For example, treating a cholesterol disorder may comprise decreasing blood cholesterol levels.
[0130] One embodiment provides a compound for administration to a patient, such as a human, as a preventative measure against cardiovascular, cholesterol or lipid related disorder. As used herein, "prevention" or "preventing" refers to a reduction of the risk of acquiring a given disease or disorder. In another embodiment, the present compositions are administered as a preventative measure to a patient, such as a human having a genetic predisposition to, for example, a cardiovascular disease, a dyslipidemia, a dyslipoproteinemia, Alzheimer's Disease, hypertension, atherosclerosis, or inflammation.
[0131] In another embodiment, the compositions of the invention are administered as a preventative measure to a patient having a non-genetic predisposition to, for example, cardiovascular disease, a dyslipidemia, a dyslipoproteinemia, Alzheimer's Disease, hypertension, atherosclerosis, or inflammation. Examples of such non-genetic predispositions include, but are not limited to, cardiac bypass surgery and percutaneous transluminal coronary angioplasty, which often leads to restenosis, an accelerated form of atherosclerosis; diabetes in women, which often leads to polycystic ovarian disease; and cardiovascular disease, which often leads to impotence.
Accordingly, the present compositions may be used for the prevention of one disease or disorder and concurrently treating another (e.g., prevention of polycystic ovarian disease while treating diabetes; prevention of impotence while treating a cardiovascular disease).
[0132] Angioplasty and open heart surgery, such as coronary bypass surgery, may be required to treat cardivascular diseases, such as atherosclerosis.
= These surgical procedures entail using invasive surgical devices and/or implants, and are associated with a high risk of restenosis and thrombosis. Accordingly, the compounds of the invention may be used as coatings on surgical devices (e.g., catheters) and implants (e.g., stents) to reduce the risk of restenosis and thrombosis associated with invasive procedures used in the treatment of cardiovascular diseases.
[0133] The present invention provides methods for the treatment or prevention of a dyslipidemia comprising administering to a patient a therapeutically effective amount of a presently disclosed compound. In one embodiment, the compound is administered as a pharmaceutically acceptable composition. As used herein, the term "dyslipidemias" refers to disorders that lead to or are manifested by aberrant levels of circulating lipids. To the extent that levels of lipids in the blood are too high, the disclosed compounds or compositions may be administered to a patient to restore normal levels. Normal levels of lipids are reported in medical treatises known to those of skill in the art.
[0134] Dyslipidemias which the disclosed compounds or compositions are useful for preventing or treating include but are not limited to hyperlipidemia and low blood levels of high density lipoprotein (HDL) cholesterol. Hyperlipidemia includes, but is not limited to, familial hypercholesterolemia; familial combined hyperlipidemia; reduced or deficient lipoprotein lipase levels or activity, including reductions or deficiencies resulting from lipoprotein lipase mutations;
hypertriglyceridemia; hypercholesterolemia; high blood levels of ketone bodies (e.g., beta-OH butyric acid); high blood levels of Lp(a) cholesterol; high blood levels of low density lipoprotein (LDL) cholesterol; high blood levels of very low density lipoprotein (VLDL) cholesterol and high blood levels of non-esterified fatty acids.
[0135] One embodiment provides methods for altering lipid metabolism in a patient, e.g., increasing the ratio of HDL to LDL in the blood of a patient, comprising administering to the patient a composition of the invention in an amount effective alter lipid metabolism.
[0136] Another embodiment provides methods for the treatment or prevention of a dyslipoproteinemia comprising administering to a patient a therapeutically effective amount of a disclosed compound or composition. As used herein, the term "dyslipoproteinemias" refers to disorders that lead to or are manifested by aberrant levels of circulating lipoproteins. To the extent that levels of lipoproteins in the blood are too high, the disclosed compounds or compositions are administered to a patient to restore normal levels. Conversely, to the extent that levels of lipoproteins in the blood are too low, the disclosed compounds or compositions are administered to a patient to restore normal levels. Normal levels of lipoproteins are reported in medical treatises known to those of skill in the art.
= [0137] Dyslipoproteinemias which the disclosed compounds or compositions are useful for preventing or treating include, but are not limited to, high blood levels of LDL; high blood levels of apolipoprotein B (apo B); high blood levels of Lp(a); high blood levels of apo(a); high blood levels of VLDL; low blood levels of HDL; reduced or deficient lipoprotein lipase levels or activity, including reductions or deficiencies resulting from lipoprotein lipase mutations; hypoalphalipo-proteinemia; lipoprotein abnormalities associated with diabetes mellitus;
lipoprotein abnormalities associated with obesity; lipoprotein abnormalities associated with Alzheimer's Disease; and familial combined hyperlipidemia.
[0138] One embodiment provides methods for elevating the levels of HDL
associated proteins, such as ApoA-I, in the blood of a mammal, comprising administering to the mammal a composition comprising a disclosed compound or composition in an amount effective to elevate levels of HDL associated proteins in the mammal.
[0139] Another embodiment provides methods for the treatment or prevention of Alzheimer's Disease, hypertension, atherosclerosis, comprising administering to a mammal a therapeutically effective amount of a disclosed compound or composition. As used herein, "treatment or prevention of Alzheimer's Disease" encompasses treatment or prevention of lipoprotein abnormalities associated with Alzheimer's Disease.
[0140] "Diseases and conditions associated with diabetes mellitus" as defined herein comprise, but are not restricted to, hyperglycemia, hyperinsulinaemia, hyperlipidaemia, insulin resistance, impaired glucose metabolism, obesity, diabetic retinopathy, macular degeneration, cataracts, diabetic nephropathy, glomerulosclerosis, diabetic neuropathy, erectile dysfunction, premenstrual syndrome, vascular restenosis, ulcerative colitis, skin and connective tissue disorders, foot ulcerations, metabolic acidosis, arthritis, osteoporosis and impaired glucose tolerance.
PREPARATION OF COMPOUNDS
[0141] Formula A represents a general formula for stilbenes and their analogs:
(Rdniv=IN W=W (Ra)n b\/\/
\/\/ __________________________ X z __ ( Y
W¨W W¨W
A
[0142] Ra may be selected from groups including, but not limited to, alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone. One of ordinary skill will appreciate that stilbene compounds and analogs thereof can be prepared according to the procedures outlined in the following schemes.
Stilbenes Comprising a -C=C- Linker 7.1¨(Ro)n [0143] In one embodiment, the synthesis of stilbenes of Formula A can be achieved by following the procedure of Scheme 1:
(Ron xw w , - w = w ,IN, II I
w.-..--... + Wittig ,w.........................õ.........õ,õ.1k,......,_ ....õ,w I II Reaction, 1 (Rahl 1 i W..wV,,,W w=:.'N,s, ../w (Ra)ni 1 X = Hal 2 3 Scheme 1 [0144] Arylalkyl halide .I can be combined with aldehyde 2 in the presence of, for example, triphenylphosphine and Na0Me, afford alkene 3. Other methods of achieving the desired coupling include, for example, a Peterson olefination.
[0145] Stilbenes of Formula A may also be prepared via the procedure of Scheme 2:
(Ra)n W
W W
II i ¨11¨(Rdri _________________________________ i w W'W + WW

Heck ,wõ......r.......õ....õ),_ I II Reaction (Ra)n¨h wyz.W....,w Br vli,. õ-vv -W
(Ra)n Scheme 2 [0146] Scheme 2 illustrates arylation of styrene 1 using a Heck reaction.
Bromobenzene 4 can be treated with a Pd catalyst, such as a palladium-triarylphosphine couple, to give stilbene 3. However, other methods known in the art that may be used include: 1) treatment of an aryl iodide with palladium acetate in the presence of a base (e.g., tributylamine, potassium acetate); or 2) reaction of an aromatic compound with palladium acetate or palladium metal and silver acetate in acetic acid.
[0147] The resuting alkene of the stilbenes of Schemes 1 and 2 can be reduced using a range of known reduction methods in the art to yield a -CH2-linker. Such methods include, but are not limited to, catalytic hydrogenation using catalysts such as Pt, Pd, or Ni, optionally in the presence of a solid support (e.g., A1203, BaSO4), in the presence of H2.
Stilbene Analogs Comprising a -C1-12-NH- Linker [0148] Formula B depicts stilbene analogs comprising a -CH2-NH- linker:
-=====
N
(Rdn I I
[0149] Stilbene analogs of Formula B may be prepared following the general procedure of Scheme 3:
(Rdn WW Reducing W"\/NWW
I 11/ Agent (Ra)n--1-1 I
W/w/W ,e Ce (Ra)11 Scheme 3 [0150] Stilbene analog 6 in Scheme 3 may be prepared using known reductive amination techniques. A Borch reaction uses NaBH3CN as a reducing agent to afford amine 6. Other reductants such as NaBH4may also be used.

Alternatively, the amine 5 can be reacted with benzaldehyde 2, followed by subjection to catalytic hydrogenation conditions (such as H2/Pd).
Stilbene Analogs Comprising a -CH2-0- Linker [0151] Formula C represents a general formula for stilbene analogs comprising a -CH2-0- linker: a)n C)WW
(Ra)n I I
[0152] Stilbenes analogs comprising an aryl ether may be synthesized following the procedure of Scheme 4:
w41 - w w II
w,..w Base L.%***w I II
(RAH¨ I
Viz,/
PVV
n(Ra) Br Scheme 4 [0153] Formic acid phenyl ester 7 may be synthesized from the corresponding benzaldehyde via traditional oxidation methods known in the art.

For example, Baeyer-Villiger oxidation using m-CPBA to form 7, followed by deformylation using a suitable base and exposure to bromide 8 affords stilbene analog 9.
Stilbenes Comprising a -(C0)-NH- Linker [0154] Formula D represents a general formula for stilbene analogs comprising a -(C0)-NH- linker:

0 ww (Nahl (Ra)n ¨f¨ I
W
[0155] Stilbene analogs of Formula D can be prepared following the procedure of Scheme 5:
r (Ra)n II I .1WWSW 0 W W 41I¨(Ra)n W\..s.,.,%W Coupling I ill (RA + Reagent 31' W
W)/W NH2 Oa) Scheme 5 [0156] The formation of stilbene derivative 12 in Scheme 5 may use an acyl halide 10 (Rb = Hal) or carboxylic acid 10 (Rb = OH). An acyl halide, optionally in the presence of a suitable base, can react with amine 11 to afford amide 12. Amide 12 can also be prepared from the carboxylic acid 10 using known peptide coupling reagents, such as EDCI.
Chalcones [0157] Formula E represents an exemplary ch:a: Icone compound:
\Ai Rc .4%
(Ra)n Rf (Ra)n ww Rd [0158] Chalcones of Formula E may be synthesized following the procedure of Scheme 6:

Rc w w n , II (main W - W
W
-F-(Rõ)õ (R Beta aL I _______________________ =
Elimination (RALH-Scheme 5 [0159] Base-catalyzed (3-elimination of flavone 13 to prepare chalcone 14 can be accomplished with a strong base such as NaOH and reflux. Other conditions include heating a reaction mixture containing flavone 14 with a metal-hydroxide or metal-alkyl species.
EXAMPLES
[0160] Examples of stilbene compounds of Formula I include, but are not limited to, the following compounds.
4,4'-dihydroxy-stilbene HO
6-[(E)-2-(4-Hydroxy-phenyl)-vinyl]-pyridin-3-ol OH
HO
5-[(E)-2-(4-Hydroxy-phenyl)-vinyl]-pyrazin-2-ol N \

HO N
5-[(E)-2-(4-Hydroxy-phenyl)-vinyl]-pyridin-2-ol \

/
HO
Nicotinic acid 4-[(E)-2-(4-hydroxy-phenyl)-vinyl]-phenyl ester N
oir0 I.
HO *
5-{[1-(4-Hydroxy-phenyl)-meth-(E)-ylideneFamino}-pyridin-2-ol N

HO N
5-(4-Hydroxy-phenylazo)-pyridin-2-ol NN
1 ' HON
[0161] Examples of Chalcone compounds of Formula 4 include but are not limited to:
(E)-3-(4-Hydroxy-phenyl)-1-phenyl-propenone OH

and its derivatives, including but not limited to:
(E)-3-(4-Hydroxy-phenyl)-1-pyridin-3-yl-propenone (E)-3-(4-Hydroxy-phenyI)-1-pyridin-4-yl-propenone (E)-3-(4-Hydroxy-phenyl)-1-pyridin-2-yl-propenone (E)-3-(5-Hydroxy-pyridin-2-yI)-1-phenyl-propenone (E)-3-(6-Hydroxy-pyridin-3-y1)-1-phenyl-propenone Nicotinic acid 4-((E)-3-oxo-3-phenyl-propenyI)-phenyl ester Nicotinic acid 4-((E)-3-oxo-3-pyridin-3-yl-propenyI)-phenyl ester Nicotinic acid 4-((E)-3-oxo-3-pyridin-4-yl-propenyI)-phenyl ester Nicotinic acid 4-((E)-3-oxo-3-pyridin-2-yl-propenyI)-phenyl ester Nicotinic acid 64(E)-3-oxo-3-phenyl-propeny1)-pyridin-3-y1 ester Nicotinic acid 54(E)-3-oxo-3-phenyl-propeny1)-pyridin-2-y1 ester [5-Hydroxy-24(E)-3-oxo-3-phenyl-propeny1)-phenylFcarbamic acid ethyl ester [5-Hydroxy-24(E)-3-oxo-3-pyridin-3-yl-propeny1)-phenylFcarbamic acid ethyl ester [5-Hydroxy-2-((E)-3-oxo-3-pyridin-4-yl-propenyI)-pheny1]-carbamic acid ethyl ester [5-Hydroxy-24(E)-3-oxo-3-pyridin-2-yl-propeny1)-phenyl]-carbamic acid ethyl ester [5-Hydroxy-2-((E)-3-oxo-3-phenyl-propeny1)-pyridin-3-y1]-carbamic acid ethyl ester [2-Hydroxy-5-((E)-3-oxo-3-phenyl-propeny1)-pyridin-4-y1]-carbamic acid ethyl ester Nicotinic acid 3-ethoxycarbonylamino-4-((E)-3-oxo-3-phenyl-propenyI)-phenyl ester Nicotinic acid 3-ethoxycarbonyla m ino-4-((E)-3-oxo-3-pyridin-3-yl-propeny1)-phenyl ester Nicotinic acid 3-ethoxycarbonylamino-4-((E)-3-oxo-3-pyridin-4-yl-propeny1)-phenyl ester Nicotinic acid 3-ethoxycarbonylamino-4-((E)-3-oxo-3-pyridin-2-yl-propenyI)-phenyl ester Nicotinic acid 5-ethoxycarbonylamino-6-((E)-3-oxo-3-phenyl-propenyI)-pyridin-3-y1 ester Nicotinic acid 4-ethoxycarbonylamino-54(E)-3-oxo-3-phenyl-propeny1)-pyridin-2-y1 ester.
[0162] The following compounds were obtained from commercially available sources (such as Indofine Chemical Company, Inc.): 4,4'-dihydroxystilbene; 4-hydroxychalcone; 4-methoxychalcone; 4,2',4'-trihydroxychalcone; 2,4-dihydroxy-4'-methoxychalcone; and 4,2',4',6'-tetramethoxychalcone.
[0163] Abbreviations used herein denote the following compounds, reagents and substituents: N-methylpyrrolidinone (NMP); dichloromethane (DCM);

dimethylaminopyridine (DMAP); dimethylformamide (DMF); dimethylsulfoxide (DMS0); ethanol (Et0H); ethyl acetate (Et0Ac); 1-ethy1-3-(3-dimethyaminopropyl)carbodiimide (EDCI); lithium aluminum hydride (LAH);
methanol (Me0H); methoxymethyl (MOM); and tetrahydrofuran (THF).
Example 1 OH
HO
OH
5[(E)-2-(4-Hydroxyphenv1)-vinvIlbenzene 1,3-diol (Resveratrol) [0164] To a solution of 4-methoxy-benzyltriphenylphosphonium bromide (12.5 g) in anhydrous tetrahydrofuran (200 mL) at -78 C was added n-butyllithium (2.44 M, 1.0 equiv), and the resulting red solution was stirred under argon for 2-4 h. A solution of 3,5-dimethoxybenzaldehyde (4.5 g) in tetrahydrofuran was added dropwise over 30 min and the mixture stirred for 6-15 h. The resulting cream suspension was poured into water and extracted with dichloromethane. The organic phase was washed with water, and removal of the solvent in vacuo afforded an oil. The oil was separated by flash column chromatography (49:1 hexane/ethyl acetate). The cis-stilbene eluted first as a clear oil followed by the trans isomer as a colorless solid or oil. Overall yield: 91%. To a solution of the trans isomer (3.1.g) in anhydrous dichloromethane (150 mL) at -78 C was added (dropwise) boron tribromide (1.0 M, 34.5 mL), and the resulting red solution was stirred under argon for 30 min. The solution was poured into water and extracted with dichloromethane. The organic phase was washed with water, and removal of the solvent in vacuo afforded an oil, which was separated by flash column chromatography (1:1 hexane/ethyl acetate) to afford a colorless solid (2.26 g, 86%): mp 260 C.

Example 2 OH
HO
5-RE)-2-(4-HydroxyphenvI)-vinyll-pvridin-2-ol [0165] (4-Methoxybenzyl)triphenylphosphonium chloride (4.79 g, 11.4 mmol) was suspended in 50 mL of dry methanol. Sodium methoxide (4.94 g of 25% solution in methanol, 22.8 mmol) was added in one portion to the above suspension and the resulting mixture was stirred at room temperature under nitrogen for 30 minutes. Pyridinecarbaldehyde (1.57 g, 11.4 mmol) was then added and the stirring was continued at room temperature overnight. Excess of sodium methoxide was quenched by addition of water and pH of the reaction mixture was adjusted to pH=4.5 with 1 N HCI, and then to pH=7.5-8.0 by addition of saturated sodium bicarbonate. Products were then extracted with ethyl acetate (500 mL). Combined extracts were washed with water and brine, and concentrated in vacuo. The crude product was purified by column chromatography using ethyl acetate in hexane (5% - 10%) to give 1.27 g (46%) of trans-stilbene.
[0166] The trans-stilbene (1.17 g, 4.85 mmol) was dissolved in dry NMP
(12 mL). Thiophenol (1.07 g, 9.7 mmol) was then added to the above solution, followed by addition of K2003 (67 mg, 0.48 mmol). The resulting mixture was heated for 8 hours at 195 C under nitrogen. The cooled reaction mixture was made alkaline with 1 N NaOH (pH=10), and the neutral components were extracted with ether (3 x 50 mL). The aqueous layer was acidified to pH=4 with N HCI and stirred at room temperature for 30 minutes, and extracted with ether (2 x 50 mL). Then pH was adjusted to pH=8 with saturated NaHCO3. The solid was isolated by filtration, washed with water (100 mL), ethyl acetate (50 mL) and ether (30 mL), and air-dried to yield 5-[(E)-2-(4-hydroxyphenyI)-vinyl]-pyridin-2-ol (715 mg, 69%). MS (ES) in/z: 214.08 (M+1); 13C-NMR (DMSO-d6): 8 162.42, 157.50, 138.21,134.07, 129.04, 127.92, 125.67, 121.59, 121.03, 116.89, and 116.18.
Example 3 ell OH
OMe HO
4-[(E)-2-(4-1-lvdroxvphenv1)-vinv11-3-methoxv-phenol [0167] In a 50 mL round-bottomed flask fitted with condenser and magnetic stirrer were placed 4-bromo-3-methoxyphenol (2.0 g, 9.85 mmol), Ac20 (1.206 g, 11.82 mmol), Et3N (1.196 g, 11.82 mmol) and catalytic amount of DMAP

and the reaction mixture was stirred for 24 h at room temperature. The reaction mixture was poured into water and extracted with ethyl acetate. The organic layer washed with water, dried and concentrated to give 4-acetoxy-3-methoxy-bromobenzene (2.4 g, 99%).
[0168] To a stirred solution of 4-acetoxy-3-methoxy-bromobenzene (3.549 g, 14.48 mmol) in acetonitrile (60 mL), were added 4-acetoxystyrene (2.47 g, 15.21 mmol), N,N-diisopropylethylamine (5.61 g, 43.45 mmol), bipheny1-2-yl-di-t-butyl phosphine (259 mg, 0.87 mmol) and palladium acetate (195 mg, 0.87 mmol) under N2. The reaction mixture was heated to 80 C for 48 h and then cooled to room temperature. Water was added and the mixture was extracted with ethyl acetate. The organic layer was separated, washed with water, brine, dried and concentrated to give crude product, which was purified by column chromatography, using 10 A ethyl acetate in hexane to give (220 mg) of diacetoxystillbene.
[0169] To a solution of diacetoxystillbene (220 mg, 0.675 mmol) in methanol: THF (10 mL: 4 mL), K2CO3 (466 mg, 3.37 mmol) was added and stirred for 25 minutes at room temperature. Then the reaction mixture was neutralized by dilute HCI and extracted by ethyl acetate, the combined organic layer was washed with water, brine, dried and concentrated to give a crude product, which was purified by column chromatography using 50% ethyl acetate in hexane, to give 140 mg (85%) of 4-[(E)-2-(4-hydroxypheny1)-vinyl]-3-methoxy-phenol. MS (ES) m/z: 243.11 (M+1), 242.07(M), 215.05, 193.08, and 182.99; 13C-NMR (CD30D): 5 158.07, 158.02, 156.50, 130.43, 127.11, 126.57, 125.62, 120.40, 118.47, 115.18, 107.33, 98.58, and 54.64; Mp. 192-194 C.
Example 4 OH OH
HO
3-HydroxymethvI-4-1(E)-2-(4-hydroxy-phenyl)-vinv11-phenol [0170] To a stirred solution of methyl-2-bromo-5-methoxy benzoate (4.0 g, 16.32 mmol) in acetonitrile (80 mL), were added 4-methoxystyrene (2.3 g, 17.14 mmol), N,N-diisopropylethylamine (6.33 g, 48.98 mmol), bipheny1-2-yl-di-t-butyl phosphine (292 mg, 0.98 mmol) and palladium acetate (220 mg, 0.98 mmol) under N2. The reaction mixture was heated to 80 C for 48 h and then cooled to rt.
Water was added and the mixture was extracted with ethyl acetate. The organic layer was separated, washed with water, brine, dried and concentrated to give the crude product, which was purified by column chromatography, using 15% ethyl acetate in hexane to give (1.5 g, 30%) of dimethoxy-stillbene ester intermediate.
[0171] To a suspension of LAH (106 mg, 2.82 mmol) in THE (20 mL), the dimethoxy-stillbene ester intermediate (700 mg, 2.34 mmol) in THE (10 mL) was added slowly at 0 C and resulting mixture was stirred at 0 C for 2 h. The reaction was quenched by adding diluted NaOH at 0 C then extracted with ethyl acetate.

The combined organic layers were washed with water, brine, dried and concentrated to give the crude product, which was purified by column chromatography using 30% ethyl acetate in hexane, to give 572 mg (90%) of the dimethoxystilbene.
[0172] To a solution of the dimethoxystillbene (572 mg, 2.12 mmol) in NMP (10 mL), thiophenol (513 mg, 4.66 mmol) and K2CO3 (29 mg, 0.212 mmol) was added and resulting mixture was heated at 190 C for 7 h. Then the reaction mixture was diluted with water, made alkaline by using 1N NaOH and extracted with ethyl acetate. The combined organic layers were washed with water, brine, dried and concentrated to give a crude product, which was purified by column chromatography using 5% methanol in ethyl acetate, to give 120 mg (23%) of 3-hydroxymethy1-4-[(E)-2-(4-hydroxy-pheny1)-vinyl]-phenol. MS (ES) rn/z: 241.15 (M), and 141.12; 13C-NMR (CD30D): 8 156.85, 156.66, 139.46, 129.84, 128.03, 127.82, 127.45, 126.46, 122.19, 115.21, 114.81, 114.51, and 62.09; Mp. 223-225 C.

Example 5 el OH

HO
HO
2-Hydroxv-5-f(E)-2-(4-hvdroxvphenyl)vinyllbenzoic acid [0173] In a 50 mL round-bottomed flask fitted with condenser and magnetic stirrer were placed methyl 5-bromosalicylate (5.0 g, 21.64 mmol), Ac20 (2.65 g, 25.97 mmol), Et3N (2.63 g, 25.97 mmol) and catalytic amount of DMAP.
The reaction mixture was stirred for 24 h at room temperature. The reaction mixture was poured into water and extracted with ethyl acetate. The organic layer was washed with water, dried and concentrated to give methyl 2-acetoxy-5-bromobenzoate (5.9 g, 98%). To a stirred solution of methyl 2-acetoxy-5-bromobenzoate (5.9 g, 21.61 mmol) in acetonitrile (80 mL) were added 4-acetoxystyrene (3.68 g, 22.69 mmol), N,N-diisopropylethylamine (8.38 g, 64.82 mmol), biphenyl-2-yl-di-t-butyl phosphine (368 mg, 1.29 mmol) and palladium acetate (291 mg, 1.29 mmol) under N2. The reaction mixture was heated to 80 C

for 48 h and then cooled to room temperature. Water was added and the mixture was extracted with ethyl acetate. The organic layer was separated, washed with water, brine, dried and concentrated to give crude product, which was purified by column chromatography, using 20% ethyl acetate in hexane to give (3.19 g, 42%) of the di-acetoxy stillbene. To a solution of the di-acetoxy stillbene (500 mg, 1.41 mmol) in acetonitrile: THF (10 mL: 10 mL), NaOH (388 mg, 8.47 mmol) was added and stirred for 24 h at room temperature. The reaction mixture was neutralized by dilute HCI and extracted with ethyl acetate. The combined organic layers were washed with water, brine, dried and concentrated to give a crude product, which was purified by crystallization in ethyl acetate and hexane to give 2-hydroxy-5-[(E)-2-(4-hydroxyphenyl)vinyl]benzoic acid (250 mg, 69%). MS (ES) m/z: 256.06 (M), 255.05 (M-1); 13C-NMR (CD30D): 5 172.3, 161.1, 157.1, 132.7, 129.6, 129.3, 128.0, 127.5, 127.1, 124.3, 117.4, 115.3, 112.7. Mp. 241-243 C.
Example 6 OH

HO
3-Nitro-4-1.(E)-2-(4-hydroxvphenv1)-vinvIlphenol [0174] To a stirred solution of 2-bromo-5-methoxynitrobenzene (5 g, 21.6 mmol) in dry acetonitrile (60 mL) were added 4-methoxystyrene (3.04 g, 22.6 mmol), N,N-diisopropylethylamine (8.36 g, 64.6 mmol), biphenyl-2-yl-di-t-butyl phosphine (385 mg, 1.3 mmol) and palladium acetate (290 mg, 1.3 mmol). The resulting mixture was heated at 80 C overnight under N2. The reaction mixture was cooled to room temperature and partitioned between ethyl acetate and water.
The organic layer was separated, washed with water, brine, and dried over magnesium sulfate. Concentration under reduced pressure gave the crude product which was purified by column chromatography using ethyl acetate in hexane (5%-10%) to give the protected nitro-stilbene (4.4 g, 72%).
[0175] The nitro-stilbene (1.45 g, 5.09 mmol) was placed in the round bottom flask under nitrogen. Pyridinium hydrochloride (8.82 g, 76.3 mmol) was then added and the mixture was heated to 190 C for 2 hours. After cooling to room temperature 50 mL of water was added and the product was extracted with ethyl acetate (3 x 80 mL). The combined extracts were washed with water and brine, and dried over magnesium sulfate. The solvent was then removed in vacuo and the crude product was purified by column chromatography using 10% - 20%
ethyl acetate in hexane to afford 3-nitro-4-[(E)-2-(4-hydroxypheny1)-vinyl]phenol (480 mg, 37%). MS (ES) m/z: 257.08 (M), 256.11 (M-1); 13C-NMR (DMSO-d6): 8 158.3, 157.5, 148.8, 131.6, 129.6, 128.8, 128.5, 123.8, 121.7, 119.9, 116.3;
111Ø
Mp. 237-240 C.
Example 7 HO
(E)-Ethyl 2-hydroxv-5-(4-hydroxystyryl) benzoate [0176] In a 50 mL round-bottomed flask fitted with condenser and magnetic stirrer were placed methyl 5-bromosalicylate (5.0 g, 21.64 mmol) , Ac20 (2.65 g, 25.97 mmol), Et3N (2.63 g, 25.97 mmol) and catalytic amount of DMAP.
The reaction mixture was stirred for 24 h at rt. The reaction mixture was poured into water and extracted with Et0Ac. The organic layer was washed with water, dried and concentrated to give the acetoxy derivative (5.9 g, 98%). To a stirred solution of methyl 2-acetoxy-5-bromobenzoate (5.9 g, 21.61 mmol) in acetonitrile (80 mL) were added 4-acetoxystyrene (3.68 g, 22.69 mmol), N,N-diisopropylethylamine (8.38 g, 64.82 mmol), biphenyl-2-yl-di-t-butyl phosphine (368 mg, 1.29 mmol) and palladium acetate (291 mg, 1.29 mmol) under N2. The reaction mixture was heated to 80 C for 48 h and then cooled to rt. Water was added and the mixture was extracted with Et0Ac. The organic layer was separated, washed with water, brine, dried and concentrated to give crude product, which was purified by column chromatography, using 20% Et0Ac in hexane to give (3.19 g, 42%) of the di-acetoxy stillbene. The di-acetoxystillbene (500 mg, 1.41 mmol) was treated with ammonia (2M solution in ethanol, 15 mL) in a sealed tube at 115 C for 12 h. The reaction mixture was cooled to it. The solvent was removed and the crude product was purified by column chromatography using 50% Et0Ac in hexane to give the product (200 mg, 50%).
MS (ES) m/z: 284.12 (M), 283.14 (M-1); 13C-NMR (DMSO-d6): 8 169.5, 159.9, 157.8, 133.2, 130.4, 129.9, 128.8, 128.4, 128.3, 127.8, 124.5, 118.6,116.2, 115.9, 113.9, 62.1, 14.7; Mp. 269-271 C.
Example 8 is OH

1.42N
HO
(E)-2-Hydroxy-5-(4-hydroxystyryl)benzamide [0177] (E)-2-Hydroxy-5-(4-hydroxystyryl)benzamide was isolated (75 mg, 20%) with (E)-ethyl 2-hydroxy-5-(4-hydroxystyryl) benzoate in the procedure set forth in Example 7. Selected data for (E)-2-hydroxy-5-(4-hydroxystyryObenzamide:
MS (ES) m/z: 254.07 (M-1); 13C-NMR (DMSO-d6): 8 172.7, 160.9, 157.8, 132.8, 129.0, 128.8, 128.1, 127.4, 125.8, 125.0, 118.3, 116.3, 115.8, 115.1. Mp. 272-274 C.

Example 9 OH

HO
44(E)-2-(4-Hydroxyphenyl)viny11-3-methanesulfonvlphenol [0178] To a solution of 3-methoxythioanisole (3.084 g, 20 mmol) in 300 mL acetone was added a solution of oxone (30.74 g, 50 mmol) in 125 mL water, resulting in the formation of a white precipitate. Stirring continued overnight, and the solid was filtered off and washed with acetone. Filtrate was concentrated to remove acetone. Aqueous layer was extracted with Et0Ac (2x200 mL) and dried over Na2SO4. The solvent was removed to give 1-methanesulfony1-3-methoxy benzene (3.68 g, 98%) as oil.
[0179] 1-Methanesulfony1-3-methoxy benzene was dissolved in 90 mL
glacial acetic acid. 100 mL water was added. A solution of bromine (4.79 g, 30 mmol) in 10 mL acetic acid was added dropwise at 10 C. The stirring was continued at rt for 15 h. The pH of the mixture was adjusted to pH 8 with aqueous NaOH solution, and extracted with Et0Ac (3x200 mL) and dried over Na2SO4.
The crude compound was purified by column chromatography (SilicaGel 230 ¨
400 mesh; 20% Et0Ac in hexanes as an eluent) to give 1-bromo-2-methanesulfony1-4-methoxy benzene (2.4 g, 45%).
[0180] To a stirred solution of 1-bromo-2-methanesulfony1-4-methoxy benzene in anhyd. acetonitrile were added 4-acetoxystyrene (1.413 g, 8.715 mmol), N,N-diisopropylethylamine (3.218 g, 24.9 mmol), bipheny1-2-yl-di-tert-butylphophine (0.149 g, 0.5 mmol) and palladium acetate (0.112 g, 0.5 mmol).

The resulting mixture was stirred at 80 C for 15 h under nitrogen. The mixture was cooled to rt, and the solvent was removed under reduced pressure. The residue was dissolved in 50 mL Et0Ac, washed with water and brine and dried over anhydrous Na2SO4. Purification by column chromatography (SilicaGel 230 ¨
400 mesh; 20% Et0Ac in hexanes as an eluent) gave 4-[2-(2-methanesulfonyI)-4-methoxyphenylvinyl] phenyl acetate 1.245 g (43% yield).
[0181] 4-[2-(2-MethanesulfonyI)-4-methoxyphenylvinyl] phenyl acetate (1.2 g, 3.5 mmol) and pyridinium hydrochloride (6.07 g, 52.5 mmol) were mixed together and stirred at 190 C for 15 h. The mixture was cooled to rt and washed with water. The residue was purified by column chromatography (SilicaGel 70 ¨
200 mesh; 25 Me0H in dichloromethane) to give 4-[(E)-2-(4-hydroxyphenyl)vinyI]-3-methanesulfonylphenol (0.35 g, 33%) as an off-white solid. MS (ES) m/z:
291.06 (M+1), 290.10 (M), 289.07 (M-1); 13C-NMR (CD30D): 8 157.8, 157.1, 137.9, 131.8, 129.2, 129.0, 128.9, 128.0, 121.0, 120.5, 120.4, 115.4, 114.9. Mp. 244-245 C.
Example 10 OH

HO
3-Amino-4-1.(E)-2-(4-hydroxy-phenyl)vinvIlphenol [0182] To a solution of the product of 3-nitro-4-[(E)-2-(4-hydroxyphenyI)-vinyl]phenol (216 mg, 0.84 mmol) in acetone (6 mL) was added a solution of ammonium chloride (89.9 mg, 1.68 mmol) in 2 mL of water. The mixture was heated to reflux, and the oil bath was removed. Zinc dust (164.8 mg, 2.5 mmol) was added in small portions to the reaction mixture to maintain a moderate reaction. After the reaction subsided an additional portion of zinc (82.4 mg, 1.25 mmol) was added. The reaction mixture was then heated to reflux for an additional 2 h. The precipitate was removed by filtration, washed with acetone (2 x 20 mL) and with ethyl acetate (2 x 30 mL). The combined washings were concentrated in vacuo and the crude product was purified by column chromatography using 5%
methanol in dichloromethane to afford 3-Amino-4-[(E)-2-(4-hydroxy-phenyl)vinyl]phenol (155 mg, 72%). MS (ES) in/z: 228.11 (M+1), 134.06; .13C-NMR (DMSO-d6): 5 158.3, 156.9, 147.9, 130.4, 130.2, 127.9, 126.9, 124.5, 122.0, 115.9, 115.5, 114.2, 105.4, 102.2. Mp. 229-232 C.
Example 11 NH el OH
HO
N-{5-Hydroxv-2-RE)-2-(4-hydroxyphenvI)vinyllphenv1}-acetamide [0183] To the solution of 3-amino-4-[(E)-2-(4-hydroxy-phenyl)vinyl]phenol (100 mg) in anhydrous THE was added 225 mg of acetic anhydride and 223 mg of Et3N. The resulting solution was stirred at rt overnight. The reaction mixture was concentrated by vacuum evaporation. The residue, which was identified as mainly desired compound by 1H-NMR, was not further purified and used directly for the next step. The crude mixture from the previous reaction was dissolved in 30 mL

of methanol. K2003 (197 mg, -12 eq.) was added as well as 20 mL of H20. The resulting solution was vigorously stirred at rt for 1 h. All volatile solvents were evaporated and the residue was loaded on a silica gel column with 5%
Me0H/dichloromethane as eluent to give N-{5-hydroxy-2-[(E)-2-(4-hydroxyphenyl)vinyl]pheny1}-acetamide (55 mg, 46.4% in two steps). MS (ES) rn/z: 270.14 (M+1); Mp. 227-229 C.
Example 12 NI
H3C/ 0 I. OH
HO
5-Hydroxy-242-(4-hydroxyphenv1)vinyll-N,N-dimethvlbenzamide [0184] To a stirred solution of methyl 2-bromo-5-methoxybenzoate (4.0 g, 16.32 mmol) in acetonitrile (80 mL), were added 4-rnethoxystyrene (2.3 g, 17.14 mmol), N,N-diisopropylethylamine (6.33 g, 48.98 mmol), biphenyl-2-yl-di-t-butyl phosphine (292 mg, 0.98 mmol) and palladium acetate (220 mg, 0.98 mmol) under N2* The reaction mixture was heated to 80 C for 48 h and then cooled to rt.
Water was added and the mixture was extracted with Et0Ac. The organic layer was separated, washed with water, brine, dried and concentrated to give the crude product, which was purified by column chromatography, using 15% Et0Ac in hexane to give (1.5 g, 30%) the stilbene methyl benzoate.
[0185] The above stilbene methyl benzoate (485 mg, 1.6 mmol) was dissolved in 20 mL of THF-acetonitrile (1:1). LiOH (hydrate, 102.4 mg, 2.4 mmol) dissolved in 1 mL of water was added to the above solution and the resulting reaction mixture was stirred at rt overnight. Most of the solvent was removed in vacuo, 10 mL of water was added, and the pH was adjusted to pH=3 by addition of 2 N HCI. The product was extracted with Et0Ac (3 x 30 mL). The extracts were washed with water, brine, and dried over MgSO4. Concentration under reduced pressure gave the crude product, which was purified by column chromatography (5% Me0H-dichloromethane) to afford 462 mg (quantitative yield) of the stilbene carboxylic acid.
[0186] The stilbene carboxylic acid (462 mg, 1.6 mmol) was dissolved in mL of dry dichloromethane. Oxalyl chloride (227 mg, 1.79 mmol) was added at rt under nitrogen atmosphere and the resulting mixture was stirred for 2 hs.
Et3N
(247 mg, 2.44 mmol) was then added, followed by addition of dimethylamine (2 M

solution in THF, 1.6 mL). The solution was stirred at rt overnight. Volatiles were removed in vacuo, and the residue was partitioned between Et0Ac and water.
The organic layer was separated, washed with water, brine, and dried over magnesium sulfate. Concentration under reduced pressure gave the crude product (510 mg, quantitative yield) which was used in the next step without further purification. The above stilbene (510 mg, 1.6 mmol) was mixed with pyridinium hydrochloride (2.82 g, 24.4 mmol) was added and the resulting mixture was heated at 190 C for 3 hs. Water (20 mL) was added and the product was extracted with Et0Ac (3 x 50 mL). The organic extracts were washed with water, brine, and dried over magnesium sulfate. Concentration in vacuo gave the crude product which was purified by column chromatography (5% Me0H-dichloromethane) to afford 210 mg of the product (46% yield). Further purification by recrystallization from Et0Ac/hexane gave 5-hydroxy-2-[2-(4-hydroxyphenyl)viny1]-N,N-dimethylbenzamide (100 mg). MS (ES) m/z: 282.94 (M), 281.92 (M-1); Mp.183.5 -187.7 C.

Example 13 = OH
I -NCI
- 0 +
=.H20 N-Oxide 4'-hydroxystilbene hydrochloride [0187] To 4-hydroxybenzaldehyde (2 g) in 40 mL of acetone was added 5.65 g of K2CO3. The resulting mixture was stirred at rt for 15 min, before the addition of 1.58 g of MOMCI. The stirring was continued overnight. After the solid was filtered off, the evaporation of the solvent afforded pure MOM-protected phenol in quantitative yield. To the mixture of 4-methyl pyridine N-oxide (1.07 g) and the above protected benzaldehyde (1 g) in 10 mL of Et0H was added Na0Et potion-wise. Gas evolution was observed. After the addition, the reaction mixture was stirred at rt overnight. Et0H was evaporated and 20 mL of water were added.
The solution was extracted by Et0Ac (3 x 30 mL). The combined Et0Ac extracts were dried over anhydrous MgSO4 and evaporated under vacuum. Column chromatography afforded 1.6 g of the pure stilbene in 62% yield. The obtained product (200 mg) was dissolved in 10 mL of THF. Then 6 mL of 1 M HCI was added. The color of the solution changed from yellow to orange. The stirring was continued for 2 h. The reaction solution was left one day without any movement.
Some precipitate formed during the day. The solid was filtered off and washed with 20% Et0Ac/hexane to afford 137 mg, of product (66%). MS (ES) miz: 214.94 (M+1), 213.95 (M); Mp. 247-249 C.

Example 14 OH

N-(4-Hydroxy-phenyl)-benzamide [0188] To a solution of 4-hydroxyaniline (2.03 g, 18.6 mmol) in dry dichloromethane (40 mL) was added Et3N (2.63 g, 26.0 mmol), followed by addition of benzoyl chloride (2.70 g, 19.1 mmol). The resulting mixture was stirred at rt under nitrogen for 17 h. Water (100 mL) was then added and the product was extracted with Et0Ac (3 x 100 mL). The extracts were combined and the organic layer was washed consecutively with 1 N HCI, water, saturated sodium bicarbonate solution, water, and brine, and dried over MgSO4. After filtration and concentration in vacuo, the crude product obtained was purified by flash column chromatography (using 20% Et0Ac in hexane) to afford 680 mg of product (17%).
Further purification by recrystallization from Et0Ac and hexane afforded 170 mg of N-(4-hydroxy-phenyl)-benzamide. MS (ES) m/z: 214.08 (M+1).
Example 15 OH
4-Hydroxv-N-phenvl-benzamide [0189] 4-Hydroxybenzoic acid (1.1 g, 7.96 mmoL) and aniline (742 mg, 7.23 mmol) were dissolved in 20 mL of dry DMF. To the above solution EDCI
(1.53 g, 7.23 mmol) was added in one portion and the resulting mixture was stirred at rt overnight under nitrogen. The reaction mixture was then partitioned between Et0Ac (200 mL) and water (200 mL). The organic layer was separated, washed consecutively with 1 N HCI, water, saturated sodium bicarbonate solution, water, and brine, and dried over MgSO4. Solvent was removed under reduced pressure and the crude product was purified by column chromatography using 10% Et0Ac in hexane to give 450 mg (29% yield) of 4-Hydroxy-N-phenyl-benzamide. MS (ES) m/z: 214.08 (M+1).
Example 16 OH
4-Hydroxy-N-(4-hydroxypheny1)-benzamide [0190] 4-Hydroxybenzoic acid (1.01 g, 7.31 mmol) and 4-aminophenol (1.19 g, 10.9 mmol) were dissolved in 15 mL of dry DMF. To the above solution EDCI (1.40 g, 7.31 mmoL) was added in one portion and the resulting mixture was stirred at rt overnight under nitrogen. The reaction mixture was then partitioned between Et0Ac (200 mL) and water (200 mL). The organic layer was separated, washed consecutively with 1 N HCI, water, saturated sodium bicarbonate solution, water, and brine, and dried over MgSO4. Solvent was removed under reduced pressure and the crude product was purified by column chromatography using Et0Ac in hexane as an eluent (10% to 20%) to give 580 mg (34% yield) of 4-hydroxy-N-(4-hydroxyphenyI)-benzamide. MS (ES) m/z :
230.04 (M+1).

Example 17 HO
OH
4[4-Hydroxy-phenethyll-phenol [0191] A solution of 4,4'-dihydroxystilbene (250 mg, 1.178 mmol) in Me0H
(10 mL) and Et0Ac (2 mL) containing 5% Pd-C (120 mg) was stirred under an atmosphere of hydrogen for 2 h. The catalyst was removed by filtration and the filtrate was concentrated. Chromatography purification of the crude product on silica gel (hexane-Et0Ac 3:1, 2:1) provided 245 mg of 4-[4-Hydroxy-phenethyI]-phenol (97%) as a white solid. MS (ES) m/z: 213.05 (M-1).
Example 18 S
HO
4-Hydroxy-thiobenzoic acid S-(4-hvdroxy-phenyl) ester [0192] A PPE (polyphosphate ester) solution (7 mL) was added to a mixture of 4-hydroxybenzoic acid (552 mg, 4 mmol) and 4-mercaptophenol (554 mg, 4.4 mmol). After stirring for 16 h, the solvents were removed by evaporation and the residue was purified by column chromatography (hexane-Et0Ac 7:1 to 1:1). 4-Hydroxy-thiobenzoic acid S-(4-hydroxy-phenyl) ester (240 mg, 24%) was obtained as a white solid. MS (ES) m/z: 247.04 (M+1), 167.03, and 122.55. 13C-NMR (DMSO-de): 6188.24, 162.85, 158.82, 135.87, 129.59, 127.10, 116.33, 115.72, and 115.34.

Example 19 el OH
CVO
\INI
HO
4-Hydroxv-N-(4-hydroxvphenyI)-benzenesulfonamide [0193] A solution of thionyl chloride (15 g, 9.2 mL) and DMF (150 mg, 0.16 mL) was added to sodium 4-hydroxybenzenesulfonate (4.9 g, 25 mmol). The resulting mixture was stirred at 60 C for 3.5 h. The solution was poured into ice and stirred for 5 min. The aqueous solution was extracted with dichloromethane, dried over sodium sulfate and evaporated to provide sulfonyl chloride (4.7 g, 97%) as an oil. The crude product (-1.9 mmol) and 4-hydroxybenzylamine (207 mg, 1.9 mmol) were stirred in pyridine (6 mL) for 2 h and evaporated. The residue was purified by two columns of silica gel (a. hexane-acetone 3:1 to 1:1, (b. Me0H
in dichloromethane 2% to 7 %) to afford 4-hydroxy-N-(4-hydroxyphenyI)-benzenesulfonamide (140 mg, 28%) as a brownish solid. MS (ES) m/z: 265.12 (M), 264.08 (M-1); 13C-NMR (CD30D): 8 161.0, 154.5, 129.5, 129.0, 128.8, 124.0, 115.5, 115.3.
Example 20 4-Hvdroxy-benzoic acid pyridin-2-vl ester [0194] To a solution of 4-hydroxybenzoic acid (2.05 g, 14.8 mmol) in dry DMF (20 mL) was added a solution of imidazole (4.04 g, 59.4 mmol) in 20 mL of DMF, followed by a solution of tert-butyldimethylsilyl chloride (4.69 g, 31.2 mmol).
The mixture was stirred at 60 C under nitrogen for 16 h and then poured over ice water (100 g). The aqueous phase was extracted with ether (3 x 100 mL). The combined extracts were washed with water and brine, and dried over magnesium sulfate. The solvent was removed in vacuo to afford quantitative yield of tert-butyldimethylsily14-((tert-butyldimethylsilyl)oxy) benzoate (5.4 g).
[0195] Tert-butyldimethylsilyl 4-((tert-butyldimethylsilyl)oxy)benzoate (5.6 g, 15.3 mmol, crude) in 20 mL of dry dichloromethane containing a few drops of DMF was treated with oxalyl chloride (2.5 g, 19.9 mmol). The resulting mixture was stirred at rt for 2 days. The solvents were removed in vacuo and the crude acyl chloride was used in the next step. The solution of the crude acyl chloride (15.3 mmol) in dry dichloromethane (20 mL) was added dropwise to a cooled (0 C) solution of 2-hydroxypyridine (1.21 g, 12.7 mmol), DMAP (156 mg, 1.3 mmol), and pyridine (3 mL) in dichloromethane (20 mL). After the addition was completed, the mixture was allowed to warm up to rt overnight. Et0Ac (150 mL) was added to the mixture and the organic layer was washed with water, brine, and dried over magnesium sulfate. Concentration under reduced pressure gave the crude product which was purified by column chromatography. Elution with 10%
Et0Ac in hexane afforded 3.52 g (69%) of the silyl protected ester.
[0196] The silyl protected ester (1.02 g, 3.1 mmol) was dissolved in dry THF (20 mL) and the solution was cooled to 0 C. Tetrabutylammonium fluoride (3.7 mL, 1 M solution in THF) was added dropwise to the above solution and the resulting mixture was stirred at 0-5 C for 45 minutes. The reaction was then quenched with saturated ammonium chloride and the product extracted with Et0Ac (3 x 100 mL). The combined organic extracts were washed with water and brine, and dried over magnesium sulfate. Solvents were removed in vacuo and the crude product was purified by column chromatography using Me0H in dichloromethane (2-10%). Further purification by recrystallization from Et0Ac/hexane afforded 546 mg (82%) of 4-hydroxy-benzoic acid pyridin-2-y1 ester. MS (ES) rn/z: 216.08 (M+1), and 121.00; 13C-NMR (CD30D): 8 164.85, 163.40, 158.57, 147.95, 140.46, 132.50, 122.37, 119.61, 117.24, and 115.37.
Example 21 OH
N
HO
4-(4-Hydroxy-phenyl)-azophenol [0197] In a 250 mL round-bottomed flask fitted with condenser and magnetic stirrer were placed 4-amino phenol (1.0 g, 9.16 mmol) and toluene (100 mL). Solid K02 was added (2.0 g, 28.1 mmol). Then the reaction mixture was stirred for 48 h at rt under nitrogen. The residual K02 was destroyed by cautious addition of water. The reaction mixture was acidified by 2 N HCI and extracted with Et0Ac. The organic layer was washed with water, dried and concentrated to give a crude product, which was purified by column chromatography using 50%
Et0Ac in hexane to a give 160 mg of 4-(4-Hydroxy-phenyl)-azophenol (8%). MS
(ES) rn/z : 215.05 (M+1).

Example 22 el OH
HO OH

(E)-3-(4-Hydroxv-Dheny1)-1-(2,4,6-trihydroxv-pheny1)-propenone [0198] In a 50 mL three neck round bottom flask fitted with a condenser, naringenine (0.5 g, 1.83 mmol), NaOH (0.59 g, 14.68 mmol, 5% aqueous solution) and Me0H (20 mL) were taken. The reaction mixture was refluxed for 2 h. then poured into excess of 2N HCI solution. The product was extracted with Et0Ac.
The organic layer was washed with water, brine and dried over Na2SO4. The solvent was removed to give crude product, which was purified by column chromatography using 30% Et0Ac in hexane to give 150 mg of (E)-3-(4-hydroxy-pheny1)-1-(2,4,6-trihydroxy-pheny1)-propenone (58 %). MS (ES) tn/z: 273.11 (M+1).
Example 23 Iso OH OH

1-(4-Hydroxv-phenv1)-3-(2-hydroxy-pheny1)-propane-1,3-dione [0199] In a 50 mL round-bottomed flask fitted with condenser and magnetic stirrer were placed 4-hydroxybenzoic acid (1.50 g, 10.86 mmol), Ac20 (1.33 g, 13.03 mmol), Et3N (1.318 g, 13.03 mmol) and catalytic amount of DMAP.

The reaction mixture was stirred for 24 h at rt. The reaction mixture was poured into water and extracted with Et0Ac. The organic layer was washed with water, dried and concentrated to give the 4-0-acetyl benzoic acid (1.66 g, 85%).
[0200] To a 100 mL round-bottomed flask fitted with condenser and magnetic stirrer were placed 2'-hydroxyacetophenone (1.25 g, 9.16 mmol), 4-0-.
acetyibenzoic acid (4.0 g, 9.16 mmol) and pyridine (26 mL). POCI3 (1.4 g, 9.16 mmol) was added slowly on cooling. The reaction mixture was stirred for 24 h at rt under nitrogen. The reaction mixture was poured into ice water and extracted with Et0Ac. The organic layer was washed with water, dried and concentrated to give product (1.7 g, 86%). To the product (1.7 g, 5.69 mmol) in THF (30 mL) was added a potassium t-butoxide (0.836 g, 6.83 mmol) and the reaction mixture was stirred for 24 h at rt under N2. The reaction mixture was poured into saturated aqueous solution of NH4CI. The organic layer was separated, washed with water, dried and concentrated to give the crude product. Purification by column chromatography using 20% Et0Ac in hexane to give 200 mg of product (14%).
MS (ES) m/z: 257.08 (M+1).
EXAMPLE 24: ApoA-I Promoter Induction in Caco-2 Intestinal cells [0201] To examine the effects of compounds of the invention to increase ApoA-1 expression, Caco-2 cells, an intestinal cell line derived from human epithelial intestinal cells were, grown under conditions recommended by the ATCC and summarized in United States Patent Application Publication 20040033480 ('Wong"), Wong demonstrated that resveratrol increases ApoA-I promoter activity by 2.5-times over untreated control. This study similarly demonstrated that compounds of the present invention induced ApoA-I promoter activity.

[0202] Briefly, the promoter region of the gene encoding human ApoA-I
was isolated and ligated upstream of the firefly luciferase gene to construct the reporter plasmid pAI.474-Luc. This reporter plasmid and pRSV-13-galactosidase (as a control for transfection efficiency) were co-transfected to Caco-2 cells. The Caco-2 cells were then incubated in a MEM selection media containing 20% fetal calf serum supplemented with G418 (final concentration: 0.5 mg/mL, Gibco) to give established strains that stably express from the reporter gene. The strains were seeded in 6-well culture plates and incubated for 48 hours at 37 C under 5% carbon dioxide. Cells were starved for 24 hrs in MEM selection media containing 0.5% FBS. Then, a solution of a compound of the invention in DMSO
or other appropriate solvent was added to the wells at a final concentration of 0 to 100 pM in MEM selection media containing 0.5% FBS.
[0203] After further incubation for 48 hours, the cells were harvested and lysed using Reporter Lysis Buffer (PROMEGA E3971) and 50 pL of luciferase assay reagent (PROMEGA E4550 Luciferase Reporter 1000 assay system) was added to measure luciferase activity with a luminometer (Fluoroskan Ascent FL
from Thermo electron Corporation). Measures of luciferase activity were normalized to lysate protein concentrations, measured using Bradford Reagent (BioRad Protein Assay reagent Cat# 500-0006). The luciferase activity of cells treated with various concentrations of test compounds was compared to that of control sample (i.e., solvent without any compound of the present invention added) and untreated samples. An increase in luciferase activity compared to untreated and or control samples indicates that the compound of the invention increases the expression of ApoA-I. cells. The results provided in Table 2 are based on average values at 15pM concentrations.

Table 2: Induction of ApoA-I Promoter in Caco-2 Intestinal Cells Compound ApoA-I Promoter Induction increased Resveratrol (Example 1) increased 4,4'-dihydroxy-stilbene increased 4-(4-Hydroxy-phenyl)-azophenol (Example 21) increased 4[4-Hydroxy-phenethyll-phenol (Example 17) increased 5-[(E)-2-(4-Hydroxy-phenyl)-vinyl]-pyridin-2-ol (Example 2) 4-[(E)-2-(4-Hydroxy-pheny1)-vinyl]-3-methoxy-phenol increased (Example 3) 3-Hydroxymethy1-4-[(E)-2-(4-hydroxy-phenyl)-vinylFphenol increased (Example 4) 2-Hydroxy-5-[(E)-2-(4-hydroxyphenyl)vinyl]benzoic acid no increase (Example 5) 4-Hydroxy-N-(4-hydroxypheny1)-benzenesulfonamide increased (Example 19) increased 3-Nitro-4-[(E)-2-(4-hydroxyphenyl)-vinyliphenol (Example 6) increased (E)-Ethyl 2-hydroxy-5-(4-hydroxystyryl) benzoate (Example 7) increased 3-Amino-4-[(E)-2-(4-hydroxy-phenyl)vinyl]phenol (Example 10) increased (E)-2-Hydroxy-5-(4-hydroxystyryl)benzamide (Example 8) increased N-oxide 4'-hydroxystilbene hydrochloride (Example 13) 5-Hydroxy-242-(4-hydroxyphenyl)viny1]-N,N-dimethyl- increased benzamide (Example 12) increased 4-Hydroxy-chalcone increased 4-Methoxy-chalcone increased 4,2',4'-Trihyd roxy-chalcone increased 2,4-Dihydroxy-4'-methoxy-chalcone increased 4,2',4',6'-Tetramethoxy-chalcone EXAMPLE 25: Kinetics of ApoA-I Promoter Induction [0204] Whereas the preceding studies showed that the compounds of the invention stimulate ApoA-I promoter activity, the duration of action was unclear.
Accordingly, the kinetics of induction of the ApoA-I promoter can be assessed as follows.
[0205] Caco-2 cells transfected with pAI.474-Luc are treated with compounds of the invention at selected time points varying from 4 to 72 hours.

pAI.474-Luc contains the human ApoA-I promoter fused to the reporter gene, firefly luciferase (Luc). Wong demonstrated that when the test compound is resveratrol a significant stimulation of ApoA-I promoter activity in Caco-2 cells was observed at 4, 8, 16 and 24 hours following administration, but maximal stimulation was observed following 16 hours of exposure.
EXAMPLE 26: Confirmation of ApoA-I Induction in Caco-2 Intestinal Cells [0206] This experiment will measure the ability of a test compound to stimulate transcriptional activity of the endogenous Apo-Al promoter in the Caco-2 cells. Such simulation will result in an increase in expression of ApoA-I
protein, which is ultimately responsible for antiatherogenic activity. A test compound, which has demonstrated an increase in the activity of the ApoA-I promoter in the pAI.474-Luc construct may be tested in this assay to confirm its effect on the activity of the ApoA-I gene endogenous to the Caco-2 cells. The Caco-2 cells are cultured as described in Wong and exposed to media containing test compound at a concentration of 5, 7.5, 10,15 or 20 1.tM for 24 or 48 hours. Longer exposure of the cells to the test compound is utilized to allow adequate time for the ApoA-I
protein to be secreted into the media from the Caco-2 cells, and detected.

Conditioned media exposed to the cells for 24 or 48 hours is assayed for its content of ApoA-I protein using Western blot analysis or enzyme-linked immunoassay (ELISA).
[0207] Results should show an increase in the amount of ApoA-I protein in the conditioned media from cells treated with test compound as compared to untreated cells. The results of these studies will demonstrate that a test compound augments expression of the ApoA-I gene, are therefore, antiatherogenic. Increased expression of the ApoA-I gene augments reverse cholesterol transport and thereby facilitates the removal of cholesterol from the body.
EXAMPLE 27: ApoA-I Promoter Induction in HepG2 Liver Cells [0208] This study determined whether compounds of the invention have an effect on ApoA-I promoter activity expression in HepG2 cells, a liver cell line.
Cells were grown under conditions recommended by the ATCC and summarized by Wong.
[0209] The promoter region of the gene encoding human ApoA-I was isolated and ligated upstream the structure gene of firefly luciferase to construct a reporter plasmid (pAI.474-Luc). The reporter plasmid, along with pRSV-8-galactosidase (as a control for transfection efficiency) were co-transfected into HepG2 cells. The cells were then incubated in an MEM selection medium containing 20% fetal calf serum supplemented with G418 (final concentration:
0.5 mg/mL, Gibco) to give established strains that stably express from the reporter gene. The strains were seeded to a 6-well culture plates and incubated for 48 hours at 37 C under 5% carbon dioxide. Cells were starved for 24 hrs in MEM

selection media containing 0.5% FBS. Then, a solution of the compounds of the invention in DMSO (or other appropriate solvent) was added to the wells at a final concentration of 0 to 100 pM in MEM selection media containing 0.5% FBS.
[0210] After further incubation for 48 hours, the cells were harvested and lysed using Reporter Lysis Buffer (PROMEGA E3971), and 50 pL of luciferase assay reagent (PROMEGA E4550 Luciferase Reporter 1000 assay system) were added to measure luciferase activity with a luminometer (Fluoroskan Ascent FL
from Thermo electron Corporation). Measures of luciferase activity were normalized to lysate protein concentrations, measured using Bradford Reagent (Biorad Protein Assay reagent Cat# 500-0006). An increase in luciferase activity compared to untreated and or control samples indicates that the compound of the invention increased the expression of ApoA-I. The results provided in Table 3 are based on average values at 15pM concentrations.
Table 3: Induction of ApoA-I Promoter in HepG2 liver cells ApoA-I Promoter Compound Induction increased Resveratrol (Example 1) increased 4[4-Hydroxy-phenethyq-phenol (Example 17) 4-[(E)-2-(4-Hydroxy-phenyl)-vinyl]-3-methoxy-phenol increased (Example 3) increased 3-Nitro-4-[(E)-2-(4-hydroxyphenyl)-vinyl]phenol (Example 6) increased 4,4'-dihydroxy-stilbene EXAMPLE 28: ApoA-I Protein Expression in HepG2 Cells [0211] This study will confirm that compounds of the invention have an effect on ApoA-I protein secretion in HepG2 (liver) cells by measuring whether stimulation of transcriptional activity of the promoter in the HepG2 cells increased the abundance and secretion of ApoA-I protein.
[0212] The HepG2 cell line is obtained from the ATCC and cultured in MEM media supplemented with 10% FBS (Gibco), with 1mM sodium pyruvate, 0.1mM MEM non-essential amino acid, 2mM L-glutamine, 100 11/m1 penicillin, 100 pg/ml streptomycin and 5 pg/ml plasmocin. Cells are maintained at 37 C in an atmosphere of 5% CO2. Cells should be grown to 85% confluency prior to initiating the experiment.
[0213] Cells are plated and allowed to adhere overnight in a phenol red-free DMEM containing 10% of Charcoal/Dextran treated FBS (Hyclone), with 1mM
sodium pyruvate, 0.1 mM MEM non-essential amino acid, 2mM L-glutamine, 100 U/ml penicillin, 100 pg/ml streptomycin and 5 pg/ml plasmocin. The medium is removed, and cells are washed in 1 x PBS. Cells are then starved in phenol red-free DMEM (serum free) for a period of 24 hours. Cells are then mock treated (untreated, DMSO), or treated with compound of the invention diluted in phenol red-free DMEM (serum free) media at a concentration of 7.5 pM. Cells and media are harvested at 0, 6 and 24 hours following compound treatment.
[0214] Medium removed from cells may be applied to a solid phase capture sandwich in an ELISA assay according to manufacture's instructions such as Total human Apolipprotein ELISA Assay -Alercheck.
[0215] Compounds of the invention activate promoter activity for ApoA-I, leading to an increase in transcription, as demonstrated in Example 24 and 27 and increase in synthesis of ApoA-I, as may be demonstrated following the procedures set forth in this Example. Thus, the compounds of the invention and pharmaceutically acceptable salts or hydrates thereof, can be expected to elevate promoter activity and ApoA-I protein levels, in intestinal and liver cells and are useful for elevating plasma ApoA-I levels in a patient to whom the compound is administered.
EXAMPLE 29: Solubility Analysis [0216] To evaluate the solubility of compounds of the invention, 1 mg of compound is added to 1 mL of PBS and sonicated for 1 hour at room temperature using the Branson 3210 Sonicator in triplicate and incubated in a water bath at 25 C for 3 hrs. Samples are then centrifuged at 14,000 rpm for 6 minutes at room temperature. The supernatant is diluted with acetonitrile and is removed for analysis. Analysis may be performed using HPLV-UV with 7 point standard curve to determine the concentration. The average concentration calculated is regarded as the solubility (pM). See e.g., Ng, et al., "Evaluation of the first-pass gluceronidation of select flavones in the gut by Caco-2 monolayer model," J.
Pharm. Pharmaceut. Sd. 8(1):1-9 (2005).
[0217] These experiments are expected to indicate that the solubility of compounds of the invention are better than naturally occurring polyphenols, such as apigenin with a solubility of 3.27 pM. The poor bioavailability of naturally occurring polyphenols is partially attributed to poor solubility. As such, solubility is unlikely to affect the validity of any in vitro tests performed on the compounds of the invention, and formulation of these compounds for in vivo work should not be technically difficult to one skilled in the art. Accordingly, the compounds of the invention and pharmaceutically acceptable derivatives thereof, are suitable for human use due to the unexpected utility of improved solubility.
EXAMPLE 30: Caco-2 Permeability [0218] The Caco-2 cell drug transport model is widely used for screening compounds in drug discovery to assess intestinal transport and predict absorption rates. For example, the fraction of drug absorbed in human could be determined by in vivo human permeability or predicted by in vitro Caco-2 permeability; if compound permeability in Caco-2 cells reaches 13.3 - 18.1 x 10-6 cm/s, its predicted in vivo permeability in humans would reach 2 x 104 cm/s, and the predicted fraction of drug absorbed would be >90%, which is defined as highly permeable. Sun et al., "In vitro testing of drug absorption for drug 'developability' assessment: forming an interface between in vitro preclinical data and clinical outcome," Curr. Opin. Drug Discov. Devel. 7(1):75-85 (2004). Therefore, in vitro absorption testing is a highly valuable tool for comparison of structural analogues for improved intestinal absorption, and to identify compounds within the decision-making for clinical studies at early-stage drug discovery and development.
[0219] The method of Hai-Zhi et al., "High-throughput Caco-2 cell permeability screening by cassette dosing and sample pooling approaches using direct injection/on-line guard cartridge extraction/tandem mass spectrometry,"

Rapid Communications in Mass Spectrometry 14:523-528 (2000) may be used with obvious modifications to someone skilled in the art. Table 4 shows the results of permeability of representative compounds of the invention in an in vitro Caco-2 intestinal transport model over time as compared to resveratrol.

%Transported Compound Paap (cm/s) Propanol 1.01 x 10-5 (E)-4,4'-(diazene-1,2-diy1)diphenol (Example 21) 8.73 x 10-6 Resveratrol (Example 1)_ 3.34 x 10-6 4-Hydroxy-benzoic acid pyridin-2-y1 ester (Example 20) 2.00 x 10-6 3-nitro-4-[(E)-2-(4-hydroxy-phenyl)-vinyl]-phenol (Example 6) 1.95 x 10-6 3-Hydroxymethy1-4-[(E)-2-(4-hydroxy-pheny1)-vinyl]-phenol (Example 4) 1.79x 10-6 [0220] These experiments indicate that the permeability of representative compounds of the invention and particularly, (E)-4,4'-(diazene-1,2-diy1)diphenol (Exampel 21), are equivalent to or greater than naturally occurring polyphenols, such as or resveratrol with a permeability 3.34 x 10-6. Accordingly, the compounds of the invention and pharmaceutically acceptable salts and hydrates thereof, are potentially suitable for human use due to the permeability of intestinal cells to these compounds.
EXAMPLE 31: Transgenic Mice [0221] To confirm that the efficacy of compounds of the invention observed in vitro extends to an in vivo model, transgenic mice carrying multiple copies of the human ApoA-I gene (C57131/6-tgn(apoa1)1rub, Jackson Laboratory, Bar Harbor, ME) are exposed to compounds of the invention. The exogenous human ApoA-I gene in these mice enables them to express the human ApoA-I
protein under the control of this promoter.
[0222] Seven to eight week old male mice, transgenic for human ApoA-I
are individually identified by numbering on tail and weighed. Mice aare pre bleed via the retro-orbital plexus and 150plof blood was collected in 1.5 ml Eppendorf tube containing 1 pl of heparin and chilled on ice. Plasma is collected after centrifuging the whole blood at 14000 rpm (TOMY highspeed microrefrigerated centrifuge NTX-150) for 10 minutes at 4 C and frozen at -80 C. Plasma is analyzed for: human ApoA1 by a human ApoAI enzyme-linked Immunoassay (Direct Sandwich ELISA Calbiochem Cat#178422, Calbiochem Cat#178452, lot #B9076, Calbiochem Cat #178470 conjugated to Horse Radish Peroxidase (Cedarlane Cat#80220)); total cholesterol (Ponte scientific reagents: # C7509-STD, #L7580-18, # C7510); triglyceride (Pointe Scientific Reagents: # 7532-STD, # L7580-18, #7532). All samples are measured in triplicates and expressed as mg/d1. Mice are grouped based on plasma parameters and average body weight.
[0223] Two days following pre-bleed, mice are dosed by oral gavage daily for 14 days using a 20 gauge, 11/2" curved disposable feeding needle [Popper &

Sons]; when BID, mice are gavaged morning and afternoon (8 am and 5 pm);
when SID, mice are gavaged in morning (8 am). Compounds are prepared each day in vehicle. Test article(s), a positive control, such as, fenofibrate, and vehicle are dosed at volume of 5 mL/kg of body weight as a suspension (0.1 mL/20 g mouse). Fenofibrate may be obtained commercially (SIGMA F 6020). Mice weights will be recorded on day 1, 4, 7, 10, 12, and 15. On day 15, mice are weighed and fasted for 4 hours, sacrificed by inhalation of CO2 and blood is obtained via cardiac puncture (0.7-1.0 ml). Plasma is collected and frozen at -80 C. Samples are assayed for ApoA-I, total cholesterol, triglyceride and HDL-C
by HPLC [Polaris 200 with an auto sampler Prostar 410 from Varian on a Superose 6 10/30 column from Amersham]. Samples are sent for NMR analysis [LipoScience] to identify particle size and subclass for lipoproteins. During necropsy, liver, brown fat, and the whole of small and large intestines were collected, cleaned with cold PBS and frozen at -80 C for further analysis of compound levels.
[0224] One adverse effect of fenofibrate treatment is liver weight gain, largely due to increased hepatic peroxisome proliferation. Compounds of the invention are expected to show reduced liver weight gain possibly because they do not act as peroxisomal proliferator activator receptor alpha ligands. This would indicate that the compounds of the invention are useful for increasing plasma ApoA-I and elevating circulating HDL, without the adverse side effect associated with liver weight gain in a patient to whom the compound is administered.
EXAMPLE 32: Measurement of AGCCCCCGC Sequence Element Induction [0225] Caco-2 or HepG2 cells are exposed to effective concentrations of compounds of the invention. The cells are first transfected using standard techniques with a reporter construct comprising one or more copies of the nine nucleotides, 5'-AGCCCCCGC-3' acting as an enhancer element (Kilbourne et al., J. Biol. Chem. 270:7004 (1995)), operably linked to a promoter (for example the thymidine kinase (TK) promoter), operably linked to a reporter gene (for example luciferase, CAT, or the ApoA-I gene) along with pRSV-R-galactosidase, which monitors transfection efficiency (as taught in Wong). Compounds of the invention are then dissolved in appropriate solvent (for example, DMSO) and then added to the culture media for 16 hours. At the end of the treatment, the cells are harvested, and the reporter gene activity is measured using standard assays.
Increased or decreased reporter gene activity indicates that compounds of the invention have the ability to modulate transcription from promoters that contain the nine nucleotide sequence 5'-AGCCCCCGC-3', which is believed to comprise an egr-1 responsive element.
EXAMPLE 33: Measurement of Antioxidant Effectiveness [0226] The antioxidant performance of compounds of the invention is demonstrated by measuring the extent of low density lipoprotein hydroxyperoxide by copper catalyzed autoxidation using a published dye based color assay. FOX
Assay, Zadeh, Methods in Enzymology, 300:58 (1999). Samples containing only LDL and copper sulfate without test materials, serve as a positive control for comparison with identical mixtures containing test materials.
[0227] Human Low Density Lipoprotein (Sigma Chemical Company L2139) in phosphate buffered saline pH 7.4 is mixed with copper sulfate.
Incubation with effective amounts of compounds of the invention at 25 C or 37 C
open to air effects oxidation, and the mixture is sampled at time zero and between 3 and 20 hours of incubation for measurement of hydroperoxide in the FOX
assay.
Samples are read in a microtitre plate reader. Decreased hydroperoxide as measured by the FOX assay reveals the anti-oxidant activity of compounds of the invention and their usefulness for the treatment or prevention of disorders, diseases or conditions associated with oxidation or benefiting from the administration of anti-oxidants. An example of such a condition that would benefit from the treatment of anti-oxidants is cardiovascular disease.

EXAMPLE 34: Measurement of Antioxidant Activity by LDL Oxidation Assay [0228] The method of Esterbauer et al., Free Radic. Res. Commun. 6:67 (1989) may be used, with some modification as follows: the compound is dissolved with an appropriate solubilizing agent in a phosphate buffer solution (PBS, 0.15 M NaCl-0.05 M Na Phosphate Buffer-pH 7.4). The exact concentration is noted (approximately 30-60 ,L/mL of extract to be measured).

To 100 lit of this solution is added to 900 tL of an oxidizing buffer (made from human LDL, 120 of 5 mg/mL solution with d=1.019-1.063 g/mL, purchased from PerImmune, Rockville, MD) and copper sulfate (20 mt 01 10 mM aqueous solution) in 8 mL PBS). A blank sample made with 1004 PBS and 900 L
oxidizing buffer is also prepared. Each solution is then transferred to a 1 cm quartz cuvette, and the cuvette is placed into thermostat (37 C). An HP-8452A
Diode Array Spectrophotometer measures optical density at 234 nm (OD sub 234), making a measurement every 5 minutes. The lag time for oxidation is calculated as the maximum of the first derivative of the optical density curve. A
standard containing ascorbic acid is run with each assay.
EXAMPLE 35: Effects on LDL-Cholesterol, HDL-Cholesterol and Triglyceride Levels in Male Sprague-Dawley Rats [0229] Compounds of the invention are administered daily at a dose of 100 mg/kg to chow fed male Sprague-Dawley rats for seven days in the morning by oral gavage in 1.5% carboxymethylcellulose/0.2 /0 Tween-20 (dosing vehicle).
Animals are weighed daily. Animals are allowed free access to rodent chow and water throughout the study. After the seventh dose, animals are sacrificed in the evening and blood serum is assayed for lipoprotein cholesterol profiles, serum triglycerides, total cholesterol VLDL, LDL, and HDL cholesterol, and the ratio of HDL cholesterol to that of VLDL plus LDL cholesterol, apolipoproteins Al, C-II, C-III, and E by immunoelectrophoresis, and percent weight gain.
[0230] Blood serum is assayed for total cholesterol and triglycerides, lipoprotein cholesterol profiles, VLDL plus LDL cholesterol combined (also referred to as Apo B containing lipoprotein cholesterol or non-HDL
cholesterol), HDL cholesterol, and the ratio of HDL cholesterol to that of VLDL plus LDL
cholesterol, serum glucose, and non-esterified fatty acids, and percent weight gain.
EXAMPLE 36: Effects on LDL-Cholesterol, HDL-Cholesterol and Triglyceride Levels in Obese Female Zucker Rats [0231] Compounds of the invention and troglitazone are administered daily at various doses to 10-week old chow fed obese female Zucker rats for 14 days in the morning by oral gavage in 1.5% carboxymethylcellulose/0.2`)/0 Tween-20 (dosing vehicle). Animals are weighed daily. Animals are allowed free access to rodent chow and water throughout the study. Blood glucose is determined after a 6-hour fast in the afternoon without anesthesia from a tail vein. Serum is also prepared from a blood sample subsequently obtained from the orbital venous plexus (with 02/CO2 anesthesia) prior to and after one week treatment and used lipid and insulin determinations. At two weeks, blood glucose is again determined after a 6-hour fast without anesthesia from a tail vein. Soon thereafter, animals are sacrificed by CO2 inhalation in the evening and cardiac blood serum is collected and assessed for various lipids and insulin. Body weight is determined daily prior to dosing and at the time of euthanasia.
[0232] Blood serum is assayed for serum non-HDL cholesterol, HDL-cholesterol, triglyceride and body weight (relative to pretreatment values) in fasted (6 hours) chow-fed obese female Zucker rats. Blood glucose and serum insulin levels are determined from fasted rats just prior to and following one and two weeks of treatment. Blood glucose is maintained at slightly elevated levels for 10-12 week old obese Zucker rats during treatment with all doses, with the exception of the doses, whereby the compounds show a tendency to lower blood glucose.
Percent liver to body weight is determined after two weeks of treatment at the time of sacrifice.
EXAMPLE 37: Effects on Lipoprotein Cholesterol Profile in LDL Receptor-Deficient Mice [0233] Homozygous familial hypercholesterolemia is a rare human disease (affecting about 1/1,000,000) characterized by absent or defective LDL

receptors, markedly elevated serum LDL cholesterol levels and very early and severe onset of atherosclerosis. The more common form of this disease in humans, heterozygous familial hypercholesterolemia, occurs in about one in every 500 humans. Patients with the heterozygous form of this disease also present with elevated LDL levels and early onset of atherosclerosis.
[0234] The effect of the compounds of the invention on LDL levels in a murine model of homozygous familial hypercholesterolemia (Ishibashi et al., J.

Clin. Invest. 92:883 (1993); Ishibashi et al., J. Clin. Invest. 93:1885 (1994)) is studied. LDL receptor-deficient mice have elevated LDL cholesterol relative to wild type mice when fed a chow diet. When fed cholesterol-enriched diets, these mice develop atherosclerosis.

EXAMPLE 38: Effect on Synthesis of Non-Saponified and Saponified Lipids in Hepatocytes Isolated From Male Sprague-Dawley Rats [0235] A male Sprague-Dawley rat is anesthetized by administration of sodium pentobarbitol by intraparitoneal injection at 50 mg/kg. In situ perfusion of the liver is performed as follows. The abdomen of the animal was opened, the portal vein canulated, and the liver perfused with WOSH solution (149 mM NaCI, 9.2 mM Na HEPES, 1.7 mM fructose, 0.5 mM EGTA, 0.029 mM phenol red, 10 U/ml heparin, pH 7.5) at a flow rate of 30 ml/min for 6 minutes. To digest the liver, DSC solution (6.7 mM KCI, 143 mM NaCI, 9.2 mM Na HEPES, 5 mM CaC12-2H20, 1.7 mM fructose, 0.029 mM Phenol red, 0.2% BSA, 100 U/ml collagenase Type 1, 93 U/mIhyaluronidase, 160 BAEE/ml trypsin inhibitor, pH 7.5) is perfused through the liver at a flow rate of 30 ml/min for 6 minutes at a temperature of 37 C. After digestion, cells are dispersed in a solution of DMEM containing 2 mM
GlutMax-1, 0.2% BSA, 5% FBS, 12 nM insulin, 1.2 t.t.M hydrocortisone to stop the digestion process. The crude cell suspension is filtered through three layers of stainless steel mesh with pore sizes of 250, 106, and 75 pm respectively.
Filtered cells are centrifuged at 50 x g for two minutes and the supernatant discarded.

The resulting cell pellet is resuspended in DMEM and centrifuged again. This final cell pellet is resuspended in DMEM+HS solution (DMEM containing 2 mM
GlutMax-1, 20 nM delta-aminolevulinic acid, 17.4 mM MEM non-essential amino acids, 20% FBS, 12 nM insulin, 1.2 pM hydrocortisone) and plated to form monolayer cultures at a density of 100 x103 cells/cm2 on collagen coated culture dishes. Four hours after initial plating, media is changed to DMEM+ (DMEM
containing 2 mM GlutMax-1, 20 nM delta-aminolevulinic acid, 17.4 mM MEM non-essential amino acids, 10% FBS, 12 nM insulin, 1.2 }tM hydrocortisone) and remained on cells overnight.
[0236] To test the effect of compounds of the invention on synthesis rates of non-saponified and saponified lipids, the monolayer cultures are exposed to }tM of lovastatin or 100 }t(V1 of test compound in DMEM+ containing 1 }ICi/m114C-acetate. Control cells are exposed to the same media lacking lovastatin or the test compounds. All are exposed to 0.1% DMSO. Metabolic labeling with 14C-acetate continued for 2 hr at 37 C. After labeling, cells are washed twice with 1 ml of PBS followed by lysing in 1 ml of deionized water. Cells are scraped from the dishes, transferred to glass tubes and sonicated. 2.5 ml of 2:1 chloroform/methanol mixture was added followed by 1.5 ml of Phosphate Buffered Saline (PBS). To correct for extraction efficiency in the upcoming extractions, 3000 dpm of 3H-cholesterol was added to each tube. Tubes are shaken for 30 min. to extract lipids into the organic phase followed by centrifugation for minutes at 1000 x g to separate the organic and aqueous phases. The lower organic phase containing total lipids is removed and placed in a new tube. The organic solution is evaporated under N2. The dry lipid extract was resuspended in 1 ml of 93% ethanol containing 1 M KOH and placed at 70 C. for 2.5 hours.
After the reaction and cooling, 2 ml of hexane and 2.5 ml of water is added to each tube followed by rigorous shaking for 10 min. Tubes are centrifuged for 10 min. at 1000 x g and the organic (top) layer containing the non-saponified lipids is transferred to a new tube followed by evaporation of the organic solvent under N2.
The aqueous phase containing the saponified lipids is also transferred to a new tube. The non-saponified lipid extract, after drying, is resuspended in toluene and an aliquot of the suspension is added to a scintillation cocktail for radioactive counting. The number of 14C counts representing the incorporation of 14C-acetate into non-saponified lipids is corrected for extraction efficiency, based on the recovery of 3H counts extracted. To isolate saponified lipids, 1.5 ml of aqueous phase solution is mixed with 400 I of 1 M HCI, and then lipids are extracted by the addition of 2.5 ml of 2:1 chloroform:methanol, 1.5 ml of PBS, and 1 ml of water followed by rigorous shaking and isolation of the organic phase. The organic phase from this extraction is evaporated under N2 and resuspended in toluene.
Its radioactivity is counted using scintillant to provide the rate of 14C-acetate incorporation into saponified lipid.
EXAMPLE 39: Measurement and Comparison of HDL, LDL, VLDL and Triglyceride Levels in Humans [0237] Compounds of the invention are administered daily to human subjects. Other dietary uptake is monitored and held constant between individuals. Blood samples are taken on the day 0, prior to commencing the administration of the compounds, and once weekly for 3 to 6 months. Blood serum is assayed for total cholesterol and triglycerides, lipoprotein cholesterol profiles, VLDL plus LDL cholesterol combined (also referred to as ApoB
containing lipoprotein cholesterol or non-HDL cholesterol), HDL cholesterol, and HDL3 cholesterol fractions, and the ratio of HDL cholesterol to that of VLDL
plus LDL cholesterol, utilizing standard, commercially available cholesterol tests, such as the VAP test (Atherotech Inc, Birmingham, AL) which can reproducibly measure these parameters from a small sample of human blood. Alternatively, HDL2 and HDL3 can be measured from blood by the method of Kulkarni et al., J.
Lipid Res. 38:2353 (1997) or by the method of Gidez et al., J. Lipid Res.
23:1206 (1982). Compounds of the invention which increase total HDL, increase HDL2, decrease total LDL, decrease VLDL, decrease triglyCeride, or increase the HDUtotal cholesterol or HDULDL ratios as determined in such a blood test are useful for the treatment of cholesterol or lipid associated disorders.
EXAMPLE 40: Measurement of Atherosclerotic Lesion Size Using Proteoglycan-Binding-Defective LDL
[0238] A nucleic acid construct may be used to generate mice expressing a proteoglycan-binding-defective LDL. The transgenic mice are fed a diet containing 1.2% cholesterol, 0.5% bile salts, and 20% fat for 17 weeks. The mice are then sacrificed, and the aortas are perfusion fixed and analyzed with the en = face procedure, in which the entire aorta is pinned out fiat, stained with Sudan IV, = and analyzed with a morphometric image-analysis system (Image-1/AT) to =
quantitate the extent of atherosclerosis.
EXAMPLE 41: Determination of ACAT Inhibition [0239] The activity of compounds of the invention as inhibitors of ACAT
may be determined by known methods, for example, those taught in US .Patent 6,165,984, summarized below.
=
[0240] First, rats are sacrificed by decapitation and the livers excised. 1 g of each of the livers is homogenized in 5 ml of homogenization medium (0,1 M
KH2 PO4, pH 7.4, 0.1 mM EDTA and 10 mM13-mercaptoethanol). The homogenate is centrifuged at 3,000 x g for 10 min. at 4 C. and the supernatant thus obtained is centrifuged at 15,000 x g for 15 min. at 4 C. to obtain a supernatant. The supernatant is put into an ultracentrifuge tube (Beckman) and .
centrifuged at 100,000 x g for 1 hour at 4 C to obtain microsomal pellets, which are then suspended in 3 ml of the homogenization medium and centrifuged at 100,000 x g for 1 hour at 4 C. The pellets thus obtained are suspended in 1 ml of the homogenization medium. The concentration of proteins in the resulting suspension is determined by Lowry's method and then adjusted to 4 to 8 mg/ml.
The resulting suspension is stored in a deep freezer (Biofreezer, Forma Scientific Inc.).
[0241] 6.67 I of 1 mg/ml cholesterol solution in acetone is mixed with 6 I
of 10% Triton WR-1339 (Sigma Co.) in acetone and, then, acetone is removed from the mixture by evaporation using nitrogen gas. Distilled water is added to the resulting mixture in an amount to adjust the concentration of cholesterol to mg/ml. To 10 I of the resulting aqueous cholesterol solution is added 10 . I
of 1 M KH2 PO4 (pH 7.4), 5 I of 0.6 mM bovine serum albumin (BSA), 10 I of microsome solution obtained in (Step 1) and 55 I of distilled water (total 90 I).
The mixture is pre-incubated in a waterbath at 37 C. for 30 min.
[0242] 10 I of (1-14C) oleoyl-CoA solution (0.05 Ci, final concentration:
M) is added to the pre-incubated mixture and the resulting mixture is incubated in a waterbath at 37 C. for 30 min. To the mixture is added 500 I
of isopropanol:heptane mixture (4:1(v/v)) 300 I of heptane and 200 I of 0.1 M
KH2PO4 (pH 7.4), and the mixture is mixed violently by using a vortex and then allowed to stand at a room temperature for 2 min. 200 I of the resulting supernatant is put in a scintillation bottle and 4 ml of scintillation fluid (Lumac) is added thereto. The mixture is assayed for radioactivity with liquid scintillation counter. ACAT activity is calculated as picomoles of cholesteryl oleate synthesized per min. per mg protein (pmoles/min/mg protein).

t-EXAMPLE 42: Determination of inhibition of HMG-CoA reductase [0243] The potency of inhibition of HMG-CoA reductase by compounds of the invention may be determined using known methods, such as that taught in US
Patent 5,877,208, summarized below.
[0244] Rats are sacrificed by decapitation and the livers are excised and , immediately placed in an ice-cold homogenization medium (50 mM KH2 PO4 (pH
7.0), 0.2M sucrose, 2 mM dithiothreitol (DTT). The livers are homogenized in the homogenization medium (2 ml medium/g of the liver) with a Waring blender for sec. (three strokes with a motor-driven Teflon pestle in a Potter-Elvehjem type glass homogenizer). The homogenate is centrifuged at 15,000 x g for 10 min.
and the supernatant thus obtained is centrifuged at 100,000 x g for 75 min. to obtain microsomal pellets, which are then resuspended in the homogenization medium containing 50 mM EDTA and centrifuged at 100,000 x g for 60 min. The supernatant containing the microsome is used as an enzyme source.
[0245] The activity of HMG-CoA reductase is determined by employing radiolabeled14C HMG-CoA, in accordance with the method of Shapiro et at.
(Shapiro et at Biochemica et Biophysica Acta 370:369 (1974)) as follows. The enzyme in the supernatant containing the microsome obtained in (Step 1) is activated at 37 C. for 30 min. Added to a reaction tube is 20 I of HMG¨CoA
reductase assay buffer (0.25M KH2 PO4 (pH 7.0), 8.75 mM EDTA, 25 mM DTI, 0.45M KCI and 0.25 mg/ml BSA), 5 p1 of 50 mM NADPH, 5 lof radiolabeled 14C
HMG-CoA (0.05 Ci/tube, final conc. 120 M), and 10 I of activated microsomal enzyme (0.03-0.04 mg), and the mixture is incubated at 37 C. for 30 min. The reaction is terminated by adding 10 I of 6M HCI to the mixture, and the mixture is incubated at 37 C. for 15 min. to allow complete lactonization of the product.
The ftio) precipitate is removed by centrifugation at 10,000 x g for 1 min. and the supernatant is applied to a Silica gel 60G TLC plate (Altech, Inc., Newark, U.S.A.) and then developed with benzene:acetone (1:1, v/v). The appropriate region is removed by scraping with a disposable cover slips and assayed for radioactivity with 1450 Microbeta liquid scintillation counter (Wallacoy, Finland). Enzyme activities are calculated as picomoles mevalonic acid synthesized per min. per mg protein (pmoles/min/mg protein). Control rats show a relatively high HMG-CoA
reductase activity, while the HMG-CoA activities observed with rats fed compounds of the invention are lower than that of the control group.
EXAMPLE 43: Method of Determining the ABCA-1 Activating Ability [0246] This test will demonstrate the effectiveness of compounds of the invention on ABCA-1 gene expression, using a known method, as taught in United States Patent 6,548,548, Briefly, the pGL3 luciferase reporter vector system (Promega, Madison, Wis.) is used to create a recombinant plasmid to measure reporter gene expression under control of the ' ABCA-1 promoter.
[0247] Plasmid pGL3-Basic (Promega, Madison, Wis.; Cat. #E1751) is!
used as a control plasmid containing the promoterless luciferase gene. The reporter construct containing the ABCA-1 promoter and luciferase gene is made by cloning a genomic fragment from the 5' flanking region of the ABCA-1 gene (hAPR1 5' promoter, corresponding to nucleotides 1080-1643 of SEQ ID NO: 3 as disclosed in United States Patent 6,548,548) into the Sac site of the GL3-Basic plasmid to generate plasmid GL-6a. Next, plasmid GL-6a is digested with Spel and Acc65I. A BsiWI-Spel fragment excised from a lambda subclone, representing the ABCA-1 genomic sequence corresponding to nucleotides 1-1534 of SEQ ID NO: 3 is ligated into the remaining vector/ABCA-I promoter fragment produced by this digestion. The resultant plasmid, pAPR1, encodes the luciferase reporter gene under transcriptional control of 1.75 kb of the human ABCA-1 promoter sequence.
[0248] The control or pAPR1 plasmid wisas transfected into confluent cultures of RAW 264.7 cells maintained in DMEM containing 10% fetal bovine serum. Each well of a 12 well dish is transfected for 5 hours with either pGL3-Basic, pGL3-Promoter or pAPR1 DNA (1 g), luciferase plasmid DNA (1 jig), and 12 I of Geneporter reagent (Gene Therapy Systems, San Diego, Calif.; Cat.
#T201007). In addition, 0.1 g of pCM\/f3 plasmid DNA (Clontech, Palo Alto, Calif., Cat. #6177-1) is added as a control for transfection efficiency. After 5 hours, the culture medium is replaced with serum-free DMEM/BSA in the presence or absence of acetylated LDL (100 g/m1) and incubated for 24 hours.
[0249] Following transfection, the cells in each well are lysed in 70 I of lx cell lysis reagent (Promega, Madison, Wis., Cat. #E3971), subjected to one freeze-thaw cycle, and the lysate cleared by centrifugation for 5 minutes at 12,000 x g. After centrifugation, 100 I of luciferase assay reagent (Promega, Madison, Wis., Cat. #E1501) is added to 10 I of lysate. The luciferase activity of each lysate is measured as light units using a luminometer. Additionally, the 13-galactosidase activity of each lysate is measured using the chemiluminescent assay reagents supplied in the Galacto-light kit according to the manufacturer's instructions (Tropix Inc., Bedford, Mass.: Cat. #BL100G). The normalized luciferase activity for each lysate is determined by dividing the luciferase activity j wo 2006/045010 value by the determined 13-galactosidase value and reported as relative light =
units. =
EXAMPLE 44: Measurement of Reduced Hypertension In Vivo [0250] A pressure transducer is connected to the right carotid artery via a catheter containing heparinized saline. The mean arterial pressure and heart rate are recorded. The rats are anesthetized with nembutal at an initial dose of 35 mg/kg body weight with additional smaller injections as necessary. The compounds are dissolved in a pharmaceutical carrier (such as Abbott's 5%
dextrose USP) and injected into the rats via a catheter in the right femoral vein.
Positive controls that may be employed include sodium nitroprusside and NaNO2, while NaNO3 may be employed as a negative control. The results will show that the compounds provided for in the invention are potent anti-hypertensives, that decreases blood pressure significantly. The peak value of the blood pressure decrease should take a short time to reach, for example approximately one minute, after injection and the blood pressure should start to rise again soon . thereafter and should have totally recovered within about approximately 10 to 15 minutes.
EXAMPLE 45: Measurement of the Reduction of Degree of Restenosis After Arterial Injury in High Cholesteric Rabbits [0251] The procedure of Tomani, as described in-U.S. Pat. No. 5,595,974 and further described by Goodman in US Patent 6,022,901, may be used to evaluate the utility of the compounds of . =
the invention to preventing restenosis in high cholesteric rabbits.

C.

EXAMPLE 46: Use in Preventing Restenosis in Humans [0252] The procedure of Tardif et al., New England J. Med. 337:365 (1997))may be carried out as described by Goodman in United States Patent 6,022,901 to examine the ability of compounds of the invention to prevent restenosis in humans.
. EXAMPLE 47: Measurement of Platelet Anti-Aggregating Activity = - [0253] Platelet anti-aggregating activity may be evaluated in vitro on human platelets stimulated by thrombin in accordance with the method described by Berlele et al., Science 220:517 (1983).
EXAMPLE 48: Measurement of the Influence on ADP-Induced Aggregation of Platelets in Rabbits [0254] Aggregation of platelet testing: Rabbit blood is sampled by cardiac puncture from rabbit with silicon-coated syringe. The blood is mixed with 3.8%

sodium citrate at 9:1 and spun at 1,000 rpm for 6 minutes. 1 ml of the platelet-rich plasma is transferred to a silicon-coated 2 ml cell, mixed and read for transmittance (Ti), with a spectrophotometer. 0.02 ml of ADP (10 mu.M) is added, stirred, and read for transmittance of the platelet-containing-plasma once per minute and the maximal transmittance (Tm) is obtained within 10 minutes. Spin the blood sample at 3000 rpm for 45 minutes and read for transmittance.
EXAMPLE 49: Measurement of the Effect on Collagen Induced Thrombo-.
cytopenia in Vivo [0255] Male rats (Charles River, CRL:CD(SD), 400-450 x g) are anesthetized with Sodium pentabarbital (65 mg/kg, Vet Labs, Limited, Inc., Lenexa, KA). Two incisions are made to expose both jugular veins. Using an infusion pump (Harvard Apparatus, South Natick, Mass.) and a 5 cc syringe with a 19 gauge butterfly, the test compound or vehicle is infused into the left jugular vein at a rate of 0.39 ml/min for 3 minutes. After 2 minutes of compound/vehicle infusion, collagen (60 g/kg) (Helena Laboratories, Beaumont, TX) is injected with a 1 ml syringe into the right jugular vein. The body cavity is opened and the vena cava is exposed for blood sampling. One minute after the collagen injection, compound infusion is stopped and blood is sampled from the vena cava (within sec) with a 3 cc syringe containing 0.3 mg of 4.5% EDTA/Tris (0.1M) (pH 7.35) plus 150 M indomethacin. Platelet rich plasma (PRP) is prepared by centrifuging the blood at 126 x g for 10 min. 5 j.t.I of PRP is counted in 20 ml of Isoton.
Ill in a Coulter Counter. Percent inhibition of collagen induced aggregation is calculated by comparison of the number of platelets counted in treated animals with numbers for animals receiving no collagen and with counts from animals receiving vehicle and collagen. Estimation of potency is based on inhibition of collagen-induced thrombocytopenia.
EXAMPLE 50: Measurement of In Vivo Anti-Psoriatic Effectiveness [0256] A topical formulation comprising a compound of the invention is administered to the affected area of human patients suffering from psoriasis.
A
control formulation, containing none of the compound of the invention, is applied to a comparable area of the patient. The effectiveness of the compound is determined by analyzing the improvement in inflammation and decrease in proliferative cells at the site at which the compound is applied compared to the site at which control formulation is applied at 3 and 7 days following administration.
EXAMPLE 51: Measurement of Protein Kinase Inhibition [0257] A compound of the invention is mixed with radio-labeled ATP, an appropriate protein kinase and an appropriate substrate in an appropriate buffer.

Following incubation the reaction is stopped by spotting onto filter paper and a scintillation counter employed to quantify the difference in ATP addition to the substrate, which measures the amount of protein kinase inhibition, when compared to control.
EXAMPLE 52: Measurement of Inhibition of Neutrophil Activation [0258] A compound of the invention is tested using the protocol of Tudan, Biochem. Pharmacol. 58:1869 (1999). This test demonstrates the ability of the test compound to inhibit the activation of neutrophils caused by crystals and by chemoattractants such as fMLP.
EXAMPLE 53: Measurement of Inhibition of TPA-Induced Inflammation [0259] A compound of the invention is tested by a modified method of Marks et at., Cancer Res. 36:2636 (1976) to demonstrate the compound's effectiveness against inflammation induced by application of 12-0-tetradecanoylphorbol-13-acetate (TPA). The compound is applied to an ear of a mouse, followed by application of TPA. Four hours later a biopsy punch of the mouse ear is weighed to measure edema, compared to a biopsy punch of the other ear which received no compound.
EXAMPLE 54: Measurement of the Inhibition of Carrageenan-Induced Inflammation [0260] A compound of the invention is tested by the method of Slowing et al., J Wrhnoph Exol. 43:9 (1994) in Wistar rats. Animals receive intradermal injections of Freund's adjuvant into the tail. Seven days later, the test compound is administered, followed one hour later by a suspension of carrageenan in saline solution into the left hind paw. Paw volume is measured by water plethysmography and compared to control.

EXAMPLE 55: Measurement of Cancer Chemopreventative Activity [0261] C3H/10T1/2 clone 8 cells (ATCC) are treated with a compound of "
the invention by the method of Mondal et al., Cancer Res. 36:2254 (1976). The cells in culture are treated with 3-methylcholanthrene for 24 hours, followed by washing a five days of incubation in fresh medium. TPA is subsequently added to the medium, with or without the test compound. Seven weeks after confiuency is reached, fixation with methanol and staining with Giemsa reveals Type II and Ill transformed foci, which are scored to demonstrate effectiveness of inhibition of two-stage transformation by the test compound.
[0262] The scope of the claims should not be limited by the embodiments set out in the examples, but should be given the broadest interpretations consistent with the description as a whole.

Claims (13)

1. The use of a effective amount of a compound of Formula I for increasing expression of ApoA-I in a mammal wherein:
X is selected from CR11 and N;
Y is selected from CR12 and N;
R3 and R5 are each hydroxyl;
R11 and R12 are each independently selected from alkoxy, aryloxy, alkenyl, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone;
R1, R5, R6, and R10 are each independently selected from alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone;

R2, R4, R7, and R9, are each independently selected from aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, nitro, phosphate, sulfide, sulfinyl, sulfonyl, sulfonic acid, sulfonamide and thioketone, or two adjacent substituents selected from R1, R2, R4, R5, R6, R7, R9, R10, R11, and R12 are connected in a 5 or 6-membered ring to form a bicyclic aryl or bicyclic heteroaryl;
each W is independently selected from C and N; and Z is a double bond;
provided that:
R2 and R4 are not each hydroxyl;
at least one W is N;
when W is N, then p is 0; and when W is C, then p is 1.
2. The use of claim 1, wherein the amount of the compound of Formula l is sufficient to establish a concentration ranging from about 0.001 µM to about 100 µM in the mammal.
3. The use of claim 2, wherein the concentration ranges from about 1 µM
to about 20 µM.
4. The use of claim 1, wherein the amount of the compound of Formula I is administered with a pharmaceutically acceptable carrier in a pharmaceutically acceptable composition.
5. The use of claim 1, further comprising treating or preventing a cardiovascular, cholesterol or lipid related disorder.
6. The use of claim 1, wherein the double bond is an E-double bond.
7. The use of a therapeutically effective amount of a compound in manufacturing a medicament for increasing expression of ApoA-I in a mammal where the compound is any one of:
4,4'-dihydroxy-stilbene;
6-[(E)-2-(4-Hydroxy-phenyl)-vinyl]-pyridin-3-ol;
5-[(E)-2-(4-Hydroxy-phenyl)-vinyl]-pyrazin-2-ol;
5-{[1-(4-Hydroxy-phenyl)-meth-(E)-ylidene]-amino}-pyridin-2-ol;
5-(4-Hydroxy-phenylazo)-pyridin-2-ol;
3-Hydroxymethyl-4-[(E)-2-(4-hydroxy-phenyl)-vinyl]-phenol;
2-Hydroxy-5-[(E)-2-(4-hydroxyphenyl)vinyl]benzoic acid;
3-Nitro-4-[(E)-2-(4-hydroxyphenyl)-vinyl]phenol;
(E)-Ethyl 2-hydroxy-5-(4-hydroxystyryl) benzoate;

4-[(E)-2-(4-Hydroxyphenyl)vinyl]-3-methanesulfonylphenol;
3-Amino-4-[(E)-2-(4-hydroxy-phenyl)vinyl]phenol;
N-{5-Hydroxy-2-[(E)-2-(4-hydroxyphenyl)vinyl]phenyl}-acetamide;
5-Hydroxy-2-[2-(4-hydroxyphenyl)vinyl]-N,N-dimethylbenzamide;
4-[4-Hydroxy-phenethyl]-phenol;
4-Hydroxy-thiobenzoic acid S-(4-hydroxy-phenyl) ester;
4-Hydroxy-benzoic acid pyridin-2-yl ester; or 4-(4-Hydroxy-phenyl)-azophenol, or pharmaceutically acceptable salts thereof.
8. The use of a therapeutically effective amount of 5-[(E)-2-(4-Hydroxyphenyl)-vinyl]-pyridin-2-ol or a pharmaceutically acceptable salt thereof in manufacturing a medicament for increasing expression of ApoA-I in a mammal.
9. The use according to claim 5, wherein the cardiovascular, cholesterol or lipid related disorder is any one of acute coronary syndrome, angina pectoris, arteriosclerosis, atherosclerosis, carotid atherosclerosis, cerebrovascular disease, cerebral infarction, congestive heart failure, congenital heart disease, coronary heart disease, coronary artery disease, coronary plaque stabilization, dyslipidemias, dyslipoproteinemias, endothelium dysfunctions, familial hypercholeasterolemia, familial combined hyperlipidemia, hypertension, hyperlipidemia, intermittent claudication, ischemia, ischemia reperfusion injury, ischemic heart diseases, multi-infarct dementia, myocardial infarction, peripheral vascular disease, restenosis, renal artery atherosclerosis, rheumatic heart disease, stroke, thrombotic disorder, transitory ischemic attacks, or lipoprotein abnormalities associated with Alzheimer's disease, obesity, diabetes mellitus, syndrome X and impotence.
10. The use according to claim 9, wherein the cardiovascular, cholesterol or lipid related disorder is any one of dyslipidemias, dyslipoproteinemias, hypertension, coronary artery disease, or atherosclerosis.
11. The use of claim 1, wherein R1, R5, R6, and R10 are each independently selected from alkoxy, aryloxy, alkyl, amide, amino, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, hydrogen, hydroxyl, nitro, phosphate, sulfonyl, sulfonic acid, and sulfonamide.
12. The use of claim 1, wherein R2, R4, R7, and R9, are each independently selected from amide, amino, carboxy, cyano, ester, ether, formyl, halogen, hydrogen, hydroxyl, phosphate, sulfide, sulfonic acid, and sulfonamide.
13. The use of claim 1, wherein only one W is N.
CA2584485A 2004-10-20 2005-10-20 Stilbenes and chalcones for the prevention and treatment of cardiovascular diseases Active CA2584485C (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US62088804P 2004-10-20 2004-10-20
US60/620,888 2004-10-20
US62681904P 2004-11-10 2004-11-10
US60/626,819 2004-11-10
US66585905P 2005-03-29 2005-03-29
US60/665,859 2005-03-29
PCT/US2005/037719 WO2006045010A2 (en) 2004-10-20 2005-10-20 Stilbenes and chalcones for the prevention and treatment of cardiovascular diseases

Publications (2)

Publication Number Publication Date
CA2584485A1 CA2584485A1 (en) 2006-04-27
CA2584485C true CA2584485C (en) 2013-12-31

Family

ID=36057631

Family Applications (2)

Application Number Title Priority Date Filing Date
CA2584507A Active CA2584507C (en) 2004-10-20 2005-10-20 Flavanoids and isoflavanoids for the prevention and treatment of cardiovascular diseases
CA2584485A Active CA2584485C (en) 2004-10-20 2005-10-20 Stilbenes and chalcones for the prevention and treatment of cardiovascular diseases

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CA2584507A Active CA2584507C (en) 2004-10-20 2005-10-20 Flavanoids and isoflavanoids for the prevention and treatment of cardiovascular diseases

Country Status (4)

Country Link
US (4) US8093273B2 (en)
EP (2) EP1838296B1 (en)
CA (2) CA2584507C (en)
WO (2) WO2006045010A2 (en)

Families Citing this family (118)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040033480A1 (en) * 2002-08-15 2004-02-19 Wong Norman C.W. Use of resveratrol to regulate expression of apolipoprotein A1
WO2004058722A1 (en) * 2002-12-24 2004-07-15 Arena Pharmaceuticals, Inc. Diarylamine and arylheteroarylamine pyrazole derivatives as modulators of 5ht2a
TW200523253A (en) 2003-07-22 2005-07-16 Arena Pharm Inc Diaryl and arylheteroaryl urea derivatives as modulators of the 5-ht2a serotonin receptor useful for the prophylaxis and treatment of disorders related thereto
AU2005207029B2 (en) 2004-01-20 2011-09-01 Brigham Young University Novel sirtuin activating compounds and methods for making the same
EP1727803B3 (en) * 2004-03-23 2014-04-23 Arena Pharmaceuticals, Inc. Processes for preparing substituted n-aryl-n'-[3-(1h-pyrazol-5-yl)phenyl] ureas and intermediates thereof
US8338648B2 (en) * 2004-06-12 2012-12-25 Signum Biosciences, Inc. Topical compositions and methods for epithelial-related conditions
WO2006045010A2 (en) 2004-10-20 2006-04-27 Resverlogix Corp. Stilbenes and chalcones for the prevention and treatment of cardiovascular diseases
SA05260357B1 (en) * 2004-11-19 2008-09-08 ارينا فارماسيتو تيكالز ، أنك 3-phenyle-pyrazole derivatives as modulators of the 5-ht 2a serotonin receptor useful for the treatment of disorders related thereto
TWI389897B (en) * 2005-02-22 2013-03-21 Chugai Pharmaceutical Co Ltd 1- (2H) -isoquinolinone derivatives
EP1872795A4 (en) 2005-04-21 2009-07-22 Astellas Pharma Inc Therapeutic agent for irritable bowel syndrome
US9133212B1 (en) 2005-06-15 2015-09-15 Vanderbilt University Inhibitors of hemeprotein-catalyzed lipid peroxidation
US8367669B2 (en) * 2005-06-15 2013-02-05 Vanderbilt University Inhibitors of hemeprotein-catalyzed lipid peroxidation
NZ594105A (en) 2005-07-25 2013-02-22 Intermune Inc Novel macrocyclic inhibitors of hepatitis c virus replication
EP1909788A2 (en) * 2005-07-29 2008-04-16 Resverlogix Corp. Pharmaceutical compositions for the prevention and treatment of complex diseases and their delivery by insertable medical devices
JO2783B1 (en) 2005-09-30 2014-03-15 نوفارتيس ايه جي 2-Amino-7,8-Dihidro-6H-Pyrido(4,3-D)Pyrimidin-5-ones
WO2007044893A2 (en) 2005-10-11 2007-04-19 Intermune, Inc. Compounds and methods for inhibiting hepatitis c viral replication
US8481535B2 (en) 2006-05-18 2013-07-09 Arena Pharmaceuticals, Inc. Crystalline forms and processes for the preparation of phenyl-pyrazoles useful as modulators of the 5-HT2A serotonin receptor
ES2536762T3 (en) 2006-05-18 2015-05-28 Arena Pharmaceuticals, Inc. Primary amines and their derivatives as 5-HT2A serotonin receptor modulators useful for the treatment of disorders related to this
CN103396412A (en) 2006-05-18 2013-11-20 艾尼纳制药公司 Modifier of 5-HT2A serotonin receptor
KR20090024834A (en) 2006-07-05 2009-03-09 인터뮨, 인크. Novel inhibitors of hepatitis c virus replication
GB0618168D0 (en) * 2006-09-15 2006-10-25 Babraham Inst Compounds
TWI415845B (en) 2006-10-03 2013-11-21 Arena Pharm Inc Pyrazole derivatives as modulators of the 5-ht2a serotonin receptor useful for the treatment of disorders related thereto
KR20090094090A (en) * 2006-12-28 2009-09-03 리머릭 바이오파르마 인코오포레이티드 Methods and compositions for therapeutic treatment
KR100879253B1 (en) 2006-12-28 2009-01-16 전북대학교산학협력단 treatment for hypertension and diabetic nephropathy using ADP-ribosyl cyclase inhibitors
US7999107B2 (en) 2007-01-31 2011-08-16 Merck Sharp & Dohme Corp. Substituted pyrano[2,3-B]pyridine derivatives as cannabinoid-1 receptor modulators
US10369222B2 (en) * 2012-01-18 2019-08-06 Biosuccess Biotech Co., Ltd. Compositions and methods of use of phorbol esters for the treatment of stroke
CN103319408B (en) * 2007-02-01 2016-04-13 雷斯韦洛吉克斯公司 For the compound of prevention and therapy cardiovascular disorder
CN101641339B (en) 2007-02-01 2013-07-17 雷斯韦洛吉克斯公司 Compounds for the prevention and treatment of cardiovascular diseases
DE102007015169A1 (en) 2007-03-27 2008-10-02 Universität des Saarlandes Campus Saarbrücken 17beta-hydroxysteroid dehydrogenase type 1 inhibitors for the treatment of hormone-dependent diseases
TWI425945B (en) 2007-05-28 2014-02-11 Seldar Pharma Inc Tetrahydroisoquinolin-1-one derivatives or salt thereof
TWI366565B (en) 2007-06-06 2012-06-21 Otsuka Pharma Co Ltd Quinolone compound and pharmaceutical composition
AU2008266856A1 (en) * 2007-06-18 2008-12-24 University Of Louisville Research Foundation, Inc. Family of PFKFB3 inhibitors with anti-neoplastic activities
CN101743242A (en) 2007-06-29 2010-06-16 苏尼西斯制药有限公司 Heterocyclic compounds useful as RAF kinase inhibitors
AR067354A1 (en) 2007-06-29 2009-10-07 Sunesis Pharmaceuticals Inc USEFUL COMPOUNDS AS INHIBITORS OF RAF QUINASA
TW200924785A (en) * 2007-07-31 2009-06-16 Limerick Biopharma Inc Phosphorylated pyrone analogs and methods
CA2693340A1 (en) * 2007-07-31 2009-02-05 Limerick Biopharma, Inc. Pyrone analog compositions and methods
WO2009018326A2 (en) * 2007-07-31 2009-02-05 Limerick Biopharma, Inc. Soluble pyrone analogs methods and compositions
WO2009023253A2 (en) 2007-08-15 2009-02-19 Arena Pharmaceuticals Inc. IMIDAZO[L,2-α]PYRIDINE DERIVATIVES AS MODULATORS OF THE 5-HT2A SEROTONIN RECEPTOR USEFUL FOR THE TREATMENT OF DISORDERS RELATED THERETO
US20090149530A1 (en) * 2007-08-17 2009-06-11 Alberte Randall S Antiinfective Flavonol Compounds and Methods of Use Thereof
WO2009049214A2 (en) * 2007-10-12 2009-04-16 Northwestern University Inhibition and treatment of prostate cancer metastasis
WO2009114875A2 (en) * 2008-03-14 2009-09-17 The Cleveland Clinic Foundation Oxidized paraoxonase 1 and paraoxonase 1/hdl particle number ratio as risk markers for cardiovascular disease
WO2009123714A2 (en) 2008-04-02 2009-10-08 Arena Pharmaceuticals, Inc. Processes for the preparation of pyrazole derivatives useful as modulators of the 5-ht2a serotonin receptor
MX2010011307A (en) * 2008-04-15 2010-11-09 Intermune Inc Novel inhibitors of hepatitis c virus replication.
PL2346837T3 (en) * 2008-06-26 2015-07-31 Resverlogix Corp Methods of preparing quinazolinone derivatives
WO2009158031A2 (en) * 2008-06-27 2009-12-30 Limerick Biopharma, Inc. Methods and compositions for therapeutic treatment
WO2010062321A1 (en) 2008-10-28 2010-06-03 Arena Pharmaceuticals, Inc. Processes useful for the preparation of 1-[3-(4-bromo-2-methyl-2h-pyrazol-3-yl)-4-methoxy-phenyl]-3-(2,4-difluoro-phenyl)-urea and crystalline forms related thereto
DK2364142T3 (en) * 2008-10-28 2018-04-30 Arena Pharm Inc COMPOSITIONS OF A 5-HT2A SEROTONIN RECEPTOR MODULATOR USED FOR TREATMENT OF DISEASES RELATED TO THE RECEPTOR
DE102009043436A1 (en) * 2008-10-29 2010-05-06 Merck Patent Gmbh liquid-crystal display
TWI492943B (en) 2008-12-05 2015-07-21 大塚製藥股份有限公司 Quinolone compound and pharmaceutical composition
US8952021B2 (en) 2009-01-08 2015-02-10 Resverlogix Corp. Compounds for the prevention and treatment of cardiovascular disease
US8455394B2 (en) 2009-01-14 2013-06-04 Dow Agrosciences, Llc Fungicidal compositions including hydrazone derivatives and copper
US20100204340A1 (en) * 2009-02-09 2010-08-12 Muhammed Majeed Orally bioavailable stilbenoids- compositions and therapeutic applications thereof
CA2754509C (en) 2009-03-18 2018-03-06 Resverlogix Corp. Novel anti-inflammatory agents
PT2421533T (en) 2009-04-22 2019-01-21 Resverlogix Corp Novel anti-inflammatory agents
TW201121968A (en) * 2009-11-09 2011-07-01 Intermune Inc Novel inhibitors of hepatitis C virus replication
WO2011075596A1 (en) 2009-12-18 2011-06-23 Arena Pharmaceuticals, Inc. Crystalline forms of certain 3-phenyl-pyrazole derivatives as modulators of the 5-ht2a serotonin receptor useful for the treatment of disorders related thereto
SI2595965T1 (en) 2010-07-20 2016-09-30 Vestaron Corporation Insecticidal triazines and pyrimidines
CN102464615B (en) * 2010-11-09 2015-08-19 中国科学院上海药物研究所 Aryl isoquinoline derivative and pharmaceutical composition, preparation method and purposes
WO2013025484A1 (en) * 2011-08-12 2013-02-21 Lapchak Paul A Polyphenol analogs to treat ischemia
AU2012295280A1 (en) 2011-08-12 2014-03-27 Salk Institute For Biological Studies Neuroprotective polyphenol analogs
ES2745471T3 (en) 2011-11-01 2020-03-02 Resverlogix Corp Oral immediate-release formulations for substituted quinazolinones
CA2865957C (en) 2012-03-01 2020-02-11 Ptc Therapeutics, Inc. Compounds for treating spinal muscular atrophy
CN104302358B (en) 2012-03-07 2017-12-05 癌症研究协会:皇家癌症医院 The assimilation compound of isoquinolin 1 and their therapeutical uses of 3 aryl 5 substitution
WO2013142236A1 (en) * 2012-03-23 2013-09-26 Ptc Therapeutics, Inc. Compounds for treating spinal muscular atrophy
US20130281396A1 (en) * 2012-04-19 2013-10-24 Rvx Therapeutics Inc. Treatment of diseases by epigenetic regulation
EP2906531B1 (en) 2012-10-15 2018-01-17 Resverlogix Corp. Compounds useful in the synthesis of benzamide compounds
WO2014080291A2 (en) 2012-11-21 2014-05-30 Rvx Therapeutics Inc. Biaryl derivatives as bromodomain inhibitors
US9073878B2 (en) 2012-11-21 2015-07-07 Zenith Epigenetics Corp. Cyclic amines as bromodomain inhibitors
US9775915B2 (en) 2012-11-26 2017-10-03 President And Fellows Of Harvard College Trioxacarcins, trioxacarcin-antibody conjugates, and uses thereof
JP2016507496A (en) 2012-12-21 2016-03-10 ゼニス・エピジェネティクス・コーポレイションZenith Epigenetics Corp. Novel heterocyclic compounds as bromodomain inhibitors
CA2915622C (en) 2013-06-21 2020-08-18 Zenith Epigenetics Corp. Novel substituted bicyclic compounds as bromodomain inhibitors
EP3010503B1 (en) 2013-06-21 2020-03-11 Zenith Epigenetics Ltd. Novel bicyclic bromodomain inhibitors
CA2919948C (en) 2013-07-31 2020-07-21 Zenith Epigenetics Corp. Novel quinazolinones as bromodomain inhibitors
US9611223B2 (en) 2013-09-11 2017-04-04 Institute Of Cancer Research: Royal Cancer Hospital (The) 3-aryl-5-substituted-isoquinolin-1-one compounds and their therapeutic use
CN104146998A (en) * 2014-08-05 2014-11-19 广东食品药品职业学院 Novel application of 5-hydroxy-7-methoxy chromone in medicine preparation
JO3589B1 (en) 2014-08-06 2020-07-05 Novartis Ag Protein kinase c inhibitors and methods of their use
CN105566306B (en) * 2014-10-14 2019-03-26 四川农业大学 A kind of application of chromocor derivative prepared and its neuraminidase is inhibited
EP3227280B1 (en) 2014-12-01 2019-04-24 Zenith Epigenetics Ltd. Substituted pyridines as bromodomain inhibitors
WO2016087936A1 (en) 2014-12-01 2016-06-09 Zenith Epigenetics Corp. Substituted pyridinones as bromodomain inhibitors
EP3230277B1 (en) 2014-12-11 2019-09-18 Zenith Epigenetics Ltd. Substituted heterocycles as bromodomain inhibitors
EP3233846A4 (en) 2014-12-17 2018-07-18 Zenith Epigenetics Ltd. Inhibitors of bromodomains
WO2016147053A1 (en) 2015-03-13 2016-09-22 Resverlogix Corp. Compositions and therapeutic methods for the treatment of complement-associated diseases
EP4119141A1 (en) 2015-06-12 2023-01-18 Axovant Sciences GmbH Nelotanserin for the prophylaxis and treatment of rem sleep behavior disorder
CN108472285A (en) 2015-07-15 2018-08-31 阿速万科学有限责任公司 Diaryl for preventing and treating illusion associated with neurodegenerative disease and aryl heteroaryl urea derivative
EP3348548A4 (en) 2015-09-07 2019-04-03 Zhejiang Huahai Pharmaceutical Co., Ltd Nitric oxide-releasing prodrug molecule
BR112019001459A2 (en) 2016-07-27 2019-05-07 Hartis-Pharma Sa combination, pharmaceutical composition, and method of preventing and / or treating a disease or disorder.
JP7076432B2 (en) 2016-09-09 2022-05-27 インサイト・コーポレイション Pyrazolopyridine derivatives as HPK1 regulators and their use for the treatment of cancer
TW201811799A (en) 2016-09-09 2018-04-01 美商英塞特公司 Pyrazolopyrimidine compounds and uses thereof
WO2018049214A1 (en) 2016-09-09 2018-03-15 Incyte Corporation Pyrazolopyridine derivatives as hpk1 modulators and uses thereof for the treatment of cancer
KR101935793B1 (en) * 2016-09-30 2019-01-08 (주)나노믹스 Stilbene derivatives and preparation method thereof
KR20180036522A (en) * 2016-09-30 2018-04-09 (주)나노믹스 Stilbene derivatives and preparation method thereof
EP3554503B1 (en) * 2016-12-16 2023-10-04 The Board of Regents of The University of Texas System Inhibitors of bromodomain-containing protein 4 (brd4)
US20180228786A1 (en) 2017-02-15 2018-08-16 Incyte Corporation Pyrazolopyridine compounds and uses thereof
EP3638801A4 (en) 2017-07-26 2021-03-24 Yacyshyn, Vincent Removing polyphenol contaminants from feedstock-based polyphenols
US10722495B2 (en) 2017-09-08 2020-07-28 Incyte Corporation Cyanoindazole compounds and uses thereof
PL3755703T3 (en) 2018-02-20 2022-11-07 Incyte Corporation N-(phenyl)-2-(phenyl)pyrimidine-4-carboxamide derivatives and related compounds as hpk1 inhibitors for treating cancer
US10745388B2 (en) 2018-02-20 2020-08-18 Incyte Corporation Indazole compounds and uses thereof
WO2019164847A1 (en) 2018-02-20 2019-08-29 Incyte Corporation Indazole compounds and uses thereof
US11299473B2 (en) 2018-04-13 2022-04-12 Incyte Corporation Benzimidazole and indole compounds and uses thereof
AU2019263176A1 (en) * 2018-04-30 2020-10-08 The Trustees Of Indiana University Compounds for modulating DDAH and ADMA levels, as well as methods of using thereof to treat disease
US10899755B2 (en) 2018-08-08 2021-01-26 Incyte Corporation Benzothiazole compounds and uses thereof
MA53726A (en) 2018-09-25 2022-05-11 Incyte Corp PYRAZOLO[4,3-D]PYRIMIDINE COMPOUNDS AS ALK2 AND/OR FGFR MODULATORS
US11173187B2 (en) 2018-11-13 2021-11-16 Immortazyme Company Ltd. Concentrated oil-based polyphenol composition and a method of producing the oil-based polyphenol composition
CR20220097A (en) 2019-08-06 2022-06-01 Incyte Corp Solid forms of an hpk1 inhibitor
US11691971B2 (en) 2020-06-19 2023-07-04 Incyte Corporation Naphthyridinone compounds as JAK2 V617F inhibitors
US11753413B2 (en) 2020-06-19 2023-09-12 Incyte Corporation Substituted pyrrolo[2,1-f][1,2,4]triazine compounds as JAK2 V617F inhibitors
AU2021300429A1 (en) 2020-07-02 2023-02-16 Incyte Corporation Tricyclic urea compounds as JAK2 V617F inhibitors
US11767323B2 (en) 2020-07-02 2023-09-26 Incyte Corporation Tricyclic pyridone compounds as JAK2 V617F inhibitors
US11661422B2 (en) 2020-08-27 2023-05-30 Incyte Corporation Tricyclic urea compounds as JAK2 V617F inhibitors
CN114133336B (en) * 2020-09-03 2024-04-02 沈阳药科大学 4-methoxy naphthalene substituted aniline compound and preparation method and application thereof
EP4213820A1 (en) * 2020-09-17 2023-07-26 Iaterion, Inc. Methods and compositions for treating viral infections
WO2022140231A1 (en) 2020-12-21 2022-06-30 Incyte Corporation Deazaguaine compounds as jak2 v617f inhibitors
KR20220162642A (en) 2021-06-01 2022-12-08 주식회사 에즈큐리스 Novel fluoro-substituted flavonoid derivative and pharmaceutical composition for preventing or treating allergic diseases comprising the same
WO2023059802A1 (en) * 2021-10-07 2023-04-13 Rutgers, The State University Of New Jersey Flavonoid derivatives with gabaa receptor activity and methods of use
CN114315780B (en) * 2021-10-14 2024-01-26 山东明化新材料有限公司 Preparation method of 4-hydroxymethylcoumarin compound
CN114394908B (en) * 2022-01-11 2024-03-01 河北师范大学 Method for preparing 2-hydroxy-3-aminoacetophenone
WO2023141469A2 (en) * 2022-01-18 2023-07-27 Tessellate Therapeutics, Inc. Therapeutic compounds and methods of use thereof
CN115504970B (en) * 2022-10-18 2023-04-18 黑龙江中医药大学 Flavonoid derivative for treating myocardial ischemia and preparation method thereof

Family Cites Families (154)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2065593A (en) 1936-12-29 Water-soluble diazoimino com
FR472489A (en) 1914-02-20 1914-12-08 Stas Motor Ges M B H Metal seal ring for pistons
FR803619A (en) 1935-03-23 1936-10-05 Ig Farbenindustrie Ag Dihydroxystilbene-dicarboxylic acid and its preparation process
US2065900A (en) 1935-03-23 1936-12-29 Gen Aniline Works Inc Dihydroxystilbene-dicarboxylic acid and a process of preparing it
DE637259C (en) 1935-03-24 1936-10-27 I G Farbenindustrie Akt Ges Process for the preparation of a dioxystilbene dicarboxylic acid
FR803201A (en) 1935-07-08 1936-09-25 Ste Ind Chim Bale Preparation of sulfonic acids
US2071329A (en) 1935-08-22 1937-02-23 Solvay Process Co Method of recovering phthalic anhydride
DE652772C (en) 1935-11-07 1937-11-08 I G Farbenindustrie Akt Ges Process for the preparation of N-dihydroazines of the anthraquinone series
GB728767A (en) 1951-10-12 1955-04-27 Wander Ag Dr A 2-substituted chromone compounds, and a method of making same
US3251837A (en) 1962-09-14 1966-05-17 Pfizer & Co C Derivatives of 1, 2, 4-benzothiadiazine-1, 1-dioxides
GB1179019A (en) 1967-05-23 1970-01-28 Produits Chimique Soc Et Polynicotinic Esters of Flavonoids
FR6928M (en) 1967-11-24 1969-05-05
US3600394A (en) 1968-05-17 1971-08-17 Searle & Co 2-aminoalkyl-3-arylisocarbostyrils
US3773946A (en) 1969-09-02 1973-11-20 Parke Davis & Co Triglyceride-lowering compositions and methods
US3930024A (en) 1969-09-02 1975-12-30 Parke Davis & Co Pharmaceutical compositions and methods
FR2244493A1 (en) 1973-08-09 1975-04-18 Pluripharm Flavonoid amino-acid salts - for treatment of haemorrhage, circulatory disorders and atherosclerosis
DE2349024A1 (en) 1973-09-26 1975-04-10 Schering Ag 6BETA, 7BETA-EPOXY-1ALPHA, 2ALPHAMETHYLENE-D-HOMO-4-PREGNEN-3,20-DIONE
IL64542A0 (en) 1981-12-15 1982-03-31 Yissum Res Dev Co Long-chain alpha,omega-dicarboxylic acids and derivatives thereof and pharmaceutical compositions containing them
JPS60136512A (en) 1983-12-26 1985-07-20 Eisai Co Ltd Remedy and preventive for hyperlipemia
DE3423166A1 (en) 1984-06-22 1986-01-02 Epis S.A., Zug ALPHA, OMEGA DICARBONIC ACIDS, METHOD FOR THE PRODUCTION THEREOF AND MEDICINAL PRODUCTS CONTAINING THESE COMPOUNDS
EP0182213B1 (en) 1984-11-08 1990-09-26 Sumitomo Pharmaceuticals Company, Limited Carbapenem compounds and production thereof
DE3515882A1 (en) 1985-05-03 1986-11-06 Dr. Karl Thomae Gmbh, 7950 Biberach MEDICINAL PRODUCTS CONTAINING PYRIDINONE WITH ANTITHROMBOTIC EFFECTS AND METHOD FOR THE PRODUCTION THEREOF
DE3601417A1 (en) 1986-01-20 1987-07-23 Nattermann A & Cie 2'-Alkyl(alkenyl)-substituted quercetins
EP0258190B1 (en) 1986-08-29 1991-11-27 Ciba-Geigy Ag Process for preparing aromatic ethers and thioethers
ES2053508T3 (en) 1986-11-24 1994-08-01 Fujisawa Pharmaceutical Co COMPOUNDS OF 3-PIRROLIDINYL-THIO-1-AZABICICLO (3.2.0) HEPT-2-ENO-2-CARBOXILICOS.
US4925838A (en) 1988-03-18 1990-05-15 Fujisawa Pharmaceutical Company, Ltd. 3-pyrrolidinylthio-1-azabicyclo[3.2.0]-hept-2-ene-2-carboxylic acid compounds
US4963544A (en) 1988-05-23 1990-10-16 Fujisawa Pharmaceutical Company, Ltd. 3-pyrrolidinylthio-1-azabicyclo[3.2.0]-hept-2-ene-2-carboxylic acid compounds
GB8926981D0 (en) 1988-12-23 1990-01-17 Ici Plc Heterocyclic derivatives
AU6062590A (en) 1989-07-07 1991-02-06 Schering Corporation Pharmaceutically active compounds
IE64358B1 (en) 1989-07-18 1995-07-26 Ici Plc Diaryl ether heterocycles
GB9018134D0 (en) 1989-09-29 1990-10-03 Ici Plc Heterocyclic derivatives
WO1991018901A1 (en) 1990-06-05 1991-12-12 Toray Industries, Inc. Indole derivative
JP2999579B2 (en) 1990-07-18 2000-01-17 武田薬品工業株式会社 DNA and its uses
IE913866A1 (en) 1990-11-28 1992-06-03 Ici Plc Aryl derivatives
US5126351A (en) 1991-01-24 1992-06-30 Glaxo Inc. Antitumor compounds
MX9200299A (en) 1991-02-07 1992-12-01 Roussel Uclaf NEW NITROGENATED BICYCLE DERIVATIVES, THEIR PROCEDURE FOR PREPARING THE NEW INTERMEDIATE COMPOUNDS OBTAINED THEIR APPLICATION AS MEDICINES AND THE PHARMACEUTICAL COMPOSITIONS THAT CONTAIN THEM.
ATE159009T1 (en) 1991-05-10 1997-10-15 Rhone Poulenc Rorer Int BIS MONO- AND BICYCLIC ARYL AND HETEROARYL DERIVATIVES WITH AN INHIBITING EFFECT ON THE EGF AND/OR PDGF RECEPTOR TYROSINKINASE
US5480883A (en) 1991-05-10 1996-01-02 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
CN1067070A (en) 1991-05-17 1992-12-16 陆国强 The method for detoxication and fresh-keeping of beverage
US5124337A (en) 1991-05-20 1992-06-23 Schering Corporation N-acyl-tetrahydroisoquinolines as inhibitors of acyl-coenzyme a:cholesterol acyl transferase
PT100905A (en) 1991-09-30 1994-02-28 Eisai Co Ltd BICYCLE HYGIENEOUS HETEROCYCLIC COMPOUNDS CONTAINING BENZENE, CYCLOHEXAN OR PYRIDINE AND PYRIMIDINE, PYRIDINE OR IMIDAZOLE SUBSTITUTES AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
FR2689127B1 (en) 1992-03-31 1994-05-06 Adir Cie NEWS 3 ', 5' -DITERTBUTYL-4'-HYDROXY FLAVONES, THEIR PREPARATION PROCESS AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
DE4215587A1 (en) 1992-05-12 1993-11-18 Bayer Ag Sulfonylbenzyl-substituted benzo- and pyridopyridones
DE4215588A1 (en) 1992-05-12 1993-11-18 Bayer Ag Biphenylmethyl-substituted pyridones
GB9218334D0 (en) 1992-08-28 1992-10-14 Ici Plc Heterocyclic compounds
JPH0725761A (en) 1993-07-09 1995-01-27 Kureha Chem Ind Co Ltd Agent for protecting cartilage
US5756763A (en) 1993-07-23 1998-05-26 Zaidan Hojin Biseibutsu Kagaku Kenkyukai Pyrrolidine derivatives
CA2184101C (en) 1994-02-25 2005-11-22 Susumu Nakagawa Carbapenem derivatives
CA2184891C (en) 1994-03-08 2000-09-26 Yasuhisa Kuroki Phosphonic diester derivative
FR2718329B1 (en) 1994-03-21 2002-09-20 Rhone Poulenc Rorer Sa Transgenic rabbit sensitized to dyslipoproteinemias.
US6048903A (en) 1994-05-03 2000-04-11 Robert Toppo Treatment for blood cholesterol with trans-resveratrol
US6168776B1 (en) 1994-07-19 2001-01-02 University Of Pittsburgh Alkyl, alkenyl and alkynyl Chrysamine G derivatives for the antemortem diagnosis of Alzheimer's disease and in vivo imaging and prevention of amyloid deposition
GB2292149A (en) 1994-08-09 1996-02-14 Ferring Res Ltd Peptide inhibitors of pro-interleukin-1beta converting enzyme
AU711426B2 (en) * 1994-11-14 1999-10-14 Warner-Lambert Company 6-aryl pyrido(2,3-d)pyrimidines and naphthyridines for inhibiting protein tyrosine kinase mediated cellular proliferation
IL115256A0 (en) * 1994-11-14 1995-12-31 Warner Lambert Co 6-Aryl pyrido (2,3-d) pyrimidines and naphthyridines and their use
US5446071A (en) 1994-11-18 1995-08-29 Eli Lilly And Company Methods for lowering serum cholesterol
JP4140981B2 (en) 1994-12-26 2008-08-27 東菱薬品工業株式会社 Anti-restenosis and arteriosclerosis drug
US5648387A (en) 1995-03-24 1997-07-15 Warner-Lambert Company Carboxyalkylethers, formulations, and treatment of vascular diseases
ES2180702T3 (en) * 1995-06-07 2003-02-16 Lilly Co Eli TREATMENT OF PATHOLOGIES FOR THE INDUCTION OF THE BEF-1 TRANSCRIPTION FACTOR.
KR100263434B1 (en) 1995-08-30 2000-08-01 오쓰카 요시미쓰 Process for producing quinazolin-4-one derivatives
AU4858596A (en) 1995-09-15 1997-04-01 Torrey Pines Institute For Molecular Studies Synthesis of quinazolinone libraries
US5783577A (en) 1995-09-15 1998-07-21 Trega Biosciences, Inc. Synthesis of quinazolinone libraries and derivatives thereof
US5756736A (en) 1996-01-26 1998-05-26 Syntex (U.S.A.) Inc. Process for preparing a 2-(2-amino-1,6-dihydro-6-oxo-purin-9-yl)methoxy-1,3-propanediol derivative
US5739330A (en) 1996-02-05 1998-04-14 Hoechst Celanese Corporation Process for preparing quinazolones
WO1997028118A1 (en) 1996-02-05 1997-08-07 Hoechst Celanese Corporation Process for preparing anthranilic acids
US5763608A (en) 1996-02-05 1998-06-09 Hoechst Celanese Corporation Process for preparing pyrimidine derivatives
US5854264A (en) 1996-07-24 1998-12-29 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
KR100213895B1 (en) 1996-10-14 1999-08-02 박원훈 Compositions for the prevention and therapy of cardiovascular disease containing extract of citrus fruit peel hesperidin or naringin
DE19651099A1 (en) 1996-12-09 1998-06-10 Consortium Elektrochem Ind Multi-component system for changing, breaking down or bleaching lignin, lignin-containing materials or similar substances as well as methods for their use
AR012634A1 (en) 1997-05-02 2000-11-08 Sugen Inc QUINAZOLINE BASED COMPOUND, FAMACEUTICAL COMPOSITION THAT UNDERSTANDS IT, METHOD TO SYNTHESIZE IT, ITS USE, METHODS OF MODULATION OF THE DESERINE / TREONIN PROTEIN-KINASE FUNCTION AND IN VITRO METHOD TO IDENTIFY COMPOUNDS THAT MODULATE
US5908861A (en) 1997-05-13 1999-06-01 Octamer, Inc. Methods for treating inflammation and inflammatory disease using pADPRT inhibitors
US6635642B1 (en) 1997-09-03 2003-10-21 Guilford Pharmaceuticals Inc. PARP inhibitors, pharmaceutical compositions comprising same, and methods of using same
US6239114B1 (en) 1997-09-26 2001-05-29 Kgk Synergize Compositions and methods for treatment of neoplastic diseases with combinations of limonoids, flavonoids and tocotrienols
WO1999021548A1 (en) 1997-10-28 1999-05-06 Korea Institute Of Science And Technology Naringin and naringenin as inhibitor of acyl coa-cholesterol-o-acyltransferase, inhibitor of macrophage-lipid complex accumulation on the arterial wall and preventive or treating agent for hepatic diseases
DE19756388A1 (en) 1997-12-18 1999-06-24 Hoechst Marion Roussel De Gmbh New 2-aryl-4-amino-6,7-di:methoxy-quinazoline derivatives useful as guanylate cyclase activators for treating cardiovascular diseases, etc.
ATE302267T1 (en) 1998-01-08 2005-09-15 Aventis Pharma Inc TRANSGENIC RABBIT THAT EXPRESSES A FUNCTIONAL HUMAN LIPOPROTEIN(A).
US6414037B1 (en) 1998-01-09 2002-07-02 Pharmascience Pharmaceutical formulations of resveratrol and methods of use thereof
US6022901A (en) 1998-05-13 2000-02-08 Pharmascience Inc. Administration of resveratrol to prevent or treat restenosis following coronary intervention
CN1326450A (en) 1998-09-24 2001-12-12 三菱化学株式会社 Hydroxyflavone derivatives as tau protein kinase inhibitors
EP1123702A1 (en) 1998-10-19 2001-08-16 Eisai Co., Ltd. Analgesics
CN1327384A (en) * 1998-10-20 2001-12-19 韩国科学技术研究院 Bioflavonoids as plasma high density lipoprotein level increasing agent
US6392349B1 (en) * 1998-10-30 2002-05-21 David B. Crenshaw Remote control test apparatus
AU2190700A (en) * 1998-12-17 2000-07-03 Tularik Inc. Tubulin-binding agents
US6291456B1 (en) 1998-12-30 2001-09-18 Signal Pharmaceuticals, Inc. Compounds and methods for modulation of estrogen receptors
US6969720B2 (en) 1999-03-17 2005-11-29 Amr Technology, Inc. Biaryl substituted purine derivatives as potent antiproliferative agents
US6054435A (en) 1999-03-19 2000-04-25 Abbott Laboratories 6-O-substituted macrolides having antibacterial activity
DE19934799B4 (en) 1999-07-28 2008-01-24 Az Electronic Materials (Germany) Gmbh Chiral smectic liquid crystal mixture and its use in high contrast active matrix displays
JP2001131151A (en) * 1999-11-02 2001-05-15 Shionogi & Co Ltd New use of olefin derivative
JP5278983B2 (en) 1999-11-17 2013-09-04 塩野義製薬株式会社 New uses of amide compounds
ATE444960T1 (en) 1999-12-06 2009-10-15 Welichem Biotech Inc POLYHYDROXYSTILBENES AND STILBENOXIDES AS ANTIPSORIASIS AGENTS AND PROTEIN KINASE INHIBITORS
FR2804679B1 (en) 2000-02-07 2002-04-26 Clariant France Sa NOVEL PHENOLIC COMPOUNDS DERIVED FROM DIALCOXYETHANALS, THEIR PREPARATION PROCESS AND THEIR APPLICATION
EP1183224B1 (en) 2000-02-17 2004-08-04 Appleton Papers Inc. Process for preparing alkoxy or arylmethoxy aroxyethanes
CA2407593C (en) 2000-04-27 2011-01-11 Yamanouchi Pharmaceutical Co. Ltd. Fused heteroaryl derivatives
US6548694B2 (en) 2000-05-23 2003-04-15 Hoffman-La Roche Inc. N-(4-carbamimidoyl-phenyl)-glycine derivatives
JP2001335476A (en) 2000-05-29 2001-12-04 Shionogi & Co Ltd New use of tricyclic compound
US6541522B2 (en) 2000-08-16 2003-04-01 Insmed Incorporated Methods of using compositions containing hypotriglyceridemically active stilbenoids
CA2356544C (en) * 2000-10-03 2006-04-04 Warner-Lambert Company Pyridotriazines and pyridopyridazines
WO2002028835A1 (en) 2000-10-05 2002-04-11 Fujisawa Pharmaceutical Co., Ltd. Benzamide compounds as apo b secretion inhibitors
WO2002030882A2 (en) 2000-10-11 2002-04-18 Esperion Therapeutics, Inc. Sulfoxide and bis-sulfoxide compounds and compositions for cholesterol management and related uses
EP1363879A2 (en) 2000-10-11 2003-11-26 Esperion Therapeutics Inc. Sulfide and disulfide compounds and compositions for cholesterol management and related uses
AU2002222566A1 (en) 2000-11-30 2002-06-11 Canon Kabushiki Kaisha Luminescent element and display
RU2003120061A (en) 2000-12-07 2004-11-27 Си Ви Терапьютикс, Инк. (Us) SUBSTITUTED 1,3,5-TRIAZINES AND PYRIMIDINES AS INCREASING ABC-1 COMPOUNDS AGAINST DISEASES OF THE CORONARY ARTERY OR ATHEROSCLEROSIS
KR100472694B1 (en) 2000-12-30 2005-03-07 한국생명공학연구원 Flavanone derivatives and composition for preventing or treating blood lipid level-related diseases comprising same
JP2002249483A (en) 2001-02-21 2002-09-06 Koei Chem Co Ltd Method of producing aryl substituted heterocyclic compound
EP1385501A2 (en) 2001-04-11 2004-02-04 Atherogenics, Inc. Probucol monoesters and their use to increase plasma hdl cholesterol levels and improve hdl functionality
WO2003007959A1 (en) * 2001-07-16 2003-01-30 Fujisawa Pharmaceutical Co., Ltd. Quinoxaline derivatives which have parp inhibitory action
JP2005504047A (en) 2001-08-13 2005-02-10 チバ スペシャルティ ケミカルズ ホールディング インコーポレーテッド UV absorber
US7429593B2 (en) * 2001-09-14 2008-09-30 Shionogi & Co., Ltd. Utilities of amide compounds
US8124625B2 (en) 2001-09-14 2012-02-28 Shionogi & Co., Ltd. Method of enhancing the expression of apolipoprotein AI using olefin derivatives
US20040235877A1 (en) * 2001-09-14 2004-11-25 Natsuki Ishizuka Novel use of tricyclic compound
US6835469B2 (en) 2001-10-17 2004-12-28 The University Of Southern California Phosphorescent compounds and devices comprising the same
US6541045B1 (en) 2002-01-04 2003-04-01 Nutraceutical Corporation Herbal composition and method for combating inflammation
DE60328461D1 (en) 2002-01-28 2009-09-03 Ube Industries PROCESS FOR PREPARING A CHINAZOLIN-4-ONDERIVATE
US7904822B2 (en) * 2002-04-23 2011-03-08 Draeger Medical Systems, Inc. System and user interface for configuring and presenting a trend indicative display of patient medical parameters
MY140680A (en) 2002-05-20 2010-01-15 Bristol Myers Squibb Co Hepatitis c virus inhibitors
TW200401770A (en) 2002-06-18 2004-02-01 Sankyo Co Fused-ring pyrimidin-4(3H)-one derivatives, processes for the preparation and uses thereof
KR20040001144A (en) 2002-06-27 2004-01-07 김대경 Novel phospholipase A2 in the cytosol of red blood cell and the antibody against it, and the use and the preparation methods thereof
US20040033480A1 (en) 2002-08-15 2004-02-19 Wong Norman C.W. Use of resveratrol to regulate expression of apolipoprotein A1
US20050080024A1 (en) 2002-08-15 2005-04-14 Joseph Tucker Nitric oxide donating derivatives for the treatment of cardiovascular disorders
US20050080021A1 (en) * 2002-08-15 2005-04-14 Joseph Tucker Nitric oxide donating derivatives of stilbenes, polyphenols and flavonoids for the treatment of cardiovascular disorders
EP1407774A1 (en) 2002-09-10 2004-04-14 LION Bioscience AG 2-Amino-4-quinazolinones as LXR nuclear receptor binding compounds
EP1398032A1 (en) 2002-09-10 2004-03-17 PheneX Pharmaceuticals AG 4-Oxo-quinazolines as LXR nuclear receptor binding compounds
AU2003301662A1 (en) 2002-10-21 2004-05-13 Bristol-Myers Squibb Company Quinazolinones and derivatives thereof as factor xa inhibitors
WO2004039795A2 (en) * 2002-10-29 2004-05-13 Fujisawa Pharmaceutical Co., Ltd. Amide compounds for the treatment of hyperlipidemia
EP1418164A1 (en) 2002-11-07 2004-05-12 Institut National De La Sante Et De La Recherche Medicale (Inserm) New stilbene derivatives and their use as aryl hydrocarbon receptor ligand antagonists
MXPA05005028A (en) 2002-11-18 2005-08-03 Hoffmann La Roche Diazinopyrimidines.
EP1562956A4 (en) * 2002-11-22 2007-10-10 Japan Tobacco Inc Fused bicyclic nitrogen-containing heterocycles
RU2340604C2 (en) 2002-12-13 2008-12-10 Ф.Хоффманн-Ля Рош Аг 3h-quinazolin-4-on derivatives and medication, possessing properties of specific monoaminooxidase b inhibitor
ITRM20020629A1 (en) * 2002-12-19 2004-06-20 Sigma Tau Ind Farmaceuti USE OF ALPHA-PHENYLTHIOCARBOXYL AND ACHYLPHOXYCARBOXYLIC ACIDS WITH HYPOGLYCEMY AND / OR HYPOLIPIDEMIZING ACTIVITY.
ES2361924T3 (en) * 2002-12-20 2011-06-24 X-Ceptor Therapeutics, Inc. DERIVATIVES OF ISOQUINOLINONA AND ITS USE AS THERAPEUTIC AGENTS.
JP2004203751A (en) 2002-12-24 2004-07-22 Pfizer Inc Substituted 6,6-heterobicyclic derivative
EP1577288B1 (en) 2002-12-26 2014-07-23 Eisai R&D Management Co., Ltd. Selective estrogen receptor modulators
WO2004065392A1 (en) 2003-01-24 2004-08-05 Smithkline Beecham Corporation Condensed pyridines and pyrimidines and their use as alk-5 receptor ligands
WO2004072042A2 (en) * 2003-02-12 2004-08-26 Carex S.A. Quinoline derivative and their use for modulation of lxr activity
JP2006522124A (en) 2003-04-03 2006-09-28 バーテックス ファーマシューティカルズ インコーポレイテッド Compositions useful as inhibitors of protein kinases
WO2004092196A2 (en) 2003-04-09 2004-10-28 Exelixis, Inc. Tie-2 modulators and methods of use
JP2004307440A (en) 2003-04-10 2004-11-04 Kyorin Pharmaceut Co Ltd 2-amino-1,3-propanediol derivative and its addition salt
ATE422895T1 (en) 2003-04-16 2009-03-15 Bristol Myers Squibb Co MACROCYCLIC ISOQUINOLINE PEPTIDE INHIBITORS OF HEPATITIS C VIRUS
WO2004113307A1 (en) 2003-06-18 2004-12-29 Ube Industries, Ltd. Process for producing pyrimidin-4-one compound
US20050043300A1 (en) 2003-08-14 2005-02-24 Pfizer Inc. Piperazine derivatives
JP2007517071A (en) 2003-12-19 2007-06-28 メルク エンド カムパニー インコーポレーテッド Mitotic kinesin inhibitor
WO2005066162A1 (en) 2003-12-23 2005-07-21 Human Biomolecular Research Institute Synthetic compounds and derivatives as modulators of smoking or nicotine ingestion and lung cancer
TW200536830A (en) 2004-02-06 2005-11-16 Chugai Pharmaceutical Co Ltd 1-(2H)-isoquinolone derivative
WO2005115993A1 (en) 2004-05-31 2005-12-08 Banyu Pharmaceutical Co., Ltd. Quinazoline derivative
WO2006012577A2 (en) 2004-07-22 2006-02-02 Bayer Pharmaceuticals Corporation Quinazolinone derivatives useful for the regulation of glucose homeostasis and food intake
US20070218155A1 (en) * 2004-08-20 2007-09-20 Kuhrts Eric H Methods and compositions for treating dyslipidaemia
WO2006045010A2 (en) 2004-10-20 2006-04-27 Resverlogix Corp. Stilbenes and chalcones for the prevention and treatment of cardiovascular diseases
KR20060079121A (en) 2004-12-31 2006-07-05 에스케이케미칼주식회사 Quinazoline derivatives for the treatment or prevention of diabetes and obesity
CR9465A (en) 2005-03-25 2008-06-19 Surface Logix Inc PHARMACOCINETICALLY IMPROVED COMPOUNDS
EP1909788A2 (en) 2005-07-29 2008-04-16 Resverlogix Corp. Pharmaceutical compositions for the prevention and treatment of complex diseases and their delivery by insertable medical devices
EP1968968A1 (en) 2005-12-21 2008-09-17 PainCeptor Pharma Corp. Compositions and methods for modulating gated ion channels
EP2005941A3 (en) 2007-06-01 2009-04-01 Henkel AG & Co. KGaA Cellular rejuvenation compounds
PL2346837T3 (en) 2008-06-26 2015-07-31 Resverlogix Corp Methods of preparing quinazolinone derivatives
US8952021B2 (en) 2009-01-08 2015-02-10 Resverlogix Corp. Compounds for the prevention and treatment of cardiovascular disease

Also Published As

Publication number Publication date
US20090259048A1 (en) 2009-10-15
EP2332527A2 (en) 2011-06-15
US8093273B2 (en) 2012-01-10
US7846915B2 (en) 2010-12-07
US20060205767A1 (en) 2006-09-14
CA2584485A1 (en) 2006-04-27
US8242144B2 (en) 2012-08-14
WO2006045010A2 (en) 2006-04-27
US20060205792A1 (en) 2006-09-14
US20110082176A1 (en) 2011-04-07
WO2006045096A3 (en) 2007-08-02
EP1838296B1 (en) 2012-08-08
WO2006045096A2 (en) 2006-04-27
CA2584507A1 (en) 2006-04-27
WO2006045010A3 (en) 2007-05-31
CA2584507C (en) 2016-04-26
US8242130B2 (en) 2012-08-14
EP1838296A2 (en) 2007-10-03
EP2332527A3 (en) 2011-11-16

Similar Documents

Publication Publication Date Title
CA2584485C (en) Stilbenes and chalcones for the prevention and treatment of cardiovascular diseases
DK2118074T3 (en) Compounds for the prevention and treatment of cardiovascular diseases
JP5368792B2 (en) Pharmaceutical compositions for the prevention and treatment of complex diseases and their delivery by insertable medical devices
MXPA06003839A (en) Nitric oxide donating derivatives for the treatment of cardiovascular disorders.
JP5265691B2 (en) Ligustrazine aromatic acid ether compound, production method and drug compound and application
EP3835298A1 (en) Novel chroman derivatives having estrogen receptor degradation activity and uses thereof
US20050080021A1 (en) Nitric oxide donating derivatives of stilbenes, polyphenols and flavonoids for the treatment of cardiovascular disorders
WO2019105234A1 (en) Aromatic compound, pharmaceutical composition thereof and use thereof
US10377695B2 (en) Metabotropic glutamate receptor positive allosteric modulators (PAMS) and uses thereof
JP6199406B2 (en) Novel compounds as diacylglycerol acyltransferase inhibitors
EP3039014A1 (en) Substituted naphthalene compounds as calcium sensing receptor modulators
BR112017019816B1 (en) COMPOUND, PHARMACEUTICAL COMPOSITION, AND ITS USES
Li et al. Synthesis and Biological Evaluation of Scutellarein Alkyl Derivatives as Preventing Neurodegenerative Agents with Improved Lipid Soluble Properties
JPS60156610A (en) Arachidonic acid 5-lipoxygenase inhibitor

Legal Events

Date Code Title Description
EEER Examination request