CA2319495A1 - Multibinding inhibitors of microsomal triglyceride transferase protein - Google Patents

Multibinding inhibitors of microsomal triglyceride transferase protein Download PDF

Info

Publication number
CA2319495A1
CA2319495A1 CA002319495A CA2319495A CA2319495A1 CA 2319495 A1 CA2319495 A1 CA 2319495A1 CA 002319495 A CA002319495 A CA 002319495A CA 2319495 A CA2319495 A CA 2319495A CA 2319495 A1 CA2319495 A1 CA 2319495A1
Authority
CA
Canada
Prior art keywords
substituted
ligand
group
linker
cycloalkyl
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.)
Abandoned
Application number
CA002319495A
Other languages
French (fr)
Inventor
John H. Griffin
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.)
Innoviva Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2319495A1 publication Critical patent/CA2319495A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B30/00Methods of screening libraries
    • C40B30/04Methods of screening libraries by measuring the ability to specifically bind a target molecule, e.g. antibody-antigen binding, receptor-ligand binding
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/55Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/55Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • A61K47/552Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds one of the codrug's components being an antibiotic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/01Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C233/34Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by amino groups
    • C07C233/35Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by amino groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom
    • C07C233/36Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by amino groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom having the carbon atom of the carboxamide group bound to a hydrogen atom or to a carbon atom of an acyclic saturated carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/64Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings
    • C07C233/77Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by amino groups
    • C07C233/78Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by amino groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/24Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atom of at least one of the carboxamide groups bound to a carbon atom of a ring other than a six-membered aromatic ring of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/06Esters of carbamic acids
    • C07C271/08Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms
    • C07C271/10Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C271/20Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms to carbon atoms of hydrocarbon radicals substituted by nitrogen atoms not being part of nitro or nitroso groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C321/00Thiols, sulfides, hydropolysulfides or polysulfides
    • C07C321/02Thiols having mercapto groups bound to acyclic carbon atoms
    • C07C321/04Thiols having mercapto groups bound to acyclic carbon atoms of an acyclic saturated carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/10Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C323/11Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and singly-bound oxygen atoms bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C323/12Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and singly-bound oxygen atoms bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being acyclic and saturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C335/00Thioureas, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups
    • C07C335/04Derivatives of thiourea
    • C07C335/06Derivatives of thiourea having nitrogen atoms of thiourea groups bound to acyclic carbon atoms
    • C07C335/08Derivatives of thiourea having nitrogen atoms of thiourea groups bound to acyclic carbon atoms of a saturated carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C335/00Thioureas, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups
    • C07C335/30Isothioureas
    • C07C335/32Isothioureas having sulfur atoms of isothiourea groups bound to acyclic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/30Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members
    • C07D207/32Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • C07D207/33Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms with substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • C07D207/333Radicals substituted by oxygen or sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/36Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D211/56Nitrogen atoms
    • C07D211/58Nitrogen atoms attached in position 4
    • 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
    • C07D215/48Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • C07D215/54Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen attached in position 3
    • C07D215/56Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen attached in position 3 with oxygen atoms in position 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/90Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/24Benzimidazoles; Hydrogenated benzimidazoles 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 in position 2
    • C07D235/30Nitrogen atoms not forming part of a nitro radical
    • 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
    • C07D401/06Heterocyclic 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 carbon chain containing only aliphatic carbon atoms
    • 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
    • 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
    • C07D401/14Heterocyclic 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 three or more hetero rings
    • 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
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms 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
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/26Heterocyclic compounds containing purine ring systems with an oxygen, sulphur, or nitrogen atom directly attached in position 2 or 6, but not in both
    • C07D473/32Nitrogen atom
    • C07D473/34Nitrogen atom attached in position 6, e.g. adenine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D475/00Heterocyclic compounds containing pteridine ring systems
    • C07D475/02Heterocyclic compounds containing pteridine ring systems with an oxygen atom directly attached in position 4
    • C07D475/04Heterocyclic compounds containing pteridine ring systems with an oxygen atom directly attached in position 4 with a nitrogen atom directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D475/00Heterocyclic compounds containing pteridine ring systems
    • C07D475/06Heterocyclic compounds containing pteridine ring systems with a nitrogen atom directly attached in position 4
    • C07D475/08Heterocyclic compounds containing pteridine ring systems with a nitrogen atom directly attached in position 4 with a nitrogen atom directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/06Peri-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D493/00Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
    • C07D493/02Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system in which the condensed system contains two hetero rings
    • C07D493/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D493/00Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
    • C07D493/02Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system in which the condensed system contains two hetero rings
    • C07D493/06Peri-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • C07H19/20Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/26Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving oxidoreductase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • C12Q1/44Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase involving esterase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/48Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving transferase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/533Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving isomerase
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/04Libraries containing only organic compounds
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/573Immunoassay; Biospecific binding assay; Materials therefor for enzymes or isoenzymes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6842Proteomic analysis of subsets of protein mixtures with reduced complexity, e.g. membrane proteins, phosphoproteins, organelle proteins
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6845Methods of identifying protein-protein interactions in protein mixtures
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/92Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving lipids, e.g. cholesterol, lipoproteins, or their receptors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2603/00Systems containing at least three condensed rings
    • C07C2603/02Ortho- or ortho- and peri-condensed systems
    • C07C2603/04Ortho- or ortho- and peri-condensed systems containing three rings
    • C07C2603/06Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members
    • C07C2603/10Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members containing five-membered rings
    • C07C2603/12Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members containing five-membered rings only one five-membered ring
    • C07C2603/18Fluorenes; Hydrogenated fluorenes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/04Endocrine or metabolic disorders
    • G01N2800/044Hyperlipemia or hypolipemia, e.g. dyslipidaemia, obesity
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S530/00Chemistry: natural resins or derivatives; peptides or proteins; lignins or reaction products thereof
    • Y10S530/807Hapten conjugated with peptide or protein

Abstract

Disclosed are multibinding compounds which inhibit microsomal triglyceride transferase protein (MTP), a protein which mediates the transfer of lipids during the assembly of lipoproteins and related biomolecules. The multibinding compounds contain from 2 to 10 ligands covalently attached to one or more linkers. The multibinding compounds of this invention are useful for lowering serum lipid, cholesterol and/or triglyceride levels, and for preventing and treating disorders associated with hyperlipemia, hyperlipidemia, hyperlipoproteinemia, hypercholestrolemia, hypertriglyceridemia and the like, such as atherosclerosis.

Description

MULTIBINDING INHIBITORS OF
MICROSOMAL TRIGLYCERiDE TRANSFERASE PROTEIN
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of U.S. Patent Application Serial No.
60/088,448, filed June 8, 1998; and U.S. Patent Application Serial No.
60/093,072, filed July 16, 1998; the disclosures of which are incorporated herein by reference in their entirety.
BACKGROUND OF THE INVENTION
Field of the Invention This invention relates to novel multibinding compounds (agents) that inhibit microsomal triglyceride transferase protein (MTP) and to pharmaceutical compositions comprising such compounds. Accordingly, the multibinding compounds and pharmaceutical compositions of this invention are useful in preventing and treating various disorders associated with hyperlipidemia and related disorders, such as atherosclerosis.
References The following publications are cited in this application as superscript numbers:
' J. R. Wetterau et al., Biochim. Biophys. Acta 1997, 1345, 136-150.

D.A. Gordon et al., Trends Cell Biol. 1995, 5, 317-321.

__2__ Robbins Pathological Basis of Disease, S" Edition (1994), pp.

473-484.

" S. L. Ohringer et al., Acta Crystallogr., Sect.
D: Biol.

S Crystallogr. 1996, D52(1), 224-225.

J. R. Wetterau et al., J. Biol. Chem. 1990, 265, 9800-9807.

J. R. Wetterau et al., Biochemistry 1991, 30, 4406-4412.

' C.C. Shoulders et al., J. Hum. Mol. Genet. 1993, 2, 2109-2116.

R. Raag et al., J. Mol. Biol. 1988, 200, 553-569.

15 9 P. A. Timmins et al., Science 1992, 257, 652-655.

' A. Atzel et al., Biochemistry 1993, 32, 10444-10450.

" A. Atzel et al., Biochemistry 1994, 33, 15382-15388.

12 H. Jamil et al., J. Biol. Chem. 1995, 270, 6549-6554.

'3 H. Jamil et al., Proc. Natl. Acad. Sci. U.S.A.
1996, 93, 11991-11995.

'a J. p. Kane et al., The Metabolic Basis of Inherited Disease, Scriver et al., Eds.; McGraw-Hill, NY, Ed. 7, 1995, pp. 1853-1885.

30 'S M. F. Linton et al., J. Lipid Res. 1993, 34, 521-541.

U.S. Patent No. 5,712,279, issued January 27, 1998 to Biller et al.

35 " U.S. Patent No. 5,739,135, issued April 14, 1998 to Biller et al.

1g U.S. Patent No. 5,760,246, issued June 2, 1998 to Biller et al.

'9 U.S. Patent No. 5,827,875, issued October 27, 1998 to Dickson 40 Jr. et al.

xo U.S. Statutory Invention Registration No. H1729, published May 5, 1998 by Biller et al.

__3__ z' WO 96/40640, published December 19, 1996.
zz WO 97/26240, published July 24, 1997.
z3 WO 97/43255, published November 20, 1997.
za Wp 98/03069, published January 29, 1998.
a WO 98/03174, published January 29, 1998.
26 WO 98/23593, published June 4, 1998.
z' WO 98/27979, published July 2, 1998.
z8 WO 98/31225, published July 23, 1998.
z9 WO 98/31366, published July 23, 1998.
so WO 98/31367, published July 23, 1998.

EP 0 643 057 A1, published March 15, 1995.
3z M. Haghpassand et al., J. Lipid Res. 1996, 37, 1468-1480.
33 F. Benoist et al. , Eur. J. Biochem. 1996, 240, 713-720.
3a J. R. Wetterau et al., Science 1998, 282, 751-754.
30 AlI of the above publications are herein incorporated by reference in their entirety to the same extent as if each individual publication was specifically and individually indicated to be incorporated by reference in its entirety.
State of the Art 35 Microsomal triglyceride transferase protein (MTP) is a lipid transfer protein which mediates the transport of lipids, such as triglycerides, cholesterol esters, and phosphatidylcholine, between membranes.'~z Accordingly, MTP is believed to play a role in the assembly of lipoproteins and related biomolecules.
In this regard, MTP has been implicated as a probable agent in the assembly of WO_99/63929 PCT/US99/11789 _.-4-_ apolipoprotein B (Apo B)-containing lipoproteins which are known to contribute to the formation of atherosclerotic lesions. Thus, effective inhibitors of MTP
would be useful in preventing the onset and progression of atherosclerosis, including myocardial infarction, stroke, peripheral vascular disease and the like, 5 which accounts for one-half of deaths in the United States 3 MTP was originally isolated from the microsomal fraction of bovine liver and has subsequently been found within the lumen of microsomes isolated from both the liver and intestine.' Since its initial isolation, MTP has been extensively 10 characterized.'.5.6 MTP is a soluble, heterodimeric protein composed of 58 and 97 kDa subunits, both of which are required for activity. The protein is localized within the lumen of the endoplasmic reticulum. The 58 kDa subunit is identical to protein disulfide isomerase (PDI), though the complex exhibits no PDI
activity and isolated PDI does not exhibit MTP activity. The noncovalent MTP
15 heterodimer does not display significant dissociation/reassociation and is either asymmetric and/or highly hydrated. The unique 97 kDa subunit bears homology to other lipid-transporting proteins, including the lipovitillin-phosvitin complex (LPC) and, to a lesser extent, plasma cholesteryl ester transfer protein (CETP).' Structural characterization of LPC reveals that it comprises a large cavity that 20 complexes multiple copies of phospholipid.8.9 Kinetic analysis of the MTP-mediated lipid transport processes have revealed ping pong bi bi kinetics which is consistent with a mechanism of action in which MTP binds and shuttles lipid molecules between membranes.'°
This 25 suggests that stable MTP-lipid complexes are formed during the transfer process, which is further supported by the observation that incubation of MTP with donor vesicles containing a variety of radio-labeled lipids followed by re-isolation affords MTP containing up to three molecules of lipid.".'z The ability of lipid molecules to occupy distinct binding sites on MTP is suggested by the 30 observation of biphasic kinetics for transfer of phosphatidyl choline, which binds with a 2:1 stoichiometry to the enzyme.l2 Moreover, an MTP inhibitor has been shown to fully ablate the MTP-mediated transfer of triglycerides and cholesterol esters but not that of phosphatidyl choline. '3 5 The ability of MTP inhibitors to prevent the onset and progression of atherosclerosis and related disorders is supported by the observation that mutations in MTP are the only known bases for abetalipoproteinemia, an autosomal recessive disorder characterized by the virtual absence of apoB-containing plasma lipoproteins.l.z.~a A~~lipoproteinemia subjects are free of 10 atherosclerosis, but they suffer from a variety of side effects as a result of the extreme nature of their condition. This suggests that non-complete inhibition of MTP would be requisite in an agent designed for human therapy. In this regard, hypobetalipoproteinemia is a relevant model for MTP inhibition. This condition is displayed by individuals who are heterozygous for mutations in 15 apolipoproteinB.lS These subjects have levels of apoB-containing lipoproteins half that of normal subjects and, as a result, they enjoy extended lifespans.
Inhibitors of MTP have been described in the patent and technical literature. See, by way of example, U.S. Patent No. 5,712,279;'6 U.S. Patent 20 No. 5,739,135;1' U.S. Patent No. 5,760,246;'8 U.S. Patent No. 5,827,875;'9 U.S. Statutory Invention Registration No. H1729;2° WO 96/40640;21 WO
97/26240;22 WO 97/43255;23 WO 98/03069;2° WO 98/03174;25 WO 98/23593;26 WO 98/27979;2' WO 98/31225;28 WO 98/31366;29 WO 98/31367;'° EP
0 643 057 A1;31 M. Haghpassand et a1.;32 F. Benoist et a1.;33 and J. R.
Wetterau 25 et a1.3" Notwithstanding such inhibitors, a need exists for effective MTP
inhibitors having improved biological and/or therapeutic effects.
It has now been discovered that MTP inhibitors having surprising and unexpected properties can be prepared by linking from 2 to 10 ligands capable of 30 binding to MTP to one or more linkers. The chemical structure of one known __6__ inhibitor of MTP, i.e. BMS-201038, is illustrated in Fig. lA.'6.s4 Without being limited to theory, a potential complex of this compound with MTP is illustrated in Figure 1B. Based on the distinct multiple binding sites believed to be present in MTP, various multibinding compounds are illustrated in Figure 1B. Such 5 multibinding compounds provide greater biological and/or therapeutic effects than the aggregate of the unlinked ligands due to their multibinding properties.
SUMMARY OF THE INVENTION
This invention is directed to novel multibinding compounds (agents) that 10 inhibit microsomal triglyceride transferase protein (MTP). The multibinding compounds of this invention are useful in the prevention and treatment of diseases associated with hyperlipemia, hyperlipidemia, hyperlipoproteinemia, hypercholestrolemia, hypertriglyceridemia and the like, such as atherosclerosis.
15 Accordingly, in one of its composition aspects, this invention provides a multibinding compound of formula I:
(L)P(X)q I
20 wherein each X is independently a linker; p is an integer of from 2 to 10;
and q is an integer of from 1 to 20; and each L is independently a ligand selected from the group consisting of:

Rs R~ ~ - ~ N~ s OH)a -N~ I R IA
R2. ~--~ R4 O R3 Rs O
R,\
- ~ -(CH)a - ~ R5 IB
W-(CH)a RZ Rs R4 R~
R~ IC

O
e- C N ~ 5 R N ( R ID

R9- I -NI \ 5 IE
(CH)b I R
Ra wherein each W is a divalent radical independently selected from the group consisting of:
R~2 A A (CR~oR»)~ p' I
Rya Q

__g__ each R' is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and a covalent bond linking the ligand to a linker;
5 each RZ is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and a covalent bond linking the ligand to a linker;
each R3 is independently selected from the group consisting of hydrogen, 10 alkyl and halo;
each R' is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl and a covalent bond linking the ligand to a linker;
each RS is independently selected from the group consisting of alkyl, 15 substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloallcyl, cycloalkenyl, substituted cycloalkenyl, alkoxy, substituted alkoxy, cycloalkoxy, substituted cycloalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, amino, substituted amino and a covalent bond linking the ligand to a linker; or R4 and RS may be joined, together with the > NC(O)-20 group to which they are attached, to form a heterocyclic ring;
each R6 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and a covalent bond linking the ligand to a linker;
25 each R' is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, a covalent bond linking the ligand to a linker and -NR'°R'S, where R'4 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, 30 substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted __9__ cycloalkenyl, aryl and heteroaryl; and R'S is a covalent bond linking the ligand to a linker;
each R8 is independently selected from the group consisting of hydrogen, acyl, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted 5 cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, a covalent bond linking the ligand to a linker each R9 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, 10 a covalent bond linking the ligand to a linker and -NR'4R'S, where R'4 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl and heteroaryl; and R'S is a covalent bond linking the ligand to a linker;
15 each R'°, R", R'2 and R'3 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, alkoxy, substituted alkoxy, cycloalkoxy, substituted cycloalkoxy, aryl, aryloxy, halo, heteroaryl, heteroaryloxy, thioalkoxy, 20 substituted thioalkoxy, thioaryloxy, thioheteroaryloxy, acyloxy, aminoacyl, aminocarbonyl, -S(O)R'6 and -SOZR'6, where each R'6 is independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, aryl and heteroaryl;
each ring A, together with the atoms to which it is attached, forms a 25 carbocyclic or heterocyclic ring selected from the group consisting of aryl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, heteroaryl and heterocyclic;
each ring B, together with the atoms to which it is attached, forms a carbocyclic or heterocyclic ring selected from the group consisting of aryl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, heteroaryl and heterocyclic;
each ring C, together with the nitrogen atom to which it is attached, forms a heterocyclic ring;
each Q is independently selected from the group consisting of a covalent bond, -O-, -S-, -S(O)-, -SOZ , alkylene, substituted alkylene, alkenylene, substituted alkenylene and -NR"-, where R" is selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl and heteroaryl;
each a is independently an integer of from 2 to 6;
each b is independently an integer of from 0 to 6;
each c is independently an integer of from 2 to 4;
and pharmaceutically-acceptable salts or pro-drugs thereof;
provided that when p is 2, q is 1 and a first ligand has formula IA or IB, where R' or Rz is a covalent bond linking the first ligand to the linker, then a second ligand does not have formula ID or IE, where R8 or R9 are a covalent bond linking the second ligand to the linker.
Preferably q is less than p in the multibinding compounds of this invention.
Preferably, W in formulae IA, IB and IC is a divalent radical having the formula:
R~8 \ I \ R~s Q' wherein WO 99/63929 PCT/fJS99/11789 each R'8 and R'9 are independently selected from the group consisting of hydrogen or halo; and Q' is a covalent bond, -O- or -S-.
Preferably, each RS group is independently selected from the group consisting of aryl, heteroaryl, heterocyclic, cycloalkyl and substituted cycloalkyl.
Additionally, each RS group is preferably substituted with from 1 to 4 substituents and one of the substituents is attached to the ring atom in the position adjacent to the atom attached to the -C(O)- group. Still more preferably, each RS is a 2-(4'-trifluoromethylphenyl)phenyl group.
Preferably, ring C in formulae IA and ID forms a piperidine or a 1,2,3,4-tetrahydroisoquinoline ring.
In another of its composition aspects, this invention provides a multibinding compound of formula II:
L'-X'-L' II
wherein X' is a linker; and each L' is a ligand independently selected from the group consisting of:
R2o ~ ~ ~ O

Q" ~''~
- N N~ 25 '~ i 1..~)a I R IIA
R22 R2a R2~

WO_99/639Z9 PCT/US99/11789 R2o ~ ~ / O its N-CH2 CFs O
Q.. IIB
~( i H)a I ( i H)a f R
-N- -N' \ 25 R2o ~ ~ O N- _CF
CFi2 3 Q IIC
( i H)b-R2~

R2~
O
28-N N~ 25 R I R

O
R29- Cti -N' \ 25 (, )b I R
R2z R24 IIE

wherein each Rz° and Rz' is independently selected from the group consisting of hydrogen and halo;
each Rzz is independently selected from the group consisting of hydrogen, alkyl and halo;
each Rz' is independently selected from the group consisting of hydrogen and a covalent bond linking the ligand to the linker;
each Rz4 is independently selected from the group consisting of hydrogen and a covalent bond linking the ligand to the linker;
each Ru is independently selected from the group consisting of aryl, heteroaryl, heterocyclic, cycloalkyl, substituted cycloalkyl and a covalent bond linking the ligand to the linker;
each Rz6 is independently selected from the group consisting of hydrogen, alkyl and a covalent bond linking the ligand to the linker;
each Rz' is independently selected from the group consisting of hydrogen, a covalent bond linking the ligand to a linker and -NR3°R3', where R3° is selected from the group consisting of hydrogen and alkyl; and R3' is a covalent bond linking the ligand to a linker;
each Rz8 is a covalent bond linking the ligand to a linker;
each Rz9 is independently selected from the group consisting of a covalent bond linking the ligand to a linker and -NR3°R3', where R3° is selected from the group consisting of hydrogen and alkyl; and R3' is a covalent bond linking the ligand to a linker;
each Q ~ is independently selected from the group consisting of a covalent bond, -O- and -S-each a is independently an integer of from 2 to 6;
each b is independently an integer of from 0 to 6;
and pharmaceutically-acceptable salts or pro-drugs thereof;
provided that in each ligand only one of Rz3, Rza, Rzs, Rzb, Rz', Rza and Rz9 is a covalent bond linking the ligand to the linker;

and further provided that when a first ligand has formula IIA or IIB, where RZ' is a covalent bond linking the first ligand to the linker, then a second ligand does not have formula IID or IIE, where R28 or R29 are a covalent bond linking the second ligand to the linker.
Preferably, each R'~ group is independently selected from the group consisting of aryl, heteroaryl, heterocyclic, cycloalkyl and substituted cycloalkyl.
Additionally, each Ru group is preferably substituted with from 1 to 4 substituents and one of the substituents is attached to the ring atom in the position adjacent to the atom attached to the -C(O)- group. More preferably, each R~ is a 2-(4'-trifluoromethylphenyl)phenyl group.
Preferably, in the multibinding compounds of this invention, each linker (i.e., X or X') independently has the formula:
_Xa_Z-(ye-Z)m ye_Z_Xe_ wherein m is an integer of from 0 to 20;
Xa at each separate occurrence is selected from the group consisting of -O-, -S-, -NR-, -C(O)-, -C(O)O-, -C(O)NR-, -C(S), -C(S)O-, -C(S)NR- or a covalent bond where R is as defined below;
Z is at each separate occurrence is selected from the group consisting of alkylene, substituted alkylene, cycloalkylene, substituted cylcoalkylene, alkenylene, substituted alkenylene, alkynylene, substituted alkynylene, cycloalkenylene, substituted cycloalkenylene, arylene, heteroarylene, heterocyclene, or a covalent bond;
Ya and Ye at each separate occurrence are selected from the group consisting of -C(O)NR'-, -NR'C(O)-, -NR'C(O)NR'-, -C(=NR')-NR'-, -NR'-C(=NR')-, -NR'-C(O)-O-, -N=C(Xa)-NR'-, -P(O)(OR')-O-, -S(O)~CR'R"-, -S(O)S NR'-, -S-S- and a covalent bond; where n is 0, 1 or 2;
and R, R' and R" at each separate occurrence are selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, cycloalkenyl, substituted cycloalkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl and heterocyclic.
In yet another of its composition aspects, this invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and an effective amount of a multibinding compound of formula I or II.
The multibinding compounds of this invention are effective inhibitors of microsomal triglyceride transferase protein (MTP), a protein which mediates the transfer of lipids during the assembly of lipoproteins and related biomolecules.
Thus, the multibinding compounds of this invention are useful for treating disorders associated with hyperlipidemia and other lipid-related conditions or disorders. Accordingly, in one of its method aspects, this invention provides a method for preventing or treating atherosclerosis in a patient, the method comprising administering to a patient with atherosclerosis or at risk for developing atherosclerosis a pharmaceutical composition comprising a pharmaceutically-acceptable carrier and a therapeutically-effective amount of a multibinding compound of formulae I or II.
In another of its method aspects, this invention provides a method for lowering serum lipid, cholesterol and/or triglyceride levels in a patient, the method comprising administering to a patient a pharmaceutical composition comprising a pharmaceutically-acceptable carrier and a therapeutically-effective amount of a multibinding compound of formulae I or II.
And, in yet another of its method aspects, this invention provides a method for preventing or treating hyperlipemia, hyperlipidemia, hyperlipoproteinemia, hypercholestrolemia, hypertriglyceridemia, pancreatitis, __ 16--diabetes and/or obsesity in a patient, the method comprising administering to a patient in need of such treatment a pharmaceutical composition comprising a pharmaceutically-acceptable carrier and a therapeutically-effective amount of a multibinding compound of formulae I or II.

This invention is also directed to general synthetic methods for generating large libraries of diverse multimeric compounds which multimeric compounds are candidates for possessing multibinding properties for microsomal triglyceride transferase protein. The diverse multimeric compound libraries provided by this 10 invention are synthesized by combining a library of linkers with a library of ligands each having complementary functional groups permitting covalent linkage. The library of linkers is preferably selected to have diverse properties such as valency, linker length, linker geometry and rigidity, hydrophilicity or hydrophobicity, amphiphilicity, acidity, basicity, polarizability and polarization.
15 The library of ligands is preferably selected to have diverse attachment points on the same ligand, different functional groups at the same site of otherwise the same ligand, and the like.
Additionally, this invention is directed to libraries of diverse multimeric 20 compounds which multimeric compounds are candidates for possessing multibinding properties for microsomal triglyceride transferase protein. These libraries are prepared via the methods described above and permit the rapid and efficient evaluation of what molecular constraints impart multibinding properties to a ligand or a class of ligands for microsomal triglyceride transferase protein.

Accordingly, in one of its method aspects, this invention is directed to a method for identifying multimeric ligand compounds possessing multibinding properties for microsomal triglyceride transferase protein, which method comprises:

__ 1 ~__ (a) identifying a ligand or a mixture of ligands wherein each ligand contains at least one reactive functionality;
(b) identifying a library of linkers wherein each linker in said library comprises at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand;
(c) preparing a multimeric ligand compound library by combining at least two stoichiometric equivalents of the ligand or mixture of ligands identified in (a) with the library of linkers identified in (b) under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said ligands; and (d) assaying the multimeric ligand compounds produced in the library prepared in (c) above to identify multimeric ligand compounds possessing multibinding properties for microsomal triglyceride transferase protein.
In another of its method aspects, this invention is directed to a method for identifying multimeric ligand compounds possessing multibinding properties for microsomal triglyceride transferase protein, which method comprises:
(a) identifying a library of ligands wherein each ligand contains at least one reactive functionality;
(b) identifying a linker or mixture of linkers wherein each linker comprises at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand;
(c) preparing a multimeric ligand compound library by combining at least two stoichiometric equivalents of the library of ligands identified in (a) with the linker or mixture of linkers identified in (b) under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said ligands; and (d) assaying the multimeric ligand compounds produced in the library prepared in (c) above to identify multimeric ligand compounds possessing multibinding properties for microsomal triglyceride transferase protein.

__lg__ Preferably, in these methods, the preparation of the multimeric ligand compound library is achieved by either the sequential or concurrent combination of the two or more stoichiometric equivalents of the ligands identified in (a) with 5 the linkers identified in (b).
Additionally, the multimeric ligand compounds comprising the multimeric ligand compound library are preferably dimeric. More preferably, the dimeric ligand compounds comprising the dimeric ligand compound iibrary are 10 heterodimeric. The heterodimeric ligand compound library is preferably prepared by sequential addition of a first and second ligand.
In a preferably embodiment of the above methods, prior to procedure (d), each member of the multimeric ligand compound library is isolated from the 15 library. More preferably, each member of the library is isolated by preparative liquid chromatography mass spectrometry (LCMS).
In the above methods, the linker or linkers employed are preferably selected from the group comprising flexible linkers, rigid linkers, hydrophobic 20 linkers, hydrophilic linkers, linkers of different geometry, acidic linkers, basic linkers, linkers of different polarization andlor polarizability and amphiphilic linkers. More preferably, the linkers comprise linkers of different chain length and/or having different complementary reactive groups. Still more preferably, the linkers are selected to have different linker lengths ranging from about 2 to 25 100.
The ligand or mixture of ligands employed in the above methods is preferably selected to have reactive functionality at different sites on said ligands.
More preferably, the reactive functionality is selected from the group consisting 30 of carboxylic acids, carboxylic acid halides, carboxyl esters, amines, halides, pseudohalides, isocyanates, vinyl unsaturation, ketones, aldehydes, thiols, alcohols, anhydrides, boronates, and precursors thereof wherein the reactive functionality on the ligand is selected to be complementary to at least one of the reactive groups on the linker so that a covalent linkage can be formed between 5 the linker and the ligand.
In one preferred embodiment of the above methods, the multimeric ligand compound library comprises homomeric ligand compounds. In another preferred embodiment, the multimeric ligand compound library comprises heteromeric 10 ligand compounds.
In one of its composition aspects, this invention is directed to a library of multimeric ligand compounds which may possess multivalent properties for microsomal triglyceride transferase protein, which library is prepared by the 15 method comprising:
(a) identifying a ligand or a mixture of ligands wherein each ligand contains at least one reactive functionality;
(b) identifying a library of linkers wherein each linker in said library comprises at least two functional groups having complementary reactivity to at 20 least one of the reactive functional groups of the ligand; and (c) preparing a multimeric ligand compound library by combining at least two stoichiometric equivalents of the ligand or mixture of ligands identified in (a) with the library of linkers identified in (b) under conditions wherein the complementary functional groups react to form a covalent linkage between said 25 linker and at least two of said ligands.
In another of its composition aspects, this invention is directed to a library of multimeric ligand compounds which may possess multivalent properties for microsomal triglyceride transferase protein, which library is prepared by the 30 method comprising:

(a) identifying a library of ligands wherein each Ligand contains at least one reactive functionality;
(b) identifying a linker or mixture of linkers wherein each Linker comprises at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand; and (c) preparing a multimeric ligand compound library by combining at least two stoichiometric equivalents of the library of ligands identified in (a) with the linker or mixture of linkers identified in (b) under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said ligands.
In a preferred embodiment, the linker or linkers employed are preferably selected from the group comprising flexible Linkers, rigid linkers, hydrophobic linkers, hydrophilic linkers, linkers of different geometry, acidic linkers, basic linkers, linkers of different polarization and/or polarizability and amphiphilic linkers. More preferably, the linkers comprise linkers of different chain length and/or having different complementary reactive groups. Still more preferably, the linkers are selected to have different linker lengths ranging from about 2 to 1001.
In the above libraries, the Ligand or mixture of ligands is preferably selected to have reactive functionality at different sites on said ligands.
Preferably, the reactive functionality is selected from the group consisting of carboxylic acids, carboxylic acid halides, carboxyl esters, amines, halides, pseudohalides, isocyanates, vinyl unsaturation, ketones, aldehydes, thiois, alcohols, anhydrides, boronates, and precursors thereof wherein the reactive functionality on the ligand is selected to be complementary to at least one of the reactive groups on the linker so that a covalent linkage can be formed between the linker and the ligand.

In one embodiment, the multimeric ligand compound library comprises homomeric ligand compounds (i.e., each of the ligands is the same, although it may be attached at different points). In another embodiment, the multimeric ligand compound library comprises heteromeric ligand compounds (i.e., at least 5 one of the ligands is different from the other ligands).
In another of its method aspects, this invention is directed to an iterative method for identifying multimeric ligand compounds possessing multibinding properties for microsomal triglyceride transferase protein, which method 10 comprises:
(a) preparing a first collection or iteration of multimeric compounds which is prepared by contacting at least two stoichiometric equivalents of the ligand or mixture of ligands which target a receptor with a linker or mixture of linkers wherein said ligand or mixture of ligands comprises at least one reactive 15 functionality and said linker or mixture of linkers comprises at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand wherein said contacting is conducted under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said ligands;
20 (b) assaying said first collection or iteration of multimeric compounds to assess which if any of said multimeric compounds possess multibinding properties for microsomal triglyceride transferase protein;
(c) repeating the process of (a) and (b) above until at least one multimeric compound is found to possess multibinding properties for microsomal 25 triglyceride transferase protein;
(d) evaluating what molecular constraints imparted or are consistent with imparting multibinding properties to the multimeric compound or compounds found in the first iteration recited in (a)- (c) above;
(e) creating a second collection or iteration of multimeric compounds 30 which elaborates upon the particular molecular constraints imparting multibinding properties to the multimeric compound or compounds found in said first iteration;
(f) evaluating what molecular constraints imparted or are consistent with imparting enhanced multibinding properties to the multimeric compound or 5 compounds found in the second collection or iteration recited in (e) above;
(g) optionally repeating steps (e) and (f) to further elaborate upon said molecular constraints.
Preferably, steps (e) and (f) are repeated from 2-50 times. More 10 preferably, steps (e) and (f) are repeated from 5-50 times.
Preferably, the ligands employed in the above methods and library compositions are selected from ligands of formula IA-IE, more preferably, from ligands of formula IIA-IIE.

BRIEF DESCRIPTION OF THE DRAWINGS
Figure lA illustrates a known inhibitor of microsomal triglyceride transferase protein (MTP). Figure 1B illustrates a various binding complexes of MTP with a known inhibitor of MTP and various multibinding compounds.

Figure 2 illustrates examples of multibinding compounds comprising 2 ligands attached in different formats to a linker.
Figure 3 illustrates examples of multibinding compounds comprising 3 25 ligands attached in different formats to a linker.
Figure 4 illustrates examples of multibinding compounds comprising 4 ligands attached in different formats to a linker.

Figure 5 illustrates examples of multibinding compounds comprising > 4 ligands attached in different formats to a linker.
Figures 6, 7 and 8 illustrate multibinding compounds of this invention where -- represents the linker.
DETAILED DESCRIPTION OF THE INVENTION
This invention is directed to multibinding compounds which inhibit microsomal triglyceride transferase protein (MTP), pharmaceutical compositions containing such multibinding compounds and methods for treating disorders associated with hyperlipidemia and other lipid-related conditions or disorders.
When discussing such compounds, compositions or methods, the following terms have the following meanings unless otherwise indicated. Any undefined terms have their art recognized meanings.
The term "alkyl" refers to a monoradical branched or unbranched saturated hydrocarbon chain preferably having from 1 to 40 carbon atoms, more preferably 1 to 10 carbon atoms, and even more preferably 1 to 6 carbon atoms.
This term is exemplified by groups such as methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, n-hexyl, n-decyl, tetradecyl, and the like.
The term "substituted alkyl" refers to an alkyl group as defined above, having from 1 to 5 substituents, and preferably 1 to 3 substituents, selected from the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, hydroxyamino, alkoxyamino, vitro, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SOZ-alkyl, -SOZ-substituted alkyl, -SOZ-aryl and -SOZ-heteroaryl.
The term "alkylene" refers to a diradical of a branched or unbranched saturated hydrocarbon chain, preferably having from 1 to 40 carbon atoms, more 5 preferably 1 to 10 carbon atoms and even more preferably 1 to 6 carbon atoms.
This term is exemplified by groups such as methylene (-CH2-), ethylene (-CHZCHZ-), the propylene isomers (e.g., -CHZCHZCHZ and -CH(CH3)CHZ-) and the like.
10 The term "substituted alkylene" refers to an alkylene group, as defined above, having from 1 to 5 substituents, and preferably 1 to 3 substituents, selected from the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloallcyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, 15 azido, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, hydroxyamino, alkoxyamino, vitro, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SOZ-alkyl, -SOZ-substituted alkyl, -SOZ-aryl and 20 -SOZ-heteroaryl. Additionally, such substituted alkylene groups include those where 2 substituents on the alkylene group are fused to form one or more cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heterocyclic or heteroaryl groups fused to the alkylene group. Preferably such fused groups contain from 1 to 3 fused ring structures.

The term "alkaryl" refers to the groups -alkylene-aryl and -substituted alkylene-aryl where alkylene, substituted alkylene and aryl are defined herein.
Such alkaryl groups are exemplified by benzyl, phenethyl and the like.

The term "alkoxy" refers to the groups alkyl-O-, alkenyl-O-, cycloalkyl-O-, cycloalkenyl-O-, and alkynyl-O-, where alkyl, alkenyl, cycloalkyl, cycloalkenyl, and alkynyl are as defined herein. Preferred alkoxy groups are alkyl-O- and include, by way of example, methoxy, ethoxy, n-propoxy, iso-5 propoxy, n-butoxy, tent-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, 1,2-dimethyibutoxy, and the like.
The term "substituted alkoxy" refers to the groups substituted alkyl-O-, substituted alkenyl-O-, substituted cycloalkyl-O-, substituted cycloalkenyl-O-, 10 and substituted alkynyl-O- where substituted alkyl, substituted alkenyl, substituted cycloalkyl, substituted cycloalkenyl and substituted alkynyl are as defined herein.
The term "alkylalkoxy" refers to the groups -alkylene-O-alkyl, 15 alkylene-O-substituted alkyl, substituted alkylene-O-alkyl and substituted alkylene-O-substituted alkyl wherein alkyl, substituted alkyl, alkylene and substituted alkylene are as defined herein. Preferred alkylalkoxy groups are alkylene-O-alkyl and include, by way of example, methylenemethoxy (-CHzOCH3), ethylenemethoxy (-CHZCHZOCH3), n-propylene-iso-propoxy 20 (-CHZCHZCH20CH(CH3)Z), methylene-t-butoxy (-CH2-O-C(CH3)3) and the like.
The term "alkylthioalkoxy" refers to the group -alkylene-S-alkyl, alkylene-S-substituted alkyl, substituted alkylene-S-alkyl and substituted alkylene-S-substituted alkyl wherein alkyl, substituted alkyl, alkylene and substituted 25 alkylene are as defined herein. Preferred alkylthioalkoxy groups are alkylene-S
alkyl and include, by way of example, methylenethiomethoxy (-CHZSCH3), ethylenethiomethoxy (-CHZCHZSCH3), n-propylene-iso-thiopropoxy (-CHZCHZCHZSCH(CH3)2), methylene-t-thiobutoxy (-CHZSC(CH3)3) and the like.

The term "alkenyl" refers to a monoradical of a branched or unbranched unsaturated hydrocarbon group preferably having from 2 to 40 carbon atoms, more preferably 2 to 10 carbon atoms and even more preferably 2 to 6 carbon atoms and having at least 1 and preferably from 1-6 sites of vinyl unsaturation.
5 Preferred alkenyl groups include ethenyl (-CH=CHZ), n-propenyl (-CHZCH=CHZ), iso-propenyl (-C(CH3)=CHZ), and the like.
The term "substituted alkenyl" refers to an alkenyl group as defined above having from 1 to 5 substituents, and preferably 1 to 3 substituents, selected from 10 the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, 15 aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, hydroxyamino, alkoxyamino, nitro, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SOZ-alkyl, -SOZ-substituted alkyl, -SOZ-aryl and -SOZ-heteroaryl.
The term "alkenylene" refers to a diradical of a branched or unbranched 20 unsaturated hydrocarbon group preferably having from 2 to 40 carbon atoms, more preferably 2 to 10 carbon atoms and even more preferably 2 to 6 carbon atoms and having at least 1 and preferably from 1-6 sites of vinyl unsaturation.
This term is exemplified by groups such as ethenylene (-CH=CH-), the propenylene isomers (e.g., -CHZCH=CH- and -C(CH3)=CH-) and the like.

The term "substituted alkenylene" refers to an alkenylene group as defined above having from 1 to 5 substituents, and preferably from 1 to 3 substituents, selected from the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, 30 acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, -_2~__ azido, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, hydroxyamino, alkoxyamino, vitro, -SO-alkyl, 5 -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SOZ-alkyl, -SOZ-substituted alkyl, -S02-aryl and -SOZ-heteroaryl. Additionally, such substituted alkenylene groups include those where 2 substituents on the alkenylene group are fused to form one or more cycloalkyl, substituted cycloalkyl, cycloalkenyi, substituted cycloalkenyl, aryl, heterocyclic or heteroaryl groups fused to the alkenylene 10 group.
The term "alkynyl" refers to a monoradical of an unsaturated hydrocarbon preferably having from 2 to 40 carbon atoms, more preferably 2 to 20 carbon atoms and even more preferably 2 to 6 carbon atoms and having at least 1 and 15 preferably from 1-6 sites of acetylene (triple bond) unsaturation.
Preferred alkynyl groups include ethynyl (-C---CH), propargyl (-CHZC=CH) and the like.
The term "substituted alkynyl" refers to an alkynyl group as defined above having from 1 to 5 substituents, and preferably 1 to 3 substituents, selected from 20 the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, 25 aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, hydroxyamino, alkoxyamino, vitro, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SOZ-alkyl, -SOZ-substituted alkyl, -S02-aryl and -SOZ-heteroaryl.
The term "alkynylene" refers to a diradical of an unsaturated hydrocarbon 30 preferably having from 2 to 40 carbon atoms, more preferably 2 to 10 carbon -_2g__ atoms and even more preferably 2 to 6 carbon atoms and having at least 1 and preferably from 1-6 sites of acetylene (triple bond) unsaturation. Preferred alkynylene groups include ethynylene (-C=C-), propargylene (-CHZC=C-) and the like.

The term "substituted alkynylene" refers to an alkynylene group as defined above having from 1 to 5 substituents, and preferably 1 to 3 substituents, selected from-the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, 10 acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, hydroxyamino, alkoxyamino, vitro, -SO-alkyl, -SO-substituted 15 alkyl, -SO-aryl, -SO-heteroaryl, -SOZ-alkyl, -S02-substituted alkyl, -SOZ-aryl and -SOZ-heteroaryi.
The term "acyl" refers to the groups HC(O)-, alkyl-C(O)-, substituted alkyl-C(O)-, cycloalkyl-C(O)-, substituted cycloalkyl-C(O)-, cycioalkenyl-C(O)-, 20 substituted cycloalkenyl-C(O)-, aryl-C(O)-, heteroaryl-C(O)- and heterocyclic-C(O)- where alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic are as defined herein.
25 The term "acylamino" or "aminocarbonyl" refers to the group -C(O)NRR
where each R is independently hydrogen, alkyl, substituted alkyl, aryl, heteroaryl, heterocyclic or where both R groups are joined to form a heterocyclic group (e.g., morpholino) wherein alkyl, substituted alkyl, aryl, heteroaryl and heterocyclic are as defined herein.

The term "aminoacyl" refers to the group -NRC(O)R where each R is independently hydrogen, alkyl, substituted alkyl, aryl, heteroaryl, or heterocyclic wherein alkyl, substituted alkyl, aryl, heteroaryl and heterocyclic are as defined herein.

The term "aminoacyloxy" or "alkoxycarbonylamino" refers to the group -NRC(O)OR where each R is independently hydrogen, alkyl, substituted alkyl, aryl, heteroaryl, or heterocyclic wherein alkyl, substituted alkyl, aryl, heteroaryl and heterocyclic are as defined herein.

The term "acyloxy" refers to the groups alkyl-C(O)O-, substituted alkyl-C(O)O-, cycloalkyl-C(O)O-, substituted cycloalkyl-C(O)O-, aryl-C(O)O-, heteroaryl-C(O)O-, and heterocyclic-C(O)O- wherein alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, heteroaryl, and heterocyclic are as 15 defined herein.
The term "aryl" refers to an unsaturated aromatic carbocyclic group of from 6 to 20 carbon atoms having a single ring (e.g., phenyl) or multiple condensed (fused) rings (e.g., naphthyl or anthryl). Preferred aryls include 20 phenyl, naphthyl and the like.
Unless otherwise constrained by the definition for the aryl substituent, such aryl groups can optionally be substituted with from 1 to 5 substituents, preferably 1 to 3 substituents, selected from the group consisting of acyloxy, 25 hydroxy, thiol, acyl, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, substituted alkyl, substituted alkoxy, substituted alkenyl, substituted alkynyl, substituted cycloalkyl, substituted cycloalkenyl, amino, substituted amino, aminoacyl, acylamino, alkaryl, aryl, aryloxy, azido, carboxyl, carboxylalkyl, cyano, halo, vitro, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, _-30_ aminoacyloxy, oxyacylamino, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioheteroaryloxy, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SOZ-alkyl, -SOZ-substituted alkyl, -SOz-aryl, -S02-heteroaryl and trihalomethyl.
Preferred aryl substituents include alkyl, alkoxy, halo, cyano, vitro, S trihalomethyl, and thioalkoxy.
The term "aryloxy" refers to the group aryl-O- wherein the aryl group is as defined above including optionally substituted aryl groups as also defined above.

The term "arylene" refers to the diradical derived from aryl (including substituted aryl) as defined above and is exemplified by 1,2-phenylene, 1,3-phenylene, 1,4-phenylene, 1,2-naphthylene and the like.
15 The term "amino" refers to the group -NHZ.
The term "substituted amino" refers to the group -NRR where each R is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, 20 cycloalkenyl, substituted cycloalkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl and heterocyclic provided that both R's are not hydrogen.
The term "carboxyalkyl" or "alkoxycarbonyl" refers to the groups "-C(O)O-alkyl", "-C(O)O-substituted alkyl", "-C(O)O-cycloalkyl", "-C(O)O-25 substituted cycloalkyl", "-C(O)O-alkenyl", "-(O)O-substituted alkenyl", "-C(O)O-alkynyl" and "-C(O)O-substituted alkynyl" where alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl and substituted alkynyl are as defined herein.

The term "cycloalkyl" refers to cyclic alkyl groups of from 3 to 20 carbon atoms having a single cyclic ring or multiple condensed rings. Such cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl, and the like, or multiple ring structures such 5 as adamantanyl, and the like.
The term "substituted cycloalkyl" refers to cycloalkyl groups having from 1 to 5 substituents, and preferably 1 to 3 substituents, selected from the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, 10 cycloalkenyl, substituted cycloalkenyi, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, 15 hydroxyamino, alkoxyamino, vitro, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SOZ-alkyl, -SOZ-substituted alkyl, -S02-aryl and -SOZ-heteroaryl.
The term "cycloalkenyl" refers to cyclic alkenyl groups of from 4 to 20 carbon atoms having a single cyclic ring and at least one point of internal 20 unsaturation. Examples of suitable cycloalkenyl groups include, for instance, cyclobut-2-enyl, cyciopent-3-enyl, cyclooct-3-enyl and the like.
The term "substituted cycloalkenyl" refers to cycloalkenyl groups having from 1 to 5 substituents, and preferably 1 to 3 substituents, selected from the 25 group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, hydroxyamino, alkoxyamino, vitro, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SOZ-alkyl, -SOZ-substituted alkyl, -SOZ-aryl and -SOZ-heteroaryl.
$ The term "halo" or "halogen" refers to fluoro, chloro, bromo and iodo.
The term "heteroaryl" refers to an aromatic group of from 1 to 15 carbon atoms and 1 to 4 heteroatoms selected from oxygen, nitrogen and sulfur within at least one ring (if there is more than one ring).

Unless otherwise constrained by the definition for the heteroaryl substituent, such heteroaryl groups can be optionally substituted with 1 to 5 substituents, preferably i to 3 substituents, selected from the group consisting of acyloxy, hydroxy, thiol, acyl, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, 15 cycloalkenyl, substituted alkyl, substituted alkoxy, substituted alkenyl, substituted alkynyl, substituted cycloalkyl, substituted cycloalkenyl, amino, substituted amino, aminoacyl, acylamino, alkaryl, aryl, aryloxy, azido, carboxyl, carboxylalkyl, cyano, halo, vitro, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, aminoacyloxy, oxyacylamino, thioalkoxy, substituted thioalkoxy, 20 thioaryloxy, thioheteroaryloxy, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SOZ-alkyl, -S02-substituted alkyl, -SOZ-aryl, -SOZ-heteroaryl and trihalomethyl. Preferred aryl substituents include alkyl, alkoxy, halo, cyano, vitro, trihalomethyl, and thioalkoxy. Such heteroaryl groups can have a single ring (e.g., pyridyl or furyl) or multiple condensed rings (e.g., indolizinyl or 25 benzothienyl). Preferred heteroaryls include pyridyl, pyrrolyl and furyl.
The term "heteroaryloxy" refers to the group heteroaryl-O-.
The term "heteroarylene" refers to the diradical group derived from heteroaryl (including substituted heteroaryl), as defined above, and is exemplified by the WO j9/63929 PCT/US99/11789 groups 2,6-pyridylene, 2,4-pyridiylene, 1,2-quinolinylene, 1,8-quinolinylene, 1,4-benzofuranylene, 2,5-pyridnylene, 2,5-indolenyl and the like.
The term "heterocycle" or "heterocyclic" refers to a monoradical saturated 5 unsaturated group having a single ring or multiple condensed rings, from 1 to 40 carbon atoms and from 1 to 10 hetero atoms, preferably 1 to 4 heteroatoms, selected from nitrogen, sulfur, phosphorus, and/or oxygen within the ring.
Unless otherwise constrained by the definition for the heterocyclic 10 substituent, such heterocyclic groups can be optionally substituted with 1 to 5, and preferably 1 to 3 substituents, selected from the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, keto, thioketo, 15 carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, hydroxyamino, alkoxyamino, vitro, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO-heteroaryl, -SOZ-alkyl, -SOZ-substituted alkyl, -SOZ-aryl and -SOz-heteroaryl. Such heterocyclic groups can have a single 20 ring or multiple condensed rings. Preferred heterocyclics include morpholino, piperidinyl, and the like.
Examples of nitrogen heterocycles and heteroaryls include, but are not limited to, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, 25 pyridazine, indolizine, isoindole, indole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthylpyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, phenanthroline, isothiazole, phenazine, isoxazole, phenoxazine, phenothiazine, imidazolidine, imidazoline, piperidine, piperazine, indoline, morpholino, piperidinyl, tetrahydrofuranyl, and the like as well as N-alkoxy-nitrogen containing heterocycles.
The term "heterocyclooxy" refers to the group heterocyclic-O-.

The term "thioheterocyclooxy" refers to the group heterocyclic-S-.
The term "heterocyclene" refers to the diradical group formed from a heterocycle, as defined herein, and is exemplified by the groups 2,6-morpholino, 10 2,5-morpholino and the like.
The term "oxyacylamino" or "aminocarbonyloxy" refers to the group -OC(O)NRR where each R is independently hydrogen, alkyl, substituted alkyl, aryl, heteroaryl, or heterocyclic wherein alkyl, substituted alkyl, aryl, heteroaryl 15 and heterocyclic are as defined herein.
The term "spiro-attached cycloalkyl group" refers to a cycloalkyl group attached to another ring via one carbon atom common to both rings.
20 The term "thiol" refers to the group -SH.
The term "thioalkoxy" refers to the group -S-alkyl.
The term "substituted thioalkoxy" refers to the group -S-substituted alkyl.

The term "thioaryloxy" refers to the group aryl-S- wherein the aryl group is as defined above including optionally substituted aryl groups also defined above.

The term "thioheteroaryloxy" refers to the group heteroaryl-S- wherein the heteroaryl group is as defined above including optionally substituted aryl groups as also defined above.
5 As to any of the above groups which contain one or more substituents, it is understood, of course, that such groups do not contain any substitution or substitution patterns which are sterically impractical and/or synthetically non-feasible. In addition, the compounds of this invention include all stereochemical isomers arising from the substitution of these compounds.

The term "pharmaceutically-acceptable salt" refers to salts which retain the biological effectiveness and properties of the multibinding compounds of this invention and which are not biologically or otherwise undesirable. In many cases, the multibinding compounds of this invention are capable of forming acid 15 and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto.
Pharmaceutically-acceptable base addition salts can be prepared from inorganic and organic bases. Salts derived from inorganic bases, include by way 20 of example only, sodium, potassium, lithium, ammonium, calcium and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of primary, secondary and tertiary amines, such as alkyl amines, dialkyl amines, trialkyl amines, substituted alkyl amines, di(substituted alkyl) amines, tri(substituted alkyl) amines, alkenyl amines, dialkenyl amines, trialkenyl amines, 25 substituted alkenyl amines, di(substituted alkenyl) amines, tri(substituted alkenyl) amines, cycloalkyl amines, di(cycloalkyl) amines, tri(cycloalkyl) amines, substituted cycloalkyl amines, disubstituted cycloalkyl amine, trisubstituted cycloalkyl amines, cycloalkenyl amines, di(cycloalkenyl) amines, tri(cycloalkenyl) amines, substituted cycloalkenyl amines, disubstituted 30 cycloalkenyl amine, trisubstituted cycloalkenyl amines, aryl amines, diaryl amines, triaryl amines, heteroaryl amines, diheteroaryi amines, triheteroaryl amines, heterocyclic amines, diheterocyclic amines, triheterocyclic amines, mixed di- and tri-amines where at least two of the substituents on the amine are different and are selected from the group consisting of alkyl, substituted alkyl, 5 alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, and the like. Also included are amines where the two or three substituents, together with the amino nitrogen, form a heterocyclic or heteroaryl group.
10 Examples of suitable amines include, by way of example only, isopropylamine, trimethyl amine, diethyl amine, tri(iso-propyl) amine, tri(n-propyl) amine, ethanolamine, 2-dimethylaminoethanol, tromethamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, N-alkylglucamines, theobromine, purines, 15 piperazine, piperidine, morpholine, N-ethylpiperidine, and the like. It should also be understood that other carboxylic acid derivatives would be useful in the practice of this invention, for example, carboxylic acid amides, including carboxamides, lower alkyl carboxamides, dialkyl carboxamides, and the like.
20 Pharmaceutically acceptable acid addition salts may be prepared from inorganic and organic acids. Salts derived from inorganic acids include hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Salts derived from organic acids include acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, tnalic acid, malonic acid, succinic acid, 25 malefic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluene-sulfonic acid, salicylic acid, and the like.
The term upharmaceutically-acceptable cation" refers to the cation of a 30 pharmaceutically-acceptable salt.

The term "protecting group" or "blocking group" refers to any group which when bound to one or more hydroxyl, thiol, amino or carboxyl groups of the compounds (including intermediates thereof) prevents reactions from 5 occurring at these groups and which protecting group can be removed by conventional chemical or enzymatic steps to reestablish the hydroxyl, thiol, amino or carboxyl group. The particular removable blocking group employed is not critical and preferred removable hydroxyl blocking groups include conventional substituents such as allyl, benzyl, acetyl, chloroacetyl, thiobenzyl, 10 benzylidine, phenacyl, t-butyl-diphenylsilyl and any other group that can be introduced chemically onto a hydroxyl functionality and later selectively removed either by chemical or enzymatic methods in mild conditions compatible with the nature of the product.
15 Preferred removable thiol blocking groups include disulfide groups, acyl groups, benzyl groups, and the like.
Preferred removable amino blocking groups include conventional substituents such as t-butyoxycarbonyl (t-BOC), benzyloxycarbonyl (CBZ), 20 fluorenylmethoxycarbonyl (FMOC), allyloxycarbonyl (ALOC), and the like which can be removed by conventional conditions compatible with the nature of the product.
Preferred carboxyl protecting groups include esters such as methyl, ethyl, 25 propyl, t-butyl etc. which can be removed by mild conditions compatible with the nature of the product.
The term "optional" or "optionally" means that the subsequently described event, circumstance or substituent may or may not occur, and that the description __3g__ includes instances where said event or circumstance occurs and instances where it does not.
The term "ligand" as used herein denotes a compound that is capable of 5 binding to microsomal triglyceride transferase protein. The specific region or regions of the ligand that is (are) recognized by the protein is designated as the "ligand domain" . A ligand may be either capable of binding to the protein by itself, or may require the presence of one or more non-ligand components for binding (e.g., Ca+2, Mg+2 or a water molecule is required for the binding of a 10 ligand to various ligand binding sites).
Examples of ligands useful in this invention are described herein. Those skilled in the art will appreciate that portions of the ligand structure that are not essential for specific molecular recognition and binding activity may be varied 15 substantially, replaced or substituted with unrelated structures (for example, with ancillary groups as defined below) and, in some cases, omitted entirely without affecting the binding interaction. The primary requirement for a ligand is that it has a ligand domain as defined above. It is understood that the term ligand is not intended to be limited to compounds known to be useful in binding to microsomal 20 triglyceride transferase protein (e.g., known drugs). Those skilled in the art will understand that the term ligand can equally apply to a molecule that is not normally associated with binding properties. In addition, it should be noted that ligands that exhibit marginal activity or lack useful activity as monomers can be highly active as multivalent compounds because of the benefits conferred by 25 multivalency .
The term "multibinding compound or agent" refers to a compound that is capable of multivalency, as defined below, and which has 2-10 ligands covalently bound to one or more linkers which may be the same or different. Multibinding 30 compounds provide a biological andlor therapeutic effect greater than the aggregate of unlinked ligands equivalent thereto which are made available for binding. That is to say that the biological and/or therapeutic effect of the ligands attached to the multibinding compound is greater than that achieved by the same amount of unlinked ligands made available for binding to the ligand binding sites (receptors). The phrase "increased biological or therapeutic effect" includes, for example: increased affinity, increased selectivity for target, increased specificity for target, increased potency, increased efficacy, decreased toxicity, improved duration of activity or action, decreased side effects, increased therapeutic index, improved bioavailibity, improved pharmacokinetics, improved activity spectrum, and the like. The multibinding compounds of this invention will exhibit at least one and preferably more than one of the above-mentioned affects.
The term "mulimeric compound" refers to a compound containing 2 to 10 ligands covalently connected through at least one linker which compound may or may not possess multibinding properties.
The term "potency" refers to the minimum concentration at which a ligand is able to achieve a desirable biological or therapeutic effect. The potency of a ligand is typically proportional to its affinity for its Iigand binding site.
In some cases, the potency may be non-linearly correlated with its affinity. In comparing the potency of two drugs, e.g., a multibinding agent and the aggregate of its unlinked ligand, the dose-response curve of each is determined under identical test conditions (e.g., in an in vitro or in vivo assay, in an appropriate animal model). The finding that the multbinding agent produces an equivalent biological or therapeutic effect at a lower concentration than the aggregate unlinked ligand is indicative of enhanced potency.
The term "univalency" as used herein refers to a single binding interaction between one ligand as defined herein with one ligand binding site as defined herein. It should be noted that a compound having multiple copies of a ligand (or ligands) exhibit univalency when only one ligand is interacting with a ligand binding site. Examples of univalent interactions are depicted below.
Mi The term "multivalency" as used herein refers to the concurrent binding of from 2 to 10 linked ligands (which may be the same or different) and two or more corresponding receptors (ligand binding sites) on one or more biomolecules i.e., proteins or enzymes, which may be the same or different.
For example, two ligands connected through a linker that bind concurrently to two ligand binding sites would be considered as bivalency;
three ligands thus connected would be an example of trivalency. An example of trivalent binding, illustrating a muitibinding compound bearing three ligands versus a monovalent binding interaction, is shown below:
Univalent Interaction _..~ 1 __ Trivalent Interaction It should be understood that all compounds that contain multiple copies of a ligand attached to a linker or to linkers do not necessarily exhibit the phenomena of multivalency, i.e., that the biological and/or therapeutic effect of the multibinding agent is greater than the sum of the aggregate of unlinked 15 ligands made available for binding to the ligand binding site (receptor).
For multivalency to occur, the ligands that are connected by a linker or linkers have to be presented to their ligand binding sites by the linkers) in a specific manner in order to bring about the desired ligand-orienting result, and thus produce a multibinding event.

The term "selectivity" or "specificity" is a measure of the binding preferences of a ligand for different ligand binding sites {receptors). The selectivity of a ligand with respect to its target ligand binding site relative to another ligand binding site is given by the ratio of the respective values of Kd 25 (i.e., the dissociation constants for each ligand-receptor complex) or, in cases where a biological effect is observed below the I~ , the ratio of the respective ECU's (i.e., the concentrations that produce 50% of the maximum response for the ligand interacting with the two distinct ligand binding sites (receptors)).

_~2__ The term "ligand binding site" denotes the sites) on the microsomal triglyceride transferase protein that recognizes a ligand domain and provides a binding partner for the ligand. The ligand binding site may be defined by monomeric or multimeric structures. This interaction may be capable of 5 producing a unique biological effect, for example, agonism, antagonism, modulatory effects, may maintain an ongoing biological event, and the like.
The terms "agonism" and "antagonism" are well known in the art. The term "modulatory effect" refers to the ability of the ligand to change the activity 10 of an agonist or antagonist through binding to a ligand binding site.
It should be recognized that the ligand binding sites of the enzyme that participate in biological multivalent binding interactions are constrained to varying degrees by their infra- and inter-molecular associations (e.g., such 15 macromolecular structures may be covalently joined to a single structure, noncovalently associated in a multimeric structure, embedded in a membrane or polymeric matrix, and so on) and therefore have less translational and rotational freedom than if the same structures were present as monomers in solution.
20 The term "inert organic solvent" or "inert solvent" means a solvent which is inert under the conditions of the reaction being described in conjunction therewith including, by way of example only, benzene, toluene, acetonitrile, tetrahydrofuran, dimethylformamide, chloroform, methylene chloride, diethyl ether, ethyl acetate, acetone, methylethyl ketone, methanol, ethanol, propanol, 25 isopropanol, t-butanol, dioxane, pyridine, and the like. Unless specified to the contrary, the solvents used in the reactions described herein are inert solvents.
The term "treatment" refers to any treatment of a pathologic condition in a mammal, particularly a human, and includes:

_.~3__ (i) preventing the pathologic condition from occurring in a subject which may be predisposed to the condition but has not yet been diagnosed with the condition and, accordingly, the treatment constitutes prophylactic treatment for the disease condition;
5 (ii) inhibiting the pathologic condition, i.e., arresting its development;
(iii) relieving the pathologic condition, i.e., causing regression of the pathologic condition; or (iv) relieving the conditions mediated by the pathologic condition.
10 The term "pathologic condition which is modulated by treatment with a ligand" covers all disease states (i.e., pathologic conditions) which are generally acknowledged in the art to be usefully treated with a ligand for microsomal triglyceride transferase protein in general, and those disease states which have been found to be usefully treated by a specific multibinding compound of our 15 invention. Such disease states include, by way of example only, the treatment of a mammal afflicted with atherosclerosis, hyperlipemia, hyperlipidemia, hyperlipoproteinemia, hypercholestrolemia, hypertriglyceridemia, pancreatitis, diabetes, obsesity and the like.
20 The term "therapeutically effective amount" refers to that amount of multibinding compound which is sufficient to effect treatment, as defined above, when administered to a mammal in need of such treatment. The therapeutically effective amount will vary depending upon the subject and disease condition being treated, the weight and age of the subject, the severity of the disease 25 condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
The term "linker", identified where appropriate by the symbol X or X', refers to a group or groups that covalently links from 2 to 10 ligands (as 30 identified above) in a manner that provides for a compound capable of multivalency. Each linker may be chiral or achiral. Among other features, the linker is a ligand-orienting entity that permits attachment of multiple copies of a ligand (which may be the same or different) thereto. In some cases, the linker may itself be biologically active. The term clinker" does not, however, extend to 5 cover solid inert supports such as beads, glass particles, fibers, and the like. But it is understood that the multibinding compounds of this invention can be attached to a solid support if desired. For example, such attachment to solid supports can be made for use in separation and purification processes and similar applications.
10 The extent to which multivalent binding is realized depends upon the efficiency with which the linker or linkers that joins the ligands presents these ligands to the array of available ligand binding sites. Beyond presenting these ligands for multivalent interactions with ligand binding sites, the linker or linkers spatially constrains these interactions to occur within dimensions defined by the 15 linker or linkers. Thus, the structural features of the linker (valency, geometry, orientation, size, flexibility, chemical composition, etc.) are features of multibinding agents that play an important role in determining their activities.
The linkers used in this invention are selected to allow multivalent binding 20 of ligands to the ligand binding sites of microsomal triglyceride transferase protein, whether such sites are located interiorly, both interiorly and on the periphery of the protein structure, or at any intermediate position thereof.
The ligands are covalently attached to the linker or linkers using 25 conventional chemical techniques providing for covalent linkage of the ligand to the linker or linkers. Reaction chemistries resulting in such linkages are well known in the art and involve the use of complementary functional groups on the linker and ligand. Preferably, the complementary functional groups on the linker are selected relative to the functional groups available on the ligand for bonding 30 or which can be introduced onto the ligand for bonding. Again, such WO ~9/b3929 PCT/US99/11789 complementary functional groups are well known in the art. For example, reaction between a carboxylic acid of either the linker or the ligand and a primary or secondary amine of the ligand or the linker in the presence of suitable, well known activating agents results in formation of an amide bond covalently linking 5 the ligand to the linker; reaction between an amine group of either the linker or the ligand and a sulfonyl halide of the ligand or the linker results in formation of a sulfonamide bond covalently linking the ligand to the linker; and reaction between an alcohol or phenol group of either the linker or the ligand and an alkyl or aryl halide of the ligand or the linker results in formation of an ether bond 10 covalently linking the ligand to the linker.
Table I below illustrates numerous complementary reactive groups and the resulting bonds formed by reaction there between.
15 Table I
Representative Co~lementarv Binding Chemistries First Reactive Group Second Reactive Grouo ~,lll~g~
hydroxyl isocyanate urethane amine epoxide b-hydroxyamine 20 sulfonyl halide amine sulfonamide carboxyl amine amide hydroxyl alkyl/aryl halide ether The linker is attached to the ligand at a position that retains ligand 25 domain-ligand binding site interaction and specifically which permits the ligand domain of the ligand to orient itself to bind to the ligand binding site. Such positions and synthetic protocols for linkage are well known in the art. The term linker embraces everything that is not considered to be part of the ligand.

The relative orientation in which the ligand domains are displayed derives from the particular point or points of attachment of the ligands to the linker, and on the framework geometry. The determination of where acceptable substitutions can be made on a ligand is typically based on prior knowledge of structure-s activity relationships (SAR) of the ligand andlor congeners and/or structural information about ligand-receptor complexes (e.g., X-ray crystallography, NMR, and the like). Such positions and the synthetic methods for covalent attachment are well known in the art. Following attachment to the selected linker (or attachment to a significant portion of the linker, for example 2-10 atoms of the 10 linker), the univalent linker-ligand conjugate may be tested for retention of activity in the relevant assay.
Suitable linkers are discussed more fully below.
15 At present, it is preferred that the multibinding agent is a bivalent compound, e.g., two ligands which are covalently linked to linker X.
The term "libraryn refers to at least 3, preferably from 102 to 109 and more preferably from 102 to 10~ multimeric compounds. Preferably, these compounds 20 are prepared as a multiplicity of compounds in a single solution or reaction mixture which permits the facile synthesis thereof. In one embodiment, the library of multimeric compounds can be directly assayed for multibinding properties. In another embodiment, each member of the library of multimeric compounds is first isolated and, optionally, characterized. This member is then 25 assayed for multibinding properties.
The term "collection" refers to a set of multimeric compounds which are prepared either sequentially or concurrently (e.g., combinatorially). The collection comprises at least 2 members; preferably from 2 to 1(~ members and 30 still more preferably from 10 to 104 members.

The term "pseudohalide" refers to a functional group which react in a displacement reaction in a manner similar to a halogen, e.g., functions as a leaving group is a displacement reaction. Such functional groups include, by way of example, mesyl, tosyl, azido, cyano and the like.

The linker, when covalently attached to multiple copies of the ligands, provides a biocompatible, substantially non-immunogenic multibinding compound. The biological activity of the multibinding compound is highly 10 sensitive to the valency, geometry, composition, size, flexibility or rigidity, etc.
of the linker and, in turn, on the overall structure of the multibinding compound, as well as the presence or absence of anionic or cationic charge, the relative hydrophobicity/hydrophilicity of the linker, and the like on the linker.
Accordingly, the linker is preferably chosen to maximize the biological activity of 15 the multibinding compound. The linker may be chosen to enhance the biological activity of the molecule. In general, the linker may be chosen from any organic molecule construct that orients two or more ligands to their ligand binding sites to permit multivalency. In this regard, the linker can be considered as a "framework" on which the ligands are arranged in order to bring about the 20 desired ligand-orienting result, and thus produce a multibinding compound.
For example, different orientations can be achieved by including in the framework groups containing mono- or polycyclic groups, including aryl and/or heteroaryl groups, or structures incorporating one or more carbon-carbon 25 multiple bonds (alkenyl, alkenylene, alkynyl or alkynylene groups). Other groups can also include oligomers and polymers which are branched- or straight-chain species. In preferred embodiments, rigidity is imparted by the presence of cyclic groups (e.g., aryl, heteroaryl, cycloalkyl, heterocyclic, etc.). In other preferred embodiments, the ring is a six or ten member ring. In still further _~g__ preferred embodiments, the ring is an aromatic ring such as, for example, phenyl or naphthyl.
Different hydrophobic/hydrophilic characteristics of the linker as well as the presence or absence of charged moieties can readily be controlled by the skilled artisan. For example, the hydrophobic nature of a linker derived from hexamethylene diamine (HZN(CHZ)6NH2) or related polyamines can be modified to be substantially more hydrophilic by replacing the alkylene group with a poly(oxyalkylene) group such as found in the commercially available "Jeffamines" .
The intersection of the framework (linker) and the ligand group, and indeed, the framework (linker) itself can have many different bonding patterns.
Examples of acceptable patterns of three contiguous atom arrangements are shown in the following diagram:
CCC NCC OCC SCC PCC

CCN NCN OCN SCN PCN

CCO NCO OCO SCO PCO

CCS NCS OCS SCS PCS

CCP NCP OCP SCP PCP

CNC NNC ONC SNC PNC

CNN NNN ONN ~ PNN

ZS CNO NNO ONO SNO PNO

CNS 1YN~ ONS SNS PNS

CNP ~Y1YE ONP SNP PNP

COC NOC QQ~ SOC POC

CON ~10N OQN SON PON

NOO o00 ~Q P~S2 COS ~S Q9~ ~ ESZ~

COP ~QE ~E

S CSC NSC OSC SSC PSC

CSN NSN OSN SSN P.~LY

CSO NSO OSO ~Q P~

CSS NSS OSS

CSP ~E

CPC NPC OPC SPC

CPN NPN OPN SPN PEN

CPS NPS OPS SPS PP.~

15 ~ IYEP QPE SEE EPE

One skilled in the art would be able to identify bonding patterns that would produce multivalent compounds. Methods for producing these bonding arrangements are described in March, "Advanced Organic Chemistry", 4th 20 Edition, Wiley-Interscience, New York, New York (1992). All of the possible arrangements for the five most preferred atoms are shown. Each atom has a variety of acceptable oxidation states. The bonding arrangements underlined are less acceptable and are not preferred.
25 Examples of molecular structures in which the above bonding patterns could be employed as components of the linker are shown below.

O
HN~C'C~
~O~O' ~ N N O N
O
w .C'' ' ~C~C'C~ ~C.O'C' ~C~N~C' C C
O C C
~C~ N ~ wC.C C' O
~N N' O N O
O O O
wS.S.N' wS.S.N~ wC.S'C~ C C ~C,S'S~
O O O
~ .O. ' O ~ .S' ' O
wC.S'C~ C N ~O~N~ C O C wC~S.C' O ~ O
wN.S.N~ ~O.C'O~ \N~N \N~N ~C,N~C' O /~- S
wC.S'O' ~g'C'S~ ~N'C'O~ ~N~N S
O ~N~N
O O
. N' , N/ wC.OwC' w N.OwC' wO.OwC' wN
The identification of an appropriate framework geometry and size for ligand domain presentation are important steps in the construction of a multibinding compound with enhanced activity. Systematic spatial searching strategies can be used to aid in the identification of preferred frameworks through an iterative process. FIG. 2 illustrates a useful strategy for determining an optimal framework display orientation for ligand domains. Various other strategies are known to those skilled in the art of molecular design and can be used for preparing compounds of this invention.
As shown in FIG. 2, display vectors around similar central core structures such as a phenyl structure and a cyclohexane structure can be varied, as can the spacing of the ligand domain from the core structure (i.e., the length of the attaching moiety). It is to be noted that core structures other than those shown here can be used for determining the optimal framework display orientation of the ligands. The process may require the use of multiple copies of the same central core structure or combinations of different types of display cores.
The above-described process can be extended to trimers (FIG. 3) and compound of higher valency.
Assays of each of the individual compounds of a collection generated as described above will lead to a subset of compounds with the desired enhanced activities (e.g., potency, selectivity, etc.). The analysis of this subset using a technique such as Ensemble Molecular Dynamics will provide a framework orientation that favors the properties desired. A wide diversity of linkers is commercially available (see, e.g., Available Chemical Directory (ACD)). Many of the linkers that are suitable for use in this invention fall into this category.
Other can be readily synthesized by methods well known in the art and/or are described below.
Having selected a preferred framework geometry, the physical properties of the linker can be optimized by varying the chemical composition thereof.
The composition of the linker can be varied in numerous ways to achieve the desired physical properties for the multibinding compound.
It can therefore be seen that there is a plethora of possibilities for the composition of a linker. Examples of linkers include aliphatic moieties, aromatic moieties, steroidal moieties, peptides, and the like. Specific examples are peptides or polyamides, hydrocarbons, aromatic groups, ethers, lipids, cationic or anionic groups, or a combination thereof.
Examples are given below, but it should be understood that various changes may be made and equivalents may be substituted without departing from the true spirit and scope of the invention. For example, properties of the linker can be modified by the addition or insertion of ancillary groups into or onto the linker, for example, to change the solubility of the multibinding compound (in water, fats, lipids, biological fluids, etc.), hydrophobicity, hydrophilicity, linker 5 flexibility, antigenicity, stability, and the like. For example, the introduction of one or more polyethylene glycol) (PEG) groups onto or into the linker enhances the hydrophilicity and water solubility of the multibinding compound, increases both molecular weight and molecular size and, depending on the nature of the unPEGylated linker, may increase the in vivo retention time. Further PEG may 10 decrease antigenicity and potentially enhances the overall rigidity of the linker.
Ancillary groups which enhance the water solubility/hydrophilicity of the linker and, accordingly, the resulting multibinding compounds are useful in practicing this invention. Thus, it is within the scope of the present invention to 15 use ancillary groups such as, for example, small repeating units of ethylene glycols, alcohols, polyols (e.g., glycerin, glycerol propoxyIate, saccharides, including mono-, oligosaccharides, etc.), carboxylates (e.g., small repeating units of glutamic acid, acrylic acid, etc.), amines (e.g., tetraethylenepentamine), and the like) to enhance the water solubility and/or hydrophilicity of the multibinding 20 compounds of this invention. In preferred embodiments, the ancillary group used to improve water solubility/hydrophilicity will be a polyether .
The incorporation of lipophilic ancillary groups within the structure of the linker to enhance the lipophilicity and/or hydrophobicity of the multibinding 25 compounds described herein is also within the scope of this invention.
Lipophilic groups useful with the linkers of this invention include, by way of example only, aryl and heteroaryl groups which, as above, may be either unsubstituted or substituted with other groups, but are at least substituted with a group which allows their covalent attachment to the linker. Other lipophilic groups useful __53_ with the linkers of this invention include fatty acid derivatives which do not form bilayers in aqueous medium until higher concentrations are reached.
Also within the scope of this invention is the use of ancillary groups 5 which result in the multibinding compound being incorporated or anchored into a vesicle or other membranous structure such as a liposome or a micelle. The term "lipid" refers to any fatty acid derivative that is capable of forming a bilayer or a micelle such that a hydrophobic portion of the lipid material orients toward the bilayer while a hydrophilic portion orients toward the aqueous phase.
10 Hydrophilic characteristics derive from the presence of phosphato, carboxylic, sulfato, amino, sulfhydryl, vitro and other like groups well known in the art.
Hydrophobicity could be conferred by the inclusion of groups that include, but are not limited to, long chain saturated and unsaturated aliphatic hydrocarbon groups of up to 20 carbon atoms and such groups substituted by one or more 15 aryl, heteroaryl, cycloalkyl, and/or heterocyclic group(s). Preferred lipids are phosphglycerides and sphingolipids, representative examples of which include phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, phosphatidic acid, palmitoyleoyl phosphatidylcholine, lysophosphatidylcholine, lysophosphatidyl-ethanolamine, 20 dipalmitoylphosphatidylcholine, dioleoylphosphatidylcholine, distearoyl-phosphatidylcholine or dilinoleoylphosphatidylcholine could be used. Other compounds lacking phosphorus, such as sphingolipid and glycosphingolipid families are also within the group designated as lipid. Additionally, the amphipathic lipids described above may be mixed with other lipids including 25 triglycerides and sterols.
The flexibility of the linker can be manipulated by the inclusion of ancillary groups which are bulky and/or rigid. The presence of bulky or rigid groups can hinder free rotation about bonds in the linker or bonds between the 30 linker and the ancillary groups) or bonds between the linker and the functional groups. Rigid groups can include, for example, those groups whose conformational lability is restrained by the presence of rings and/or multiple bonds within the group, for example, aryl, heteroaryl, cycloalkyl, cycloalkenyl, and heterocyclic groups. Other groups which can impart rigidity include polypeptide groups such as oligo- or polyproline chains.
Rigidity may also be imparted by internal hydrogen bonding or by hydrophobic collapse.
Bulky groups can include, for example, large atoms, ions (e.g., iodine, sulfur, metal ions, etc.) or groups containing large atoms, polycyclic groups, including aromatic groups, non-aromatic groups and structures incorporating one or more carbon-carbon multiple bonds (i.e., alkenes and alkynes). Bulky groups can also include oligomers and polymers which are branched- or straight-chain species. Species that are branched are expected to increase the rigidity of the structure more per unit molecular weight gain than are straight-chain species.
In preferred embodiments, rigidity is imparted by the presence of cyclic groups (e.g., aryl, heteroaryl, cycloalkyl, heterocyclic, etc.). In other preferred embodiments, the linker comprises one or more six-membered rings. In still further preferred embodiments, the ring is an aryl group such as, for example, phenyl or naphthyl.
Rigidity can also be imparted electrostatically. Thus, if the ancillary groups are either positively or negatively charged, the similarly charged ancillary groups will force the presenter linker into a configuration affording the maximum distance between each of the like charges. The energetic cost of bringing the like-charged groups closer to each other will tend to hold the linker in a configuration that maintains the separation between the like-charged ancillary groups. Further ancillary groups bearing opposite charges will tend to be ' PCT/US99/11789 attracted to their oppositely charged counterparts and potentially may enter into both inter- and intramolecular ionic bonds. This non-covalent mechanism will tend to hold the linker into a conformation which allows bonding between the oppositely charged groups. The addition of ancillary groups which are charged, 5 or alternatively, bear a latent charge when deprotected, following addition to the linker, include deprotectation of a carboxyl, hydroxyl, thiol or amino group by a change in pH, oxidation, reduction or other mechanisms known to those skilled in the art which result in removal of the protecting group, is within the scope of this invention.

In view of the above, it is apparent that the appropriate selection of a linker group providing suitable orientation, restricted/unrestricted rotation, the desired degree of hydrophobicity/hydrophilicity, etc. is well within the skill of the art. Eliminating or reducing antigenicity of the multibinding compounds 15 described herein is also within the scope of this invention. In certain cases, the antigenicity of a multibinding compound may be eliminated or reduced by use of groups such as, for example, polyethylene glycol).
As explained above, the multibinding compounds described herein 20 comprise 2-10 ligands attached to a linker that links the ligands in such a manner that they are presented to the target receptor for multivalent interactions with ligand binding sites thereon/therein. The linker spatially constrains these interactions to occur within dimensions defined by the linker. This and other factors increases the biological activity of the multibinding compound as 25 compared to the same number of ligands made available in monobinding form.
The compounds of this invention are preferably represented by the empirical formula (L)p(X)q where L, X, p and q are as defined above. This is intended to include the several ways in which the ligands can be linked together in order to achieve the objective of multivalency, and a more detailed explanation is described below.
As noted previously, the linker may be considered as a framework to 5 which ligands are attached. Thus, it should be recognized that the iigands ca>z be attached at any suitable position on this framework, for example, at the termini of a linear chain or at any intermediate position.
The simplest and most preferred multibinding compound is a bivalent 10 compound which can be represented as L-X-L, where each L is independently a ligand which may be the same or different and each X is independently the linker.
Examples of such bivalent compounds are provided in FIG. 2 where each shaded circle represents a ligand. A trivalent compound could also be represented in a linear fashion, i.e., as a sequence of repeated units L-X-L-X-L, in which L is a 15 ligand and is the same or different at each occurrence, as can X. However, a trimer can also be a radial multibinding compound comprising three ligands attached to a central core, and thus represented as (LAX, where the linker X
could include, for example, an aryl or cycloalkyl group. Illustrations of trivalent and tetravalent compounds of this invention are found in FIG.s 3 and 4 20 respectively where, again, the shaded circles represent ligands.
Tetravalent compounds can be represented in a linear array, e.g., L-X-L-X-L-X-L
25 in a branched array, e.g., L-X-L-X-L

(a branched construct analogous to the isomers of butane -- n-butyl, iso-butyl, sec-butyl, and t-butyl) or in a tetrahedral array, e.g., L~ ~L
X
... ,, L 'L
where X and L are as defined herein. Alternatively, it could be represented as an 5 alkyl, aryl or cycloalkyl derivative as above with four (4) ligands attached to the core linker.
The same considerations apply to higher multibinding compounds of this invention containing 5-10 ligands as illustrated in FIG. 5 where, as before, the 10 shaded circles represent ligands. However, for multibinding agents attached to a central linker such as aryl or cycloalkyl, there is a self-evident constraint that there must be sufficient attachment sites on the linker to accommodate the number of ligands present; for example, a benzene ring could not directly accommodate more than 6 ligands, whereas a mufti-ring linker (e.g., biphenyl) could 15 accommodate a larger number of ligands.
Certain of the above described compounds may alternatively be represented as cyclic chains of the form:
L
X X
-L
and variants thereof.

__Sg__ All of the above variations are intended to be within the scope of the invention defined by the formula (L)P(X)q.
With the foregoing in mind, a preferred linker may be represented by the 5 following formula:
-Xa-Z-(Ya-Z)m Yb_Z-Xa_ in which:
10 m is an integer of from 0 to 20;
Xa at each separate occurrence is selected from the group consisting of -O-, -S-, -NR-, -C(O)-, -C(O)O-, -C(O)NR-, -C(S), -C(S)O-, -C(S)NR- or a covalent bond where R is as defined below;
Z is at each separate occurrence is selected from the group consisting of 15 alkylene, substituted alkylene, cycloalkylene, substituted cylcoalkylene, alkenylene, substituted alkenylene, alkynylene, substituted alkynylene, cycloalkenylene, substituted cycloalkenylene, arylene, heteroarylene, heterocyclene, or a covalent bond;
Ya and Yb at each separate occurrence are selected from the group 20 consisting of:

~O O O
~N~ ~
N' R' R R~ R.
R' N iR, ~N, \ ~ -O
/ \ N' \ P-O-O R' R
Xa -S(O)S-CR'R"-N O N N~
I -S(O)~-NR'-R' -S-S- or a covalent bond;
in which:
n is 0, 1 or 2; and R, R' and R" at each separate occurrence are selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, cycloalkenyl, substituted cycloalkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl and heterocyclic.
Additionally, the linker moiety can be optionally substituted at any atom therein by one or more alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, cycloalkenyl, substituted cycloalkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl and heterocyclic group.
In one embodiment of this invention, the linker (i.e., X or X') is selected from those shown in Table II:

-_(lp-_ Table II
Linker -HN-(CHZ) ,-NH-C(O)-(CHZ)-C{O)-NH-(CHZ) z-NH--HN-(CHZ) 2-NH-C(O)-(CHI) 2-C(O)-NH-(CHI) 2-NH--HN-(CHz),-NH-C(O)-(CHZ),-C{O)-NH-(CHZ) z-NH--HN-(CHZ) Z-NH-C(O)-(CHZ),-C(O)-NH-(CHZ) Z-NH--HN-(CHZ) Z-NH-C(O)-(CHZ)5-C(O)-NH-(CHz) 2-NH--HN-(CH,) ~-NH-C(O)-(CH,)6-C(O)-NH-(CH,) ,-NH--HN-(CHZ) ,-NH-C(O)-(CHz),-C(O)-NH-(CHI) ,-NH--HN-(CHZ) 2-NH-C(O)-(CH2)s-C(O)-NH-(CHZ) z-NH--HN-(CHZ) 2-NH-C(O)-(CHZ)9-C(O)-NH-(CHZ) 2-NH--HN-(CHZ) Z-NH-C(O)-{CHZ),o-C(O)-NH-(CHZ) 2-NH--HN-(CHZ) Z-NH-C(O)-(CH2)"-C(O)-NH-(CHZ) Z-NH--HN-(CHz) 2-NH-C(O)-(CHZ),2-C(O)-NH-(CHz) z-NH--HN-(CHZ) 2-NH-C(O)-Z-C(O)-NH-(CHI) i-NH- where Z is 1,2-phenyl -HN-(CHI) 2-NH-C(O)-Z-C(O)-NH-(CH2) 2-NH- where Z is 1,3-phenyl -HN-(CH,) 2-NH-C(O)-Z-C(O)-NH-(CHz) 2-NH- where Z is 1,4-phenyl -HN-(CHZ) Z-NH-C(O)-Z-O-Z-C(O)-NH-(CHZ) Z-NH- where Z is 1,4-phenyl -HN-(CHZ) 2-NH-C(O)-(CHz) 2-CH(NH-C(O)-(CHZ)8-CH3)-C(O)-NH-(CHZ)2-NH--HN-(CHZ) 2-NH-C(O)-(CHZ)-O-(CHz)-C(O)-NH-(CHZ)z-NH--HN-(CHz) 2-NH-C(O)-Z-C(O)-NH-(CHz) 2-NH-where Z is 5-(n-octadecyloxy)-1,3-phenyl -HN-{CH,) z-NH-C(O)-(CHZ) ~-CH(NH-C(O)-Z)-C{O}-NH-(CH2) where Z is 4-biphenyl -HN-(CHZ) 2-NH-C(O)-Z-C(O)-NH-(CH~)z-NH-where Z is 5-{n-butyloxy)-1,3-phenyl Linker -HN- _(CHz) ,-NH-C(O)-(CH,)g-traps-(CH=CH)-C(O)-NH-(CH,) z-NH--HN-(CH,) _ _~-NH-C(O)-(CHZ),-CH(NH-C(O)-(CH,),~-CH3)-C(O)-NH-(CH~),-NH--HN-(CH,),-NH-C(O)-(CHZ) ~-CH(NH-C(O)-Z)-C(O)-NH-(CHZ) Z-NH-where Z is 4-(n-octyl)-phenyl -HN-(CH,)-Z-O-(CHZ)6-O-Z-(CHI)-NH- where Z is 1,4-phenyl -HN-( _CHZ)2-NH-C(O)-(CHZ)2-NH-C(O)-(CHZ)3-C(O)-NH-(CH,)2-C(O)-NH-(CH~)z-NH--HN-(CH~) ,-NH-C(O)-(CHI) 2-CH(NH-C(O)-Ph)-C(O)-NH-(CH,) ~-NH-- -HN-(CHZ) ~-NH-C(O)-(CHZ)-N+((CHZ)9-CH3)(CH~-C(O)-NH-(CHI) 2-NH,)-(CH,)-C(O)-NH-(CHZ) Z-NH--HN-(CHZ) z-NH-C(O)-(CH,)-N((CH2~-CH3)-(CHZ)-C(O)-NH-(CHz) z-NH--HN-{CHz) ,-NH-C(O)-(CHZ) z-NH-C(O)-(CHZ),-NH-C(O)-(CHZ) 3-C(O)-NH-(CHZ) 2-C(O)-NH-(CHZ) 2-C(O)-NH-(CHZ)2-NH--HN-(CHZ) Z-NH-C(O)-Z-C(O)-NH-(CHZ) Z-NH-where Z is 5-hydroxy-1,3-phenyl In another embodiment of this invention, the linker (i.e., X, X' or X") has the formula:
Ib Ib Re O-CH-CH O-Ra n' wherein each Ra is independently selected from the group consisting of a covalent bond, alkylene, substituted alkylene and aryiene;
each Rb is independently selected from the group consisting of hydrogen, alkyl and substituted alkyl; and n' is an integer ranging from 1 to about 20.

In yet another embodiment, the linker (i.e., X or X') has the formula:
-(CH2)n~-, where n' is an integer ranging from 1 to about 20, preferably from to 6.
In view of the above description of the linker, it is understood that the term "linker" when used in combination with the term "multibinding compound"
includes both a covalently contiguous single linker (e.g., L-X-L) and multiple covalently non-contiguous linkers (L-X-L-X-L) within the multibinding compound.
The multibinding compounds of this invention can be prepared from readily available starting materials using the following general methods and procedures. It will be appreciated that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.
Additionally, as will be apparent to those skilled in the art, conventional protecting groups may be necessary to prevent certain functional groups from undergoing undesired reactions. The choice of a suitable protecting group for a particular functional group as well as suitable conditions for protection and deprotection are well known in the art. For example, numerous protecting groups, and their introduction and removal, are described in T. W. Greene and G. M. Wuts, Protecting Groups in Organic Synthesis, Second Edition, Wiley, New York, 1991, and references cited therein.

Any compound which inhibits or binds to microsomal triglyceride transferase protein can be used as a ligand in this invention. As discussed in further detail below, numerous such compounds are known in the art and any of these known compounds or derivatives thereof may be employed as ligands in this invention. Typically, a compound selected for use as a ligand will have at least one functional group, such as an amino, amido, hydroxyl, thiol or carboxyl group and the like, which allows the compound to be readily coupled to the linker. Compounds having such functionality are either known in the art or can be prepared by routine modification of known compounds using conventional reagents and procedures. The patents and publications set forth below provide numerous examples of suitably functionalized compounds with inhibit or bind to microsomal triglyceride transferase protein and intermediates thereof which may be used as ligands in this invention.
The ligand can be covalently attached to the linker through any available position on the Iigand, provided that when the ligand is attached to the linker, the ligand retains its ability to inhibit or bind to microsomal triglyceride transferase protein.
A preferred group of ligands for use in this invention are those ligands having formulae IA-IE. A more preferred group of ligands are those having formulae IIA-IIE. Examples of multibinding compounds of this invention having specific ligands are illustrated in Figures 6-8, wherein - represents the linker.
Ligands of formula IA-IE and IIA-IIE are either known in the art or can be readily prepared using art-recognized starting materials, reagents and reaction conditions. By way of illustration, the following patents and publications disclose compounds, intermediates and procedures useful in the preparation of ligands of formulae IA-IE and IIA-IIE or related compounds suitable for use in this invention: U.S. Patent No. 5,712,279, issued January 27, 1998 to Biller et al.;

U.S. Patent No. 5,739,135, issued April 14, 1998 to Biller et al.; U.S. Patent No. 5,760,246, issued June 2, 1998 to Biller et al.; U.S. Patent No.
5,827,875, issued October 27, 1998 to Dickson Jr. et al.; U.S. Statutory Invention Registration No. H1729, published May 5, 1998 by Biller et al.; WO 96/40640, published December 19, 1996; WO 97/26240, published July 24, 1997; WO
97/43255, published November 20, 1997; WO 98/03069, published January 29, 1998; WO 98/03174, published January 29, 1998; WO 98/23593, published June 4, 1998; WO 98/27979, published July 2, 1998; WO 98/31225, published July 23, 1998; WO 98/31366, published July 23, 1998; WO 98/31367, published July 23, 1998; and EP 0 643 057 A1, published March 15, 1995. Each of these patents and publications is incorporated herein by reference in its entirety to the same extent as if each individual patent or publication was specifically and individually indicated to be incorporated by reference in its entirety. The syntheses of specific ligands or ligand precusors (i.e., Synthons A-I) are described in further detail in the Examples set forth below.
The compounds of formula I are typically prepared by coupling two or more ligands to a linker using conventional coupling procedures. Such coupling reactions are typically conducted by reacting to complimentary functional groups, such as carboxylic acid and an amine, to form a stable covalent bond, e.g. an amide. Suitable complimentary functional groups are described herein above.
By way of example, two or more ligands containing a carboxylic acid functional group can be coupled with a polyamine to form a polyamide as illustrated in Scheme 1 (where for purposes of illustration, R represents the ligand and R' represents the linking group).

O O O
-R OH + HZN- R NH2 R ~ R p R
Typically, this reaction is conducted by first activating the carboxylic acid, i.e., by conversion to a carboxylic acid anhydride or acid halide, and then coupling the activated carboxylic acid with a polyamine. For example, a carboxylic acid may be converted into a mixed trifluoroacetic anhydride by contacting the carboxylic acid with one molar equivalent of trifluoroacetic anhydride in an inert diluent, such as THF, at ambient temperature for about 0.5 to 6 hours. The resulting mixed anhydride intermediate is typically not isolated, but is contacted in situ with a polyamine having one molar equivalent of amine groups. This reaction is typically conducted in an inert diluent, such as THF, at a temperature ranging from about 0°C to about 100°C for about 1 to 48 hours.
Upon completion of the reaction, the compound of formula I is typically isolated and purified using conventional techniques, extraction, precipitation, chromatography, filtration, and the like.
Additionally, if desired, the carboxylic acid can be converted into an acid halide and the acid halide coupled with a polyamine to provide compounds of formula I. Acid halides can be prepared by contacting the carboxylic acid with an inorganic acid halide, such as thiony! chloride, phosphorous trichloride, phosphorous tribromide or phosphorous penta-chloride, or preferably, with oxalyl chloride under conventional conditions. Generally, this reaction is conducted using about 1 to 5 molar equivalents of the inorganic acid halide or oxalyl chloride, either neat or in an inert solvent, such as dichloromethane or carbon tetrachloride, at temperature in the range of about 0°C to about 80°C for about 1 to about 48 hours. A catalyst, such as DMF, may also be used in this reaction.
The acid halide is then contacted with a polyamine containing one molar equivalent of amino groups in an inert diluent, such as dichloromethane, at a temperature ranging from about -70°C to about 40°C for about 1 to about 24 hours. Preferably, this reaction is conducted in the presence of a suitable base to scavenge the acid generated during the reaction. Suitable bases include, by way of example, tertiary amines, such as triethylamine, diisopropylethylamine, N
methylmorpholine and the like. Alternatively, the reaction can be conducted under Schotten-Baumann-type conditions using aqueous alkali, such as sodium hydroxide and the like. Upon completion of the reaction, the compound of formula I is recovered by conventional methods including neutralization, extraction, precipitation, chromatography, filtration, and the like.
Alternatively, the carboxylic acid may be directed coupled with the polyamine using well-known coupling reagents such as carbodiimides, BOP
reagent (benzotriazol-1-yloxy-tris(dimethylamino)phosphonium hexafluorophosphonate) and the like. Suitable carbodiimides include, by way of example, dicyclohexylcarbodiimide (DCC), 1-(3-dimethylamino-propyl)-3-ethylcarbodiimide (EDC) and the like. If desired, polymer supported forms of carbodiimide coupling reagents may also be used including, for example, those described in Tetrahedron Levers, 34(48), 7685 {1993). Additionally, well-known coupling promoters, such as N-hydroxysuccinimide, 1-hydroxybenzotriazole and the like, may be used to facilitate the coupling reaction.
This coupling reaction is typically conducted by contacting the carboxylic acid with about 1 to about 2 equivalents of the coupling reagent and with a polyamine containing one molar equivalent of amino groups in an inert diluent, such as dichloromethane, chloroform, acetonitrile, tetrahydrofuran, N,N

__67__ dimethylformamide and the like. Generally, this reaction is conducted at a temperature ranging from about 0°C to about 37°C for about 12 to about 24 hours. Upon completion of the reaction, the compound of formula I is recovered by conventional methods including neutralization, extraction, precipitation, 5 chromatography, filtration, and the like.
Compounds of formula I can also be prepared by reacting two or more ligands containing a primary or secondary amine functional group with an alkyl poly(halide) to form a polyamine as shown in Scheme 2 (where for purposes of 10 illustration, R represents a portion of the ligand or hydrogen and R' represents the linking group).
O O
R-NH ~ - --.~ ~N- ~ -N/
+ Br- R Br R ( R I R
R R R
15 This coupling reaction is typically conducted by contacting the amine compound with a suitable alkali or alkaline earth metal base, such as potassium carbonate, in an inert diluent, such as DMF and the like, in the presence of a alkyl poly(halide) having one molar equivalent halide groups. Generally, this reaction is conducted at a temperature ranging from about 25°C to about 100°C
20 for about 24 to about 72 hours. Optionally, a catalytic amount of sodium or potassium iodide may be added to the reaction mixture when an alkyl poly(chloride) or poly(bromide) is employed in the reaction. Upon completion of the reaction, the compound of formula I is recovered by conventional methods including neutralization, extraction, precipitation, chromatography, filtration, and 25 the like.

__6g__ Other methods and reaction conditions for preparing compounds of formula I are described in further detail in the Examples set forth below. As will be readily apparent to those of ordinary skill in the art, the synthetic procedures described herein or those known in the art may be readily modified to afford a 5 wide variety of compounds within the scope of this invention.
The methods described herein lend themselves to combinatorial approaches for identifying multimeric compounds which possess multibinding 10 properties.
Specifically, factors such as the proper juxtaposition of the individual ligands of a multibinding compound with respect to the relevant array of binding sites on a target or targets is important in optimizing the interaction of the 15 muitibinding compound with its targets) and to maximize the biological advantage through multivalency. One approach is to identify a library of candidate multibinding compounds with properties spanning the multibinding parameters that are relevant for a particular target. These parameters include: (1) the identity of ligand(s), (2) the orientation of ligands, (3) the valency of the 20 construct, (4) linker length, {5) linker geometry, (6) linker physical properties, and (7) linker chemical functional groups.
Libraries of multimeric compounds potentially possessing multibinding properties (i.e., candidate multibinding compounds) and comprising a multiplicity 25 of such variables are prepared and these libraries are then evaluated via conventional assays corresponding to the ligand selected and the multibinding parameters desired. Considerations relevant to each of these variables are set forth below:

A single ligand or set of ligands is (are) selected for incorporation into the libraries of candidate multibinding compounds which library is directed against a particular biological target or targets, i.e., inhibition of MTP. The only requirement for the ligands chosen is that they are capable of interacting with the selected target(s). Thus, ligands may be known drugs, modified forms of known drugs, substructures of known drugs or substrates of modified forms of known drugs (which are competent to interact with the target), or other compounds.
Ligands are preferably chosen based on known favorable properties that may be projected to be carried over to or amplified in multibinding forms. Favorable properties include demonstrated safety and efficacy in human patients, appropriate PK/ADME profiles, synthetic accessibility, and desirable physical properties such as solubility, loge, etc. However, it is crucial to note that ligands which display an unfavorable property from among the previous list may obtain a more favorable property through the process of multibinding compound formation; i.e., ligands should not necessarily be excluded on such a basis.
For example, a ligand that is not sufficiently potent at a particular target so as to be efficacious in a human patient may become highly potent and efficacious when presented in multibinding form. A ligand that is potent and efficacious but not of utility because of a non-mechanism-related toxic side effect may have increased therapeutic index (increased potency relative to toxicity) as a multibinding compound. Compounds that exhibit short in vivo half lives may have extended half lives as multibinding compounds. Physical properties of ligands that limit their usefulness (e.g. poor bioavailability due to low solubility, hydrophobicity, hydrophilicity) may be rationally modulated in multibinding forms, providing compounds with physical properties consistent with the desired utility.
Several points are chosen on each iigand at which to attach the ligand to the linker. The selected points on the ligand/linker for attachment are functionalized to contain complementary reactive functional groups. This permits probing the effects of presenting the ligands to their target binding sites) in multiple relative orientations, an important multibinding design parameter.
The only requirement for choosing attachment points is that attaching to at least 5 one of these points does not abrogate activity of the ligand. Such points for attachment can be identified by structural information when available. For example, inspection of a co-crystal structure of a ligand bound to its target allows one to identify one or more sites where linker attachment will not preclude the ligand/target interaction. Alternatively, evaluation of ligand/target binding by 10 nuclear magnetic resonance will permit the identification of sites non-essential for ligand/target binding. See, for example, Fesik, et al., U.S. Patent No.
5,891,643, the disclosure of which is incorporated herein by reference in its entirety. When such structural information is not available, utilization of structure-activity relationships (SAR) for ligands will suggest positions where 15 substantial structural variations are and are not allowed. In the absence of both structural and SAR information, a library is merely selected with multiple points of attachment to allow presentation of the ligand in multiple distinct orientations.
Subsequent evaluation of this library will indicate what positions are suitable for attachment.

It is important to emphasize that positions of attachment that do abrogate the activity of the monomeric ligand may also be advantageously included in candidate multibinding compounds in the library provided that such compounds bear at least one ligand attached in a manner which does not abrogate intrinsic 25 activity. This selection derives from, for example, heterobivalent interactions within the context of a single target molecule. For example, consider a ligand bound to its target, and then consider modifying this ligand by attaching to it a second copy of the same ligand with a linker which allows the second ligand to interact with the same target at sites proximal to the first binding site, which 30 include elements of the target that are not part of the formal ligand binding site __~1__ and/or elements of the matrix surrounding the formal binding site, such as the membrane. Here, the most favorable orientation for interaction of the second ligand molecule may be achieved by attaching it to the linker at a position which abrogates activity of the ligand at the first binding site. Another way to consider 5 this is that the SAR of individual ligands within the context of a multibinding structure is often different from the SAR of those same ligands in momomeric form.
The foregoing discussion focused on bivalent interactions of dimeric 10 compounds bearing two copies of the same ligand joined to a single linker through different attachment points, one of which may abrogate the binding/activity of the monomeric ligand. It should also be understood that bivalent advantage may also be attained with heterodimeric constructs bearing two different ligands that bind to common or different targets.

Once the ligand attachment points have been chosen, one identifies the types of chemical linkages that are possible at those points. The most preferred types of chemical linkages are those that are compatible with the overall structure of the ligand (or protected forms of the ligand) readily and generally formed, 20 stable and intrinsically innocuous under typical chemical and physiological conditions, and compatible with a large number of available linkers. Anude bonds, ethers, amines, carbamates, ureas, and sulfonamides are but a few examples of preferred linkages.
25 Linker Selection In the library of linkers employed to generate the library of candidate multibinding compounds, the selection of linkers employed in this library of linkers takes into consideration the following factors:

_-~2__ Valency In most instances the library of linkers is initiated with divalent linkers. The choice of ligands and proper juxtaposition of two ligands relative to their binding sites permits such molecules to exhibit target binding affinities and specificities more than sufficient to confer biological advantage.
Furthermore, divalent linkers or constructs are also typically of modest size such that they retain the desirable biodistribution properties of small molecules.
Linker Len~~ Linkers are chosen in a range of lengths to allow the spanning of a range of inter-ligand distances that encompass the distance preferable for a given divalent interaction. In some instances the preferred distance can be estimated rather precisely from high-resolution structural information of targets. In other instances where high-resolution structural information is not available, one can make use of simple models to estimate the maximum distance between binding sites either on adjacent receptors or at different locations on the same receptor. In situations where two binding sites are present on the same target (or target subunit for multisubunit targets), preferred linker distances are 2-20 ~, with more preferred linker distances of 3-12 ~.
In situations where two binding sites reside on separate target sites, preferred linker distances are 20-100 ~, with more preferred distances of 30-70 f~.
I,,'Ln_k_er Geometry and Rigidity: The combination of ligand attachment site, linker length, linker geometry, and linker rigidity determine the possible ways in which the ligands of candidate multibinding compounds may be displayed in three dimensions and thereby presented to their binding sites. Linker geometry and rigidity are nominally determined by chemical composition and bonding pattern, which may be controlled and are systematically varied as another spanning function in a multibinding array. For example, linker geometry is varied by attaching two ligands to the ortho, meta, and para positions of a benzene ring, or in cis- or traps-arrangements at the 1,1- vs. 1,2- vs. 1,3-vs.
1,4- positions around a cyclohexane core or in cis- or traps-arrangements at a __73__ point of ethylene unsaturation. Linker rigidity is varied by controlling the number and relative energies of different conformational states possible for the linker. For example, a divalent compound bearing two ligands joined by 1,8-octyl linker has many more degrees of freedom, and is therefore less rigid than a compound in which the two ligands are attached to the 4,4' positions of a biphenyl linker.
l~in_ker Phvsicai_ Propgrties: The physical properties of linkers are nominally determined by the chemical constitution and bonding patterns of the linker, and linker physical properties impact the overall physical properties of the candidate multibinding compounds in which they are included. A range of linker compositions is typically selected to provide a range of physical properties (hydrophobicity, hydrophilicity, amphiphilicity, polarization, acidity, and basicity) in the candidate multibinding compounds. The particular choice of linker physical properties is made within the context of the physical properties of the ligands they join and preferably the goal is to generate molecules with favorable PK/ADME properties. For example, linkers can be selected to avoid those that are too hydrophilic or too hydrophobic to be readily absorbed and/or distributed in vivo.
1 inker Chemical Flan .rinna rouns: Linker chemical functional groups are selected to be compatible with the chemistry chosen to connect linkers to the ligands and to impart the range of physical properties sufficient to span initial examination of this parameter.
Having chosen a set of n ligands (n being determined by the sum of the number of different attachment points for each ligand chosen) and m linkers by the process outlined above, a library of (n!)m candidate divalent multibinding compounds is prepared which spans the relevant multibinding design parameters for a particular target. For example, an array generated from two ligands, one which has two attachment points (A1, A2) and one which has three attachment points (B1, B2, B3) joined in all possible combinations provide for at least possible combinations of multibinding compounds:
S
A1-A1 Al-A2 A1-B1 A1-B2 Al-B3 A2-A2 A2-B1 A2-B2 When each of these combinations is joined by 10 different linkers, a library of 1S0 candidate multibinding compounds results.
Given the combinatorial nature of the library, common chemistries are preferably used to join the reactive functionalies on the ligands with complementary reactive functionalities on the linkers. The library therefore lends 1S itself to efficient parallel synthetic methods. The combinatorial library can employ solid phase chemistries well known in the art wherein the ligand and/or linker is attached to a solid support. Alternatively and preferably, the combinatorial libary is prepared in the solution phase. After synthesis, candidate multibinding compounds are optionally purified before assaying for activity by, ZO for example, chromatographic methods (e.g., HPLC).
Anal3rsis of the Library Various methods are used to characterize the properties and activities of the candidate multibinding compounds in the library to determine which 2S compounds possess multibinding properties. Physical constants such as solubility under various solvent conditions and IogD/clogD values are determined. A
combination of NMR spectroscopy and computational methods is used to determine low-energy conformations of the candidate multibinding compounds in fluid media. The ability of the members of the library to bind to the desired 30 target and other targets is determined by various standard methods, which include radioligand displacement assays for receptor and ion channel targets, and kinetic inhibition analysis for many enzyme targets. In vitro efficacy, such as for receptor agonists and antagonists, ion channel blockers, and antimicrobial activity, are also determined. Pharmacological data, including oral absorption, 5 everted gut penetration, other pharmacokinetic parameters and efficacy data are determined in appropriate models. In this way, key structure-activity relationships are obtained for multibinding design parameters which are then used to direct future work.
10 The members of the library which exhibit multibinding properties, as defined herein, can be readily determined by conventional methods. First those members which exhibit multibinding properties are identified by conventional methods as described above including conventional assays (both in vitro and in vivo).

Second, ascertaining the structure of those compounds which exhibit multibinding properties can be accomplished via art recognized procedures. For example, each member of the library can be encrypted or tagged with appropriate information allowing determination of the structure of relevant members at a later 20 time. See, for example, Dower, et al., International Patent Application Publication No. WO 93/06121; Brenner, et al., Proc. Natl. Acad. Sci., USA, 89:5181 (1992); Gallop, et al., U.S. Patent No. 5,846,839; each of which are incorporated herein by reference in its entirety. Alternatively, the structure of relevant multivalent compounds can also be determined from soluble and 25 untagged libaries of candidate multivalent compounds by methods known in the art such as those described by Hindsgaul, et al. , Canadian Patent Application No.
2,240,325 which was published on July 11, 1998. Such methods couple frontal amity chromatography with mass spectroscopy to determine both the structure and relative binding affinities of candidate multibinding compounds to receptors.

__ The process set forth above for dimeric candidate multibinding compounds can, of course, be extended to trimeric candidate compounds and higher analogs thereof.
5 Follow-up Synthesis and Analysis of Additional Libraries Based on the information obtained through analysis of the initial library, an optional component of the process is to ascertain one or more promising multibinding "lead" compounds as defined by particular relative ligand orientations, linker lengths, linker geometries, etc. Additional libraries can then 10 be generated around these leads to provide for further information regarding structure to activity relationships. These arrays typically bear more focused variations in linker structure in an effort to further optimize target affinity and/or activity at the target (antagonism, partial agonism, etc.), and/or alter physical properties. By iterative redesign/analysis using the novel principles of 15 multibinding design along with classical medicinal chemistry, biochemistry, and pharmacology approaches, one is able to prepare and identify optimal multibinding compounds that exhibit biological advantage towards their targets and as therapeutic agents.
20 To further elaborate upon this procedure, suitable divalent linkers include, by way of example only, those derived from dicarboxyIic acids, disulfonylhalides, dialdehydes, diketones, dihalides, diisocyanates,diamines, diols, mixtures of carboxylic acids, sulfonyihalides, aldehydes, ketones, halides, isocyanates, amines and diols. In each case, the carboxylic acid, sulfonylhalide, 25 aldehyde, ketone, halide, isocyanate, amine and diol functional group is reacted with a complementary functionality on the ligand to form a covalent linkage.
Such complementary functionality is well known in the art as illustrated in the following table:

Representative Complementa_rv Binding Chemicties First Reactive Group Second Reactive Group~j~g hydroxyl isocyanate carbamate 5 amine epoxide ~3-hydroxyamine sulfonyl halide amine sulfonamide carboxyl acid amine amide hydroxyl alkyl/aryl halide ether aldehyde amine( + reducing amine agent) 10 ketone amine(+ reducing agent)amine amine isocyanate urea Exemplary linkers include the following linkers identified as X-1 through X-418 as set forth below:

W0.99/63929 PCT/US99/11789 _ 78 _ D~ i 0 0 . a~ o . « al o all °~ a_ll ~~1''~ ~o woo ~o~o uo -~ ~: o~ay . o" . rtc w'QO ~~ . aS
xai x.71 x-fi xai x.fl x 0 oN o ~ ° ~ ~
.1 p~.M~~o : Ipso ~ ' °~° ~ wo 0 . ~ . Ho a5 ya a4 p Xa x.ti .-~° xwl xaol x.111 x.l _. I
p ~ ~ NO o ND - I NO i C~O ~ G
°" °II NO
O ~F
X.171 x.l~I X.IfI X.IeI X.t71 X~1 _ ~ N . a~, o, O
o~x a .~..... .w~ew "i"1 .o o r no -aJ
i x.ls~ x-7oi ~ xan ~o xa71 xs7i o _ ~ I °
° ~ ~ o~
I
I i xal Xat ~"' xr7 X.t, xari o a I
' ~ ~S
xall x-7z ~ x-77 x.7d _ xafl ~ I
r1~ i.° ~ ~ ° i oN
.,h/ a ow ~ a eK
X.711 X.7tl Xaf xa1 v'"'~~. Xall X.11 0 0" . p~--.~ p4 I no~o ~ ° ' O . o~o ', I p~ ~ _..
p file .« o ~ °) x,,7i x."I x.xsl xm 'x~ xrn x.n _ _.
a., ~ "°~ro ~-all -,_ _ ~ ' ~» ..
_J
a. i ~ -mw ~ a : ow ew o N0 a N0 W C

X~, "° a x-fe x-fl X-ril X-ffi r ~ ~ _ i ~~ a ° ° °~
all I o ' w e'~'-!~'~'~i'~ xf7 xat xsf x.7s w p~v"~° -.. I o / 1 0ll i .w t °~o~ ~ ~° ° 1 o j MD
1a~ i i o w i a °
X~11 xIf X.t7 X.fl1 xif, ° a~ i o1 0~ ~ 0- ; ee ~ o .~ . a p o : p~aH i ~~°~ j I
i p I a X.nV xat p X-w x.x xnl x-° rrrrrrrr ~p ; ~.,-~ , o o" ~
j-r-~-T_'rC ; p~~ ~ j HoMS~o Wa~ I ow w r s ° s s r f s o~ ~ i i wo al i ~...J..J~e.. °
X.n~ x-vv x-» x.»i x-n~ x~»
a °° ~ o . $ , ,!', ~' ' r ~.w p w wr ° ~°" ° ° .
No i a °",.°°a. ; °~"~j.°, pas eo"'ae X-»' x-al x-ul x-m x.t71 x-a WO'.99/63929 _ ,~9 - PCT/US99/11789 ~ ,~ " , .
~ ~' o ~'t r~ o 1~ o r, ~'~'~ ~'/~!~ "o~~....~o" " l'~ ' ~
1~/ ~."i e q ~
"
..., A ~ ~

w x.u. '~ x-IIi xall xall x.w a ~ o '~ 0 o 0 /~a ~ \ .. /l 1 s ~ ' ~ W
"o O
e~e~0 w o~/'~~~ w , " .
!

x ~. ." xa1 xall xall xas al o rr r.ry ~ w " In o it it rr " " ~ o ~~'"
~"
w I

I i t 0 xa1' x.111 74111 X-1111 X.111 Xr Fr Fc cc o o f ' Fr .~e'. o" ~> '.
"o~4~a" ~ ; i ~ .~~y~
i c f F ~o i F O ~
O f x.lm .~'~ x.lwi x.lasl x.1011 7a1 g 'gyp x.loi I y..~~ ~ ' !
we la r~ "~
o ' I
' I
w ~~w ' e o/.
i x.IMI y,/~j x.1111 X.113' 70.117X-1 - o ~~

o'~

" " '~ .
0 ~ W
C 11 ~ ~--O" 0 " I~ dN

.M " ~ I ~
x.lu7 ~ x-us x.ul1 I x.lnx.
_._~__- 0 ._ x.u1 a x.111; 7a17~x x-ml X.111 x.u1 I Info ~o~~e I a" " ... en ~/~" . w a x.lm x-111 x.lle x-ul x.lI

Dinll(onyl . I
Holides O O r o r ~ ' : ew-~-~~-a ~ ,~~/'~. I o ~ , o L." ~~r-.,a a a ~ o, ~ ir o 'e ,o a I I sl ' r- 1 . . .
0 , F
a o a o o a X.t771 741N %-1I ~~ x.171 x.111141 F g o a . ISe 0 a "v"~s O Oae~ 41~p ~s0 ",~1 a4 a , a I o : ( O a o '!/ i ~~~ ;.
~
o a o ~a x-1i x.la ~ x.ln~ 74.10x-t x-tall "P a r r ~,,e~ F ~o P or ,a ol, ~ p so ! g so " ~rl o;so , '~
~J~, I a ~
~ I

,o ~ p4 w l '~1~

X.IHI o~~r X.llia4 7f-1171 X.111 X.1117F1 a ' o,a ; 0. ~~~a 1's . iy o~ ~0 0 o v i x.uli x.lu ' s DVWeYyda ' 0~0 n I 6 r ~

,., x.l w x.ls x.lss' x.ISl1 x.ls1o 7c-1 W0 ,99/63929 PCT/US99/11789 ~.~/~!/ a :~ ~e~ M a I ~
~ ~ _.
~
~
/

~ ,. e r o~ a ~e O ~ a , ~
ef' ~e p o .
'~ a4 X.Ilf: X.11e1 74111 x.llil X.IpI
~. - oho ~ 741 _ ~ .
.~y'0 _ 0a o y ~e o a eN

X.1e71 %Ifl1 141111 x1111 x-1I
_ xl _ a ~~o I ~
o~o ' I

.,.. x.lnl x.lnl x.lnl x.n~i piY~er a p ~ ;
./\i'. i 1\~'n lr~h ul a~a . I
j i al I

e~~/
a a x.lr1 xln x.lw x.lls x-In erg" : ai\,io~a~,/~a~ ( W ai\~a ~\/\/~~I /"~h i ~
a4 a i x-ltll x.111 X.10 X-IN 161tf %

h . , r~ 1\/\~
a ~ a x-m x.la x-to X-ISO x-1n _ ~ 1 I
at I ' ~ I h r ~~

i I

x.1s71 x.lsi x.lss X.ISI x.lafX.I,t _-.. -h\/\/\/~h 1\/\/\W ~~ , ~
o o" 4~ON :
h ~h Ir ~
lMlr n I~

I N Q
N,G
a4.
C

X.INI x.701 X-7111 x-7ltl x,7171 - %

her h~p~er ' 1 1 ~
~~0 a a~a ~
h i ~
I ~u '\/~Ir l4Oro ,y, i 1 I 1.
h X.711 X.7til X71'1 X.Itl1 XJtl1 7Ftl \/\~'~/~/\rr . r ~ a ' _ i H~ _.-k x.7111 76717r Xd171 X-711 I I
suW I
-I

o a e~ ~ ~ ~ I
~ ~ O~N~N~O ~ , t ~

Np-a e-er,~ o x.7ls) x.7ls xals x.7ui xalsi ~.

~ . N
a ~ ~~ i ~ !
~ I o ~
o ~ ~e ~N~ ~ -~N~N~

o~Y~ ~lr h I ep ar, u4 : I
~ ~
i X-7tli x.77! X-171 X.1711 x-1tt '1' N a4 ~ e~ _.-f N
s,. - N~' ~ e~
:. e~ I a a iW /~./\~ ~
o~' ~
o ~
~
~e ~
a~aS

~ ~ r.
x.7711 i b .
1(.7771 X.77f1 x.77e1 x.7711 X.771 I Z S
~ ~e ~ oy N~
o~N~N~o , e~
I I ' ~
N
y .' ~ ' ~ 1~ I ~~
a ~
; 'K
' N ~ Xilfl X.y1 X.b71 X-fill X-itf a ~o p I
N .G~'N~" ~
''~o o I dV.
I N .
ww i ~ ~ ~

e , .
w : a p W I
~ I

X.as~ x-1u1 xs.l x-:.11 wx w xaal ~ ~N6 ~ ~'~
N - T
o~~~~ ~e N....

~ I I
_ ~

~ 1 f xs~el x.177 x.lu1 x.1f x-fwd i i ~ , ' I I
I
Di~oines . i i :~ ~'~N ~ ~ ~WMNM~K Mfl~
~,~.e~e~l'../'w/'NJ ~N~/ ~ Q4 aN i ~ ~''~.~~' I j ~ 1 X41! %-1~~ 7L771 X-1~ X3f1 N>!~/~' s ~ I ~~ ~ I
wwe ~

N I
X.tff X.7ff~ X.b7 7CJf1 X.111 ws~/~e~MneMp,y N~~/'vpw~ i I ~4 N/~~./~e~
I

wt 1(-011 Xdu ' X-~1 XJf1 Xy!

WI~N~GI~ MVP ~ ~V~J'~
M

Nfl e xu7 xafl xall xxn ~ xm ~ ~ e~~e~p~
i ~~ ~N~le~
I ~ Kp ~ i W a. : Y~~S

I I f I i I

x.1~ x.lsv xml clrei X.ln et lei ' ' I I
~~ ~/~ql KN~N
~ ~'~~ ~
. I
~J

I
l x-m x-ao xan Xm. x.la y'we.~.yow"". ~~~~ ~ ~yN.~w"4 yT ; "!~".~".
i ; we w xsu x.1,1 xln ~ Xaai x Xal1 nrwp~p~~;, ~,-~.~ "~"..~NN, i ~? ~ i I
~

X7f4 XJI! X.717 X-1f1 X.llf Xdlf q KovN~NNVw4 ' '4N~/~/W~ ~Y' 'I

I ~
X311 7(.7N X-If! X.701 XJ11 ~ I
.~fp~/ofNN~ ~'' ~~IN~
e~ ; i I
J ~l ' ~

"r, ~ .
r, i ;

X7m X.7111 X-lof XJel1 X.fC1 74Jef I I ~..~w, . P'~ -o a4 ~K
x.wl 1 xalel Xall l x.n71 xal xau WO 99/63929 _ g2 PCT/US99/11789 ~ . ' :
N~N/\/\N i F~t i KO dS ' I

Ko~Q1 i ~

X7111 Xa111 1(a171 X-111j X-7111 N,C\/NV/'~/~N~qyWlv/~./W : ~
y K0.N/~,~/Nvo4 NFvNMNnCK

j KN\~\O/~NK

I
xa:l x7a~ x-777 xalf xaal '' /~~ W" "0N
N ~ . /v./N~ . ~
C /\/~ ~

y ' ~ ~
n N0 oN
..
N

. i ON ' J .'!N/
NO~O~ ~pN 0 C

N ~ abl X1711 X1711 X.1711 ~t~ U xall 14777!

NO ~ O M .
~ ~0~~ W

o~ i ~o N,t~~. ~
K~~prf~oK . ~.,-, I
~"

~N~pN v\
~

X.I)11 Xal1 X-174 X.171 xa711 ~,~tNO~f~~ ~i.\i\s/~\o~ /a aN a r . ' I ~ aN
~ I ISP~~

,~ I '~' I' 'K

x.1111 IcnrJ . . xaa xaa xalli x.H7 N.

No oN "NI~sNN yVj oN j No~~'aN
' No N ~ "'o~~
N~~eN eN

x-7w xau xaw Iwl7 xaa ",N'~sNO~oN ~ o~aN Net Ne oN ~ No aN
NO

X.)1p x-711 Xal1 X)f) xaHl IIONO~ Np~ ON I , . ~OII
I ~~ON .
ND~~

I : pN ND\
KC~oN

I .
\ON

)lf~ Xal7~ X.1111 X.lff1 X.Hp1 xafl _.__. , aN qN
~ o v ~~aN
No~~w o Nod ' \\/i ", , vy/ Nod n ~
I " I
~

.. 1 ~

xx1 Xap x-1sn xa11 xxsf X.ln NoNO~o~o~oN ~ I No~oN aN
I N I
I ~ 1 1 No Ko eK
o~ I J I

ew l Xa"i x.H1 x.ne x.m xa711 x.

oN I ~oN j ~ ; ~ - a fK Ko ~ ~,~.~'~t"
a4 No n xn xn1xa71 xm xrn pN0\~/0\i\~ ~ ~N0~0NQ1 ~NO\~\oN I Hp/~oN1 pNf\fNON

X.111 Xal1 X->tt x.Jt1 X-f11 Xalf I I ~ i 1 I i I
i ' ~ ~ ~ NI ! NIfW
M~tN I . ' i I ' W' a Ns w L

f I
X.)1,1 xanl x.1111 x.Inl xafo~ x-I

~ ; w~~ ' y ~ IN ~
w~~0~~ w ~ ~ 111 MI 1 ~ ~ ~ 1 ' II

x-mi xan x.i xarsl x.~
.

w.w/~o~~aw ~ - i S i w~; ;
~ ~ wwall w oh w w I c~

i w~/ 1 1 i XJ111 X-~1~ X.11I \al 1c.111 74IQ

w ~ M.... ~ w~~~ wow 1 w~~l I

x,app x.IN 7GIOf Xal7 X.IOI
.

! ~ ~ w W
w w~~N

~s ' He : I A~
off N1 I il I

xmo~ xrlli x.sul x.mi xals~ x.W

"' : w - _.
w ~ w~N ~ 1 ~

n 1 i xp~~ 74117 X-111 -_gø-Representative ligands for use in this invention include, by way of example, ligands of formula IA-IE and IIA-IIE as defined herein.
Combinations of ligands (L) and linkers (X) per this invention include, by way example only, homo- and hetero-dimers wherein a first ligand is selected from formula IA through IE above and the second ligand and linker is selected from the following:

iA/X-169-IA/X-170- IA/X-171- IA/X-172-IA/X-209-IA/X-210- iAIX-211- IA/X-212- IA/X-213- IA/X-214-_-g6_ ~g'7_-IB/X-115-IB/X-116- IB/X-117- IB/X-118- IBlX-119- IB/X-120-__gg_ __g9__ IC/X-173-IC/X-174- IC/X-175- IC/X-1?6- IC/X-177- IC/X-178-IC/X-191-IC/X-192- IC/X-193- IC/X-194- iC/X-195- IC/X-196-IC/X-341- IC/X-342- IC/X-343- IC/X-344- IC/X-345- IC/X-34Cr IC/X-377- IC/X-378- IC/X-379- IC/X-380- IC/X-381- ' IC/X-382-ID/X-3?- ID/X-38-~ ID/X-39- ID/X-40- ID/X-41- ID/X-42-ID/X-43- ID/X-44- ID/X-45- ID/X-46- ID/X-4.7- ID/X-48-ID/X-163- ID/X-i64- ID/X-165- ID/X-166- ID/X-167- ID/X-168-ID/X-239- ID/X-240- ID/X-241- ID/X-242- IDlX-243- ID/X-244-iD/X-245- ID/X-246- ID/X-247- ID/X-248- ID/X-249- ID/X-250-ID/X-269- ID/X-270- ID/X-271- ID/X-2?2-ID/X-273- ID/X-274-iD/X-407- ID/X-408- ID/X-409- ID/X-410-ID/X-411- ID/X-412-IE/X-7- IE/X-8-~ IE/X-9- IE/X-10- IE/X-11- IE/X-12-IE/X-37- IE/X-38- IE/X-39- IE/X~0- IE/X-41- IE/X-42-IE/X-4.3-IE/X-44- IE/X-45- IE/X-46- IE/X-47- IE/X-48-IE/X-91- IE/X-92- IE/X-93- IE/X-94- IE/X-95- ~IE/X-96-~

provided that when the first ligand has formula IA or IB, where R' or R2 is a covalent bond linking the first ligand to the linker, then a second ligand does not have formula ID or IE, where R8 or R9 are a covalent bond linking the second ligand to the linker.

Eat ~ atiQn~
When employed as pharmaceuticals, the compounds of this invention are usually administered in the form of pharmaceutical compositions. These compounds can be administered by a variety of routes including oral, rectal, 10 transdermal, subcutaneous, intravenous, intramuscular, and intranasal.
These compounds are effective as both injectable and oral compositions. Such compositions are prepared in a manner well known in the pharmaceutical art and comprise at least one active compound.
15 This invention also includes pharmaceutical compositions which contain, as the active ingredient, one or more of the compounds described herein associated with pharmaceutically acceptable carriers. In making the compositions of this invention, the active ingredient is usually mixed with an excipient, diluted by an excipient or enclosed within such a carrier which can be in the form of a 20 capsule, sachet, paper or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), 25 ointments containing, for example, up to 10 % by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
In preparing a formulation, it may be necessary to mill the active compound to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it ordinarily is milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size is normally adjusted by milling to provide a substantially uniform distribution in the formulation, e.g. about 40 mesh.
Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, sterile water, syrup, and methyl cellulose.
The formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents;
preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents. The compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
The compositions are preferably formulated in a unit dosage form, each dosage containing from about 0.001 to about 1 g, more usually about 1 to about mg, of the active ingredient. The term "unit dosage forms" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material 25 calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Preferably, the compound of formula I above is employed at no more than about 20 weight percent of the pharmaceutical composition, more preferably no more than about 15 weight percent, with the balance being pharmaceutically inert carrier(s).
w 30 The active compound is effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It, will be understood, however, that the amount of the compound actually administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered and its relative activity, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid preformulation is then, subdivided into unit dosage forms of the type described above containing from, for example, 0.1 to about 500 mg of the active ingredient of the present invention.
The tablets or pills of the present invention may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.

__9g_ The liquid forms in which the novel compositions of the present invention may be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as corn oil, cottonseed oil, sesame oil, coconut 5 oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain 10 suitable pharmaceutically acceptable excipients as described supra.
Preferably the compositions are administered by the oral or nasal respiratory route for local or systemic effect. Compositions in preferably pharmaceutically acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be inhaled directly from the nebulizing device or the nebulizing device may be 15 attached to a face mask tent, or intermittent positive pressure breathing machine.
Solution, suspension, or powder compositions may be administered, preferably orally or nasally, from devices which deliver the formulation in an appropriate manner.
20 The following formulation examples illustrate representative pharmaceutical compositions of the present invention.
Hard gelatin capsules containing the following ingredients are prepared:
25 Quantity ID,gI~h~.~ ,(mQ/ca~sulel Active Ingredient 30.0 Starch 305.0 30 Magnesium stearate 5.0 __9g_ The above ingredients are mixed and filled into hard gelatin capsules in 340 mg quantities.
A tablet formula is prepared using the ingredients below:
Quantity 1~la~
Active Ingredient 25.0 Cellulose, microcrystalline 200.0 Colloidal silicon dioxide 10.0 Stearic acid 5.0 The components are blended and compressed to form tablets, each weighing 240 mg.
EQImulailQtiEX~mpl~ ~
A dry powder inhaler formulation is prepared containing the following components:
IDgI~d~llt Weigh Active Ingredient 5 Lactose 95 The active ingredient is mixed with the lactose and the mixture is added to a dry powder inhaling appliance.
Formulation Example 4 Tablets, each containing 30 mg of active ingredient, are prepared as follows:

__ 10~_ Quantity jng~ _(,mg/tablet) Active Ingredient 30.0 mg 5 Starch 45.0 mg Microcrystalline cellulose 35.0 mg Polyvinylpyrrolidone (as 10 % solution in sterile water) 4.0 mg Sodium carboxymethyl starch 4.5 mg 10 Magnesium stearate 0.5 mg Talc 1.

Total 120 mg 15 The active ingredient, starch and cellulose are passed through a No. 20 mesh U.S. sieve and mixed thoroughly. The solution of polyvinylpyrrolidone is mixed with the resultant powders, which are then passed through a 16 mesh U.S.
sieve. The granules so produced are dried at 50° to 60°C and passed through a 16 mesh U.S. sieve. The sodium carboxymethyl starch, magnesium stearate, and 20 talc, previously passed through a No. 30 mesh U.S. sieve, are then added to the granules which, after mixing, are compressed on a tablet machine to yield tablets each weighing 120 mg.
25 Capsules, each containing 40 mg of medicament are made as follows:
Quantity In~i~nt Active Ingredient 40.0 mg Starch 109.0 mg 30 Magnesium stearate 1.
Total 150.0 mg The active ingredient, starch, and magnesium stearate are blended, passed through a No. 20 mesh U.S. sieve, and filled into hard gelatin capsules in 150 mg 35 quantities WO_99/63929 PCT/US99/11789 Suppositories, each containing 25 mg of active ingredient are made as follows:
Ingr~i~nt A~m~unt Active Ingredient 25 mg Saturated fatty acid glycerides to 2,000 mg The active ingredient is passed through a No. 60 mesh U.S. sieve and suspended in the saturated fatty acid glycerides previously melted using the minimum heat necessary. The mixture is then poured into a suppository mold of nominal 2.0 g capacity and allowed to cool.
Suspensions, each containing 50 mg of medicament per 5.0 mL dose are made as follows:
IuErsi~i~nt AmQUnI
Active Ingredient 50.0 mg Xanthan gum 4.0 mg Sodium carboxymethyl cellulose (1196) Microcrystalline cellulose (8996) 50.0 mg Sucrose 1.75 g Sodium benzoate I0.0 mg Flavor and Color q.v.

Purified water to 5.0 mL

The active ingredient, sucrose and xanthan gum are blended, passed through a No. 10 mesh U.S. sieve, and then mixed with a previously made solution of the microcrystalline cellulose and sodium carboxymethyl cellulose in water. The sodium benzoate, flavor, and color are diluted with some of the water and added with stirring. Sufficient water is then added to produce the required volume.
A formulation may be prepared as follows:

Q~uri IngLC~ ~g(~nsulel Active Ingredient 15.0 mg Starch 407.0 mg S Magnesium stearate 3.
Total 425.0 mg The active ingredient, starch, and magnesium stearate are blended, passed through a No. 20 mesh U.S. sieve, and filled into hard gelatin capsules in 425.0 mg quantities.
Formulation Example 9 A formulation may be prepared as follows:
ID,gt~j~t ~ti>x Active Ingredient 5.0 mg Corn Oil 1.0 mL
A topical formulation may be prepared as follows:
Active Ingredient 1-10 g Emulsifying Wax 30 g Liquid Paraffin 20 g White Soft Paraffin to 100 g The white soft paraffin is heated until molten. The liquid paraffin and emulsifying wax are incorporated and stirred until dissolved. The active ingredient is added and stirring is continued until dispersed. The mixture is then cooled until solid.
Another preferred formulation employed in the methods of the present invention employs transdermal delivery devices ("patches"). Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Patent 5,023,252, issued June 11, 1991, herein incorporated by reference in its entirety. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
Other suitable formulations for use in the present invention can be found in Remington's Pharmaceutical Sciences, Mace Publishing Company, Philadelphia, PA, 17th ed. (1985).
Ut The multibinding compounds of this invention inhibit microsomal triglyceride transferase protein (MTP), a protein which mediates the transfer of lipids during the assembly of lipoproteins and related biomolecules.
Accordingly, the multibinding compounds and phannaceutical compositions of this invention are useful for lowering serum lipid, cholesterol and/or triglyceride levels, and for preventing and treating disorders associated with atherosclerosis, hyperlipemia, hyperlipidemia, hyperlipoproteinemia, hypercholestrolemia, hypertriglyceridemia, pancreatids, diabetes and/or obsesity and the like.
When used in treating or ameliorating such conditions, the compounds of this invention are typically delivered to a patient in need of such treatment by a pharmaceutical composition comprising a pharmaceutically acceptable diluent and an effective amount of at least one compound of this invention. The amount of compound administered to the patient will vary depending upon what compound and/or composition is being administered, the purpose of the administration, such as prophylaxis or therapy, the state of the patient, the manner of administration, and the like. In therapeutic applications, compositions are administered to a patient already suffering from, for example, hypertriglyceridemia or hypercholestrolemia in an amount sufficient to at least partially reduce the patient's triglyceride or cholesterol levels. Amounts effective for this use will depend on the judgment of the attending clinician depending upon factors such as the degree or severity of the disorder in the patient, the age, weight and general condition of the patient, and the like. The pharmaceutical compositions of this invention may contain more than one compound of the present invention.

As noted above, the compounds administered to a patient are in the form of pharmaceutical compositions described above which can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intravenous, intramuscular, etc.. These compounds are effective as both injectable and oral 10 deliverable pharmaceutical compositions. Such compositions are prepared in a manner well known in the pharmaceutical art and comprise at least one active compound.
The multibinding compounds of this invention can also be administered in 15 the form of pro-drugs, i.e., as derivatives which are converted into a biologically active compound in vivo. Such pro-drugs will typically include compounds in which, for example, a carboxylic acid group, a hydroxyl group or a thiol group is converted to a biologically liable group, such as an ester, lactone or thioester group which will hydrolyze in vivo to reinstate the respective group.

The following synthetic and biological examples are offered to illustrate this invention and are not to be construed in any way as limiting the scope of this invention.. Unless otherwise stated, all temperatures are in degrees Celsius.

W099/63929 PC'T/US99/11789 EXAMPLES
In the examples below, the following abbreviations have the following meanings. If an abbreviation is not defined, it has its generally accepted meaning.

- Angstroms cm - centimeter DCC - dicyclohexylcarbodiimide DMF - N,N-dimethylfornaamide DMSO - dimethylsulfoxide EDTA - ethylenediaminetetraacetic acid g _ g~

HPLC - high performance liquid chromatography MEM - minimal essential medium mg - milligram MIC - minimum inhibitory concentration min - minute mL - milliliter mm - millimeter mmol - millimol N - normal THF - tetrahydrofuran ~cL - microliters ,um - microns Example A
Preparation of Synthon A

off N
/ p O ~CFs Synthon A is prepared by aikylation of Synthon F with [1-bromobutylJ-9H-fluorene-9-carboxylic acid, the preparation of which is described as part of WO,Q9/63929 PCT/US99/11789 -106-_ Example 11 in U.S. Patent No. 5,712,279. A solution of Synthon F
hydrochloride (18.0 g, 49 mmol) in 100 mL dimethylformamide is stirred under argon at room temperature and treated with potassium carbonate (12.6 g, 49 mmol) followed by [1-bromobutyl]-9H-fluorene-9-carboxylic acid (16.9 g, 49 5 mmol). The reaction is heated to 50°C for 24 h. After cooling, the reaction is filtered to remove potassium carbonate, and the filter cake is rinsed with ethyl acetate. The solvents are removed in vacuo to afford a solid from which Synthon A is obtained as an off white solid after recrystallization from ethanol (24.1 g, 38.7 mmol, 79%).

Example B
Preparation of Synthon B
~-C F3 \

NH ~ \
O v 'CF3 20 Synthon B is prepared from [1-bromobutyl]-N-(2,2,2-trifluoroethyl)-9H-fluorene-9-carboxamide (Example 11 in U.S. Patent No. 5,712,279), 1,3-diaminopropane, and commercially available Synthon G.
The acid chloride derivative of Synthon G is generated as described in 25 Example 10 of U.S. Patent No. 5,712,279. To a slurry of Synthon G (50.0 g, 190 mmol) in 500 mL methylene chloride is added oxalyl chloride (28.7 mL, 330 mmol) followed by five drops of dimethylformamide. The reaction bubbles vigorously and is stirred at room temperature under argon for 2 h. At this time all solid has dissolved and gas evolution has ceased. The solvent is removed in 30 vacuo, and the residue is dissolved in 400 mL methylene chloride. This solution is then added dropwise to a solution of 1,3-diaminopropane (31.7 mL, 380 mmol) and triethylamine (65.4 mL, 470 mmol) in 300 mL methylene chloride cooled in an ice/brine bath. After the addition is complete, a lot of solid has precipitated from the reaction. Additional methylene chloride (200 mL) is added and the reaction is stirred at room temperature under argon for 18 h. The reaction is then diluted with 600 mL methylene chloride and washed twice with saturated sodium bicarbonate solution, once with brine, and once with 1N
potassium hydroxide. The organic layer is dried over sodium sulfate, and the solvent is removed in vacuo to give a white solid. This solid is recrystallized from hot ethanol and washed with heptane to afford 3"-(aminopropyl)-4'-(trifluoromethyl)-2-biphenylcarboxamide (45.8 g, 142 mmol, 75 % ) as a white solid.
To a stirred solution of [1-bromobutyl]-N-(2,2,2-trifluoroethyl)-9H-fluorene-9-carboxamide (29.5 g, b9.2 mmol) in 100 mL dimethylformamide under argon is added anhydrous potassium carbonate (9.55 g, 69.2 mmol) followed by 3 "-(aminopropyl)-4'-(trifluoromethyl)-2-biphenyicarboxamide (22.3 g, 69.2 mmol). The reaction mixture is then heated to 50°C and stirred under argon for 24 h. After cooling, the reaction is filterred to remove potassium carbonate, and the filter cake is rinsed with ethyl acetate. The filtrate is partitioned between 20% heptane in ethyl acetate and water. The organic layer is washed five times with water and once with brine. The organic layer is then dried over sodium sulfate and the solvent is removed in vacuo to give a beige solid. This solid is recrystallized from 300 mL 25 % ethyl acetate in heptane to provide Synthon B as an off white solid ( 36.5 g, 54.7 mmol, 79% ).

Example C
Preparation of Synthon C

Synthon C is prepared from [1-bromobutyl]-N-(2,2,2-trifluoroethyl)-9H-fluorene-9-carboxamide (Example 11 in U.S. Patent No. 5,712,279), tent-butyl N (2-aminoethyl)carbamate, 1-benzyl-4-piperidone, and commercially available Synthon G.
Potassium hydroxide pellets (16 g, 0.25 mole) are added to a stirred solution of tert-butyl N (2-aminoethyl)carbamate (160 g, 1.00 mole) in 1 L of methanol. Once the pellets are completely dissolved, 1-benzyl-4-piperidone (185 mL, 1.00 mole) is added in one portion and the resulting suspension is stirred under reflux for 1 h. The reaction is then cooled in an ice bath and treated dropwise with sodium cyanoborohydride (22.0 g, 0.35 mole) in 250 mL
methanol. This mixture is allowed to warm to room temperature and is then refluxed for 1 h. After cooling to room temperature, the mixture is treated with potassium hydroxide pellets (60 g, 1.5 mole) and stirred until the pellets are completely dissolved. The reaction mixture is suction filtered and then concentrated to 250 mL on a rotary evaporator. The residue is then diluted with 500 mL half saturated brine and extracted with two 500 mL portions of diethyl ether. The combined organic layers are in turn extracted with two 500 mL
portions of 2 N sodium hydrogen sulfate and then discarded. The combined aqueous extracts are adjusted to pH 10 by the addition of 6 M sodium hydroxide __ 109__ and then back-extracted with two 500 mL portions of ethyl acetate. The combined ethyl acetate extracts are extracted with 500 mL brine, dried over sodium sulfate, and dried to afford the crude 1-benryl-4-[(2-aminoethylcarbamato)amino]-piperidine as an oil.
The acid chloride derivative of Synthon G is generated as described in Example 10 of U.S. Patent No. 5,712,279. To a slurry of Synthon G (50.0 g, 190 mmol) in 500 mL methylene chloride is added oxalyl chloride (28.7 mL, 330 rnmol) followed by five drops of dimethylformamide. The reaction bubbles vigorously and is stirred at room temperature under argon for 2 h. At this time all solid has dissolved and gas evolution has ceased. The solvent is removed in vacuo, and the residue is dissolved in 400 mL methylene chloride. This solution is then added dropwise to a solution of the crude 1-benzyl-4-[(2-aminoethylcarbamato)amino]-piperidine (63.3 g, 190 mmol) prepared in the previous step and triethylamine (65.4 mL, 470 mmol) in 300 mI. methylene chloride cooled in an ice/brine bath. After the addition is complete, a lot of solid has precipitated from the reaction. Additional methylene chloride (200 mL) is added and the reaction is stirred at room temperature under argon for 18 h.
The reaction is then diluted with 600 mL methylene chloride and washed twice with saturated sodium bicarbonate solution, once with brine, and once with 1N
potassium hydroxide. The organic layer is dried over sodium sulfate, and the solvent is removed in vacuo to give a white solid. This solid is recrystallized from hot ethanol and washed with heptane to afford 1-benzyl-4-[[4'-(trifluoromethyl)[ 1,1'-biphenyl]-2-yl]carbonyl] (2-aminoethylcarbamato)amino]-piperidine (74.8 g, 142 mmol, 75 % ) as a white solid.
A solution of 1-benzyl-4-[[4'-(trifluoromethyl)[1,1'-biphenyl]-2-yl]carbonyl](2-aminoethylcarbamato)amino]-piperidine (52.5 g, 100 mmol) in a mixture of 200 mL ethanol plus 10 mi, glacial acetic acid is treated with 10 palladium on activated carbon (2.6 g) and then subjected to hydrogenation on a Parr apparatus (initial pressure 40 psi) for 19 h. The reaction is then filtered through Celite and the filtrate is concentrated to dryness. The residue is dissolved in 500 mL chloroform and washed once with 100 mL 1 N potassium hydroxide and three times with 100 mL brine. The aqueous layers are combined and back-exctracted with three 80 mL portions of chloroform. The combined organic extracts are then dried over sodium sulfate and evaporated to afford 4-[[4'-(trifluoromethyl)[ 1,1'-biphenyl]-2-yl]carbonyl] (2-aminoethylcarbamato)amino]-1-piperidine (39.2 g, 90 mmol, 90% ) as a white solid.
To a stirred solution of [1-bromobutyl]-N-(2,2,2-trifluoroethyl}-9H-fluorene-9-carboxamide (29.5 g, 69.2 mmol) in 100 mL dimethylformamide under argon is added anhydrous potassium carbonate (9.55 g, 69.2 mmol) followed by 4-[[4'-(trifluoromethyl)[1,1'-biphenyl]-2-yl]carbonyl](2-aminoethylcarbamato)amino]-1-piperidine prepared in the previous step (30.1 g, 69.2 mmol). The reaction mixture is then heated to 50°C and stirred under argon for 24 h. After cooling, the reaction is filterred to remove potassium carbonate, and the filter cake is rinsed with ethyl acetate. The filtrate is partitioned between 20% heptane in ethyl acetate and water. The organic layer is washed five times with water and once with brine. The organic layer is then dried over sodium sulfate and the solvent is removed in vacuo to give a beige solid. This solid is recrystallized from 300 mL 25 % ethyl acetate in heptane to provide 9-[4-[4-[[[4'-(trifluoromethyl)(1,1'-biphenyl]-2-yl]carbonyl](2-aminoethylcarbamato)amino]-1-piperidinyl]butyl]-N-(2,2,2-trifluoroethyl)-9H-fluorene-9-carboxamide, the Boc-protected form of Synthon C (40.5 g, 52 mmol, 75 % ) as a white solid.
To a solution of 9-(4-[4-[[[4'-(trifluoromethyl)[1,1'-biphenyl]-2-yl]carbonyl] (2-aminoethylcarbamato)amino]-1-piperidinyl]butyl]-N-(2,2,2-trifluoroethyl)-9H-fluorene-9-carboxamide (30.0 g, 38.5 mmol) in 100 mI.

dioxane was added 75 mL 4 N HCl in dioxane (300 mmol). The reaction was stirred at room temperature for 4 h, then concentrated in vacuo to afford 9-[4-[4 [[[4'-(trifluoromethyl)[ 1,1'-biphenyl)-2-yl]carbonyl)amino]-1-piperidinyl]butyl) N-(2,2,2-trifluoroethyl)-9H-fluorene-9-carboxamide (27.8 g, 37 mmol, 96%)) as the white solid dihydrochloride.
Example D
Preparation of Synthon D
/ \

~OH

\ /

Synthon D is commercially available, for example, from Aldrich Chemical Company, Milwaukee, WI 53201 USA.
Example E
Preparation of Synt6on E
/ \ O
NH
v ~ ~NH
\ / ~H3 Synthon E is prepared from [1-bromobutyl]-N-(2,2,2-trifluoroethyl)-9H-fluorene-9-carboxamide (Example 11 in U.S. Patent No. 5,712,279) and methylamine. [1-bromobutyl]-N-(2,2,2-trifluoroethyl)-9H-fluorene-9-carboxamide (29.5 g, 69.2 mmol), methylamine hydrochloride (5.06 g, 75.0 W0,~9/63929 PCT/US99/I 1789 mmol), and anhydrous potassium carbonate (23.0 g, 166 mmol) are placed in a glass pressure vessel. Dimethylformamide (100 mL) is added, and the vessel is then sealed and heated at 50°C for 48 h, cooled and concentrated to dryness, and taken up in 500 mL methylene chloride. The solution is washed with three 80 mL portions of saturated sodium bicarbonate solution and then two 80 mL
portions of brine, followed by drying over magnesium sulfate and evaporation of solvent. The crude is fractionated by flash chromatography on 600 g silica gel, loading the mixture in methylene chloride and then eluting with a step gradient of 2 % to 3 % methanol in methylene chloride (4 L total solvent volume).
Fractions containing pure compound are combined and evaporated to yield Synthon E (22.5 g, 59.5 mmol, 86 % ) as a white foamy gum.
Example F
Preparation of Synthon F
HN
O V 'C F9 The preparation of Synthon F as the dihydrochloride salt is described as part of Example 10 in U.S. Patent No. 5,712,279.
Example G
Preparation of Synt6on G
i HO O v 'CFs WO _99/63929 Synthon G is commercially available, for example, from Aldrich Chemical Company, Milwaukee, WI 53201 USA.
Example H
Preparation of Synthon H
~-C F3 NH
-N
10 ~ / NH2 The preparation of Synthon H as the dihydrochloride salt is described as part of Example 11 in U.S. Patent No. 5,712,279.
15 Example I
Preparation of Synthon I
O OH
I \
N / I \
2~ p O ~CFs Synthon I is prepared by alkylation of Synthon F with 6-bromohexanoic acid. A solution of Synthon F hydrochloride (18.0 g, 49 mmol) in 100 mL
25 dimethylformamide is stirred under argon at room temperature and treated with potassium carbonate ( 12.6 g, 49 mmol) followed by 6-bromohexanoic acid (9.6 g, 49 mmol). The reaction is heated to 50°C for 24 h. After cooling, the reaction is filtered to remove potassium carbonate, and the filter cake is rinsed with ethyl acetate. The solvents are removed in vacuo to afford a solid from __ 114-- __ which Synthon I can be obtained as an off white solid after recrystallization from ethyl acetate (18.6 g, 38.7 mmol, 79%).
Example 1 Synthesis of FsC CFa A solution of 100 mmols of Synthon A in 200 mL of THF under IVz is treated at room temperature with 100 mmols of trifluoroacetic anhydride and stirred for lhr. To the resultant mixture is added a solution of 50 mmols of 1,3-diaminopropane and 200 mmols of triethylamine in 200 mL of THF. The temperature is raised to 60°C and the reaction followed by TLC. When the reaction is essentially complete by TLC, the solvent is removed in vacuo and the residue partitioned between ethyl acetate and water. The organic layer is washed with water, dried over sodium sulfate, filtered and the solvent removed. The final product is purified by crystallization or chromatography.
Using this procedure, other compounds of formula I may be prepared by employing alternative diamine linker molecules and analogs of Synthon A.

Example 2 Synthesis of N_ 10 FaC
F3C~

15 A mixture of 100 mmols of phthalic anhydride and 120 mmols of 1-amino-4-butanoi in 200 mL of toluene is refluxed with azeotropic removal of water. The reaction is followed by TLC and when judged complete, is cooled and washed with 1~ HCl and water and dried over sodium sulfate. To this solution is added 100 mmols of triethylamine followed by 100 mmols of ZO methanesulfonyl chloride with cooling. After lhr., the mixture is washed with water, dried over sodium sulfate and the solvent removed in vacuo. The resulting phthalimide mesylate is dissolved in 100 mL of DMF and 100 mmols of Synthon E, 100 mmols of potassium carbonate and 50 mmols of potassium iodide are added. The mixture is maintained at 60°C under 1Vz for 24hrs. then diluted 25 with water. The product is washed with water and purified if necessary. The phthalimide is removed by refluxing a solution of the above material in i00 mL
of ethanol with 120 mmols of hydrazine hydrate (TLC). When complete, the reaction is filtered and the solvent removed. The residue is purified as required.

A solution of 100 mmols of Synthon A in 100 mL of THF is treated at room temperature with 100 mmols of trifluoroacetic anhydride and after lhr the resulting solution is added to a solution of the above amine in 100 mL of THF
with 200 mmols of triethylamine. The reaction is followed by TLC and when 5 judged complete, the solvent is removed and the residue partitioned between ethyl acetate and water. The organic phase is repeatedly washed with water, dried over sodium sulfate, filtered and the solvent removed. The final product is purified by crystallization or chromatography.
10 In a similar manner, other compounds of formula I may be prepared by using alternative amino-alcohol linker molecules and analogs of the Synthons.

Example 3 Synthesis of A solution of 100 mmols of Synthon A in 200 mL of THF under NZ is treated at room temperature with 100 mmols of trifluoroacetic anhydride and stirred for 1 hr. To the resultant solution is added a mixture of 100 mmols Synthon C and 200 mmols triethylamine in 100 mL THF. The temperature is raised to 60°C and the reaction is followed by TLC. When is essentially complete by TLC, the solvent is removed in vacuo and the residue partitioned between ethyl acetate and water. The organic layer is washed with water, dried over sodium sulfate, filtered and the solvent removed. The final product is purified by crystallization or chromatography.
In a similar manner, other compounds of formula I may be prepared by using analogs of Synthon A or C.

Example 4 Synthesis of 5 F C~ ~CFs v 10 ~ O NH HN O w CF / ~ \ CFs s \ ~ /
15 A mixture of 100 mmols of Synthon B in 100 mL of DMF with SO mmols of 1,4-dibromobutane and 100 mmols of potassium carbonate is stirred under NZ
at 60°C for 24hr. The reaction is cooled and 500 rnL of water added.
The resultant final product is isolated, washed with water and purified by crystallization or chromatography.
In a similar manner, other compounds of formula I may be prepared by using alternative dihalo linker molecules and analogs of Synthon B.

Example 5 Synthesis of F'C~~
~CF3 O
N~ 'N

HN O w ~ \ CF3 A solution of 100 mmols of Synthon C in I00 mL of THF is treated with I00 mmols of trifluoroacetic anhydride and 100 mmols of triethylamine. After lhr., the solvent is removed in vacuo and the residue partitioned between ethyl acetate and water. The organic layer is washed with water, dried over sodium sulfate, filtered and the solvent removed to afford the crude trifluoroacetamide.
A solution of 100 mmols of Synthon G in 200 mL methylene chloride is treated with 150 mmols of oxalyl chloride and a few drops of DMF. After 2hrs., the solvent is removed and the residue is dissolved in 100 mL of methylene chloride and added to a mixture of 100 mmols of 3-bromopropylamine hydrobromide and 250 mmols of triethylamine in 100 mL of methylene chloride.
After lhr., the solvent is removed in vacuo and the residue partitioned between ethyl acetate and water. The organic layer is washed with water, dried over sodium sulfate, filtered and the solvent removed to afford the crude bromopropylamide which is purified by chromatography or crystallization as necessary.
A solution of the above crude trifluoroacetamide in 200 mL of THF is cooled under NZ to -78°C and 100 mL of ljy LDA in THF is added. The temperature is raised to -20°C and a solution of the above brompropylamide in 100mL of THF is added. The temperature is slowly raised as required and the reaction followed by TLC. When the reaction is essentially complete by TLC, 150 mL of 1~1 NaOH solution is added and the temperature is raised to 60°C
until the trifluoraoacteamide is removed (TLC). The solvent is removed in vacuo and the residue partitioned between ethyl acetate and water. The organic layer is washed with water, dried over sodium sulfate, filtered and the solvent removed.
The product is purified by crystallization or chromatography to afford Intermediate A.
A solution of 50 mmols of Intermediate A in 50 mL of DMF with 50 mmols of Synthon J and 50 mmols of potassium carbonate is maintained at 60°C
for 24hrs. The reaction is cooled and 500 mL of water added. The resultant final product is isolated, washed with water and purified by crystallization or chromatography.
In a similar manner, other compounds of formula I may be prepared by using other halo-amine linkers and analogs of the Synthons.

Example 6 Synthesis of v F3C~~' n ~ ~~' ~CF
F3C~ O. ~ _ ,.._~ ., ~/CF3 A solution of 100 mmols of Synthon H in 100 mL of ethyl acetate is treated with 100 mmols of trifluoroacetic anhydride. After lhr., the reaction is made basic with NaHC03 and washed with water, dried over sodium sulfate, filtered and the solvent removed to afford crude trifluoroacetamide. This material is dissolved in 100 mL of THF and the solution cooled to -78°C.
Following addition of 100 mL of 1L~1 LDA in THF the temperature is raised to -20°C and 50 mmols of 1,4-dibromobutane is added. The temperature is slowly raised as required and the reaction followed by TLC. When the reaction is essentially complete by TLC, 150 mL of 1.~ NaOH solution is added and the temperature is raised to 60°C until the trifluoraoacteamide is removed (TLC).
The solvent is removed in vacuo and the residue partitioned between ethyl acetate and water. The organic layer is washed with water, dried over sodium sulfate, filtered and the solvent removed. The product is purified by crystallization or chromatography to afford Intermediate B.

A solution of 100 mmols of Synthon G in 200 mL of methylene chloride is treated with 150 mmols of oxalyl chloride and a few drops of DMF. After 2hrs., the solvent is removed and the residue is dissolved in 200 mL of methylene chloride and added to a solution of intermediate 2 in 100 mL of methylene chloride with 150 mmols of triethylamine. After lhr., the solvent is removed in vacuo and the residue partitioned between ethyl acetate and water.
The organic layer is washed with water, dried over sodium sulfate, filtered and the solvent removed to afford the final product which may be purified by crystallization or chromatography.
In a similar manner, other compounds of formula I may be prepared by using alternative dihalo linker molecules and analogs of the Synthons.
Example 7 Synthesis of ~a o r A solution of 100 mmols of Synthon D in methylene chloride is treated with 150 mmols of oxalyl chloride and a few drops of DMF. After 2hrs. the solvent is removed and the residue dissolved in 200 mL methylene chloride and added slowly to an excess of neat 1,3-diaminopropane and 100 mmols of triethylamine in 200 mL of methylene chloride. After lhr. the solvent is removed and the residue stirred with water which is discarded. The remaining material is partitioned between ethyl acetate and 1Z[ HCI. The aqueous phase is 5 thoroughly extracted with ethyl acetate to remove any bis-amide and then made basic. This aqueous mixture is extracted with ethyl acetate which is dried over sodium sulfate, filtered and the solvent removed. The crude amino amide is purified by crystallization or chromatography.
10 A solution of 100 mmols of Synthon A in 100 mL of THF is treated with 100 mmols of trifluoroacetic anhydride. After lhr. this solution is added to a solution of the above amide and 200 mmols of triethylamine in 200 mL of THF.
After lhr., the solvent is removed in vacuo and the residue partitioned between ethyl acetate and water. The organic layer is washed with water, dried over 15 sodium sulfate, filtered and the solvent removed to afford the final product which may be purified by crystallization or chromatography.
In a similar manner, other compounds of formula I may be prepared by using alternative diamino linker molecules and analogs of the Synthons.

Example 8 Synthesis of A solution of 100 mmols of Synthon D in 200 mL methylene chloride is treated with 150 mmols of oxalyl chloride and a few drops of DMF. After 2hrs., the solvent is removed and the residue is dissolved in 200 mL of methylene chloride and added to 100 mmols of 5-aminopentanol and 120 mmols of triethylamine. After lhr., the solvent is removed in vacuo and the residue partitioned between ethyl acetate and water. The organic layer is washed with 1~ HCl followed by water, dried over sodium sulfate, filtered and the solvent removed to afford crude hydroxyamide. This material is dissolved in 100 mL of THF with 100 mmols of triethylamine, and 100 mmols of methanesulfonyl chloride added. After lhr., the solvent is removed in vacuo and the residue partitioned between ethyl acetate and water. The organic layer is washed with water, dried over sodium sulfate, filtered and the solvent removed to afford Intermediate C.
A solution of 100 mmols of Intermediate C, 100 mmols of Synthon B, 100 mmols of potassium carbonate and 50 mmols of potassium iodide in 100 mL
of DMF is kept at 60°C under NZ for 24hrs. The mixture is cooled and 500 mL

of water is added and the product is isolated and washed with water. The final product is purified by crystallization or chromatography.
In a similar manner, other compounds of formula I may be prepared by S using alternative amino alcohol linker molecules and analogs of the Synthons.
Example 9 Synthesis of NH
A solution of 100 mmols of 6-bromo-1-hexanol and 120 mL of freshly distilled dihydropyran in 150 mL of anhyd. ether is treated with 1 drop of POCK.
After lhr. the mixture is washed with sat. NaHC43, dried over sodium sulfate, filtered and the solvent removed. The residue is dissolved in 100 mL of DMF
and 100 mmol of Synthon E and 100 mmols of potassium carbonate is added.
The mixture is kept a 60°C under NZ for 24hrs. then cooled and partitioned between ethyl acetate and water. The organic phase is repeatedly washed with water and 50 mL of trifluoroacetic acid added. When TLC indicates that THP
removal is complete, the solution is washed with water then sat. NaHCOj until basic, dried over sodium sulfate and filtered. To this solution is added 100 mmols of methanesulfonyl chloride in 200 mL of ethyl acetate. After lhr. the reaction mixture is made basic with sat. NaHC03 and the organic phase washed with water, dried over sodium sulfate, filtered and the solvent removed to afford the mesylate Intermediate D.
A mixture of 100 mmols of Intermediate D, 100 mmols of Synthon B, 100 mmols of potassium carbonate and 50 mmols of sodium iodide in 100 mL
of DMF is kept at 60°C under NZ for 24hrs. The mixture is cooled and 500 mL
of water is added. The material is isolated and washed with water. The final product is purified by crystallization or chromatography.
In a similar manner, other compounds of formula I may be prepared by using alternative halo alcohol linker molecules and analogs of the Synthons.
Example 10 Synthesis of O
F~
2s p A solution of 100 mmols of Synthon H in 100 mL of ethyl acetate is treated with 100 mmols of trifluoroacetic anhydride. After lhr., the reaction is __127-made basic with NaHC03 and washed with water, dried over sodium sulfate, filtered and the solvent removed. The residue is dissolved in 100 mL of THF
and cooled under NZ to -78°C. After addition of 100 mL of a 1rJ LDA
/THF
solution, the temperature is raised to -20°C and 100 mmols of Intermediate C
5 added. The temperature is raised as required and the reaction followed by TLC. When judged complete, 100 mL of 1~T NaOH solution is added and the reaction warmed to remove the trifluoroacetamide. After removal of the solvent, extractive workup with ethyl acetate/water affords Intermediate E.
10 A solution of 100 mmols of Synthon G in 100 mL of THF is treated with 100 mL of trifluoroacetic anhydride. After lhr., the resultant solution is added to a solution of 100 mmols of Intermediate E and 200 mmols of triethylamine in 100 mL of THF. The temperature is warmed as required and when TLC indicates that the coupling is complete the solvent is removed and 15 ' the residue is partitioned between water and ethyl acetate. The organic phase is washed with water, dried over sodium sulfate, filtered and the solvent removed. The final product is purified by crystallization or chromatography.
In a similar manner, other compounds of formula I may be prepared by 20 using alternative amino alcohol linker molecules and analogs of the Synthons.

W0~99/63929 PCTNS99/11789 Example 11 Synthesis of A solution of 100 mmols of Synthon H in 100 mL of ethyl acetate is treated with 100 mmols of trifluoroacetic anhydride. After lhr., the reaction is made basic with NaHC03 and washed with water, dried over sodium sulfate, filtered and the solvent removed. The residue is dissolved in 100 mL of THF
and cooled under NZ to -78°C. After addition of 100 mL of a lst LDA
/THF
solution, the temperature is raised to -20°C and 100 mmols of Intermediate D
added. The temperature is raised as required and the reaction followed by TLC. When judged complete, 100 mL of 1rI NaOH solution is added and the reaction warmed to remove the trifluoroacetamide. After removal of the solvent, extractive workup with ethyl acetate / water afforded Intermediate F.
A solution of 100 mmols of Synthon G in 100 mL of THF is treated with 100 mL of trifluoroacetic anhydride. After ihr., the resultant solution is added to a solution of 100 mmols of Intermediate F and 200 mmols of triethylamine in 100 mL of THF. The temperature is warmed as required and when TLC indicates that the coupling is complete the solvent is removed and the residue is partitioned between water and ethyl acetate. The organic phase is washed with water, dried over sodium sulfate, filtered and the solvent removed. The final product is purified by crystallization or chromatography.
In a similar manner, other compounds of formula I may be prepared by using alternative bromo alcohol linker molecules and analogs of the Synthons.
Example 12 Synthesis of F3C~
O
C ' /
O Whl A solution of 100 mmols of 6-bromo-1-hexanol and 120 mL of freshly distilled dihydropyran in 150 mL of anhyd. ether is treated with 1 drop of POC13. After lhr., the mixture is washed with sat. NaHC03, dried over sodium sulfate, filtered and the solvent removed. The residue is dissolved in 100 mL of DMF and 100 mmol of Synthon F and 100 mmols of potassium carbonate added The mixture is kept a 60°C under NZ for 24hrs. then cooled and partitioned between ethyl acetate and water. The organic phase is repeatedly washed with water and 50 mL of trifluoroacetic acid added. When TLC indicates that THP removal is complete, the solution is washed with water then sat. NaHC03 until basic, dried over sodium sulfate and filtered. To this solution is added 100 mmols of methanesulfonyl chloride in 200 mL of ethyl acetate. After lhr., the reaction mixture is made basic with sat. NaHC03 and the organic phase washed with water, dried over sodium sulfate, filtered and 5 the solvent removed to afford the mesylate Intermediate G.
A mixture of 100 mmols of Intermediate G, 100 mmols of Synthon B, 100 mmols of potassium carbonate and 50 mmols of sodium iodide in 100 mL
of DMF is kept at 60 ° C under NZ for 24hrs. The mixture is cooled and 10 mL of water added. The material is isolated and washed with water. The final product is purified by crystallization or chromatography.
In a similar manner, other compounds of formula I may be prepared by using alternative bromo alcohol linker molecules and analogs of the Synthons.

Example 13 Synthesis of ~~ ~

A solution of 100 mmols of Synthon G in 200 mL methylene chloride is treated with 150 mmols of oxalyl chloride and a few drops of DMF. After 2hrs., the solvent is removed and the residue is dissolved in 200 mL of methylene chloride and added to 100 mmols of 5-aminopentanol and 120 5 mmols of triethylamine. After lhr., the solvent is removed in vacuo and the residue partitioned between ethyl acetate and water. The organic layer is washed with 1~I HCl followed by water, dried over sodium sulfate, filtered and the solvent removed to afford crude hydroxyamide. This material is dissolved in 100 mL of THF with 100 mmols of triethylamine, and 100 mmols 10 of methanesulfonyl chloride added. After lhr., the solvent is removed in vacuo and the residue partitioned between ethyl acetate and water. The organic layer is washed with water, dried over sodium sulfate, filtered and the solvent removed to afford Intermediate H.
15 A solution of 100 mmols of Intermediate H, 100 mmols of Synthon B, 100 mmols of potassium carbonate and 50 mmols of potassium iodide in 100 mL of DMF is kept at 60°C under NZ for 24 hrs. The mixture is cooled and 500 mL of water is added and the product is isolated and washed with water.
The final product is purified by crystallization or chromatography.

In a similar manner, other compounds of formula I may be prepared by using alternative amino alcohol linker molecules and analogs of the Synthons.

Example 14 Synthesis of to F~ \ /
J
A solution of 100 mmols of Synthon H in 100 mL of ethyl acetate is treated with 100 mmols of trifluoroacetic anhydride. After lhr., the reaction is made basic with NaHC03 and washed with water, dried over sodium sulfate, filtered and the solvent removed. The residue is dissolved in 100 mL of THF
and cooled under N2 to -78°C. After addition of 100 mL of a lay LDA
/THF
solution, the temperature is raised to -20 C and 100 mmols of Intermediate G
added. The temperature is raised as required and the reaction followed by TLC. When judged complete, 100 mL of 1~1 NaOH solution is added and the reaction warmed to remove the trifluoroacetamide. After removal of the solvent, extractive worlcup with ethyl acetate/water afforded Intermediate I.
A solution of 100 mmols of Synthon G in 100 mL of THF is treated with 100 mL of trifluoroacetic anhydride. After lhr., the resultant solution is added to a solution of 100 mmols of Intermediate I and 200 mmols of triethylamine in 100 mL of THF. The temperature is warmed as required and when TLC indicates that the coupling is complete the solvent is removed and the residue is partitioned between water and ethyl acetate. The organic phase is washed with water, dried over sodium sulfate, filtered and the solvent removed. The final product is purified by crystallization or chromatography.
5 In a similar manner, other compounds of formula I may be prepared by using alternative bromo alcohol linker molecules and analogs of the Synthons.
Example 15 Synthesis of A solution of 100 mmols of Synthon H in 100 mL of ethyl acetate is treated with 100 mmols of trifluoroacetic anhydride. After lhr., the reaction is made basic with NaHC03 and washed with water, dried over sodium sulfate, 25 filtered and the solvent removed. The residue is dissolved in 100 mL of THF
and cooled under NZ to -78 ° C . After addition of 100 mL of a 1~ LDA
/THF
solution, the temperature is raised to -20°C and 100 mmols of Intermediate H
added. The temperature is raised as required and the reaction followed by TLC. When judged complete, 100 mL of 1~1 NaOH solution is added and the reaction warmed to remove the trifluoroacetamide. After removal of the solvent, extractive workup with ethyl acetate/water afforded Intermediate J.
A solution of 100 mmols of Synthon G in 100 mL of THF is treated S with 100 mL of trifluoroacetic anhydride. After lhr., the resultant solution is added to a solution of 100 mmols of Intermediate E and 200 mmols of triethylamine in 100 mL of THF. The temperature is warmed as required and when TLC indicates that the coupling is complete the solvent is removed and the residue is partitioned between water and ethyl acetate. The organic phase is washed with water, dried over sodium sulfate, filtered and the solvent removed. The final product is purified by crystallization or chromatography.
In a similar manner, other compounds of formula I may be prepared by using alternative amino alcohol linker molecules and analogs of the Synthons.
Example 16 Synthesis of 25 A mixture of 100 mmols of Synthon E and 100 mmols of Synthon J in 100 mL of DMF with 100 mmols of potassium carbonate is kept at 60°C
under N2 for 24 hrs. The reaction is cooled and 500 mL of water added. The final product is isolated, washed with water and purified by crystallization or chromatography.

Example 17 Synthesis of \ I Hs / I
I / a~N~b \
O
/ / I
\ I \

A solution of 100 mmols of Synthon G in 100 mL of THF is treated with 100 mmols of trifluoroacetic anhydride at room temperature. After lhr., this solution is added to a solution of 50 mmols of 3,3'-diamino-N-methyldipropylamine and 200 mmols of triethylamine in 100 mL of THF. The temperature is raised as necessary and the reaction followed by TLC. When the reaction is judged to be complete by TLC, the solvent is removed and the residue partitioned between ethyl acetate and water. The organic layer is washed repeatedly with water, dried over sodium sulfate, filtered and the solvent removed. The final product is purified by crystallization or chromatography.
In a similar manner, other compounds of formula I may be prepared by using alternative diamino linker molecules and analogs of the Synthons.

Example 18 Synthesis of N
I

10 A mixture of 100 mmols of Synthon E and 50 mmols of 1,4-dibromobutane in 100 mL of DMF with 100 mmols of potassium carbonate is kept at 60°C under NZ for 24 hrs. The reaction is cooled and 500 mL of water added. The final product is isolated, washed with water and purified by crystallization or chromatography.

In a similar manner, other compounds of formula I may be prepared by using alternative dihalo linker molecules and analogs of the Synthons.
Example 19 20 Synthesis of y a N N~--b ' ° ° ' 2s A mixture of 100 rnmols of Synthon F and 50 mmols of 1,6-30 dibromohexane in 100 mL of DMF with 100 mmois of potassium carbonate is kept at b0°C under NZ for 24 hrs. The reaction is cooled and 500 mL of water added. The final product is isolated, washed with water and purified by crystallization or chromatography.
In a similar manner, other compounds of formula I may be prepared by using alternative dihalo linker molecules and analogs of the Synthons.
Example 20 Synthesis of Fs C~~ O ~ ~ O~~~CF3 A solution of 50 mmols of glutaryl dichloride in 100 mL of ethyl acetate is added to a solution of 100 mmols of Synthon H and 100 mmols of triethylamine in 100 mL of ethyl acetate. After lhr., the solution is thoroughly washed with water, dried over sodium sulfate, filtered and the solvent removed. The resulting final product is purified by chromatography or crystallization.
In a similar manner, other compounds of formula I may be prepared by using alternative diacid chloride linker molecules and analogs of the Synthons.

Example 21 Synthesis of O
N

HN O O NH
A solution of 100 mmols of Synthon I in 100 mL of THF is treated with 100 mmols of trifluoroacetic anhydride at room temperature. After lhr., the reaction mixture is added to a solution of 50 mmols of 1,5-diaminopentane in 100 mL of THF with 200 mmols of triethylanune. The reaction is followed by TLC and when judged to be complete, the solvent is removed and the residue partitioned between water and ethyl acetate. The organic phase is washed with water, dried over sodium sulfate, filtered and the solvent removed. The final product is purified by crystallization or chromatography.
In a similar manner, other compounds of formula I may be prepared by using alternative diamino linker molecules and analogs of the Synthons.

WO 99/63929 PCT1US99/11'789 Bioassay Example 1 Inhibition of Triglyceride Transfer MTP activity is routinely determined by measuring the rate of transfer of radiolabeled triglyceride from donor small unilamellar vesicles (SUV) to acceptor SW. In this bioassay, the inhibition of radiolabled triglyceride transfer between donor and acceptor vesicles is determined using the procedure described in J. R. Wetterau et al., Biochim. Biophys. Acta 1997, 1345, 136-150.
A typical transfer reaction mixture contains donor vesicles (40 nmol egg phosphatidyl choline, 0.25 mol % radiolabeled triglyceride (['4C]txiolein), and 7.5 mol % cardiolipin), acceptor vesicles (240 nmol egg phosphatidyl choline and 0.25 % unlabeled triglyceride) and 5 mg bovine serum albumin in a total of 0.9 mL buffer. The negative charge in the donor vesicles due to the presence of cardiolipin facilitates the separation of donor and acceptor membranes. Following a transfer reaction, a DEAF-cellulose suspension is added to selectively bind the negatively charged donor vesicles. The DEAE-cellulose and bound donor membranes are then pelleted by low speed centrifugation. The acceptor SUV concentration in the assay mixture is kept in excess over that of the donor SW to minimize back transfer from acceptor to donor vesicles. First-order kinetics are used to quantify total lipid transfer.
This corrects for the dilution of labeled lipid in the donor vesicles as the transfer reaction proceeds. The transfer activity is generally expressed as a percent of the donor lipid transferred per unit of time.

Bioassay Example 2 Inltibition of Lipoprotein Secretion by HepG2 Cells In this bioassy, the inhibition of lipoprotein secretion by cultured human hepatoma cells (HepG2) is determined using the procedure described in H. Jamil et al., Proc. Natl. Acad. Sci. U.S.A. 1996, 93, 11991-11995.
HepG2 cells are seeded at a density of approximately 50 % confluency in 48-well plates and allowed to grow for 48 h before treatment. At this time, the medium is replaced with fresh medium containing 0.5 % DMSO and a range of concentrations of the inhibitor. After a 16 h incubation under standard cell culture conditions, the medium is diluted with fresh tissue culture medium 30-fold for an apoB ELISA, and 60-fold for an apoAI ELISA. A
sandwich ELISA is used to measure apoB in the media. A similar assay is used to quantitate apoAI. For the apoAI assay, the primary and secondary antibodies are a monoclonal anti-human apoAI (1:500 dilution) and a goat anti-human apoAI polyclonal antibody (1:500). The concentration of the respective proteins is measured against a 2-fold dilution standard curve from 1.25-40 ng/mL of the purified proteins. In this range of concentrations, both assays show a linear response. Each inhibitor concentration is tested in duplicate cultures, and apoB and apoAI are measured by ELISA in each culture in triplicate.
Bioassay Example 3 Inhibition of Triglyceride Secretion in Rats In this bioassay, the inhibition of triglyceride secretion in fasted and fed rats is determined using the procedure described in J. R. Wetterau et al., Nature 1998, 282, 751-754.

Sprague-Dawley rats ( ~ 200 g each, four per treatment group) are adapted to a reversed diurnal light cycle for two weeks. Before the experiment, the rats either are fasted or have free access to food for 18 hours.
The animals are dosed with a test compound one hour before receiving an intravenous injection of Triton WR1339 (250 mg/kg), which prevents the catabolism of triglyceride-rich lipoproteins produced during the course of the experiment. The triglyceride secretion rate is determined by calculating the amount of triglyceride that is accumulated in plasma during the 2.5 hours after the Triton injection. The standard assay is linear for at least 5 hours after the Triton injection. Plasma triglyceride levels are determined with a Roche Cobas blood chemistry autoanalyzer.
Bioassay Example 4 Diminution of Serium Cholesterol Levels in Hamsters In this bioassay, the diminution of serum cholesterol levels in hamsters is determined using the procedure described in J. R. Wetterau et al., Nature 1998, 282, 751-754.
Male Golden Syrian hamsters ( --140 g each, four per treatment group) are adapted to and maintained on a reverse diurnal light cycle. They are dosed once a day with a test compound and are allowed i~oe access to a standard hamster diet. After seven days of treatment, hamsters are fasted for 18 hours, after which plasma lipid levels (triglycerides and cholesterol) and chemistries are determined with a Roche Cobas blood chemistry autoanalyzer. Hamster lipoprotein fractions (VLDL, LDL, HDL) are quantitated after the precipitation of apoB-containing lipoproteins with phosphotungstate and magnesium chloride.

' PCT/US99/11789 Bioassay Example 5 Amelioration of Hyperlipidemia in WHHL Rabbits In this bioassay, the amelioration of hyperlipidemia in Watanabe-S heritable hyperlipidemic (WHHL) rabbits is determined using the procedure described in J. R. Wetterau et al., Nature 1998, 282, 751-754.
Five WHHL rabbits are treated with a test compound for 14 days.
Plasma lipid levels (triglycerides and cholesterol) and chemistries are 10 determined with a Roche Cobas blood chemistry autoanalyzer 18 hours after the last dose.

Claims (56)

WHAT IS CLAIMED IS:
1. A multibinding compound of formula I:
(L)p(X)q I
wherein each X is independently a linker; p is an integer of from 2 to 10;
and q is an integer of from 1 to 20; and each L is independently a ligand selected from the group consisting of:
wherein each W is a divalent radical independently selected from the group consisting of:
each R1 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and a covalent bond linking the ligand to a linker;
each R2 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and a covalent bond linking the ligand to a linker;
each R3 is independently selected from the group consisting of hydrogen, alkyl and halo;
each R4 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl and a covalent bond linking the ligand to a linker;
each R5 is independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, alkoxy, substituted alkoxy, cycloalkoxy, substituted cycloalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, amino, substituted amino and a covalent bond linking the ligand to a linker; or R4 and R5 may be joined, together with the > NC(O)-group to which they are attached, to form a heterocyclic ring;
each R6 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and a covalent bond linking the ligand to a linker;
each R' is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, a covalent bond linking the ligand to a linker and -NR14R15, where R14 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl and heteroaryl; and R15 is a covalent bond linking the ligand to a linker;
each R8 is independently selected from the group consisting of hydrogen, acyl, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, a covalent bond linking the ligand to a linker each R9 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, a covalent bond linking the ligand to a linker and -NR14R15, where R14 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl and heteroaryl; and R15 is a covalent bond linking the ligand to a linker;
each R10, R11, R12 and R13 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, alkoxy, substituted alkoxy, cycloalkoxy, substituted cycloalkoxy, aryl, aryloxy, halo, heteroaryl, heteroaryloxy, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioheteroaryloxy, acyloxy, aminoacyl, aminocarbonyl, -S(O)R16 and -SO2R16, where each R16 is independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, aryl and heteroaryl;
each ring A, together with the atoms to which it is attached, forms a carbocyclic or heterocyclic ring selected from the group consisting of aryl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, heteroaryl and heterocyclic;
each ring B, together with the atoms to which it is attached, forms a carbocyclic or heterocyclic ring selected from the group consisting of aryl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, heteroaryl and heterocyclic;
each ring C, together with the nitrogen atom to which it is attached, forms a heterocyclic ring;
each Q is independently selected from the group consisting of a covalent bond, -O-, -S-, -S(O)-, -SO2-, alkylene, substituted alkylene, alkenylene, substituted alkenylene and -NR17-, where R17 is selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl and heteroaryl;
each a is independently an integer of from 2 to 6;
each b is independently an integer of from 0 to 6;
each c is independently an integer of from 2 to 4;
and pharmaceutically-acceptable salts or pro-drugs thereof;
provided that when p is 2, q is 1 and a first ligand has formula IA or IB, where R1 or R2 is a covalent bond linking the first ligand to the linker, then a second ligand does not have formula ID or IE, where R8 or R9 are a covalent bond linking the second ligand to the linker.
2. The multibinding compound of Claim 1, wherein q is less than p.
3. The multibinding compound of Claim 1, wherein W is a divalent radical having the formula:
wherein each R18 and R19 are independently selected from the group consisting of hydrogen or halo; and Q' is a covalent bond, - O- or - S-
4. The multibinding compound of Claim 1, wherein each R5 group is independently selected from the group consisting of aryl, heteroaryl, heterocyclic, cycloalkyl and substituted cycloalkyl.
5. The multibinding compound of Claim 4, wherein each R5 group is substituted with from 1 to 4 substituents and one of the substituents is attached to the ring atom in the position adjacent to the atom attached to the -C(O)-group.
6. The multibinding compound of Claim 5, wherein each R5 is a 2-(4'-trifluoromethylphenyl)phenyl group.
7. The multibinding compound of Claim 1, wherein each ring C
forms a piperidine or a 1,2,3,4-tetrahydroisoquinoline ring.
8. The multibinding compound of Claim 1, wherein each linker independently has the formula:

-X a-Z-(Y a-Z)m Y-b-Z-X a-wherein m is an integer of from 0 to 20;
X a at each separate occurrence is selected from the group consisting of -O-, -S-, -NR-, -C(O)-, -C(O)O-, -C(O)NR-, -C(S), -C(S)O-, -C(S)NR- or a covalent bond where R is as defined below;
Z is at each separate occurrence is selected from the group consisting of alkylene, substituted alkylene, cycloalkylene, substituted cylcoalkylene, alkenylene, substituted alkenylene, alkynylene, substituted alkynylene, cycloalkenylene, substituted cycloalkenylene, arylene, heteroarylene, heterocyclene, or a covalent bond;
Y a and Y b at each separate occurrence are selected from the group consisting of -C(O)NR'-, -NR'C(O)-, -NR'C(O)NR'-, -C(=NR')-NR'-, -NR'-C(=NR')-, -NR'-C(O)-O-, -N=C(X a)-NR'-, -P(O)(OR')-O-, -S(O)n CR'R"-, -S(O)n NR'-, -S-S- and a covalent bond; where n is 0, 1 or 2;
and R, R' and R" at each separate occurrence are selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, cycloalkenyl, substituted cycloalkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl and heterocyclic.
9. A multibinding compound of formula II:
L'- X'- L' II
wherein X' is a linker; and each L' is a ligand independently selected from the group consisting of:

wherein each R20 and R21 is independently selected from the group consisting of hydrogen and halo;
each R22 is independently selected from the group consisting of hydrogen, alkyl and halo;
each R23 is independently selected from the group consisting of hydrogen and a covalent bond linking the ligand to the linker;
each R24 is independently selected from the group consisting of hydrogen and a covalent bond linking the ligand to the linker;
each R25 is independently selected from the group consisting of aryl, heteroaryl, heterocyclic, cycloalkyl, substituted cycloalkyl and a covalent bond linking the ligand to the linker;
each R26 is independently selected from the group consisting of hydrogen, alkyl and a covalent bond linking the ligand to the linker;
each R27 is independently selected from the group consisting of hydrogen, a covalent bond linking the ligand to a linker and -NR30R31, where R30 is selected from the group consisting of hydrogen and alkyl; and R31 is a covalent bond linking the ligand to a linker;

each R28 is a covalent bond linking the ligand to a linker;
each R29 is independently selected from the group consisting of a covalent bond linking the ligand to a linker and -NR30R31, where R30 is selected from the group consisting of hydrogen and alkyl; and R31 is a covalent bond linking the ligand to a linker;
each Q" is independently selected from the group consisting of a covalent bond, -O- and -S-each a is independently an integer of from 2 to 6;
each b is independently an integer of from 0 to 6;
and pharmaceutically-acceptable salts or pro-drugs thereof;
provided that in each ligand only one of R23, R24, R25, R26, R27, R28 and R29 is a covalent bond linking the ligand to the linker;
and further provided that when a first ligand has formula IIA or IIB, where R23 is a covalent bond linking the first ligand to the linker, then a second ligand does not have formula IID or IIE, where R28 or R29 are a covalent bond linking the second ligand to the linker.
10. The multibinding compound of Claim 9, wherein each R25 group is independently selected from the group consisting of aryl, heteroaryl, heterocyclic, cycloalkyl and substituted cycloalkyl.
11. The multibinding compound of Claim 10, wherein each R25 group is substituted with from 1 to 4 substituents and one of the substituents is attached to the ring atom in the position adjacent to the atom attached to the -C(O)-group.
12. The multibinding compound of Claim 11, wherein each R25 is a 2-(4'-trifluoromethylphenyl)phenyl group.
13. The multibinding compound of Claim 9, wherein each linker independently has the formula:
-X a-Z-(Y a-Z)m-Y b-Z-X a-wherein m is an integer of from 0 to 20;
X a at each separate occurrence is selected from the group consisting of -O-, -S-, -NR-, -C(O)-, -C(O)O-, -C(O)NR-, -C(S), -C(S)O-, -C(S)NR- or a covalent bond where R is as defined below;
Z is at each separate occurrence is selected from the group consisting of alkylene, substituted alkylene, cycloalkylene, substituted cylcoalkylene, alkenylene, substituted alkenylene, alkynylene, substituted alkynylene, cycloalkenylene, substituted cycloalkenylene, arylene, heteroarylene, heterocyclene, or a covalent bond;
Y a and Y b at each separate occurrence are selected from the group consisting of -C(O)NR'-, -NR'C(O)-, -NR'C(O)NR'-, -C(=NR')-NR'-, -NR'-C(=NR')-, -NR'-C(O)-O-, -N=C(X a)-NR'-, -P(O)(OR')-O-, -S(O)n CR'R"-, -S(O)n-NR'-, -S-S- and a covalent bond; where n is 0, 1 or 2;
and R, R' and R" at each separate occurrence are selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, cycloalkenyl, substituted cycloalkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl and heterocyclic.
14. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and an effective amount of a multibinding compound of formula I:
(L)p(X)q~~~I

wherein each X is independently a linker; p is an integer of from 2 to 10;
and q is an integer of from 1 to 20; and each L is independently a ligand selected from the group consisting of:
wherein each W is a divalent radical independently selected from the group consisting of:
each R1 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and a covalent bond linking the ligand to a linker;
each R2 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and a covalent bond linking the ligand to a linker;
each R3 is independently selected from the group consisting of hydrogen, alkyl and halo;
each R4 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl and a covalent bond linking the ligand to a linker;
each R5 is independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, alkoxy, substituted alkoxy, cycloalkoxy, substituted cycloalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, amino, substituted amino and a covalent bond linking the ligand to a linker; or R4 and R5 may be joined, together with the > NC(O)-group to which they are attached, to form a heterocyclic ring;
each R6 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and a covalent bond linking the ligand to a linker;
each R7 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, a covalent bond linking the ligand to a linker and -NR14R15, where R14 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl and heteroaryl; and R15 is a covalent bond linking the ligand to a linker;
each R8 is independently selected from the group consisting of hydrogen, acyl, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, a covalent bond linking the ligand to a linker each R9 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, a covalent bond linking the ligand to a linker and -NR14R15, where R14 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl and heteroaryl; and R15 is a covalent bond linking the ligand to a linker;
each R10, R11, R12 and R13 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, alkoxy, substituted alkoxy, cycloalkoxy, substituted cycloalkoxy, aryl, aryloxy, halo, heteroaryl, heteroaryloxy, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioheteroaryloxy, acyloxy, aminoacyl, aminocarbonyl, -S(O)R16 and -SO2R16, where each R16 is independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, aryl and heteroaryl;
each ring A, together with the atoms to which it is attached, forms a carbocyclic or heterocyclic ring selected from the group consisting of aryl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, heteroaryl and heterocyclic;
each ring B, together with the atoms to which it is attached, forms a carbocyclic or heterocyclic ring selected from the group consisting of aryl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, heteroaryl and heterocyclic;
each ring C, together with the nitrogen atom to which it is attached, forms a heterocyclic ring;
each Q is independently selected from the group consisting of a covalent bond, -O-, -S-, -S(O)-, -SO2-, alkylene, substituted alkylene, alkenylene, substituted alkenylene and -NR17-, where R17 is selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl and heteroaryl;
each a is independently an integer of from 2 to 6;
each b is independently an integer of from 0 to 6;
each c is independently an integer of from 2 to 4;
and pharmaceutically-acceptable salts or pro-drugs thereof;
provided that when p is 2, q is 1 and a first ligand has formula IA or IB, where R1 or R2 is a covalent bond linking the first ligand to the linker, then a second ligand does not have formula ID or IE, where R8 or R9 are a covalent bond linking the second ligand to the linker.
15. The pharmaceutical composition of Claim 14, wherein q is less than p.
16. The pharmaceutical composition of Claim 14, wherein W is a divalent radical having the formula:
wherein each R18 and R19 are independently selected from the group consisting of hydrogen or halo; and Q' is a covalent bond, -O- or -S-.
17. The pharmaceutical composition of Claim 14, wherein each R5 group is independently selected from the group consisting of aryl, heteroaryl, heterocyclic, cycloalkyl and substituted cycloalkyl.
18. The pharmaceutical composition of Claim 17, wherein each R5 group is substituted with from 1 to 4 substituents and one of the substituents is attached to the ring atom in the position adjacent to the atom attached to the -C(O)- group.
19. The pharmaceutical composition of Claim 18, wherein each R5 is a 2-(4'-trifluoromethylphenyl)phenyl group.
20. The pharmaceutical composition of Claim 14, wherein each ring C
forms a piperidine or a 1,2,3,4-tetrahydroisoquinoline ring.
21. The pharmaceutical composition of Claim 14, wherein each linker independently has the formula:

-X a-Z-(Y a-Z)m-Y b-Z-X a-wherein m is an integer of from 0 to 20;
X a at each separate occurrence is selected from the group consisting of -O-, -S-, -NR-, -C(O)-, -C(O)O-, -C(O)NR-, -C(S), -C(S)O-, -C(S)NR- or a covalent bond where R is as defined below;
Z is at each separate occurrence is selected from the group consisting of alkylene, substituted alkylene, cycloalkylene, substituted cylcoalkylene, alkenylene, substituted alkenylene, alkynylene, substituted alkynylene, cycloalkenylene, substituted cycloalkenylene, arylene, heteroarylene, heterocyclene, or a covalent bond;
Y a and Y b at each separate occurrence are selected from the group consisting of -C(O)NR'-, -NR'C(O)-, -NR'C(O)NR'-, -C(=NR')-NR'-, -NR'-C(=NR')-, -NR'-C(O)-O-, -N=C(X a)-NR'-, -P(O)(OR')-O-, -S(O)n CR'R"-, -S(O)n-NR'-, -S-S- and a covalent bond; where n is 0, 1 or 2;
and R, R' and R" at each separate occurrence are selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, cycloalkenyl, substituted cycloalkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl and heterocyclic.
22. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and an effective amount of a multibinding compound of formula II:
L'-X'-L' ~~~II
wherein X' is a linker; and each L' is a ligand independently selected from the group consisting of:

wherein each R20 and R21 is independently selected from the group consisting of hydrogen and halo;
each R22 is independently selected from the group consisting of hydrogen, alkyl and halo;
each R23 is independently selected from the group consisting of hydrogen and a covalent bond linking the ligand to the linker;
each R24 is independently selected from the group consisting of hydrogen and a covalent bond linking the ligand to the linker;
each R25 is independently selected from the group consisting of aryl, heteroaryl, heterocyclic, cycloalkyl, substituted cycloalkyl and a covalent bond linking the ligand to the linker;
each R26 is independently selected from the group consisting of hydrogen, alkyl and a covalent bond linking the ligand to the linker;
each R27 is independently selected from the group consisting of hydrogen, a covalent bond linking the ligand to a linker and -NR30R31, where R30 is selected from the group consisting of hydrogen and alkyl; and R31 is a covalent bond linking the ligand to a linker;

each R28 is a covalent bond linking the ligand to a linker;
each R29 is independently selected from the group consisting of a covalent bond linking the ligand to a linker and -NR30R31, where R30 is selected from the group consisting of hydrogen and alkyl; and R" is a covalent bond linking the ligand to a linker;
each Q" is independently selected from the group consisting of a covalent bond, -O- and -S-each a is independently an integer of from 2 to 6;
each b is independently an integer of from 0 to 6;
and pharmaceutically-acceptable salts or pro-drugs thereof;
provided that in each ligand only one of R23, R24, R25, R26, R27, R28 and R29 is a covalent bond linking the ligand to the linker;
and further provided that when a first ligand has formula IIA or IIB, where R23 is a covalent bond linking the first ligand to the linker, then a second ligand does not have formula IID or IIE, where R28 or R29 are a covalent bond linking the second ligand to the linker.
23. The pharmaceutical composition of Claim 22, wherein each R25 group is independently selected from the group consisting of aryl, heteroaryl, heterocyclic, cycloalkyl and substituted cycloalkyl.
24. The pharmaceutical composition of Claim 23, wherein each R25 group is substituted with from 1 to 4 substituents and one of the substituents is attached to the ring atom in the position adjacent to the atom attached to the -C(O)- group.
25. The pharmaceutical composition of Claim 24, wherein each R25 is a 2-(4'-trifluoromethylphenyl)phenyl group.
26. The pharmaceutical composition of Claim 22, wherein each linker independently has the formula:
-X a-Z-(Y a-Z)m-Y b-Z-X a-wherein m is an integer of from 0 to 20;
X a at each separate occurrence is selected from the group consisting of -O-, -S-, -NR-, -C(O)-, -C(O)O-, -C(O)NR-, -C(S), -C(S)O-, -C(S)NR- or a covalent bond where R is as defined below;
Z is at each separate occurrence is selected from the group consisting of alkylene, substituted alkylene, cycloalkylene, substituted cylcoalkylene, alkenylene, substituted alkenylene, alkynylene, substituted alkynylene, cycloalkenylene, substituted cycloalkenylene, arylene, heteroarylene, heterocyclene, or a covalent bond;
Y a and Y b at each separate occurrence are selected from the group consisting of -C(O)NR'-, -NR'C(O)-, -NR'C(O)NR'-, -C(=NR')-NR'-, -NR'-C(=NR')-, -NR'-C(O)-O-, -N=C(X a)-NR'-, -P(O)(OR')-O-, -S(O)n CR'R"-, -S(O)n NR'-, -S-S- and a covalent bond; where n is 0, 1 or 2;
and R, R' and R" at each separate occurrence are selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, cycloalkenyl, substituted cycloalkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl and heterocyclic.
27. A method for preventing or treating atherosclerosis in a patient, the method comprising administering to a patient with atherosclerosis or at risk for developing atherosclerosis a pharmaceutical composition comprising a pharmaceutically-acceptable carrier and a therapeutically-effective amount of a multibinding compound of Claims 1 or 9.
28. A method for lowering serum lipid, cholesterol and/or triglyceride levels in a patient, the method comprising administering to a patient a pharmaceutical composition comprising a pharmaceutically-acceptable carrier and a therapeutically-effective amount of a multibinding compound of Claims 1 or 9.
29. A method for preventing or treating hyperlipemia, hyperlipidemia, hyperlipoproteinemia, hypercholestrolemia, hypertriglyceridemia, pancreatitis, diabetes and/or obsesity in a patient, the method comprising administering to a patient in need of such treatment a pharmaceutical composition comprising a pharmaceutically-acceptable carrier and a therapeutically-effective amount of a multibinding compound of Claims 1 or 9.
30. A method for identifying multimeric ligand compounds possessing multibinding properties for microsomal triglyceride transferase protein, which method comprises:
(a) identifying a ligand or a mixture of ligands wherein each ligand contains at least one reactive functionality;
(b) identifying a library of linkers wherein each linker in said library comprises at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand;
(c) preparing a multimeric ligand compound library by combining at least two stoichiometric equivalents of the ligand or mixture of ligands identified in (a) with the library of linkers identified in (b) under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said ligands; and (d) assaying the multimeric ligand compounds produced in the library prepared in (c) above to identify multimeric ligand compounds possessing multibinding properties for microsomal triglyceride transferase protein.
31. A method for identifying multimeric ligand compounds possessing multibinding properties for microsomal triglyceride transferase protein, which method comprises:
(a) identifying a library of ligands wherein each ligand contains at least one reactive functionality;
(b) identifying a linker or mixture of linkers wherein each linker comprises at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand;
(c) preparing a multimeric ligand compound library by combining at least two stoichiometric equivalents of the library of ligands identified in (a) with the linker or mixture of linkers identified in (b) under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said ligands; and (d) assaying the multimeric ligand compounds produced in the library prepared in (c) above to identify multimeric ligand compounds possessing multibinding properties for microsomal triglyceride transferase protein.
32. The method according to Claim 30 or 31, wherein the preparation of the multimeric ligand compound library is achieved by either the sequential or concurrent combination of the two or more stoichiometric equivalents of the ligands identified in (a) with the linkers identified in (b).
33. The method according to Claim 32, wherein the multimeric ligand compounds comprising the multimeric ligand compound library are dimeric.
34. The method according to Claim 33, wherein the dimeric ligand compounds comprising the dimeric ligand compound library are heterodimeric.
35. The method according to Claim 34, wherein the heterodimeric ligand compound library is prepared by sequential addition of a first and second ligand.
36. The method according to Claim 30 or 31, wherein, prior to procedure (d), each member of the multimeric ligand compound library is isolated from the library.
37. The method according to Claim 36, wherein each member of the library is isolated by preparative liquid chromatography mass spectrometry (LCMS).
38. The method according to Claim 30 or Claim 31, wherein the linker or linkers employed are selected from the group comprising flexible linkers, rigid linkers, hydrophobic linkers, hydrophilic linkers, linkers of different geometry, acidic linkers, basic linkers, linkers of different polarization and/or polarizability and amphiphilic linkers.
39. The method according to Claim 38, wherein the linkers comprise linkers of different chain length and/or having different complementary reactive groups.
40. The method according to Claim 39, wherein the linkers are selected to have different linker lengths ranging from about 2 to 100.ANG..
41. The method according to Claim 30 or 31, wherein the ligand or mixture of ligands is selected to have reactive functionality at different sites on said ligands.
42. The method according to Claim 41, wherein said reactive functionality is selected from the group consisting of carboxylic acids, carboxylic acid halides, carboxyl esters, amines, halides, pseudohalides, isocyanates, vinyl unsaturation, ketones, aldehydes, thiols, alcohols, anhydrides, boronates, and precursors thereof wherein the reactive functionality on the ligand is selected to be complementary to at least one of the reactive groups on the linker so that a covalent linkage can be formed between the linker and the ligand.
43. The method according to Claim 30 or Claim 31, wherein the multimeric ligand compound library comprises homomeric ligand compounds.
44. The method according to Claim 30 or Claim 31, wherein the multimeric ligand compound library comprises heteromeric ligand compounds.
45. A library of multimeric ligand compounds which may possess multivalent properties for microsomal triglyceride transferase protein, which library is prepared by the method comprising:
(a) identifying a ligand or a mixture of ligands wherein each ligand contains at least one reactive functionality;
(b) identifying a library of linkers wherein each linker in said library comprises at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand; and (c) preparing a multimeric ligand compound library by combining at least two stoichiometric equivalents of the ligand or mixture of ligands identified in (a) with the library of linkers identified in (b) under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said ligands.
46. A library of multimeric ligand compounds which may possess multivalent properties for microsomal triglyceride transferase protein, which library is prepared by the method comprising:
(a) identifying a library of ligands wherein each ligand contains at least one reactive functionality;
(b) identifying a linker or mixture of linkers wherein each linker comprises at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand; and (c) preparing a multimeric ligand compound library by combining at least two stoichiometric equivalents of the library of ligands identified in (a) with the linker or mixture of linkers identified in (b) under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said Ligands.
47. The library according to Claim 45 or Claim 46, wherein the linker or linkers employed are selected from the group comprising flexible linkers, rigid linkers, hydrophobic linkers, hydrophilic linkers, linkers of different geometry, acidic linkers, basic linkers, linkers of different polarization and/or polarizability and amphiphilic linkers.
48. The library according to Claim 47, wherein the linkers comprise linkers of different chain length and/or having different complementary reactive groups.
49. The library according to Claim 48, wherein the linkers are selected to have different linker lengths ranging from about 2 to 100.ANG..
50. The library according to Claim 45 or 46, wherein the ligand or mixture of ligands is selected to have reactive functionality at different sites on said ligands.
51. The library according to Claim 50, wherein said reactive functionality is selected from the group consisting of carboxylic acids, carboxylic acid halides, carboxyl esters, amines, halides, pseudohalides, isocyanates, vinyl unsaturation, ketones, aldehydes, thiols, alcohols, anhydrides, boronates, and precursors thereof wherein the reactive functionality on the ligand is selected to be complementary to at least one of the reactive groups on the linker so that a covalent linkage can be formed between the linker and the ligand.
52. The library according to Claim 45 or Claim 46, wherein the multimeric ligand compound library comprises homomeric ligand compounds.
53. The library according to Claim 45 or Claim 46, wherein the multimeric ligand compound library comprises heteromeric ligand compounds.
54. An iterative method for identifying multimeric ligand compounds possessing multibinding properties for microsomal triglyceride transferase protein, which method comprises:
(a) preparing a first collection or iteration of multimeric compounds which is prepared by contacting at least two stoichiometric equivalents of the ligand or mixture of ligands which target a receptor with a linker or mixture of linkers wherein said ligand or mixture of ligands comprises at least one reactive functionality and said linker or mixture of linkers comprises at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand wherein said contacting is conducted under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said ligands;
(b) assaying said first collection or iteration of multimeric compounds to assess which if any of said multimeric compounds possess multibinding properties for microsomal triglyceride transferase protein;

(c) repeating the process of (a) and (b) above until at least one multimeric compound is found to possess multibinding properties for microsomal triglyceride transferase protein;
(d) evaluating what molecular constraints imparted or are consistent with imparting multibinding properties to the multimeric compound or compounds found in the first iteration recited in (a)- (c) above;
(e) creating a second collection or iteration of multimeric compounds which elaborates upon the particular molecular constraints imparting multibinding properties to the multimeric compound or compounds found in said first iteration;
(f) evaluating what molecular constraints imparted or are consistent with imparting enhanced multibinding properties to the multimeric compound or compounds found in the second collection or iteration recited in (e) above;
(g) optionally repeating steps (e) and (f) to further elaborate upon said molecular constraints.
55. The method according to Claim 54, wherein steps (e) and (f) are repeated from 2-50 times.
56. The method according to Claim 55, wherein steps (e) and (f) are repeated from 5-50 times.
CA002319495A 1998-06-08 1999-06-04 Multibinding inhibitors of microsomal triglyceride transferase protein Abandoned CA2319495A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US8844898P 1998-06-08 1998-06-08
US60/088,448 1998-06-08
US9307298P 1998-07-16 1998-07-16
US60/093,072 1998-07-16
PCT/US1999/011789 WO1999063929A2 (en) 1998-06-08 1999-06-04 Multibinding inhibitors of microsomal triglyceride transferase protein

Publications (1)

Publication Number Publication Date
CA2319495A1 true CA2319495A1 (en) 1999-12-16

Family

ID=26778665

Family Applications (11)

Application Number Title Priority Date Filing Date
CA002319495A Abandoned CA2319495A1 (en) 1998-06-08 1999-06-04 Multibinding inhibitors of microsomal triglyceride transferase protein
CA002321162A Abandoned CA2321162A1 (en) 1998-06-08 1999-06-07 Novel antibacterial compounds
CA2319080A Expired - Lifetime CA2319080C (en) 1998-06-08 1999-06-07 Multivalent antibacterial agents
CA002319120A Abandoned CA2319120A1 (en) 1998-06-08 1999-06-07 Phosphodiesterase-v modulator drugs and their uses
CA002320241A Abandoned CA2320241A1 (en) 1998-06-08 1999-06-08 Multivalent macrolide antibiotics
CA002319477A Abandoned CA2319477A1 (en) 1998-06-08 1999-06-08 Inhibitors of h+k+-atpase
CA002316999A Abandoned CA2316999A1 (en) 1998-06-08 1999-06-08 Inhibitors of hiv reverse transcriptase
CA002321190A Abandoned CA2321190A1 (en) 1998-06-08 1999-06-08 Novel therapeutic agents that modulate enzymatic processes
CA002321166A Abandoned CA2321166A1 (en) 1998-06-08 1999-06-08 Multibinding inhibitors of topoisomerase
CA002319474A Abandoned CA2319474A1 (en) 1998-06-08 1999-06-08 Inhibitors of nitric oxide synthase
CA002321274A Abandoned CA2321274A1 (en) 1998-06-08 1999-06-08 Novel antiviral compounds

Family Applications After (10)

Application Number Title Priority Date Filing Date
CA002321162A Abandoned CA2321162A1 (en) 1998-06-08 1999-06-07 Novel antibacterial compounds
CA2319080A Expired - Lifetime CA2319080C (en) 1998-06-08 1999-06-07 Multivalent antibacterial agents
CA002319120A Abandoned CA2319120A1 (en) 1998-06-08 1999-06-07 Phosphodiesterase-v modulator drugs and their uses
CA002320241A Abandoned CA2320241A1 (en) 1998-06-08 1999-06-08 Multivalent macrolide antibiotics
CA002319477A Abandoned CA2319477A1 (en) 1998-06-08 1999-06-08 Inhibitors of h+k+-atpase
CA002316999A Abandoned CA2316999A1 (en) 1998-06-08 1999-06-08 Inhibitors of hiv reverse transcriptase
CA002321190A Abandoned CA2321190A1 (en) 1998-06-08 1999-06-08 Novel therapeutic agents that modulate enzymatic processes
CA002321166A Abandoned CA2321166A1 (en) 1998-06-08 1999-06-08 Multibinding inhibitors of topoisomerase
CA002319474A Abandoned CA2319474A1 (en) 1998-06-08 1999-06-08 Inhibitors of nitric oxide synthase
CA002321274A Abandoned CA2321274A1 (en) 1998-06-08 1999-06-08 Novel antiviral compounds

Country Status (17)

Country Link
US (6) US6288234B1 (en)
EP (11) EP1085846A2 (en)
JP (3) JP2004512255A (en)
AR (3) AR019634A1 (en)
AT (1) ATE322910T1 (en)
AU (11) AU4543899A (en)
CA (11) CA2319495A1 (en)
CY (1) CY1105087T1 (en)
DE (1) DE69930818T2 (en)
DK (1) DK1005356T3 (en)
ES (1) ES2263274T3 (en)
HK (1) HK1028737A1 (en)
NZ (1) NZ505979A (en)
PT (1) PT1005356E (en)
SG (3) SG106036A1 (en)
TW (1) TWI239959B (en)
WO (11) WO1999063929A2 (en)

Families Citing this family (157)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2330611A1 (en) * 1998-05-22 1999-12-02 The Board Of Trustees Of The Leland Stanford Junior University Bifunctional molecules and therapies based thereon
AUPP913999A0 (en) 1999-03-12 1999-04-01 Biota Scientific Management Pty Ltd Novel chemical compounds and their use
US20020048571A1 (en) * 1999-07-19 2002-04-25 Jeno Gyuris Chimeric polypeptides of serum albumin and uses related thereto
DE19933926A1 (en) 1999-07-20 2001-01-25 Boehringer Ingelheim Pharma Biphenyl derivatives, their preparation and their use as medicines
AU5424500A (en) 2000-02-24 2001-09-03 Kopran Res Lab Ltd Orally administrable acid stable anti-ulcer benzimidazole derivatives
EP1328658A2 (en) * 2000-06-22 2003-07-23 San Diego State University Foundation Recombination modulators and methods for their production and use
DE10033337A1 (en) * 2000-07-08 2002-01-17 Boehringer Ingelheim Pharma Biphenylcarboxamides, their preparation and their use as medicaments
BRPI0003386B8 (en) 2000-08-08 2021-05-25 Cristalia Produtos Quim Farmaceuticos Ltda homo- or heterodimeric prodrugs useful in treating diseases or disorders mediated by phosphodiesterases; pharmaceutical compositions containing the prodrug or its pharmaceutical acceptable salts; process of obtaining these prodrugs
US6947844B2 (en) 2000-08-09 2005-09-20 Yale University Modulators of ribosomal function and identification thereof
US6638908B1 (en) 2000-08-09 2003-10-28 Yale University Crystals of the large ribosomal subunit
DE10042853A1 (en) * 2000-08-30 2002-04-25 Florian Lang Detection and influencing of the expression or function of CD95 / CD95L in infections
UA80393C2 (en) 2000-12-07 2007-09-25 Алтана Фарма Аг Pharmaceutical preparation comprising an pde inhibitor dispersed on a matrix
DE10061137B4 (en) * 2000-12-07 2016-10-06 Takeda Gmbh New pharmaceutical preparation
HRP20010018A2 (en) * 2001-01-09 2002-12-31 Pliva D D Novel anti-inflammatory compounds
BRPI0210383B1 (en) * 2001-06-12 2015-08-04 Wellstat Therapeutics Corp Biologically active agent, process for its production, use and pharmaceutical composition comprising the same
TNSN03146A1 (en) 2001-06-28 2005-12-23 Pfizer Prod Inc TRIAMIDE SUBSTITUTED INDOLES, BENZOFURANS AND BENZOTHIOPHENES, USEFUL AS SECRETION INHIBITORS OF MICROSOMAL TRIGLYCERIDE TRANSFER PROTEIN AND / OR APOLIPO-PROTEIN B (APO B).
IL151012A0 (en) 2001-08-03 2003-02-12 Ribosomes Structure And Protei Ribosomes structure and protein synthesis inhibitors
US6952650B2 (en) 2001-08-03 2005-10-04 Yale University Modulators of ribosomal function and identification thereof
US20030158093A1 (en) * 2001-08-17 2003-08-21 Binyuan Sun Bifunctional glycopeptide antibiotics and combinatorial libararies thereof
NZ531621A (en) 2001-09-11 2005-06-24 Astrazeneca Ab Oxazolidinone and/or isoxazoline as antibacterial agents
KR20040041177A (en) 2001-09-26 2004-05-14 바이엘 파마슈티칼스 코포레이션 1,6-Naphthyridine Derivatives as Antidiabetics
TWI335332B (en) 2001-10-12 2011-01-01 Theravance Inc Cross-linked vancomycin-cephalosporin antibiotics
AUPR879601A0 (en) * 2001-11-09 2001-12-06 Biota Scientific Management Pty Ltd Novel chemical compounds and their use
WO2003053420A1 (en) * 2001-12-20 2003-07-03 Pharmacia Corporation Multiple-pulse extended release formulations of clindamycin
IL162734A0 (en) * 2002-02-01 2005-11-20 Ariad Gene Therapeutics Inc Phosphorus-containing compounds & uses thereof
NZ535355A (en) 2002-02-15 2007-09-28 Merckle Gmbh Antibiotic conjugates
US7579324B2 (en) 2002-02-15 2009-08-25 C-A-I-R Biosciences Gmbh Conjugates of biologically active compounds, methods for their preparation and use, formulation and pharmaceutical applications thereof
EP1483579A4 (en) 2002-02-15 2006-07-12 Merckle Gmbh Conjugates of biologically active compounds, methods for their preparation and use, formulation and pharmaceutical applications thereof
MY140561A (en) 2002-02-20 2009-12-31 Nycomed Gmbh Dosage form containing pde 4 inhibitor as active ingredient
WO2003072532A1 (en) * 2002-02-28 2003-09-04 Japan Tobacco Inc. Ester compound and medicinal use thereof
US20040171062A1 (en) * 2002-02-28 2004-09-02 Plexxikon, Inc. Methods for the design of molecular scaffolds and ligands
US6946472B2 (en) * 2002-04-30 2005-09-20 Hoffman-La Roche Inc. Polycyclic compounds exhibiting anti-tumor activities
AU2003231833A1 (en) 2002-05-24 2003-12-12 Theravance, Inc. Cross-linked glycopeptide-cephalosporin antibiotics
CA2489398A1 (en) * 2002-07-08 2004-01-15 Pliva-Istrazivacki Institut D.O.O. Novel nonsteroidal anti-inflammatory substances, compositions and methods for their use
EP1551865B1 (en) * 2002-07-08 2009-04-22 GlaxoSmithKline istrazivacki centar Zagreb d.o.o. Hybrid molecules of macrolides with steroidal/non-steroidal anti-inflammatory molecules
JP4617449B2 (en) 2002-07-11 2011-01-26 ヴィキュロン ファーマシューティカルズ インコーポレイテッド N-hydroxyamide derivative having antibacterial activity
NZ538141A (en) 2002-08-15 2007-06-29 Vicuron Pharm Inc Lincomycin derivatives possessing antibacterial activity
US7199105B2 (en) 2002-08-15 2007-04-03 Vicuron Pharmaceuticals, Inc. Lincomycin derivatives possessing antibacterial activity
US7091196B2 (en) * 2002-09-26 2006-08-15 Rib-X Pharmaceuticals, Inc. Bifunctional heterocyclic compounds and methods of making and using same
JP2007524374A (en) * 2003-02-28 2007-08-30 プレキシコン,インコーポレーテッド PYK2 crystal structure and use
EP1599491A1 (en) * 2003-03-05 2005-11-30 Rib-X Pharmaceuticals, Inc. Bifunctional heterocyclic compounds and methods of making and using the same
RS52908B (en) 2003-03-10 2014-02-28 Takeda Gmbh Novel process for the preparation of roflumilast
US20040216174A1 (en) * 2003-03-14 2004-10-28 Siegfried Hekimi Screening assays for targets and drugs useful in treatment and prevention of lipid metabolism disorders
KR101127390B1 (en) * 2003-04-15 2012-03-23 웰스태트 테러퓨틱스 코포레이션 Compounds for the treatment of metabolic disorders
US7271252B2 (en) * 2003-04-22 2007-09-18 Roche Diagnostics Operations, Inc. Reagents for detecting efavirenz
AU2004237602B2 (en) * 2003-04-30 2009-05-28 Wellstat Therapeutics Corporation Compounds for the treatment of metabolic disorders
ES2335013T3 (en) 2003-05-23 2010-03-18 Theravance, Inc. GLUCOPEPTIDE-CEPHALOSPORIN ANTIBIOTICS RETICULATED.
US7199106B2 (en) 2003-06-17 2007-04-03 Vicuron Pharmaceuticals, Inc. Lincomycin derivatives possessing antimicrobial activity
US7256177B2 (en) 2003-06-17 2007-08-14 Vicuron Pharmaceuticals, Inc. Lincomycin derivatives possessing antibacterial activity
US20050079548A1 (en) * 2003-07-07 2005-04-14 Plexxikon, Inc. Ligand development using PDE4B crystal structures
DE602004012269T2 (en) 2003-07-11 2009-04-30 Theravance, Inc., South San Francisco CROSS-LINKED GLYCOPEPTIDE-CEPHALOSPORIN ANTIBIOTICS
US7202266B2 (en) * 2003-07-17 2007-04-10 Plexxikon, Inc. PPAR active compounds
US7348338B2 (en) * 2003-07-17 2008-03-25 Plexxikon, Inc. PPAR active compounds
JP4832897B2 (en) 2003-08-29 2011-12-07 日本たばこ産業株式会社 Ester derivatives and their pharmaceutical uses
CA2537182A1 (en) * 2003-08-29 2005-03-10 Dynogen Pharmaceuticals, Inc. Compositions useful for treating gastrointestinal motility disorders
US20050164300A1 (en) * 2003-09-15 2005-07-28 Plexxikon, Inc. Molecular scaffolds for kinase ligand development
US20070149463A1 (en) * 2003-10-30 2007-06-28 Oyelere Adegboyega K Bifunctional macrolide heterocyclic compounds and methods of making and using the same
US20070270357A1 (en) * 2003-11-18 2007-11-22 Farmer Jay J Bifunctional Macrolide Heterocyclic Compounds and Methods of Making and Using the Same
EP1696920B8 (en) 2003-12-19 2015-05-06 Plexxikon Inc. Compounds and methods for development of ret modulators
US20070066641A1 (en) * 2003-12-19 2007-03-22 Prabha Ibrahim Compounds and methods for development of RET modulators
CA2551566A1 (en) * 2004-02-04 2005-08-18 Active Biotech Ab Diurea derivatives
US7361743B2 (en) 2004-02-11 2008-04-22 Pfizer Inc Lincomycin derivatives possessing antibacterial activity
WO2005085266A2 (en) 2004-02-27 2005-09-15 Rib-X Pharmaceuticals, Inc. Macrocyclic compounds and methods of making and using the same
CA2565965A1 (en) 2004-05-06 2006-07-27 Plexxikon, Inc. Pde4b inhibitors and uses therefor
US7498342B2 (en) 2004-06-17 2009-03-03 Plexxikon, Inc. Compounds modulating c-kit activity
US20060030623A1 (en) * 2004-07-16 2006-02-09 Noboru Furukawa Agent for the treatment or prevention of diabetes, obesity or arteriosclerosis
AU2005281359A1 (en) * 2004-08-26 2006-03-16 Apparao Satyam Prodrugs and codrugs containing novel bio-cleavable disulfide linkers
TW200616604A (en) 2004-08-26 2006-06-01 Nicholas Piramal India Ltd Nitric oxide releasing prodrugs containing bio-cleavable linker
AU2005279795A1 (en) * 2004-09-03 2006-03-09 Plexxikon, Inc. Bicyclic heteroaryl PDE4B inhibitors
US8101774B2 (en) * 2004-10-18 2012-01-24 Japan Tobacco Inc. Ester derivatives and medicinal use thereof
RU2375046C2 (en) * 2004-10-25 2009-12-10 Джапан Тобакко Инк. Solid composition with improved solubility and stability and method of obtaining it
EP1819673A2 (en) * 2004-11-30 2007-08-22 Plexxikon, Inc. Indole derivatives for use as ppar active compounds
EP1835913A2 (en) 2004-12-06 2007-09-26 Avigen, Inc. Ibudilast for treating neuropathic pain and associated syndromes
US20060160135A1 (en) * 2004-12-08 2006-07-20 Weiru Wang SF-1 and LRH-1 modulator development
EP1861074B1 (en) * 2005-03-16 2013-04-24 Takeda GmbH Taste masked dosage form containing roflumilast
AU2006235483B2 (en) * 2005-04-12 2010-11-25 Elan Pharma International Limited Controlled release compositions comprising a cephalosporin for the treatment of a bacterial infection
WO2007013896A2 (en) * 2005-05-17 2007-02-01 Plexxikon, Inc. Pyrrol (2,3-b) pyridine derivatives protein kinase inhibitors
EP1890767A2 (en) * 2005-05-27 2008-02-27 Pfizer Products Inc. Combination of a cannabinoid-1- receptor-antagonist and a microsomal triglyceride transfer protein inhibitor for treating obesity or mainataining weight loss
HUE027370T2 (en) * 2005-06-22 2016-10-28 Plexxikon Inc Pyrrolo [2,3-b]pyridine derivatives as protein kinase inhibitors
JP2009506063A (en) 2005-08-24 2009-02-12 リブ−エックス ファーマシューティカルズ,インコーポレイテッド Triazole compounds and methods for making and using the same
JP2009506066A (en) 2005-08-24 2009-02-12 リブ−エックス ファーマシューティカルズ,インコーポレイテッド Triazole compounds and methods for making and using the same
RU2419618C2 (en) * 2005-09-07 2011-05-27 Плекссикон, Инк. Compounds, active towards ppar (peroxisome proliferator-activated receptor)
US7915285B2 (en) 2005-09-26 2011-03-29 The Regents Of The University Of Colorado Method for treating drug and behavioral addictions
CA2653345A1 (en) 2006-05-31 2007-12-13 Avigen, Inc. Ibudilast for inhibiting macrophage migration inhibitory factor (mif) activity
US20080161335A1 (en) * 2006-11-14 2008-07-03 Vladyka Ronald S Oral formulations
WO2008063888A2 (en) 2006-11-22 2008-05-29 Plexxikon, Inc. Compounds modulating c-fms and/or c-kit activity and uses therefor
WO2008079909A1 (en) * 2006-12-21 2008-07-03 Plexxikon, Inc. Pyrrolo [2,3-b] pyridines as kinase modulators
PE20121126A1 (en) * 2006-12-21 2012-08-24 Plexxikon Inc PIRROLO [2,3-B] PYRIDINES COMPOUNDS AS KINASE MODULATORS
RU2009122670A (en) 2006-12-21 2011-01-27 Плекссикон, Инк. (Us) COMPOUNDS AND METHODS FOR MODULATION OF KINASES AND INDICATIONS FOR THEIR USE
US20080181876A1 (en) * 2007-01-30 2008-07-31 Johnson Kirk W Methods for treating acute and subchronic pain
WO2008095063A1 (en) 2007-01-31 2008-08-07 Dana-Farber Cancer Institute, Inc. Stabilized p53 peptides and uses thereof
US9260476B2 (en) 2007-02-23 2016-02-16 The Research Foundation For The State University Of New York RNA targeting compounds and methods for making and using same
EP2129388B1 (en) * 2007-02-23 2012-09-12 The Research Foundation Of State University Of New York Rna targeting compounds and methods for making and using same
PE20090159A1 (en) * 2007-03-08 2009-02-21 Plexxikon Inc INDOL-PROPIONIC ACID DERIVED COMPOUNDS AS PPARs MODULATORS
ES2430067T3 (en) 2007-03-28 2013-11-18 President And Fellows Of Harvard College Sewn polypeptides
JP5119543B2 (en) * 2007-04-06 2013-01-16 独立行政法人国立がん研究センター Screening method for drugs used in the treatment of colorectal cancer
US20080287402A1 (en) * 2007-05-03 2008-11-20 Johnson Kirk W Use of a glial attenuator to prevent amplified pain responses caused by glial priming
NZ582772A (en) 2007-07-17 2012-06-29 Plexxikon Inc Compounds and methods for kinase modulation, and indications therefor
KR20100093532A (en) 2007-10-25 2010-08-25 아카오젠, 인코포레이티드 CARBACEPHEM β-LACTAM ANTIBIOTICS
CA2723358A1 (en) * 2008-05-05 2009-11-12 Allison B. Reiss Method for improving cardiovascular risk profile of cox inhibitors
CN103251631A (en) * 2008-05-13 2013-08-21 根梅迪卡治疗公司 Salicylate conjugates useful for treating metabolic disorders
US20100239552A1 (en) * 2009-03-16 2010-09-23 Genmedica Therapeutics Sl Combination Therapies for Treating Metabolic Disorders
CA2755069A1 (en) * 2009-03-16 2010-09-23 Genmedica Therapeutics Sl Anti-inflammatory and antioxidant conjugates useful for treating metabolic disorders
TWI404719B (en) * 2009-04-03 2013-08-11 Hoffmann La Roche Compositions and uses thereof
NZ624963A (en) 2009-04-29 2016-07-29 Amarin Pharmaceuticals Ie Ltd Pharmaceutical compositions comprising epa and a cardiovascular agent and methods of using the same
WO2011007367A1 (en) * 2009-07-15 2011-01-20 Lupin Limited An improved process for preparation of efavirenz
US8329724B2 (en) 2009-08-03 2012-12-11 Hoffmann-La Roche Inc. Process for the manufacture of pharmaceutically active compounds
CN110483499A (en) 2009-11-05 2019-11-22 圣母大学 Imidazo [1,2-a] pyridine compounds and their and its synthesis and application method
CN106220623A (en) 2009-11-06 2016-12-14 普莱希科公司 Compounds and methods for and indication thereof for kinases regulation
US8992897B2 (en) 2010-01-06 2015-03-31 Elc Management Llc Skin lightening compositions
US8722026B2 (en) 2010-01-06 2014-05-13 Elc Management, Llc Skin lightening compositions
ES2711526T3 (en) 2010-08-13 2019-05-06 Aileron Therapeutics Inc Peptidomimetic macrocycles
EP3246026A3 (en) 2010-08-18 2018-02-21 Del Mar Pharmaceuticals Compositions and methods to improve the therapeutic benefit of suboptimally administered chemical compounds including substituted hexitols such as dianhydrogalactitol and diacetyldianhydrogalactitol
US8466197B2 (en) 2010-12-14 2013-06-18 Genmedica Therapeutics Sl Thiocarbonates as anti-inflammatory and antioxidant compounds useful for treating metabolic disorders
WO2012109075A1 (en) 2011-02-07 2012-08-16 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
AR085279A1 (en) 2011-02-21 2013-09-18 Plexxikon Inc SOLID FORMS OF {3- [5- (4-CHLORINE-PHENYL) -1H-PIRROLO [2,3-B] PIRIDINA-3-CARBONIL] -2,4-DIFLUOR-PHENIL} -AMIDE OF PROPANE ACID-1- SULFONIC
US9221760B2 (en) 2011-05-09 2015-12-29 Van Andel Research Institute Autophagy inhibitors
ES2597034T3 (en) * 2011-05-30 2017-01-13 Astellas Pharma Inc. Imidazopyridine Compound
CA2863679A1 (en) 2011-08-17 2014-05-30 Dennis Brown Compositions and methods to improve the therapeutic benefit of suboptimally administered chemical compounds including substituted hexitols such as dibromodulcitol
WO2013036783A2 (en) 2011-09-09 2013-03-14 Cubist Pharmaceuticals, Inc. Methods for treating intrapulmonary infections
KR20140100937A (en) 2011-10-18 2014-08-18 에일러론 테라퓨틱스 인코포레이티드 Peptidomimetic macrocycles
BR112014017833A2 (en) 2012-01-20 2021-03-23 Dennis Brown USE OF REPLACED HEXITOLS INCLUDING DIANIDROGALACTITOL AND ANALOGS TO TREAT NEOPLASTIC DISEASE AND CANCER STEM CELLS INCLUDING MULTIFORM GLIOBLASTOMA AND MEDULOBLASTOMA
RU2017145921A (en) 2012-02-15 2019-02-21 Эйлерон Терапьютикс, Инк. PEPTIDOMIMETIC MACROCYCLES
WO2013123267A1 (en) 2012-02-15 2013-08-22 Aileron Therapeutics, Inc. Triazole-crosslinked and thioether-crosslinked peptidomimetic macrocycles
US20130231372A1 (en) * 2012-03-01 2013-09-05 Mary Matsui Small Molecule Inhibitors Of P-type ATPases
WO2013169600A1 (en) 2012-05-09 2013-11-14 Delmar Pharmaceuticals Veterinary use of dianhydrogalactitol, diacetyldianhydrogalactitol, and dibromodulcitol to treat malignancies
US9150570B2 (en) 2012-05-31 2015-10-06 Plexxikon Inc. Synthesis of heterocyclic compounds
CN104797267A (en) 2012-06-26 2015-07-22 德玛医药 Methods for treating tyrosine-kinase-inhibitor-resistant malignancies in patients with genetic polymorphisms or ahi1 dysregulations or mutations employing dianhydrogalactitol, diacetyldianhydrogalactitol, dibromodulcitol, or analogs or derivatives thereof
CN104812384B (en) 2012-11-01 2020-09-18 爱勒让治疗公司 Disubstituted amino acids and methods of making and using the same
PL2927231T3 (en) * 2012-11-30 2018-01-31 Astellas Pharma Inc Imidazopyridine compounds
US8883857B2 (en) 2012-12-07 2014-11-11 Baylor College Of Medicine Small molecule xanthine oxidase inhibitors and methods of use
US20140274993A1 (en) 2013-03-15 2014-09-18 Cubist Pharmaceuticals, Inc. Ceftolozane-tazobactam pharmaceutical compositions
PE20160048A1 (en) 2013-03-15 2016-02-12 Merck Sharp & Dohme ANTIBIOTIC COMPOSITIONS OF CEFTOLOZANE
US9872906B2 (en) 2013-03-15 2018-01-23 Merck Sharp & Dohme Corp. Ceftolozane antibiotic compositions
CN105492011A (en) 2013-04-08 2016-04-13 丹尼斯·M·布朗 Therapeutic benefit of suboptimally administered chemical compounds
CA2938626A1 (en) 2013-07-26 2015-01-29 John Rothman Compositions to improve the therapeutic benefit of bisantrene
CN103439503B (en) * 2013-08-03 2016-12-28 河南省农业科学院 The enzyme linked immunological kit of Sparfloxacin and establishment thereof and detection method
WO2015022335A1 (en) * 2013-08-12 2015-02-19 Katholieke Universiteit Leuven Vancomycin analogs
EP3043797B1 (en) 2013-09-09 2020-04-08 Merck Sharp & Dohme Corp. Treating infections with ceftolozane/tazobactam in subjects having impaired renal function
US20150094293A1 (en) 2013-09-27 2015-04-02 Calixa Therapeutics, Inc. Solid forms of ceftolozane
CN103755728B (en) * 2013-12-24 2016-06-29 深圳华润九新药业有限公司 Cefazolin derivatives and preparation method thereof, oral antibiotic preparation
WO2015114666A2 (en) 2014-01-29 2015-08-06 Vyome Biosciences Pvt. Ltd. Treatments for resistant acne
SG11201608303QA (en) 2014-04-04 2016-11-29 Del Mar Pharmaceuticals Use of dianhydrogalactitol and analogs or derivatives thereof to treat non-small-cell carcinoma of the lung and ovarian cancer
WO2016012934A1 (en) * 2014-07-21 2016-01-28 Mylan Laboratories Ltd Process for making lomitapide mesylate
JP2017533889A (en) 2014-09-24 2017-11-16 エルロン・セラピューティクス・インコーポレイテッドAileron Therapeutics,Inc. Peptidomimetic macrocycles and uses thereof
CN107106642B (en) 2014-09-24 2021-02-26 艾瑞朗医疗公司 Peptidomimetic macrocycles and formulations thereof
EP3215490A1 (en) * 2014-11-05 2017-09-13 Hetero Research Foundation Process for the preparation of lomitapide
US10253067B2 (en) 2015-03-20 2019-04-09 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
BR102015025502B1 (en) * 2015-04-30 2022-06-21 Aegerion Pharmaceuticals, Inc Composition of lomitapide, tablet, lomitapide product, methods for analyzing a sample composition of lomitapide and for determining an amount of an impurity in a sample of the composition
CN105671110B (en) * 2015-05-05 2019-02-01 重庆乾泰生物医药有限公司 A method of producing Dalbavancin precursor A40926
JP2018528217A (en) 2015-09-10 2018-09-27 エルロン・セラピューティクス・インコーポレイテッドAileron Therapeutics,Inc. Peptidomimetic macrocycles as modulators of MCL-1
PE20181352A1 (en) 2015-09-17 2018-08-22 Marvin J Miller HETEROCYCLIC COMPOUNDS CONTAINING BENZYLAMINE AND USEFUL COMPOSITIONS AGAINST MYCOBACTERIAL INFECTION
WO2017084973A1 (en) * 2015-11-16 2017-05-26 Evonik Röhm Gmbh Injection solution comprising a non-nucleoside reverse-transcriptase inhibitor and poly(lactide-co-glycolide)
IL270686B (en) * 2017-05-22 2022-09-01 Insmed Inc Glycopeptide derivative compounds and uses thereof
CN107652268A (en) * 2017-08-01 2018-02-02 广州加德恩医药有限公司 The preparation method of 4 5-Hydroxyomeprazoles
JP2020537636A (en) * 2017-09-22 2020-12-24 マンセル、ジョン Compositions and Methods for the Treatment of Sepsis-Related Disorders
JPWO2022059700A1 (en) * 2020-09-15 2022-03-24

Family Cites Families (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4358525A (en) * 1978-10-10 1982-11-09 Eastman Kodak Company Blocked photographically useful compounds and photographic compositions, elements and processes employing them
BE886467A (en) * 1980-12-03 1981-04-01 Dong A Pharm Co Ltd PROCESS FOR THE PREPARATION OF ANTIBIOTICS DERIVATIVES FROM THE BETA-LACTAM SERIES, NEW INTERMEDIATE PRODUCTS AND PROCESS FOR THE PREPARATION THEREOF
US4693992A (en) * 1985-08-26 1987-09-15 International Minerals & Chemical Corp. Bacitracin-metal-polyether antibiotic complexes
US5023252A (en) 1985-12-04 1991-06-11 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery of drugs
US4996143A (en) * 1985-12-23 1991-02-26 Syngene, Inc. Fluorescent stokes shift probes for polynucleotide hybridization
US4879220A (en) * 1986-11-18 1989-11-07 State Of Oregon Acting By And Through The State Board Of Higher Education On Behalf Of The University Of Oregon Crosslinking receptor-specific probes for electron microscopy
US5686428A (en) * 1989-04-07 1997-11-11 Aktiebolaget Astra Pharmaceutical composition
JPH0429930A (en) * 1990-05-24 1992-01-31 Fujisawa Pharmaceut Co Ltd Antibacterial agent for prevention or therapy of enterococcus bacterium infectious disease
DE69126124T2 (en) * 1990-09-28 1997-08-28 Neorx Corp POLYMERS CARRIER FOR RELEASING COVALENT-RELATED ACTIVE SUBSTANCES
US5312837A (en) * 1991-01-29 1994-05-17 Genelabs Technologies, Inc. Method of treating viral infections with aryl macrocyclic compounds
ATE148889T1 (en) 1991-09-18 1997-02-15 Affymax Tech Nv METHOD FOR SYNTHESIS OF VARIOUS COLLECTIONS OF OLIGOMERS
US5595872A (en) 1992-03-06 1997-01-21 Bristol-Myers Squibb Company Nucleic acids encoding microsomal trigyceride transfer protein
US5739135A (en) 1993-09-03 1998-04-14 Bristol-Myers Squibb Company Inhibitors of microsomal triglyceride transfer protein and method
MY115155A (en) * 1993-09-09 2003-04-30 Upjohn Co Substituted oxazine and thiazine oxazolidinone antimicrobials.
US5519134A (en) * 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
WO1995019974A2 (en) * 1994-01-24 1995-07-27 Harris Stephen J Calixarene-based compounds having antibacterial, antifungal, anticancer-hiv activity
US5508289A (en) * 1994-03-14 1996-04-16 The United States America As Represented By The Department Of Health And Human Services Bis-acridone chemotherapeutic derivatives
US5807886A (en) * 1994-05-07 1998-09-15 Astra Aktiebolag Bicyclic amidine dervatives as inhibitors of nitric oxide synthetase
DE4418096A1 (en) * 1994-05-24 1995-11-30 Cassella Ag Use of pteridine derivatives as inhibitors of NO synthase
US5463564A (en) 1994-09-16 1995-10-31 3-Dimensional Pharmaceuticals, Inc. System and method of automatically generating chemical compounds with desired properties
US5718915A (en) * 1994-10-31 1998-02-17 Burstein Laboratories, Inc. Antiviral liposome having coupled target-binding moiety and hydrolytic enzyme
KR100517210B1 (en) * 1994-12-12 2006-06-07 오메로스 코포레이션 Infusion solution for pain, inflammation and spasm
US5693791A (en) 1995-04-11 1997-12-02 Truett; William L. Antibiotics and process for preparation
EP0832069B1 (en) 1995-06-07 2003-03-05 Pfizer Inc. BIPHENYL-2-CARBOXYLIC ACID-TETRAHYDRO-ISOQUINOLIN-6-YL AMIDE DERIVATIVES, THEIR PREPARATION AND THEIR USE AS INHIBITORS OF MICROSOMAL TRIGLYCERIDE TRANSFER PROTEIN AND/OR APOLIPOPROTEIN B (Apo B) SECRETION
US5698401A (en) * 1995-11-14 1997-12-16 Abbott Laboratories Use of nuclear magnetic resonance to identify ligands to target biomolecules
US5846839A (en) 1995-12-22 1998-12-08 Glaxo Group Limited Methods for hard-tagging an encoded synthetic library
EP0904262B1 (en) 1996-01-16 2004-04-21 Bristol-Myers Squibb Company Conformationally restricted aromatic inhibitors of microsomal triglyceride transfer protein and method
WO1997035195A1 (en) * 1996-03-19 1997-09-25 The Salk Institute For Biological Studies In vitro methods for identifying modulators of members of the steroid/thyroid superfamily of receptors
AU2609297A (en) * 1996-04-12 1997-11-07 Eli Lilly And Company Glycopeptide compounds
USH1729H (en) 1996-05-09 1998-05-05 Bristol-Myers Squibb Company Method for preparing compounds employing solid phase synthesis and novel linker-resin
US5827875A (en) * 1996-05-10 1998-10-27 Bristol-Myers Squibb Company Inhibitors of microsomal triglyceride transfer protein and method
US5885983A (en) 1996-05-10 1999-03-23 Bristol-Myers Squibb Company Inhibitors of microsomal triglyceride transfer protein and method
WO1998003174A1 (en) 1996-07-24 1998-01-29 Bristol-Myers Squibb Company Method for treating tumors having high ldl requirements employing mtp inhibitors
US5883109A (en) 1996-07-24 1999-03-16 Bristol-Myers Squibb Company Method for lowering serum lipid levels employing an MTP inhibitor in combination with another cholesterol lowering drug
US6030429A (en) * 1996-08-06 2000-02-29 Toyo Kohan Co., Ltd. Hard sintered alloy
US5723451A (en) * 1996-08-09 1998-03-03 Ontogen Corporation Nitric oxide synthase (NOS) inhibitors
US6121283A (en) 1996-11-27 2000-09-19 Pfizer Inc Apo B-secretion/MTP inhibitory amides
US5760246A (en) * 1996-12-17 1998-06-02 Biller; Scott A. Conformationally restricted aromatic inhibitors of microsomal triglyceride transfer protein and method
EP0946549A1 (en) * 1996-12-18 1999-10-06 Eli Lilly And Company Combinatorial process for preparing fused pyrimidine libraries
AU5513298A (en) 1996-12-20 1998-07-17 Bristol-Myers Squibb Company Heterocyclic inhibitors of microsomal triglyceride transfer protein and method
US6319894B1 (en) * 1997-01-08 2001-11-20 The Picower Institute For Medical Research Complexes and combinations of fetuin with therapeutic agents
AU6023298A (en) 1997-01-17 1998-08-07 Bristol-Myers Squibb Company A method of inhibiting or treating phytosterolemia with an mtp inhibitor
CA2276467A1 (en) 1997-01-17 1998-07-23 Bristol-Myers Squibb Company Method for treating atherosclerosis with an mpt inhibitor and cholesterol lowering drugs
US6066653A (en) 1997-01-17 2000-05-23 Bristol-Myers Squibb Co. Method of treating acid lipase deficiency diseases with an MTP inhibitor and cholesterol lowering drugs
CA2240325A1 (en) 1998-03-27 1998-11-14 Synsorb Biotech, Inc. Methods for screening compound libraries
US6437119B1 (en) * 1998-05-07 2002-08-20 William Lawrence Truett Compounds formed from two or three antibiotics and their processes of preparation
US20020131972A1 (en) * 1998-05-21 2002-09-19 Daniel Sem Multi-partite ligands and methods of identifying and using same

Also Published As

Publication number Publication date
WO1999063929A3 (en) 2000-02-03
CA2319080C (en) 2011-05-31
WO1999063940A3 (en) 2001-06-07
SG90053A1 (en) 2002-07-23
SG80631A1 (en) 2001-05-22
ES2263274T3 (en) 2006-12-01
US20070134729A1 (en) 2007-06-14
HK1028737A1 (en) 2001-03-02
WO1999064033A9 (en) 2000-07-27
CA2321166A1 (en) 1999-12-16
WO1999064033A1 (en) 1999-12-16
WO1999064054A1 (en) 1999-12-16
EP1085891A1 (en) 2001-03-28
AU4551899A (en) 1999-12-30
CA2319120A1 (en) 1999-12-16
US7179794B2 (en) 2007-02-20
EP1005356A1 (en) 2000-06-07
WO1999063940A2 (en) 1999-12-16
WO1999063937A9 (en) 2001-06-28
AU764401B2 (en) 2003-08-21
CA2316999A1 (en) 1999-12-16
AU4677199A (en) 1999-12-30
US6566509B1 (en) 2003-05-20
WO1999063988A1 (en) 1999-12-16
AR019634A1 (en) 2002-02-27
US20030176670A1 (en) 2003-09-18
TWI239959B (en) 2005-09-21
WO1999063929A2 (en) 1999-12-16
EP0989857A4 (en) 2001-01-17
EP1124528A1 (en) 2001-08-22
DK1005356T3 (en) 2006-08-14
WO1999064049A1 (en) 1999-12-16
ATE322910T1 (en) 2006-04-15
US20020028943A1 (en) 2002-03-07
AU4818099A (en) 1999-12-30
SG106036A1 (en) 2004-09-30
CA2319477A1 (en) 1999-12-16
US20040023290A1 (en) 2004-02-05
WO1999064047A8 (en) 2001-03-08
CA2321274A1 (en) 1999-12-16
CA2321162A1 (en) 1999-12-16
WO1999064049A9 (en) 2001-01-18
CA2320241A1 (en) 1999-12-16
CA2319080A1 (en) 1999-12-16
JP2004512255A (en) 2004-04-22
JP2002517465A (en) 2002-06-18
AU4677399A (en) 1999-12-30
EP0989857A1 (en) 2000-04-05
WO1999063937A3 (en) 2000-03-02
WO1999064047A1 (en) 1999-12-16
WO1999064048A9 (en) 2000-07-20
DE69930818T2 (en) 2006-12-07
AR019635A1 (en) 2002-02-27
CY1105087T1 (en) 2009-11-04
EP1143991A2 (en) 2001-10-17
US6288234B1 (en) 2001-09-11
JP2002517422A (en) 2002-06-18
EP1005356A4 (en) 2001-08-22
AU4430599A (en) 1999-12-30
NZ505979A (en) 2003-09-26
WO1999064047A9 (en) 2000-03-16
WO1999064054A9 (en) 2000-03-02
WO1999063937A2 (en) 1999-12-16
CA2321190A1 (en) 1999-12-16
WO1999064051A1 (en) 1999-12-16
EP1085861A1 (en) 2001-03-28
WO1999064048A1 (en) 1999-12-16
PT1005356E (en) 2006-07-31
EP1003541A1 (en) 2000-05-31
AU4551699A (en) 1999-12-30
EP1083920A1 (en) 2001-03-21
EP1083919A1 (en) 2001-03-21
EP1085893A1 (en) 2001-03-28
AU4543899A (en) 1999-12-30
AU4426699A (en) 1999-12-30
DE69930818D1 (en) 2006-05-24
AU4426399A (en) 1999-12-30
WO1999063988A9 (en) 2000-05-04
AR018631A1 (en) 2001-11-28
AU4674599A (en) 1999-12-30
EP1085846A2 (en) 2001-03-28
CA2319474A1 (en) 1999-12-16
WO1999064037A1 (en) 1999-12-16
EP1005356B1 (en) 2006-04-12
WO1999063929A9 (en) 2000-03-16
AU4551799A (en) 1999-12-30

Similar Documents

Publication Publication Date Title
US6288234B1 (en) Multibinding inhibitors of microsomal triglyceride transferase protein
US6288055B1 (en) Analgesic agents
US6395724B1 (en) Multibinding inhibitors of cyclooxygenase-2
US20040110229A1 (en) Muscarinic receptor antagonists
WO1999064043A1 (en) Muscarinic receptor antagonists
CA2319761A1 (en) Multibinding inhibitors of hmg-coa reductase
US6420560B1 (en) H1—histamine receptor antagonists
WO1999064039A1 (en) Bradykinin antagonists
CA2321191A1 (en) Multivalent agonists, partial agonists, inverse agonists and antagonists of the 5-ht3 receptors

Legal Events

Date Code Title Description
FZDE Discontinued