US20070134729A1 - Novel antibacterial agents - Google Patents

Novel antibacterial agents Download PDF

Info

Publication number
US20070134729A1
US20070134729A1 US11/603,287 US60328706A US2007134729A1 US 20070134729 A1 US20070134729 A1 US 20070134729A1 US 60328706 A US60328706 A US 60328706A US 2007134729 A1 US2007134729 A1 US 2007134729A1
Authority
US
United States
Prior art keywords
linker
substituted
ligands
ligand
compound
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
US11/603,287
Inventor
Burton Christensen
Edmund Moran
John Griffin
J. Judice
YongQi Mu
John Pace
Mathai Mammen
James Aggen
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.)
Theravance Biopharma Antibiotics IP LLC
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
Priority to US11/603,287 priority Critical patent/US20070134729A1/en
Publication of US20070134729A1 publication Critical patent/US20070134729A1/en
Assigned to THERAVANCE BIOPHARMA ANTIBIOTICS IP, LLC reassignment THERAVANCE BIOPHARMA ANTIBIOTICS IP, LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: THERAVANCE, INC.
Abandoned legal-status Critical Current

Links

Images

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

Definitions

  • the multibinding compounds of the invention comprise from 2-10 ligands covalently connected by a linker or linkers, wherein each of said ligands in their monovalent (i.e., unlinked) state have the ability to bind to an enzyme involved in cell wall biosynthesis and metabolism, a precursor used in the synthesis of the bacterial cell wall and/or the cell surface and thereby interfere with the synthesis and or metabolism of the cell wall.
  • the ligands are selected from the beta lactam and/or glycopeptide class of antibacterial agents.
  • the invention also relates to pharmaceutical compositions comprising a pharmaceutically acceptable excipient and a therapeutically effective amount of one or more compound(s) of the invention, methods of using such compounds and methods of preparing such compounds.
  • the cell wall maintains the shape of the microorganism which has a high internal osmotic pressure. Damage to the cell wall (e.g. by lysozyme) or inhibition of the cell wall's formation leads to lysis of the cell.
  • the cell wall contains a chemically distinct complex polymer “mucopeptide” (“murein”, “peptidoglycan”) consisting of polysaccharides and a highly cross-linked polypeptide.
  • the polysaccharides comprise an alternating copolymer of the amino sugars N-acetylglucosamine and N-acetylmuramic acid, the latter being found only in bacteria.
  • pentapeptides To the N-acetylmuramic residues are attached pentapeptides.
  • the polysaccharide backbone of the cell wall is formed by oligomerization of disaccharide pentapeptide precursors (lipid intermediate II) and is catalyzed an enzyme known as transglycosylase.
  • the final rigidity of the cell wall is imparted by cross-linking of the peptide chains as a result of transpeptidation reactions by several bacterial enzymes one of which is known as peptidoglycan transpeptidase.
  • antibacterial agents exert their antibacterial activity is by inhibiting the transglycosylase enzyme, thus interfering with the penultimate step in the synthesis of the bacterial cell wall.
  • a glycopeptide for example vancomycin, binds with high affinity and specificity to N-terminal sequences (L-lysyl-D-alanyl-D-alanine in vancomycin sensitive organisms) of peptidoglycan precursors known as lipid intermediate II. By binding to and sequestering these precursors, vancomycin prevents their utilization by the cell wall biosynthesis machinery.
  • vancomycin inhibits the bacterial transglycosylase that is responsible for adding lipid intermediate II subunits to growing peptidoglycan chains. This step preceeds the cross-linking transpeptidation step which is inhibited by beta lactam antibiotics. It is believed that the ⁇ -lactam antibiotics bind to certain cell receptors (the penicillin binding proteins, “PBPs”) which catalyze the transpeptidation reaction and other cell wall metabolic processes. The incomplete cell wall likely serves as a substrate for autolytic enzymes in the cell wall and results in lysis if the environment is isotonic.
  • PBPs penicillin binding proteins
  • Antibacterial agents have proved to be important weapons in the fight against pathogenic bacteria.
  • an increasing problem with respect to the effectiveness of antibacterial agents relates to the emergence of strains of bacteria that are highly resistant to such agents. It would therefore be highly desirable to find antibacterial agents that are active against a broad spectrum of bacteria, in particular resistant strains. It would also be advantageous to discover antibacterial agents that demonstrate high activity and selectivity toward their targets, and are of low toxicity.
  • the multibinding compounds of the present invention fulfill this need.
  • the multibinding compounds of the invention comprise from 2-10 ligands covalently connected by a linker or linkers, wherein each of said ligands in their monovalent (i.e., unlinked ) state have the ability to bind to an enzyme involved in cell wall biosynthesis and metabolism, a precursor used in the synthesis of the bacterial cell wall and/or the bacterial cell surface and thereby interfere with the synthesis and/or metabolism of the cell wall.
  • the ligands are selected from the beta lactam and glycopeptide classes of antibacterial agents.
  • the invention also relates to pharmaceutical compositions comprising a pharmaceutically acceptable excipient and a therapeutically effective amount of one or more compound(s) of the invention, methods of using such compounds and methods of preparing such compounds.
  • this invention provides a multibinding compound of Formula (I): (L) p (X) q (I) wherein:
  • p is an integer of from 2 to 10;
  • q is an integer of from 1 to 20;
  • each ligand, L comprises a ligand domain capable of binding to penicillin binding proteins, a transpeptidase enzyme, a substrate of a transpeptidase enzyme, a beta-lactamase enzyme, pencillinase enzyme, cephalosporinase enzyme, a transglycoslase enzyme, or a transglycosylase enzyme substrate; and
  • X is a linker that may be the same or different at each occurrence; and pharmaceutically acceptable salts thereof provided that:
  • q is less thanp in the multibinding compounds of this invention.
  • this invention provides a multibinding compound of Formula (I):
  • p is an integer of from 2 to 10;
  • q is an integer of from 1 to 20;
  • each ligand, L is a beta lactam antibiotic, an optionally substituted glycopeptide antibiotic, or an aglycone derivative of an optionally substituted glycopeptide antibiotic;
  • X is a linker that may be same or different at each occurrence provided that:
  • q is less than p
  • each ligand that is a beta lactam antibiotic is selected from the group consisting of penems, penams, cephems, carbapenems, oxacephems, carbacephems, and monobactam ring systems; and
  • each ligand that is a glycopeptide antibiotic is selected from the group consisting of Actaplanin, Actinodidin, Ardacin, Avoparcin, Azureomycin, A477, A35512, A40926, A41030, A42867, A47934, A80407, A82846, A83850, A84575, A84428, AB-65, Balhimycin, Chloroeremomycin, Chloroorienticin, Chloropolysporin, Decaplanin, N-demethylvancomycin, Eremomycin, Galacardin, Helvecardin.
  • each ligand that is a beta lactam antibiotic is selected from the group consisting of:
  • R is substituted alkyl, aryl aralkyl, or heteroaryl wherein each of said substituent optionally links (a) to a linker via a covalent bond or R is a covalent bond that links (a) to a linker;
  • R 1 and R 2 are, independently of each other, alkyl or at least one of R 1 and R 2 is a covalent bond linking (a) to a linker;
  • one of P and Q is O, S, or —CH 2 — and the other is —CH 2 —;
  • R 3 is substituted alkyl, heteroarylalkyl, aralkyl, heterocyclylalkyl, or —C(R 6 ) ⁇ NOR 7 (where R 6 is aryl, heteroaryl, or substituted alkyl; and R 7 is alkyl or substituted alkyl) wherein each of said substituent optionally links (b) to a linker or R 3 is a covalent bond that links (b) to a linker; and
  • R 4 is hydrogen, alkyl, alkenyl, substituted alkenylene, substituted alkyl, halo, heteroarylalkyl, heterocyclylalkyl, —SR a (where R a is aryl, heteroaryl, heterocyclyl, or cycloalkyl) or —CH 2 SR a (where R a is aryl, heteroaryl, heterocyclyl, or cycloalkyl) wherein each of said substituent optionally links (b) to a linker or R 4 is a covalent bond that links (b) to a linker;
  • R 5 is hydrogen, hydroxy, or alkoxy
  • T is S or CH 2 ;
  • R 8a is alkyl
  • W is O, S, —OCH 2 —, or CH 2 ; and R 8 is -(alkylene)-NHC(R b ) ⁇ NH where R b is a covalent bond linking (c) to a linker; or —W—R 8 is a covalent bond that links (c) to a linker;
  • R 9 and R 9a are alkyl
  • R 10 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, halo, aryl, heteroaryl, heterocyclyl, aralkyl, heteroaralkyl, heterocyclylalkyl or —CH 2 SR a (where R a is aryl, heteroaryl, heterocyclyl, or cycloalkyl) wherein each of said substituent optionally links (d) to a linker or at, least one of R 9 and R 10 is a covalent bond that links (d) to a linker; or
  • R 9 and R 10 together with the carbon atoms to which they are attached form an aryl, heteroaryl, cycloalkyl, substituted cycloalkyl, or heterocyclyl ring of 4 to 7 ring atoms wherein one of the ring atoms optionally links (d) to a linker; or
  • R 11 is —SO 3 H or -(alkylene)-COOH
  • R 12 is alkyl, substituted alkyl, haloalkyl, alkoxy, aryl, aralkyl, heteroaryl, heteroaralkyl, cycloalkyl, substituted cycloalkyl, or heterocyclyl wherein each of said substituent optionally binds (e) to a linker or R 12 is a covalent bond that links (e) to a linker; and
  • R 13 is alkyl, acyl, or —COC(R 14 ) ⁇ N—OR 15 wherein R 14 is aryl, heteroaryl which optionally links (e) to a linker, and R 15 is -(alkylene)-COOR 6 wherein R 16 is hydrogen or optionally links (e) to a linker or R 13 is a covalent bond that links (e) to a linker; and pharmaceutically acceptable salts thereof;
  • each ligand that is a beta lactam antibiotic is selected from the group consisting of:
  • R is: where:
  • R 17 is a covalent bond that links the (a) group to a linker
  • R 18 and R 19 is hydrogen and the other is a covalent bond that links the (a) group to a linker;
  • R 1 and R 2 are methyl
  • R 3 and R 4 are: R 3 R 4 —CH 2 OCOCH 3 —CH 3 —CH 3 —CH 2 OCONHR 19 —CH 2 OCOCH 3 —Cl —CH 2 OCONHR 19 —CH 2 OCOCH 3 , —CH 2 OCH 3 , H (Note: the R 3 group in the left column is paired with the R 4 in the right column) wherein:
  • R 23 is a covalent bond that links (d) to a linker
  • R 24 and R 25 is hydrogen, alkyl substituted alkyl, or aralkyl, and other is a covalent bond that hnks (d) to a linker;
  • R 26 is alkyl; or
  • X is selected from a compound of formula:. —X a —Z—(Y a —Z) m —X a — wherein
  • n 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—, —OC(O)—, —C(O)NR—, —NRC(O)—, C(S), —C(S)O—, —C(S)NR—, —NRC(S)—, or a covalent bond where R is as defined below;
  • Z at each separate occurrence is selected from the group consisting of alkylene, substituted alkylene, cycloalkylene, substituted cylcoalkylene, alkenylene, substituted alkenylene, alkynylene, substituted alynylene, cycloalkenylene, substituted cycloalkenylene, arylene, heteroarylene, heterocyclene, or a covalent bond;
  • each Y a at each separate occurrence is selected from the group consisting of —O—, —C(O)—, —OC(O)—, —C(O)O—, —NR—, —S(O)n—, —C(O)NR′—,—NR′ C(O)—, —NR′C(O)NR′—, —NR′C(S)NR′—, —C( ⁇ NR′)—NR′—, —NR′—C( ⁇ NR′)—, —OC(O)—NR′—, —NR′—C(O)—O—, —N ⁇ C(X a )—NR′—, —NR′—C(X a ) ⁇ N—,—P(O)(OR′)—O—, —O—P(O)(OR′)—, —S(O) n CR′R′′—, —S(O) n —NR′—, —NR′—S(O) n
  • the invention in a second aspect, relates to a method of treatment of mammals having a disease state that is treatable by antibacterial agents, comprising administering a therapeutically effective amount of a compound of Formula (I), or a mixture of compounds of Formula (I), thereto.
  • the invention in a third aspect, relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of one or more compounds of Formula (I) or a pharmaceutically acceptable salt thereof, admixed with at least one pharmaceutically acceptable excipient.
  • this invention is directed to general synthetic methods for generating large libraries of diverse multimeric compounds which multimeric compounds are candidates for possessing multibinding properties.
  • the diverse multimeric compound libraries provided by this invention are synthesized by combining a linker or linkers with a ligand or ligands to provide for a library of multimeric compounds wherein the linker and ligand each have 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 and polarization.
  • the library of ligands is preferably selected to have diverse attachment points on the same ligand, different finctional groups at the same site of otherwise the same ligand, and the like.
  • this invention is directed to libraries of diverse multimeric compounds which multimeric compounds are candidates for possessing multibinding properties. 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 targeting a receptor.
  • this invention is directed to a method for identifying multimeric ligand compounds possessing multibinding properties which method comprises:
  • 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;
  • this invention is directed to a method for identifying multimeric ligand compounds possessing multibinding properties which method comprises:
  • each linker comprises at least two functional groups .having complementary reactivity to at least one of the reactive flnctional groups of the ligand;
  • 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). Sequential addition is preferred when a mixture of different ligands is employed to ensure heterodimeric or multimeric compounds are prepared. Concurrent addition of the ligands occurs when at least a portion of the multimer compounds prepared are homomultimeric compounds.
  • the assay protocols recited in (d) can be conducted on the multimeric ligand compound library produced in (c) above, or preferably, each member of the library is isolated by preparative liquid chromatography mass spectrometry (LCMS).
  • LCMS preparative liquid chromatography mass spectrometry
  • this invention is directed to a library of multimeric ligand compounds which may possess multivalent properties which library is prepared by the method comprising:
  • each linker in said library comprises at least two finctional groups having complementary reactivity to at least one of the reactive functional groups of the ligand;
  • this invention is directed to a library of multimeric ligand compounds which may possess multivalent properties which library is prepared by the method comprising:
  • each linker comprises at least two functional groups having complementary reactivity to at least one of the reactive finctional groups of the ligand;
  • the library of linkers employed in either the methods or the library aspects of this invention is 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.
  • each of the linkers in the linker library may comprise linkers of different chain length and/or having different complementary reactive groups. Such linker lengths can preferably range from about 2 to 100 ⁇ .
  • the ligand or mixture of ligands is selected to have reactive functionality at different sites on said ligands in order to provide for a range of orientations of said ligand on said multimeric ligand compounds.
  • Such reactive functionality includes, by way of example, carboxylic acids, carboxylic acid halides, carboxyl esters, .amines, halides, pseudohalides, isocyanates, vinyl unsaturation, ketones, aldehydes, thiols, alcohols, anhydrides, boronates, and precursors thereof. It is understood, of course, that 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.
  • the multimeric ligand compound is homomeric (i.e., each of the ligands is the same, although it may be attached at different points) or heteromeric (i.e., at least one of the ligands is different from the other ligands).
  • this invention provides for an iterative process for rationally evaluating what molecular constraints impart multibinding properties to a class of multimeric compounds or ligands targeting a receptor.
  • this method aspect is directed to a method for identifying multimeric ligand compounds possessing multibinding properties which method comprises:
  • steps (e) and (f) are repeated at least two times, more preferably at from 2-50 times, even more preferably from 3 to 50 times, and still more preferably at least 5-50 times.
  • FIG. 1 illustrates examples of multibinding compounds comprising 2 ligands attached in different formats to a linker.
  • FIG. 2 illustrates examples of multibinding compounds comprising 3 ligands attached in different formats to a linker.
  • FIG. 3 illustrates examples of multibinding compounds comprising 4 ligands attached in different formats to a linker.
  • FIG. 4 illustrates examples of multibinding compounds comprising >4 ligands attached in different formats to a linker.
  • FIGS. 5, 6A , and 6 B disclose some representative compounds of formula (a) and (b).
  • FIGS. 7-10 disclose examples of multibinding compounds comprising 2ligands attached in different formats.
  • FIGS. 11-23 illustrate synthesis of compounds of Formula (I).
  • This invention is directed to multibinding compounds that are antibacterial agents and pharmaceutical compositions containing such compounds.
  • pharmaceutical compositions containing such compounds.
  • alkyl refers to a monoradical branched or unbranched saturated hydrocarbon chain preferably having from 1 to 40 carbon atoms, more preferably 1 to 1 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.
  • 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,
  • This term is exemplified by groups such as hydroxymethyl, hydroxyethyl, hydroxypropyl, 2—aminoethyl, 3-aminopropyl, 2-methylaminoethyl, 3-dimethylaminopropyl, 2—sulfonamidoethyl, 2-carboxyethyl, and the like.
  • al kylene refers to a diradical of a 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 methylene (—CH 2 —), ethylene (—CH 2 CH 2 —), the propylene isomers (e.g., —CH 2 CH 2 CH 2 — and —CH(CH 3 )CH 2 —) and the like.
  • substituted alkylene refers to:
  • alkaryl or “aralkyl” 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.
  • alkoxy refers to the groups alkyl-O—, alkenyl-O—, cycloaikyl-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-propoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, 1,2-dimethylbutoxy, and the like.
  • substituted alkoxy refers to the groups substituted alkyl-O—, substituted alkenyl-O—, substituted cycloalkyl-O—, substituted cycloalkenyl-O—, and substituted alkynyl-O— where substituted alkyl, substituted alkenyl, substituted cycloalkyl, substituted cycloalkenyl and substituted alkynyl are as defined herein.
  • 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.
  • Preferred alkenyl groups include ethenyl (—CH ⁇ CH 2 ), n-propenyl.(—CH 2 CH ⁇ CH 2 ), iso-propenyl (—C(CH 3 ) ⁇ CH 2 ), and the like.
  • substituted alkenyl refers to an alkenyl 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
  • alkenylene refers to a diradical 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. This term is exemplified by groups such as ethenylene (—CH ⁇ CH—), the propenylene isomers (e.g., —CH 2 CH ⁇ CH—, —C(CH 3 ) ⁇ CH—, and the like.
  • 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, 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
  • substituted alkenylene groups include those where 2 substituents on the alkenylene group are fused to form one or more cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heterocyclic or heteroaryl groups fused to the alkenylene group.
  • 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 preferably from 1-6 sites of acetylene (triple bond) unsaturation.
  • Preferred alkynyl groups include ethynyl (—C—CH), propargyl (—CH 2 C ⁇ CH) and the like.
  • substituted alkynyl refers to an alkynyl 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, alkoxya
  • alkynylene refers to a diradical of an unsaturated hydrocarbon 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 acetylene (triple bond) unsaturation.
  • Preferred alkynylene groups include ethynylene (—C ⁇ C—), propargylene (—CH 2 C ⁇ C—) and the like.
  • 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, 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, alkoxya
  • acyl refers to the groups HC(O)—, alkyl-C(O)—, substituted alkyl-C(O)—, alkenyl-C(O)—, substituted alkenyl-C(O)—, cycloalkyl-C(O)—, substituted cycloalkyl-C(O)—, cycloalkenyl-C(O)—, substituted cycloalkenyl-C(O)—, aryl-C(O)—, heteroaryl-C(O)— and heterocyclic-C(O)— where alkyl, substituted alkyl,.alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic are as defined herein.
  • 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.
  • sulfonylamino refers to the group —NRSO 2 R a where R is hydrogen, alkyl, substituted alkyl, aralkyl, or heteroaralkyl, and R a is alkyl, substituted alkyl, amino, or substituted amino wherein alkyl, substituted alkyl, aralkyl, heteroaralkyl and substituted amino are as defined herein.
  • aminoacyl refers to the group —NRC(O)R where each R is independently hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, amino, substituted amino, aryl, heteroaryl, or heterocyclic wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, aryl, heteroaryl and heterocyclic are as defined herein.
  • 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.
  • 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 defined herein.
  • 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).
  • the aryl group may optionally be fuised to a heterocyclic or cycloalkyl group.
  • such aryl groups can optionally be substituted with from 1 to 5 suhstituents, preferably 1 to 3 substituents, selected from the group consisting of acyloxy, 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, sulfonylamino, alkaryl, aryl, aryloxy, azido, carboxyl, carb6xylalkyl, cyano, halo, nitro, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, aminoacyloxy, oxyacylanino, thioalk
  • aryloxy refers to the group aryl-O— wherein the aryl group is as defined above including optionally substituted aryl groups as also defined above.
  • amino refers to the group —NH 2 .
  • substituted amino refers to the group —NRR where each R is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, acyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, cycloalkenyl, substituted cycloalkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl and heterocyclic provided that both R's are not hydrogen.
  • carboxyalkyl or “alkoxycarbonyl” refers to the groups “—C(O)O-alkyl”, “—C(O)O-substituted alkyl”, “—C(O)O-cycloalkyl”, “—C(O)O-substituted cycloalkyl”, “—C(O)O-alkenyl”, “-C(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.
  • cycloalkyl refers to cyclic alkyl groups of from 3 to 20 carbon atoms having a single cyclic ring or multiple condensed rings, said cycloalkyl group may optionally be fused to an aryl or heteroaryl group.
  • cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl, and the like, or multiple ring structures such as adamantanyl, and the like.
  • substituted cycloalkyl refers to cycloalkyl groups having from 1 to 5 substituents, and preferably 1 to 3 substituents, selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, 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, substituted thio
  • 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 unsaturation.
  • suitable cycloalkenyl groups include, for instance, cyclobut-2-enyl, cyclopent-3-enyl, cyclooct-3-enyl and the like.
  • substituted cycloalkenyl refers to cycloalkenyl groups having from 1 to 5 substituents, and preferably 1 to 3 substituents, selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, 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, heteroary
  • halo or “halogen” refers to fluoro, chloro, bromo and iodo.
  • 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).
  • the heteroaryl ring may optionally be fused to a cycloalkyl or heterocyclyl ring.
  • heteroaryl groups can be optionally substituted with 1 to 5 substituents, preferably 1 to 3 substituents, selected from the group consisting of acyloxy, 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, nitro, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, aminoacyloxy, oxyacylamino, thioalkoxy, substituted thioalkoxy, thioaryloxy
  • Preferred heteroaryl substituents include alkyl, alkoxy, halo, cyano, nitro, trihalomethyl, and thioalkoxy.
  • Such heteroaryl groups can have a single ring (e.g., pyridyl or furyl) or multiple condensed rings (e.g., indolizinyl or benzothienyl).
  • Preferred heteroaryls include pyridyl, pyrrolyl and furyl.
  • heteroaryloxy refers to the group heteroaryl-O—.
  • heterocycle refers to a monoradical saturated or 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 and further wherein one, two, or three of the ring carbon atoms may optionally be replaced with a carbonyl group (i.e., a keto group).
  • the heterocycle group may optionally fused to an aryl or heteroaryl ring.
  • heterocyclic groups can be optionally substituted with 1 to 5, and preferably 1 to 3 substituents, selected from the group consisting of alkyl, acyloxy, alkyl, substituted alkyl, 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, hetero
  • heteroaryls and heterocycles include, but are not limited to, pyrrole, thiophene, furan imidazole, pyrazole, pyridine, pyrazine, pyrimidine, 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, pyrrolidine, piperidine, piperazine, indoline, morpholine, tetrahydrofuiranyl, tetrahydrothioph
  • Heterocycloamino means a saturated, unsaturated, bridged or unbridged, monovalent cyclic group of 4 to 8 ring atoms, wherein at least one ring atom is N and optionally contains one or two additional ring heteroatoms selected from the group consisting of N, O , or S(O)n (where n is an integer from 0 to 2), the remaining ring atoms being C, where one or two C atoms may optionally be replaced by a carbonyl group.
  • the heterocycloamino ring may be fused to a cycloalkyl, aryl or heteroaryl ring, and it may be optionally substituted with one or more substituents, preferably one or two substituents, selected from alkyl, substituted alkyl, cycloaLkyl, aryl, araiiyl, heteroaryl, heteroaralkyl, halo, cyano, acyl, amino, substituted amino, acylamino, —OR (where R is hydrogen, alkyl, alkenyl, cycloalkyl, acyl, aryl, heteroaryl, aralkyl, or heteroaralkyl), or —S(O)nR [where n is an integer from 0 to 2 and R is hydrogen (provided that n is 0), alkyl, alkenyl, cycloalkyl, amino, heterocyclo, aryl, heteroaryl, aralkyl, or heteroaralkyl].
  • heterocycloamino includes, but is not limited to, pyrrolidino, piperidino, morpholino, piperazino, indolino, quinuclidine, or thiomorpholino, and the derivatives thereof.
  • heterocyclooxy refers to the group heterocyclic-O—.
  • thioheterocyclooxy refers to the group heterocyclic-S—.
  • 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 and heterocyclic are as defined herein.
  • spiro-attached cycloalkyl group refers to a cycloalkyl group joined to another ring via one carbon atom common to both rings.
  • thiol refers to the group —SH.
  • thioalkoxy or “alkylthio” refers to the group —S-alkyl.
  • substituted thioalkoxy refers to the group —S-substituted alkyl.
  • thioaryloxy refers to the group aryl-S— wherein the aryl group is as defined above including optionally substituted aryl-groups also defined above.
  • heteroaryloxy refers to the group heteroaryl-S— wherein the heteroaryl group is as defined above including optionally substituted aryl groups as also defined above.
  • 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.
  • the compounds-of this invention include all stereochemical isomers arising from the substitution of these compounds.
  • 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.
  • the multibinding compounds of this invention are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto.
  • Pharnaceutically-acceptable base addition salts can be prepared from inorganic and organic bases.
  • Salts derived from inorganic bases include by way 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 arnines, trialkenyl amines, substituted alkenyl amines, di(substituted alkenyl) amines, tri(substituted alkenyl) amines, cycloalkyl amines, di(cycloalkyl) amines, tri(cycloalkyl) amines, substituted
  • amiines where the two or three substituents, together with the amino nitrogen, form a heterocyclic or heteroaryl group.
  • 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, piperazine, piperidine, morpholine, N-ethylpiperidine, and the like.
  • 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
  • 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, mtalic acid, malonic acid, succinic acid, maleic 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.
  • pharmaceutically-acceptable cation refers to the cation of a pharmaceutically-acceptable salt.
  • protecting 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 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 (See., T. W. Greene and P. G. H. Wuts, “ Protective Groups in Organic Synthesis”, 2 nd Ed.).
  • removable blocking group employed is not critical and preferred removable hydroxyl blocking groups include conventional substituents such as allyl, benzyl, acetyl, chloroacetyl, thiobenzyl, 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.
  • 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), fluorenylmethoxy-carbonyl (FMOC), allyloxycarbonyl (ALOC), and the like which can be removed by conventional conditions compatible with the nature of the product.
  • t-BOC t-butyoxycarbonyl
  • CBZ benzyloxycarbonyl
  • FMOC fluorenylmethoxy-carbonyl
  • ALOC allyloxycarbonyl
  • Preferred carboxyl protecting groups include esters such as methyl, ethyl, propyl, t-butyl etc. which can be removed by mild conditions compatible with the nature of the product.
  • ligand or “ligands” as used herein denotes a compound that is a binding partner for penicillin binding proteins, a pencillinase enzyme, a cephalosporinase enzyme, a transpeptidase enzyme, a substrate of a transpeptidase enzyme, a beta-lactamase enzyme, a transglycoslase enzyme, or a transglycosylase enzyme substrate and is bound thereto by complementarity.
  • a ligand may be either capable of binding to its target 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 ligand to various ligand binding sites). Examples. of ligands useful in this invention are described herein.
  • portions of the ligand structure that are not essential for specific molecular recognition and binding activity may be varied 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.
  • ligand is not intended to be limited to compounds known to be usefuil in binding to penicillin binding proteins, a pencillinase enzyme, a cephalosporinase enzyme, a transpeptidase enzyme, a substrate of a transpeptidase enzyme, a beta-lactamase enzyme, a transglycoslase enzyme, or a transglycosylase enzyme substrate (e.g., known drugs).
  • ligand can equally apply to a molecule that is not normally associated with penicillin binding proteins, a transglycoslase enzyme, or a taansglycosylase enzyme substrate binding properties.
  • ligands that exhibit marginal activity or lack useful activity as monomers can be highly active as multivalent compounds because of the benefits conferred by multivalency.
  • the term “ligand” or “ligands” as used herein is intended to include the racemic forms of the ligands as well as individual enantiomers and diasteromers and non-racemic mixtures thereof.
  • ⁇ -lactam antibiotic refers to antibiotics, having a ⁇ -lactam ring core which can be depicted as follows:
  • the ⁇ -lactam antibiotics are classified into the penicillins, cephalosporins, carbapenems, oxacephems, carbacephems, and monobactams and include drugs such as Penicillin G, Penicillin V, Methicillin, Oxacillin, Cloxacillin, Dicloxacillin, Nafcillin, Ampicillin, Amoxicillin, Carbenicillin, Carbenicillin indanyl, Ticarcillin, Mezlocillin, Piperacillin Cephalothin, Cefazolin, Cephalexin, Cefadroxil, Cefamandole, Cefoxitin, Cefaclor, Cefuroxime, Cefuroxime axetil, Loracarbef, Cefonicid, Cefotetan, Ceforanide, Cefotaxime, Cefpodoxime proxetil, Ceftizoxime, Ceftriaxone, Cefoperazone, Ceftazidime, Cef
  • Preferred ⁇ -lactam antibiotics are Amoxicillin, Nafcillin, Cefadroxil, Ceftriaxone, Cefaclor, Aztreonam, Ceftazidime, Imipenem, Meropenem, Ritipenem, Ceftazidine, Pipericillin, Clauvlinic acid, Cefepime, Cefoxitin, Cefotaximne, Cefixime, Lefluzidine and derivatives thereof
  • glycopeptide antibiotics are characterized by a multi-ring peptide core and at least one sugar attached at various sites, of which vancomycin is an important example.
  • Examples of the glycopeptide class of ligands included in this definition may be found in “Glycopeptides Classification, Occurrence, and Discovery” by Rao, R. C. and Crandall, L. W., (Drugs and the Pharmaceutical Sciences” Vol. 63, edited by Ramakrishnan Nagarajan, published by Marcal Dekker, Inc.) which is hereby incorporated by reference.
  • Another preferred class of ligands is the general class of glycopeptides disclosed above on which the sugar moiety is absent.
  • vancomycin aglycone For example removal of the disaccharide moiety appended to the phenol on vancomycin (as shown below as Formula II) by mild hydrolysis gives vancomycin aglycone.
  • a further preferred class are glycopeptides that have been further appended with additional saccharide residues,.especially aminoglycosides, in a manner similar to vancosamine.
  • “Vancomycin” refers to the antibacterial compound whose structure is reproduced below as Formula Ia.
  • “Optional” or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not.
  • “Optionally substituted glycopeptide” with respect to a compound of Formula (I) refers to a ligand as defined above in which those positions that are not linked to X may or may not be substituted by various groups as defined below.
  • the term also includes those instances in which one amino acid of the basic core structure is replaced by another amino acid, for example as described in “Preparation and conformational analysis of vancomycin hexapeptide and aglucovancomycin hexapeptide”, by Booth, P. M., Williams, D. H., Univ. Chem.
  • Optionally substituted vancomycin with respect to the multibinding agents of the invention refers to vancomycin in which the hydroxy group at any position, the [R] position the carboxyl groups at the [C] position, or the amine groups at the [V].or [N] position that are not attached to the linker X may or may not be substituted by various groups.
  • Such groups include: R a where R a at each occurrence is chosen from alkyl, alkyl optionally interrupted by 1-5 atoms-chosen from O, S, or —NR b — where R b is alkyl, aryl, or heteroaryl, all of which are optionally.substituted, haloalkyl, alkenyl, alkynyl, alkylamino, alkylaminoalkyl, cycloalkyl, alkanoyl, aryl, heteroaryl, heterocyclic, additional saccharide residues, especially ammoglycosides, all of which are optionally substituted as defined above; and: NRWd in which R c and R d are independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkanoyl, aryl, heteroaryl, arylalkyl, or heteroarylalkyl, or R c and R d when taken together with the nitrogen atom to
  • An example of a preferred [C] substitution is dimethylaminopropylamino and glucosamino; and example of a preferred [V] substitution is alkyl, for example n-decyl, or alkylaminoalkyl, for example n-decylaminoethyl.
  • Optionally substituted vancomycin aglycone with respect to the multibinding agents of the invention refers to vancomycin aglycone in which the hydroxy group at any position, particularly the hydroxy group at the [O] position, the [R] position, the carboxy groups at the [C] position, or the amine group at the [N] position, that are not attached to the linker X may or may not be substituted by various groups —R a as defined above.
  • Transglycosylase enzyme substrate denotes the molecular target of the transglycosylase enzyme.
  • the substrate binds to the enzyme and eventually results in the synthesis of the bacterial cell wall.
  • the action of this enzyme is inhibited by a ligand domain that binds to the enzyme itself and/or the enzyme substrate.
  • a ligand such as vancomycin binds to this substrate and in effect “sequesters” the substrate to prevent its recognition by the enzyme and subsequent use in the construction of the bacterial cell wall.
  • some glycopeptides or derivatives thereof may directly bind to and inhibit the transglycolase.
  • 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.
  • each ligand and linker in the multibinding compound is independently selected such that the multibinding compound includes both symmetric compounds (i.e., where each ligand as well as each linker is identical) and asymmetric compounds ((i.e., where at least one of the ligands is different from the other ligand(s) and/or at least one linker is different from the other linker(s)).
  • Multibinding compounds provide a biological and/or therapeutic effect greater than the aggregate of unlinked ligands equivalent thereto which are made available for binding.
  • 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, increased ability to kill cells such as fungal pathogens, cancer cells, etc., 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.
  • univalency 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.
  • multivalency refers to the concurrent binding of from 2 to 10 linked ligands (which may be the same or different) and two or more corresponding ligand binding sites which May be the same or different.
  • 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 ligand binding site. In some cases, the potency may be non-linearly correlated with its affinity.
  • 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 multibinding agent produces an equivalent biological or therapeutic effect at a lower concentration than the aggregate unlinked ligand is indicative of enhanced potency.
  • selectivity 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 K d (i.e., the dissociation constants for each ligand-receptor complex) or, in cases where a biological effect is observed below the Y , the ratio of the respective EC 50 's (i.e., the concentrations that produce 50% of the maximum response for the ligand interacting with the two distinct ligand binding sites (receptors)).
  • ligand binding site denotes the site on a penicillin binding proteins, a transpeptidase enzyme, penicillinase enzyme, cephalosporinase enzyme, beta lactamase enzyme, a transpeptidase enzyme substrate, a transglycosylase enzyme and/or transglycosylase enzyme substrate 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 producing a unique biological effect, for example, agonism, antagonism, and modulatory effects, or it may maintain an ongoing biological event, and the like.
  • ligand binding sites of the enzyme or the receptor that participate in biological multivalent binding interactions are constrained to varying degrees by their intra- and inter-molecular associations.
  • ligand binding sites 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.
  • 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, isopropanol, t-butanol, dioxane, pyridine, and the like.
  • treatment refers to any treatment of a pathologic condition in a mammal, particularly a human, and includes:
  • 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 that is an antibacterial agent, and those disease states which have been found to be usefully treated by a specific multibinding compound of our invention.
  • 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 condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • linker refers to a group or groups that covalently attaches from 2 to 10 ligands (as identified above) in a manner that provides for a compound capable of multivalency.
  • the linker is a ligand-orienting entity that permits attachment of at least two copies of a ligand (which may be the same or different) thereto. In some cases, the linker may itself be biologically active.
  • the term “linker” does not, however, extend to 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.
  • 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 linker or linkers.
  • 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 of ligands to the ligand binding sites of an enzyme involved in cell wall biosynthesis and metabolism, a precursor used in the synthesis of the bacterial cell wall and/or the cell surface, whether such sites are located interiorly, both interiorly and on the periphery of the enzyme structure, or at any intermediate position thereof.
  • glycopeptides are depicted in a simplified form as a shaded box that shows only the carboxy terminus, labeled [C], the sugar amine terminus (e.g., vancosamine), labeled [V], and the “non-sugar” amino terminus, labeled [N] as follows: where R is hydrogen (as N-desmethylvancomycin) or methyl (as in vancomycin).
  • one class of multivalent compounds that fall within the scope of the definition of Formula (I) include compounds wherein the glycopeptide ligand is connected by one or more linkers at the [C], [V], or [N] terminus.
  • Another class of multivalent compounds that fall within the scope of the definition of Formula (I) include compounds where the aglycone derivatives of glycopeptides. depicted as a triangle that shows only the carboxyl terminus, labeled [C], the aglycone hydroxy terminus labeled [9], and the “non-sugar” amino terminus, labeled [N] as follows: where R is hydrogen (as in N-desmethylvancomycin aglycone) or methyl (as in vancomycin aglycone) wherein the aglycone derivatives ligand is connected by one or more linkers at the [C], [V], or [N] terminus.
  • a third class of compounds falling within the scope of the invention are those in which the glycopeptides, or aglycone derivatives thereof, are linked via the [R] position.
  • Reaction schemes that exemplify this linking strategy depict the ligands in a simplified form as above, i.e., as a shaded box in which the carboxyl terminus is labeled [C], the vancosamine amino terminus is labeled [V], and the “non-sugar” amino terminus is labeled [N], with the addition of the [R] position as a resorcinol derivative as shown below: where R is hydrogen or methyl.
  • L a is a beta lactam antibiotic is selected from the group consisting of:
  • R is substituted alkyl, aryl, aralkyl, or heteroaryl wherein each of said substituent optionally links (a) to a linker via a covalent bond or R is a covalent bond that links (a) to a linker;
  • R 1 and R 2 are, independently of each other, alkyl or at least one of R 1 and R 2 is a covalent bond linking (a) to a linker;
  • one of P and Q is O, S, or —CH 2 — and the other is —CH 2 —;
  • R 3 is substituted alkyl, heteroarylalkyl, aralkyl, heterocyclylalkyl, or —C 6 ) ⁇ NOR 7 (where R 6 is aryl, heteroaryl, or substituted alkyl; and R 7 is alkyl or substituted alkyl) wherein each of said substituent optionally links (b) to a linker or R 3 is a covalent bond that links (b) to a linker; and
  • R 4 is hydrogen, alkyl, alkenyl, substituted alkenylene, substituted alkyl, halo, heteroarylalkyl, heterocyclylalkyl, —SR a (where R a is aryl, heteroaryl, heterocyclyl, or cycloalkyl) or —CH 2 SR a (where R a is aryl, heteroaryl, heterocyclyl, or cycloalkyl) wherein each of said substituent optionally links (b) to a linker or R 4 is a covalent bond that links (b) to a linker;
  • R 5 is hydrogen, hydroxy, or alkoxy
  • T is S or CH 2 ;
  • R 8a is alkyl
  • R 9 and R 9a are alkyl
  • R 10 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, halo, aryl, heteroaryl, heterocyclyl, aralkyl, heteroaralkyl, heterocyclylalkyl or —CH 2 SR a (where R 1 is aryl, heteroaryl, heterocyclyl, or cycloalkyl) wherein each of said substituent optionally links (d) to a linker or at least one of R 9 and R 10 is a covalent bond that links (d) to a linker; or
  • R 9 and R 10 together with the carbon atoms to which they are attached form an aryl, heteroaryl, cycloalkyl, substituted cycloalkyl, or heterocyclyl ring of 4 to 7 ring atoms wherein one of the ring atoms optionally links (d) to a linker; or
  • R 11 is —SO 3 H or -(alkylene)-COOH
  • R 12 is alkyl, substituted alkyl, haloalkyl, alkoxy, aryl, aralkyl, heteroaryl, heteroaralkyl, cycloalkyl, substituted cycloalkyl, or heterocyclyl wherein each of said substituent optionally binds (e) to a linker or R 12 is a covalent bond that links (e) to a linker; and
  • R 13 is alkyl, acyl, or —COC(R 14 ) ⁇ N—OR 15 wherein R 14 is aryl, heteroaryl which optionally links (e) to a linker, and R 15 is -(alkylene)-COOR 16 wherein R 16 is hydrogen or optionally links (e) to a linker or R 13 is a covalent bond that links (e) to a linker, preferably
  • L a is selected from the group consisting of.
  • R is: where:
  • R 17 is a covalent bond that links the (a) group to a linker
  • R 18 and R 19 is hydrogen and the other is a covalent bond that links the (a) group to a linker;
  • R 1 and k 2 are methyl
  • R 3 and R 4 are: R 3 R 4 —CH 2 OCOCH 3 —CH 3 —CH 3 —CH 2 OCONHR 19 —CH 2 OCOCH 3 —Cl —CH 2 OCONHR 19 —CH 2 OCOCH 3 , —CH 2 OCH 3 , H (Note: the R 3 group in the left column is paired with the R 4 in the right column) wherein:
  • n 0 or 1; mis 1-5; Z is CH or N; Y is H or halo; R is alkyl; R 17 is a covalent bond that links the (b) group to a linker; one of R 18 and R 19 is hydrogen or alkyl; R 30 and R 31 are, independently of each other, hydrogen or alkyl; or together with the nitrogen atom to which they are attached form a heterocycloamino group; and R, R 32 and R 33 are independently alkyl wherein one of R 18 , R 19 , R 30 —R 33 is a covalent bond that links the (b) group to a linker;
  • R 23 is a covalent bond that links (d) to a linker
  • L b is an optionally substituted vancomycin which is linked to a linker via any hydroxyl group, carboxyl group or amino group;
  • X is a linker and is selected from a compound of formula: —X a —Z—(Y a —Z) m —X a — wherein
  • n 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—, —OC(O)—, —C(O)NR—, —NRC(O)—, C(S), —C(S)O—, —C(S)NR—, —NRC(S)—, or a covalent bond where R is as defined below;
  • Z 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;
  • each ya at each separate occurrence is selected from the group consisting of —O—, —C(O)—, —OC(O)—, —C(O)O—, —NR—, —S(O)n—, —C(O)NR′—, —NR′C(O)—, —NR′C(O)NR′—, —NR′C(S)NR′—, —C( ⁇ NR′)—NR′—, —NR′—C( ⁇ NR′)—, —OC(O)—NR′—, —NR′—C(O)—O—, —N ⁇ C(X a )—NR′—,—NR′—C(X a Y—N—,—P(O)(OR′)—O—, —O—P(O)(OR′)—, —S(O) n CR′R′′—, —S(O) n —NR′—, —NR′—S(O) n
  • ligands, L c and L d are a beta lactam antibiotic and are independently selected from the group consisting of:
  • R is substituted alkyl, aryl, aralkyl, or heteroaryl wherein each of said substituent optionally. links (a) to a linker via a covalent bond or R is a covalent bond that links (a) to a linker;
  • R 1 and R 2 are, independently of each other, alkyl or at least one of R 1 and R 2 is a covalent bond linking (a) to a linker;
  • one of P and Q is O, S, or —CH 2 — and the other is —CH 2 —;
  • R 3 is substituted alkyl, heteroarylalkyl, aralkyl, heterocyclylalkyl, or —C 6 ) ⁇ NOR 7 (where R 6 is aryl, heteroaryl, or substituted alkyl; and R 7 is alkyl or substituted alkyl) wherein each of said substituient optionally links (b) to a linker or R 3 is a covalent bond that links (b) to a linker; and
  • R 4 is hydrogen, alkyl, alkenyl, substituted alkenylene, substituted alkyl, halo, heteroarylalkyl, heterocyclylalkyl, —SR a (where R a is aryl, heteroaryl, heterocyclyl, or cycloalkyl) or —CH 2 SR a (where R a is aryl, heteroaryl, heterocyclyl, or cycloalkyl) wherein each of said substituent optionally links (b) to a linker or R 4 is a covalent bond that links (b) to a linker;
  • R 5 is hydrogen, hydroxy, or alkoxy
  • T is S or CH 2 ;
  • R 8 is alkyl
  • W is O, S, —OCH 2 —, or CH 2 ; and R 8 is -(alkylene)-NHC(R b ) ⁇ NH where R b is a covalent bond linking (c) to a linker; or —W—R 8 is a covalent bond that links (c) to a linker;
  • R 9 and R 9a are alkyl
  • R 10 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, halo, aryl, heteroaryl, heterocyclyl, aralkyl, heteroaralkyl, heterocyclylalkyl or —CH 2 SR a (where R a is aryl, heteroaryl, heterocyclyl, or cycloalkyl) wherein each of said substituent optionally links (d) to a linker or at least one of R 9 and R 10 is a covalent bond that links (d) to a linker; or
  • R 9 and R 10 together with the carbon atoms to which they are attached form an aryl, heteroaryl, cycloalkyl, substituted cycloalkyl, or heterocyclyl ring of 4 to 7 ring atoms wherein one of the ring atoms optionally links (d) to a linker; or
  • R 11 is —SO 3 H or -(alkylene)-COOH
  • R 12 is alkyl, substituted alkyl, haloalkyl, alkoxy, aryl, aralkyl, heteroaryl, heteroaralkyl, cycloalkyl, substituted cycloalkyl, or heterocyclyl wherein each of said substituent optionally binds (e) to a linker or R 12 is a covalent bond that links (e) to a linker; and
  • R 13 is alkyl, acyl, or —COC(R 14 ) ⁇ N—OR 15 wherein R 14 is aryl, heteroaryl which optionally links (e) to a linker, and R 15 is -(alkylene)-COOR 16 wherein R 16 is hydrogen or optionally links (e) to a linker or R 13 is a covalent bond that links (e) to a linker, preferably
  • L c and L d are independently selected from the group consisting of:
  • R is: where:
  • R 17 is a covalent bond that. links the (a) group to a linker, one of R 18 and R 19 is hydrogen and the other is a covalent bond that links the (a) group to a linker;
  • R 3 and R 4 are: R 3 R 4 —CH 2 OCOCH 3 —CH 3 —CH 3 —CH 2 OCONHR 19 —CH 2 OCOCH 3 —Cl —CH 2 OCONHR 19 —CH 2 OCOCH 3 , —CH 2 OCH 3 , H (Note: the R 3 group in the left column is paired with the R 4 in the right column) wherein:
  • R 17 is a covalent bond that links the (b) group to a linker; one of R 18 and R 19 is hydrogen or alkyl; R 30 and R 31 are, independently of each other, hydrogen or alkyl; or together with the nitrogen atom to which they are attached form a heterocycloamino group; and R, R 32 and R 33 are independently alkyl wherein one of R 18 , R 19 , R 30 —R 33 is a covalent bond that links the (b) group to a linker;
  • R 23 is a covalent bond that liniks (d) to a linker
  • X is a linker is selected from a compound of formula: —X a —Z—(Y a —Z) m —X a — wherein
  • n 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—, —OC(O)—, —C(O)NR—, —NRC(O)—, C(S), —C(S)O—, —C(S)NR—, —NRC(S)—, or a covalent bond where R is as defined below;
  • Z at each separate occurrence is selected from the group consisting of alkylene, substituted alkylene, cycloalkylene, substituted cylcoallylene, alkenylene, substituted alkenylene, alkynylene, substituted alkynylene, cycloalkenylene, substituted cycloalkenylene, arylene, heteroarylene, heterocyclene, or a covalent bond;
  • each Y a at each separate occurrence is selected from the group consisting of —O—, —C(O)—, —OC(O)—, —C(O)O—, —NR—, —S(O)n—, —C(O)NR′—, —NR′C(O), —NR′C(O)NR′—, —NR′C(S)NR′—, —C( ⁇ NR′)—NR′—, —NR′—C( ⁇ NR′)—, —OC(O)—NR′—, —NR′—C(O)—O—, —N ⁇ C(X a )—NR′—, —NR′—C(X a ) ⁇ N—, —P(O)(OR′)—O—, —O—P(O)(OR′)—,—S(O) n CR′R′′—,—S(O) n —NR′—, —NR′—S(O) n —
  • L a , L c , and L d are independently selected from the group consisting of:
  • L b is selected from the group consisting of: wherein the atom carrying the bond with the dashed line indicates the point of attachment of the ligand to the linker;
  • linker is selected from the group consisting of: LINKERS DERIVED FROM AMINOALDEHYDES Linkers Derived from Aminoacids
  • the starting materials and reagents used in preparing these compounds are either available from commercial suppliers such as Aldrich Chemical Co., (Milwaukee, Wis., USA), Bachem (Torrance, Calif., USA), Emka-Chemie, or Sigma (St.
  • the starting materials and the intermediates of the reaction may be isolated and purified if desired using conventional techniques, including but not limited to filtration, distillation, crystallization, chromatography, and the like. Such materials may be characterized using conventional means, including physical constants and spectral data.
  • 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.
  • a multibinding compound of Formula (I) can be prepared as illustrated and described in Scheme A below.
  • a multibinding compound of Formula (I) can be prepared by covalently attaching the ligands, L, to a linker, X, as shown in Scheme A below.
  • a multibinding compound of Formula (I) is prepared in one step, by covalently attaching the ligands, L, to a linker, X, where FG 1 and FG 2 represent a functional group such as halo, pseudohalides, boronates, amino, hydroxy, thio, aldehyde, ketone, carboxy, carboxy derivatives such as acid halide, ester, amido, and the like.
  • This method is preferred for preparing compounds of Formula (I) where both the ligands are identical.
  • the compounds of Formula (I) are prepared in a stepwise manner by covalently attaching one equivalent of a ligand, L 1 , with a ligand X where FG 1 and FG 2 represent a functional group as defined above, and FG 2 PG is a protected finctional group to give an intermediate of formula (II).
  • Deprotection of the second functional group on the ligand, followed by reaction with a ligand L 2 , which may be same or different than ligand L 1 then provides a compound of Formula (I).
  • This method is suitable for preparing compounds of Formula (I) where the ligands are the non-identical.
  • the ligands are covalently attached to the linker using conventional chemical techniques providing for covalent linkage of the ligand to the linker. 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 as shown in Table I below.
  • reaction between a carboxylic acid of either the linker or the ⁇ -lactam and a primary or secondary amine of the ⁇ -lactam or the linker in the presence of suitable, well-known activating agents such as dicyclohexylcarbodiimide results in formation of an amide bond covalently linking the ⁇ -lactam to the linker
  • reaction between an amine group of either the linker or the ⁇ -lactam and a sulfonylbhalide of the ⁇ -lactarn or the linker in the presence of a base such as triethylamine, pyridine, an the like results in formation of a sulfonamide bond covalently linking the ⁇ -lactam to the linker
  • any-compound which is an antibacterial agent can be used as a ligand in this invention.
  • a compound selected for use as a ligand will have at least one functional group, such as an amino, 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.
  • Linkers can be attached to different positions on the ligand molecule to achieve different orientations of the ligand domains, and thereby facilitate multivalency. While a number of positions on the ligands are synthetically practical for linking, it is:preferred to preserve those ligand substructures which are most important for ligand-receptor binding.
  • the linker is attached to the ligand at a position that retains ligand 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.
  • 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-activity relationships (SAR) of the ligand and/or congeners and/or structural information about ligand-receptor complexes (e.g., X-ray crystallography, NMR, and the like).
  • SAR structure-activity relationships
  • 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.
  • the univalent linker-ligand conjugate may be tested for retention of activity in the relevant assay.
  • 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 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 the multibinding compound.
  • the linker may be chosen to enhance the biological activity of the molecule.
  • the linker may be chosen from any organic molecule construct that orients two or more ligands to their ligand binding sites to permit multivalency.
  • the linker can be considered as a “framework” on which the ligands are arranged in order to bring about the desired ligand-orienting result, and thus produce a multibinding compound.
  • 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 multiple bonds (alkenyl, alkenylene, alikyyl or alkynylene groups).
  • Other groups can also include oligomers and polymers which are branched- or straight-chain species.
  • rigidity is imparted by the presence of cyclic groups (e.g., aryl, heteroaryl, cycloalkyl, heterocyclic, etc.).
  • the ring is a six or ten member ring.
  • the ring is an aromatic ring such as, for example, phenyl or naphthyl.
  • linker As well as the presence or absence of charged moieties can readily be controlled by the skilled artisan.
  • hydrophobic nature of a linker derived from hexamethylene diamine (H 2 N(CH 2 ) 6 NH 2 ) 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”.
  • frameworks can be designed to provide preferred orientations of the ligands.
  • Such frameworks may be represented by using an array of dots (as shown below) wherein each dot may potentially be an atom, such as C, O, N, S, P, H, F, Cl, Br, and F or the dot may alternatively indicate the absence of an atom at that position.
  • the framework is illustrated as a two dimensional array in the following diagram, although clearly the framework is a three dimensional array in practice:
  • Each dot is either an atom, chosen from carbon, hydrogen, oxygen, nitrogen, sulfur, phosphorus, or halogen, or the dot represents a point in space (i.e., an absence of an atom).
  • the ligands namely, C, O, N, S and P.
  • Atoms can be connected to each other via bonds (single, double or triple bonds with acceptable resonance and tautomeric forms), with regard to the usual constraints of chemical bonding.
  • Ligands may be attached to the framework via single, double or triple bonds (with chemically acceptable tautomeric and resonance forms).
  • Multiple ligand groups (2 to 10) can be attached to the framework such that the minimal, shortest path distance between adjacent ligand groups does not exceed 100 atoms.
  • the linker connections to the ligand is selected such that the maximum spatial distance between two adjacent ligands is no more than 100 ⁇ .
  • Nodes ( 1 , 2 ) and ( 9 , 4 ) are attachment points. Hydrogen atoms are affixed to nodes ( 2 , 4 ), ( 4 , 4 ), ( 4 , 0 ), ( 2 , 0 ), ( 7 , 4 ), ( 10 , 2 ) and ( 7 , 0 ). Nodes ( 5 , 2 ) and ( 6 , 2 ) are connected single bond.
  • the carbon atoms present are connected by either a single or double bonds, taking into consideration the principle of resonance and/or tautomerism.
  • FIG. 1 illustrates a usefuil 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.
  • di splay vectors around similar central core structures such as a phenyl structure (Panel A) and a cyclohexane structure (Panel B) can be varied, as can the spacing of the ligand domain from the core structure (i.e., the length of the attaching moiety).
  • 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 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.
  • 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.
  • 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 flexibility, antigenicity, stability, and the like.
  • the introduction of one or more poly(ethylene 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 decrease antigenicity and potentially enhances the overall rigidity of the linker.
  • PEG poly(ethylene glycol)
  • ancillary groups which enhance the water solubility/hydrophilicity of the linker and, accordingly, the resulting multibinding compounds are useful in practicing this invention.
  • ancillary groups such as, for example, small repeating units of ethylene glycols, alcohols, polyols (e.g., glycerin, glycerol propoxylate, saccharides, including mono-, oligosaccharides, etc.), carboxylates (e.g., small repeating units of glutamic acid, acrylic acid, etc.), arnines (e.g., tetraethylenepentamine), and the like) to enhance the water solubility and/or hydrophilicity of the multibinding compounds of this invention.
  • the ancillary group used to improve water solubility/hydrophilicity will be a polyether.
  • lipophilic ancillary groups within the structure of the linker to enhance the lipophilicity and/or hydrophobicity of the multibinding 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 with the linkers of this invention include fatty acid derivatives which do not form bilayers in aqueous medium until higher concentrations are reached.
  • 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. Hydrophilic characteristics derive from the presence of phosphate, carboxylic, sulfate, amino, sulfhydryl, nitro 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 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, dipalmitoylphosphatidylcholine, dioleoylphosphatidylcholirne, distearoyl-phosphatidylcholine or dilinoleoylphosphatidylcholine could be used.
  • lipid 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 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 linker and the ancillary group(s) 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 can also be imparted electrostatically.
  • 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 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.
  • ancillary groups which are charged, 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.
  • 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.
  • rigidity is imparted by the presence of cyclic groups (e.g., aryl, heteroaryl, cycloalkyl, heterocyclic, etc.).
  • the linker comprises one or more six-membered rings.
  • the ring is an aryl group such as, for example, phenyl or naphthyl.
  • the multibinding compounds described herein comprise 2-10 ligands attached to a linker-that attaches the ligands in such a manner that they are presented to the enzyme 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 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.
  • the linker may be considered as a framework to which ligands are attached.
  • the ligands can 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 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. 1 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 ligand and is the same or different at each occurrence, as can X.
  • a trimer can also be a radial multibinding compound comprising three ligands attached to a central core, and thus represented as (L) 3 X, 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 FIGS. 2 and 3 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 in a branched array e.g., (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., where X and L are as defined herein.
  • a branched array e.g., (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., where X and L are as defined herein.
  • it could be represented as an alkyl, aryl or cycloalkyl derivative as above with four (4) ligands attached to the core linker.
  • 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.
  • 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.
  • 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 multibinding compound with its target(s) 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: (I) the identity of ligand(s), (2) the orientation of ligands, (3) the valency of the construct, (4) linker length, (5) linker geometry, (6) linker physical properties, and (7) linker chemical functional groups.
  • 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.
  • the only requirement for the ligands chosen is that they are capable of interacting with the selected target(s).
  • 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, log P, etc.
  • 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.
  • 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
  • each ligand 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 receptor(s) in multiple relative orientations, an important multibinding design parameter.
  • the only requirement for choosing attachment points is that attaching to at least 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 protease inhibitor bound to its target allows one to identify one or more sites where linker attachment will not preclude the enzyme:inhibitor interaction.
  • 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 activity. This selection derives from, for example, heterobivalent interactions within the context of a single target molecule.
  • a receptor antagonist ligand bound to its target receptor 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 receptor molecule at sites proximal to the antagonist binding site, which include elements of the receptor that are not part of the formal antagonist binding site and/or elements of the matrix surrounding the receptor such as the membrane.
  • the most favorable orientation for interaction of the second ligand molecule with the receptor/matrix may be achieved by attaching it to the linker at a position which abrogates activity of the ligand at the formal antagonist binding site.
  • Another way to consider 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.
  • bivalent interaction focused on bivalent interactions of dimeric 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.
  • a 5HT 4 receptor antagonist and a bladder-selective muscarinic M 3 antagonist may be joined to a linker through attachment points which do not abrogate the binding affinity of the monomeric ligands for their respective receptor sites.
  • the dimeric compound may achieve enhanced affinity for both receptors due to favorable interactions between the 5HT 4 ligand and elements of the M 3 receptor proximal to the formal M 3 antagonist binding site and between the M 3 ligand and elements of the 5HT 4 receptor proximal to the formal 5 HT 4 antagonist binding site.
  • the dimeric compound may be more potent and selective antagonist of overactive bladder and a superior therapy for urinary urge incontinence.
  • 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, stable and intrinsically inocuous under typical chemical and physiological conditions, and compatible with a large number of available linkers. Amide bonds, ethers, amines, carbamates, ureas, and sulfonamides are but a few examples of preferred linkages.
  • Linkers Spanning Relevant Multibinding Parameters Through Selection of Valency, Linker Length, Linker Geometry, Rigidity, Physical Properties, and Chemical Functional Groups
  • linkers 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.
  • divalent linkers or constructs are also typically of modest size such that they retain the desirable biodistribution properties of small molecules.
  • 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.
  • the preferred distance can be estimated rather precisely from high-resolution structural information of targets, typically enzymes and soluble receptor targets.
  • high-resolution structural information is not available (such as 7TM G-protein coupled receptors)
  • preferred linker distances are 2-20 ⁇ , with more preferred linker distances of 3-12 ⁇ .
  • preferred linker distances are 20-100 ⁇ , with more preferred distances of 30-70 ⁇ .
  • 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 cds- or trans-arrangements at the 1,1- vs. 1,2- vs. 1,3- vs.
  • Linker rigidity is varied by controlling the number and relative energies of different conformational states possible for the linker.
  • 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.
  • 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.
  • linkers can be selected to avoid those that are too hydrophilic or too hydrophobic to be readily absorbed and/or distributed in vivo.
  • 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.
  • n being determined by the sum of the number of different attachment points for each ligand chosen
  • 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.
  • an array generated from two ligands, one which has two attachment points (A1, A2) and one which has three attachment points (B 1, B2, B3) joined in all possible combinations provide for at least 15 possible combinations of multibinding compounds: A1—A1 A1-A2 A1-B1 A1-B2 A1-B3 A2—A2 A2-B1 A2-B2 A2-B3 B1—B1 B1-B2 B1-B3 B2—B2 B2-B3 B3—B3
  • combinatorial library Given the combinatorial nature of the library, common chemistries are preferably used to join the reactive functionalities on the ligands with complementary reactive functionalities on the linkers.
  • the library therefore lends 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.
  • the combinatorial libary is prepared in the solution phase.
  • candidate multibinding compounds are optionally purified before assaying for activity by, for example, chromatographic methods (e.g., HPLC).
  • Various methods are used to characterize the properties and activities of the candidate multibinding compounds in the library to determine which compounds possess multibinding properties. Physical constants such as solubility under various solvent conditions and logD/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 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 determined. Pharmacological data, including oral absorption, 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.
  • the members of the library which exhibit multibinding properties 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).
  • each member of the library can be encrypted or tagged with appropriate information allowing determination of the structure of relevant members at a later time.
  • each member of the library can be encrypted or tagged with appropriate information allowing determination of the structure of relevant members at a later 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. Pat. No. 5,846,839; each of which are incorporated herein by reference in its entirety.
  • the structure of relevant multivalent compounds can also be determined from soluble and 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 Jul. 11, 1998. Such methods couple frontal affinity chromatography with mass spectroscopy to determine both the structure and relative binding affinities of candidate multibinding compounds to receptors.
  • 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 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.
  • iterative redesign/analysis using the novel principles of 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.
  • suitable divalent linkers include, by way of example only, those derived from dicarboxylic acids, disulfonylhalides, dialdehydes, diketones, dihalides, diisocyanates,diamines, diols, mixtures of carboxylic acids, sulfonylhalides, aldehydes, ketones, halides, isocyanates, amines and diols.
  • carboxylic acid, sulfonylhalide, aldehyde, ketone, halide, isocyanate, amine and diol functional group is reacted with a complementary functionality on the ligand to form a covalent linkage.
  • complementary functionality is well known in the art as illustrated in the following table:
  • First Reactive Group Second Reactive Group Linkage carboxyl amine amide sulfonyl halide amine sulfonamide hydroxyl alkyl/aryl halide ether hydroxyl isocyanate urethane amine epoxide ⁇ -hydroxyamine amine alkyl/aryl halide alkylamine hydroxyl carboxyl ester amine aldehyde/NaCNBH 3 amine hydroxylamine sulfonyl halide sulfonamide aldehyde amine/NaCHBH 3 amine aldehyde amine/NaCHBH 3 amine amine isocynate urea
  • the following table illustrates, by way of examples, starting materials (identified as X-1 through X-418) that can be used to prepare linkers incorporated in the multibinding compounds of this invention utilizing the chemistry described above.
  • 1,10-decanedicarboxylic acid, X1 can be reacted with 2 equivalents of a ligand carrying an amino group in the presence of a coupling reagent such as DCC to provide a multibinding compound of formula (I) wherein the ligands are linked via 1,10-decanediamido linking group.
  • beta lactam antibiotic ligands represented as L 1 for use in this invention include, by way of example, L 1 -1 through L 1 -5, the ligands L 1 -1 through L 1 -5 having been selected from the compounds of formula (a)-(e) disclosed in the Summary of the invention: compound (a) (L 1 -1), compound (b) (L 1 -2), compound (c) (L 1 -3), compound (d) (L 1 -4) and compound (e) (L 1 -5).
  • glycopeptide ligands represented as L 2 for use in this invention include, by way of example, L 2 -1 through L 2 -2: L 2 -1 being an optionally substituted vancomycin and L 2 -2 being an aglycone derivative of an optionally substituted vancomycin.
  • Combinations of-ligands (L 1 and L 2 ) and linkers (X) per this invention include, by way example only, heterodimers wherein a first ligand, L 1 , selected from L 1 -1 through L 1 -5 above, and a second ligand, L 2 , and a linker, X, are selected from the following: L 2 -1/X-1- L 2 -1/X-2- L 2 -1/X-3- L 2 -1/X-4- L 2 -1/X-5- L 2 -1/X-6- L 2 -1/X-7- L 2 -1/X-8- L 2 -1/X-9- L 2 -1/X-10- L 2 -1/X-11- L 2 -1/X-12- L 2 -1/X-13- L 2 -1/X-14- L 2 -1/X-15- Lz-1/X-16- L 2 -1/X-17- L 2 -1/X-18- L 2 -1/X-19- L 2 -1/X-20- L 2 -1/X-21
  • the compounds of the invention are useful in medical treatments and exhibit biological activity, including antibacterial activity, which can be demonstrated in the tests described in the Examples.
  • the antibacterial activity of the instant compounds may be determined by testing in standardized in vitro dilution tests for minimum inhibitory concentration (MICs). Such tests are well known to those skilled in the art, and are referenced and described in the fourth edition of “Antibiotics in Laboratory Medicine”, by Victor Lorian, M. D., published by Williams and Wilkins, which is hereby incorporated by reference.
  • the compounds of this invention will be found to exhibit activity against gram-positive and gram-negative bacteria such as Staphylococcus aureus, Escherichia coli and Pseudomonas aeruginosa at test levels.
  • gram-positive and gram-negative bacteria such as Staphylococcus aureus, Escherichia coli and Pseudomonas aeruginosa at test levels.
  • the compounds of the present invention are useful in the treatment in mammals of bacterial infections, by both gram-positive and gram-negative bacteria.
  • the compounds may be administered to the mammals in the form of a pharmaceutical composition comprising the compounds of the invention admixed with a pharmaceutically acceptable excipient.
  • 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, transdermnal, subcutaneous, intravenous, intramuscular, and intranasal. These compounds are effective as injectable intranasal and oral compositions. Such compositions are prepared in a manner well known in the pharmaceutical art and comprise at least one active compound.
  • compositions which contain, as the active ingredient, one or more of the compounds described herein associated with pharmaceutically acceptable carriers.
  • 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 capsule, sachet, paper or other container.
  • 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.
  • 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), 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.
  • the active compound 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.
  • 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.
  • 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 30 mg, of the active ingredient.
  • 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 calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • 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).
  • 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.
  • 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.
  • a solid preformulation composition containing a homogeneous mixture of a compound of the present invention.
  • 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.
  • 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.
  • 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.
  • 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 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 suitable pharmaceutically acceptable excipients as described supra.
  • 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 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.
  • Diacid 4 is dissolved in 10 mL anhydrous dimethylformamide and treated sequentially with hydroxybenzotriazole (5.0 mmol), diisopropylethyl amine (4.0 mmol) and PyBOP (4.0 mmol). After stirring for 15 minutes at room temperature, the activated diacid is treated with (+)-6-aminopenicillanic acid 5 (4.0 mmol) and the coupling reaction is stirred overnight at room temperature. Volatiles are removed under vacuum and the crude is fractionated by reverse-phase HPLC using a linear gradient of acetonitrile in water (both buffered with 0.1% trifluoracetic acid) to afford 6 after lyopholization of the appropriate fractions.
  • Diacid 6 (1.0 mmol) is dissolved in 10 mL anhydrous tetrahydrofuran and stirred under nitrogen at room temperature and treated sequentially with pyrrolidine (3.0 mmol) and tetrakis(triphenylphosphine)palladium[0] (0.15 mmol). After 2 hours, the mixture is evaporated to dryness and then fractionated by reverse-phase HPLC using a linear gradient of acetonitrile in water (both buffered with 0.1% trifluoracetic acid) to afford the desired amoxicillin dimer 7 after lyopholization of the appropriate fractions.
  • 4,4′-Dipiperidine hydrochloride (10 mmol) is dissolved in water (100 mL), stirred at room temperature, and treated sequentially with triethylamine (40 mmol) and 2-iminothiolane hydrochloride (20 mmol). After two hours the reaction mixture is frozen and lyopholized and the diamidine dithiol 8 is recovered as the dihydrochloride after crystallization from HCl/diethyl ether.
  • This material is then dissolved in a mixture of tetrahydrofuran and water buffered to pH 7.0 with morpholinopropane sulfonic acid, treated with 10% palladium on carbon (200 mg) and subjected to 40 psi H 2 for 4 hours.
  • the mixture is filtered through a pad of celite to remove catalyst and chromatographed at 4° C. on a column of Dowex 50 ⁇ 4 (Na+cycle, 200-400 mesh) resin eluted with deionized water.
  • the desired compound 10 is recovered upon lyopholization of the appropriate fractions.
  • Thienamycin 11 (2.0 numol) is dissolved in aqueous buffer (morpholinopropane sulfonic acid, pH 8.2) and stirred in an ice/water bath.
  • Dimethyloctanediimidate dihydrochloride 12 (1.0 mmol) is added as a solid and the reaction is stirred one hour in the cooling bath and then two hours at room temperature.
  • the mixture is then chromatographed at 4° C. on a column of Dowex 50 ⁇ 4 (Na+cycle, 200-400 mesh) resin eluted with deionized water.
  • the desired compound 13 is recovered upon lyopholization of the appropriate fractions.
  • Vancomycin-2-aminoethanamide 18 (compound 18 prepared as described in Example 5 below, 1.0 mmol) is dissolved in 5.0 mL anhydrous dimethylformamide, stirred at room temperature, and treated sequentially with diisopropylethyl amine (4.0 mmol) and the solution of the imidazolide 16 (prepared in Step 3 above). After 2 hours, volatiles are removed under vacuum and the residue is triturated with acetonitrile.
  • Vancomycin hydrochloride 17 (10 mmol) is slurried in 100 mL 1:1 methanol:anhydrous dimethylformamide, stirred at room temperature, and treated sequentially with diisopropylethyl amine (20 mmol) and Fmoc glycinal (prepared as described by Salvi et al. Tetrahedron Lett. 1994, 35, 1181-1184). After 2 hours the reaction mixture is cooled in an ice water bath and treated further with sodium cyanoborohydride (4.0 mmol) and trifluoroacetic acid (30 mmol).
  • the crude product is precipitated by dropwise addition to a ten-fold volume of acetonitrile, and then fractionated by reverse-phase HPLC using a linear gradient of acetonitrile in water (both buffered with 0.1% trifluoracetic acid) to afford the adducts reductively alkylated on the N-methylamino terminus 20 and on the N′-amino group of the vancosamine residue 21 .
  • Vancomycin hydrochloride 17 (10 mmol) is) is dissolved in water (100 mL), stirred at room temperature, and treated sequentially with triethylamine (40 mmol) and 2-iminothiolane hydrochloride (10 mmol). After two hours the reaction mixture is fractionated by reverse-phase HPLC using a linear gradient of acetonitrile in water (both buffered with 0. 1% trifluoracetic acid) to afford the imine adducts modified on the N-methylamino terminus compound 26 and on the N′-amino group of the vancosamine residue compound 27 .
  • the crude product is dissolved in a mixture of tetrahydrofuran and water buffered to pH 7.0 with morpholinopropane sulfonic acid, treated with 10% platinum oxide (20 mg) and subjected to 40 psi H 2 for 4 hours.
  • the mixture is filtered through a pad of celite to remove catalyst and chromatographed at 4° C. on a column of Dowex 50 ⁇ 4 (Na+cycle, 200-400 mesh) resin eluted with deionized water.
  • the desired compound 29 is recovered upon lyopholization of the appropriate fractions.
  • Vancomycin hydrochloride 17 (10 mmol) is dissolved in 100 mL 1:1 anhydrous dimethylsulfoxide:dimethylformamide, stirred at room temperature, and treated sequentially with ethylenediamine (20 mmol), hydroxybenzotriazole (10 rnmol), and PyBOP (10 mmol). After two hours, the crude product is precipitated by dropwise addition to 1 vigorously stirred acetonitrile, and then fractionated by reverse-phase HPLC using a linear gradient of acetonitrile in water (both buffered with 0.1% trifluoracetic acid) to afford compound 31 after lyopholization of the appropriate fractions.
  • the crude product is dissolved in a mixture of tetrahydrofuran and water buffered to pH 7.0 with morpholinopropane sulfonic acid, treated with 10% platinum oxide (20 mg) and subjected to 40 psi H 2 for 4 hours.
  • the mixture is filtered through a pad of celite to remove catalyst and chromatographed at 4° C. on a column of Dowex 50 ⁇ 4 (Na+cycle, 200-400 mesh) resin eluted with deionized water.
  • the desired compound 33 is recovered upon lyopholization of the appropriate fractions.
  • compound 42 may be prepared in a similar manner from the PNB ester of commercially available Amoxicillin.
  • the cooling bath is removed and the reaction followed by TLC.
  • the mixture is partitioned between water and ethyl acetate and the aqueous phase back extracted with ethyl acetate.
  • the combined organic extracts are washed with water followed by sat. sodium bicarbonate, dried over sodium sulfate and the solvent removed in vacuo.
  • the crude product is purified by chromatography and dissolved in a mixture of THF and water buffered to pH 7.0 with morpholinopropane sulfonic acid. Add 10% platinum oxide (20 mg) and subject the reaction mixture to 40 psi H 2 for four hours.
  • the reaction mixture is filtered and purified by chromatography.
  • the product is dissolved in a mixture of THF (5 mL) and trifluoroacetic acid (0.5 mL). After the reaction is complete volatiles are removed under vacuum and the crude product is purified by HPLC to afford the desired compound 51 .
  • hexadecanedioic acid (12 mg, 0.05 rnmol) in dimethylformamide (0.3 mL) was added HATU (40 mg, 0.11 mmol). After 1 h, compound 55 (75 mg, 0.10 mmol) was added and stirring was continued. After 4 h, 0.5% aqueous trifluoroacetic acid (0.5 mL) was added and the reaction mixture was purified by reverse phase HPLC to give the desired compound 56 .
  • Hard gelatin capsules containing the following ingredients are prepared: Quantity Ingredient (mg/capsule) Active Ingredient 30.0 Starch 305.0 Magnesium stearate 5.0
  • 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 Ingredient (mg/tablet) 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.
  • a dry powder inhaler formulation is prepared containing the following components: Ingredient Weight % Active Ingredient 5 Lactose 95
  • the active ingredient is mixed with the lactose and the mixture is added to a dry powder inhaling appliance.
  • Tablets each containing 30 mg of active ingredient, are prepared as follows: Quantity Ingredient (mg/tablet) Active Ingredient 30.0 mg Starch 45.0 mg Micro-crystalline cellulose 35.0 mg Polyvinylpyrrolidone 4.0 mg (as 10% solution in sterile water) Sodium carboxymethyl starch 4.5 mg Magnesium stearate 0.5 mg Talc 1.0 mg Total 120 mg
  • 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 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.
  • Capsules each containing 40 mg of medicament are made as follows: Quantity Ingredient (mg/capsule) Active Ingredient 40.0 mg Starch 109.0 mg Magnesium stearate 1.0 mg 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 quantities.
  • Suppositories each containing 25 mg of active ingredient are made as follows: Ingredient Amount 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: Ingredient Amount Active Ingredient 50.0 mg Xanthan gum 4.0 mg Sodium carboxymethyl cellulose (11%) Microcrystalline cellulose (89%) 50.0 mg Sucrose 1.75 g Sodium benzoate 10.0 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: Quantity Ingredient (mg/capsule) Active Ingredient 15.0 mg Starch 407.0 mg Magnesium stearate 3.0 mg 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.
  • a formulation may be prepared as follows: Ingredient Quantity Active Ingredient 5.0 mg Corn Oil 1.0 mL
  • a topical formulation may be prepared as follows: Ingredient Quantity 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.
  • transdermal delivery devices 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. Pat. No. 5,023,252, issued Jun. 11, 1991, herein incorporated by reference in its entirety.
  • patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • ⁇ -lactam resistant bacteria are obtained and phenotyped based on their sensitivity.
  • Minimal inhibitory concentrations are measured in a microdilution broth procedure under NCCLS guidelines. The compounds are serially diluted into Mueller-Hinton broth in 96-well microtiter plates. Overnight cultures of bacterial strains are diluted based on absorbance at 600 nm so that the final concentration in each well was 5 ⁇ 105 cfu/ml. Plates are returned to a 35° C. incubator. The following day (or 24 hours in the case of Enterococci strains), MICs are determined by visual inspection of the plates.
  • Bacterial strains which may be tested in this model include, but are not limited to, those found in Table I and Table II below. Growth conditions may be modified as necessary for each particular strain. Growing conditions and growth media for the strains listed in Table I and Table II are known in the art.
  • CFUs colony forming units
  • the compounds of Formula (I) are administered either intravenously or subcutaneously and observed for 5-15 minutes. If there are no adverse effects, the dose is increased in a second group of mice. This dose incrementation continues until mortality occurs, or the dose is maximized. Generally, dosing begins at 20 mg/kg and increases by 20 mg/kg each time until the maximum tolerated dose (MTD) is achieved.
  • MTD maximum tolerated dose
  • mice are administered the compound of Formula (I) either intravenously or subcutaneously at a therapeutic dose (in general, approximately 50 mg/kg).
  • a therapeutic dose in general, approximately 50 mg/kg.
  • Groups of animals are placed in metabolic cages so that urine and feces may be collected for analysis.
  • Blood is collected by cardiac puncture and the following organs are harvested: lung, liver, heart, brain, kidney, and spleen.
  • Tissues were weighed and prepared for HPLC analysis. HPLC analysis on the tissue homogenates and fluids is used to determine the concentration of the compound of Formula (I). Metabolic products resulting from changes to the compound of Formula (I) are also determined.
  • the bacteria was combined with hog gastric mucin to enhance virulence.
  • the dose of bacteria (normally 10 5 -10 7 ) is that which is sufficient to induce mortality in all of the mice over a three day period.
  • the compound of Formula (I) is administered in a single dose, either IV or subcutaneously.
  • Each dose is administered to groups of 5 to 10 mice, at doses that typically range from a maximum of about 20 mg/kg to a minimum of less than 1 mg/kg.
  • a positive control (normally ⁇ -lactam with ⁇ -lactam sensitive strains) is administered in each experiment. The dose at which approximately 50% of the animals are saved is calculated from the results.
  • mice are initially rendered neutropenic by administration of cyclophosphamide at 200 mg/kg on day 0 and day 2. On day 4, they are infected in the left anterior thigh by an IM injection of a single dose of bacteria. The mice are administered the compound of Formula (I) one hour after the administration of bacteria. At various later times (normally 1, 2.5, 4 and 24 hours) the mice are sacrificed (3 per time point). The thigh is excised, homogenized and the number of CFUs (colony forming units) is determined by plating. Blood is also plated to determine the CFUs in the blood.
  • CFUs colony forming units
  • the rate at which the compound of Formula (I) is removed from the blood can be determined in either rats or mice.
  • rats the test animals are cannulated in the jugular vein.
  • a compound of Formula (I) is administered via tail vein injection, and at various time points (normally 5, 15, 30, 60 minutes and 2, 4, 6 and 24 hours) blood is withdrawn from the cannula.
  • mice a compound of Formula (I) is also administered via tail vein injection, and at various time points. Blood is normally obtained by cardiac puncture.
  • the concentration of the remaining compound of Formula (I) is determined by HPLC.

Abstract

This invention relates to novel multibinding compounds (agents) that are antibacterial agents. The multibinding compounds of the invention comprise from 2-10 ligands covalently connected by a linker or linkers, wherein each of said ligands in their monovalent (i.e., unlinked) state have the ability to bind to a an enzyme involved in cell wall biosynthesis and metabolism, a precursor used in the synthesis of the bacterial cell wall and/or the bacterial cell surface thereby interfere with the synthesis and/or metabolism of the cell wall. In particular the multibinding compounds of the invention comprise from 2-10 ligands covalently connected by a linker or linkers, wherein each of said ligands has a ligand domain capable of binding to penicillin binding proteins, a transpeptidase enzyme, a substrate of a transpeptidase enzyme, a beta-lactamase enzyme, pencillinase enzyme, cephalosporinase enzyme, a transglycoslase enzyme, or a transglycosylase enzyme substrate; Preferably, the ligands are selected from the beta lactam or glycopeptide class of antibacterial agents.

Description

    BACKGROUND OF THE INVENTION
  • 1. Field of the Invention
  • This invention relates to novel multibinding compounds (agents) that are antibacterial agents. The multibinding compounds of the invention comprise from 2-10 ligands covalently connected by a linker or linkers, wherein each of said ligands in their monovalent (i.e., unlinked) state have the ability to bind to an enzyme involved in cell wall biosynthesis and metabolism, a precursor used in the synthesis of the bacterial cell wall and/or the cell surface and thereby interfere with the synthesis and or metabolism of the cell wall. Preferably, the ligands are selected from the beta lactam and/or glycopeptide class of antibacterial agents.
  • The invention also relates to pharmaceutical compositions comprising a pharmaceutically acceptable excipient and a therapeutically effective amount of one or more compound(s) of the invention, methods of using such compounds and methods of preparing such compounds.
  • 2. Background
  • Bacteria possess a rigid outer layer, the cell wall. The cell wall maintains the shape of the microorganism which has a high internal osmotic pressure. Injury to the cell wall (e.g. by lysozyme) or inhibition of the cell wall's formation leads to lysis of the cell.
  • The cell wall contains a chemically distinct complex polymer “mucopeptide” (“murein”, “peptidoglycan”) consisting of polysaccharides and a highly cross-linked polypeptide. The polysaccharides comprise an alternating copolymer of the amino sugars N-acetylglucosamine and N-acetylmuramic acid, the latter being found only in bacteria. To the N-acetylmuramic residues are attached pentapeptides. The polysaccharide backbone of the cell wall is formed by oligomerization of disaccharide pentapeptide precursors (lipid intermediate II) and is catalyzed an enzyme known as transglycosylase. The final rigidity of the cell wall is imparted by cross-linking of the peptide chains as a result of transpeptidation reactions by several bacterial enzymes one of which is known as peptidoglycan transpeptidase.
  • One method by which antibacterial agents exert their antibacterial activity is by inhibiting the transglycosylase enzyme, thus interfering with the penultimate step in the synthesis of the bacterial cell wall. Although not wishing to be bound by theory, it is believed that a glycopeptide, for example vancomycin, binds with high affinity and specificity to N-terminal sequences (L-lysyl-D-alanyl-D-alanine in vancomycin sensitive organisms) of peptidoglycan precursors known as lipid intermediate II. By binding to and sequestering these precursors, vancomycin prevents their utilization by the cell wall biosynthesis machinery. In a formal sense, therefore, vancomycin inhibits the bacterial transglycosylase that is responsible for adding lipid intermediate II subunits to growing peptidoglycan chains. This step preceeds the cross-linking transpeptidation step which is inhibited by beta lactam antibiotics. It is believed that the β-lactam antibiotics bind to certain cell receptors (the penicillin binding proteins, “PBPs”) which catalyze the transpeptidation reaction and other cell wall metabolic processes. The incomplete cell wall likely serves as a substrate for autolytic enzymes in the cell wall and results in lysis if the environment is isotonic.
  • Antibacterial agents have proved to be important weapons in the fight against pathogenic bacteria. However, an increasing problem with respect to the effectiveness of antibacterial agents relates to the emergence of strains of bacteria that are highly resistant to such agents. It would therefore be highly desirable to find antibacterial agents that are active against a broad spectrum of bacteria, in particular resistant strains. It would also be advantageous to discover antibacterial agents that demonstrate high activity and selectivity toward their targets, and are of low toxicity.
  • The multibinding compounds of the present invention fulfill this need.
  • SUMMARY OF THE INVENTION
  • This invention relates to novel multibinding compounds (agents) that are antibacterial agents. The multibinding compounds of the invention comprise from 2-10 ligands covalently connected by a linker or linkers, wherein each of said ligands in their monovalent (i.e., unlinked ) state have the ability to bind to an enzyme involved in cell wall biosynthesis and metabolism, a precursor used in the synthesis of the bacterial cell wall and/or the bacterial cell surface and thereby interfere with the synthesis and/or metabolism of the cell wall. Preferably, the ligands are selected from the beta lactam and glycopeptide classes of antibacterial agents.
  • The invention also relates to pharmaceutical compositions comprising a pharmaceutically acceptable excipient and a therapeutically effective amount of one or more compound(s) of the invention, methods of using such compounds and methods of preparing such compounds.
  • Accordingly, in one aspect, this invention provides a multibinding compound of Formula (I):
    (L)p(X)q  (I)
    wherein:
  • p is an integer of from 2 to 10;
  • q is an integer of from 1 to 20;
  • each ligand, L, comprises a ligand domain capable of binding to penicillin binding proteins, a transpeptidase enzyme, a substrate of a transpeptidase enzyme, a beta-lactamase enzyme, pencillinase enzyme, cephalosporinase enzyme, a transglycoslase enzyme, or a transglycosylase enzyme substrate; and
  • X is a linker that may be the same or different at each occurrence; and pharmaceutically acceptable salts thereof provided that:
    • (i) all the ligands in a multibinding compound of Formula (I) cannot be either an optionally substituted glycopeptide antibiotic, or an aglycone derivative of an optionally substituted glycopeptide antibiotic;
    • (ii) when p is 2 and q is 1 then at least one of the ligands is a beta lactam antibiotic; and
    • (iii) when p is 2, q is 1, and one of the ligands is vancomycin attached to a linker via the [C] terminus, then the other ligand cannot be cefalexin attached to the linker via acylation of its alpha amino group.
  • Preferably, q is less thanp in the multibinding compounds of this invention.
  • In a second aspect, this invention provides a multibinding compound of Formula (I):
  •   (L)p(X)q  (I)
  • wherein:
  • p is an integer of from 2 to 10;
  • q is an integer of from 1 to 20;
  • each ligand, L, is a beta lactam antibiotic, an optionally substituted glycopeptide antibiotic, or an aglycone derivative of an optionally substituted glycopeptide antibiotic;
  • X is a linker that may be same or different at each occurrence provided that:
    • (i) all the ligands in a multibinding compound of Formula (I) cannot be. either an optionally substituted glycopeptide antibiotic, or an aglycone derivative of an optionally substituted glycopeptide antibiotic;
    • (ii) when p is 2 and q is 1 then at least one of the ligands is a beta lactam antibiotic; and
    • (iii) when p is 2, q is 1, and one of the ligands is vancomycin attached to a linker via the [C] terminus, then the other ligand cannot be cefalexin attached to the linker via acylation of its alpha amino group.
  • Preferably, q is less than p;
  • each ligand that is a beta lactam antibiotic is selected from the group consisting of penems, penams, cephems, carbapenems, oxacephems, carbacephems, and monobactam ring systems; and
  • each ligand that is a glycopeptide antibiotic is selected from the group consisting of Actaplanin, Actinodidin, Ardacin, Avoparcin, Azureomycin, A477, A35512, A40926, A41030, A42867, A47934, A80407, A82846, A83850, A84575, A84428, AB-65, Balhimycin, Chloroeremomycin, Chloroorienticin, Chloropolysporin, Decaplanin, N-demethylvancomycin, Eremomycin, Galacardin, Helvecardin. Izupeptin, Kibdelin, LL-AM374, Mannopeptin, MM45289, MM47756, MM47761, MM47921, MM47766, MM55260, MM55266, MM55270, MM56579, MM56598, OA-7653, Oreenticin, Parvodicin. Ristocetin, Ristomycin, Synmonicin, Teicoplanin, UK-68597, UK-69542, UK-72051, optionally substituted Vancomycin, and aglycone derivatives thereof.
  • More preferably, each ligand that is a beta lactam antibiotic is selected from the group consisting of:
    • (i) a compound of formula (a):
      Figure US20070134729A1-20070614-C00001

      wherein:
  • R is substituted alkyl, aryl aralkyl, or heteroaryl wherein each of said substituent optionally links (a) to a linker via a covalent bond or R is a covalent bond that links (a) to a linker; and
  • R1 and R2 are, independently of each other, alkyl or at least one of R1 and R2 is a covalent bond linking (a) to a linker;
    • (ii) a compound of formula (b):
      Figure US20070134729A1-20070614-C00002

      wherein:
  • one of P and Q is O, S, or —CH2— and the other is —CH2—;
  • R3 is substituted alkyl, heteroarylalkyl, aralkyl, heterocyclylalkyl, or —C(R6)═NOR7 (where R6 is aryl, heteroaryl, or substituted alkyl; and R7 is alkyl or substituted alkyl) wherein each of said substituent optionally links (b) to a linker or R3 is a covalent bond that links (b) to a linker; and
  • R4 is hydrogen, alkyl, alkenyl, substituted alkenylene, substituted alkyl, halo, heteroarylalkyl, heterocyclylalkyl, —SRa (where Ra is aryl, heteroaryl, heterocyclyl, or cycloalkyl) or —CH2SRa (where Ra is aryl, heteroaryl, heterocyclyl, or cycloalkyl) wherein each of said substituent optionally links (b) to a linker or R4 is a covalent bond that links (b) to a linker;
  • R5is hydrogen, hydroxy, or alkoxy;
    • (iii) a compound of formula (c):
      Figure US20070134729A1-20070614-C00003

      wherein:
  • T is S or CH2;
  • R8a is alkyl;
  • W is O, S, —OCH2—, or CH2; and R8 is -(alkylene)-NHC(Rb)═NH where Rb is a covalent bond linking (c) to a linker; or —W—R8 is a covalent bond that links (c) to a linker;
    • (iv) a compound of formula (d):
      Figure US20070134729A1-20070614-C00004

      wherein:
  • R9 and R9a are alkyl;
  • R10 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, halo, aryl, heteroaryl, heterocyclyl, aralkyl, heteroaralkyl, heterocyclylalkyl or —CH2SRa (where Ra is aryl, heteroaryl, heterocyclyl, or cycloalkyl) wherein each of said substituent optionally links (d) to a linker or at, least one of R9 and R10 is a covalent bond that links (d) to a linker; or
  • R9 and R10 together with the carbon atoms to which they are attached form an aryl, heteroaryl, cycloalkyl, substituted cycloalkyl, or heterocyclyl ring of 4 to 7 ring atoms wherein one of the ring atoms optionally links (d) to a linker; or
    • (v) a compound of formula (e):
      Figure US20070134729A1-20070614-C00005

      wherein:
  • R11 is —SO3H or -(alkylene)-COOH;
  • R12 is alkyl, substituted alkyl, haloalkyl, alkoxy, aryl, aralkyl, heteroaryl, heteroaralkyl, cycloalkyl, substituted cycloalkyl, or heterocyclyl wherein each of said substituent optionally binds (e) to a linker or R12 is a covalent bond that links (e) to a linker; and
  • R13 is alkyl, acyl, or —COC(R14)═N—OR15 wherein R14 is aryl, heteroaryl which optionally links (e) to a linker, and R15 is -(alkylene)-COOR6 wherein R16 is hydrogen or optionally links (e) to a linker or R13 is a covalent bond that links (e) to a linker; and pharmaceutically acceptable salts thereof;
  • Even more preferably, each ligand that is a beta lactam antibiotic is selected from the group consisting of:
    • (i) a compound of formula (a):
      Figure US20070134729A1-20070614-C00006

      wherein:
  • R is:
    Figure US20070134729A1-20070614-C00007

    where:
  • R17 is a covalent bond that links the (a) group to a linker;
  • one of R18 and R19 is hydrogen and the other is a covalent bond that links the (a) group to a linker; and
  • R1 and R2 are methyl;
    • (ii) a compound of formula (b)):
      Figure US20070134729A1-20070614-C00008

      where:
  • R3 and R4 are:
    R3 R4
    Figure US20070134729A1-20070614-C00009
    —CH2OCOCH3
    Figure US20070134729A1-20070614-C00010
    Figure US20070134729A1-20070614-C00011
    Figure US20070134729A1-20070614-C00012
    —CH3
    Figure US20070134729A1-20070614-C00013
    —CH3
    Figure US20070134729A1-20070614-C00014
    Figure US20070134729A1-20070614-C00015
    Figure US20070134729A1-20070614-C00016
    —CH2OCONHR19 —CH2OCOCH3
    Figure US20070134729A1-20070614-C00017
    —Cl
    Figure US20070134729A1-20070614-C00018
    —CH2OCONHR19
    Figure US20070134729A1-20070614-C00019
    Figure US20070134729A1-20070614-C00020
    Figure US20070134729A1-20070614-C00021
    —CH2OCOCH3, —CH2OCH3, H
    Figure US20070134729A1-20070614-C00022
    Figure US20070134729A1-20070614-C00023
    Figure US20070134729A1-20070614-C00024
    Figure US20070134729A1-20070614-C00025
    Figure US20070134729A1-20070614-C00026
    Figure US20070134729A1-20070614-C00027
    Figure US20070134729A1-20070614-C00028
    Figure US20070134729A1-20070614-C00029
    Figure US20070134729A1-20070614-C00030
    Figure US20070134729A1-20070614-C00031
    Figure US20070134729A1-20070614-C00032
    Figure US20070134729A1-20070614-C00033

    (Note:

    the R3 group in the left column is paired with the R4 in the right column) wherein:
  • n is 0 or 1; m is 1-5; Z is CH or N; Y is H or halo; R is alkyl; R17 is a covalent bond that links the (b) group to a linker; one of R18 and R19 is hydrogen or alkyl; R30 and R31 are, independently of each other, hydrogen or aLlcyl; or together with the nitrogen atom to which they are attached form a heterocycloamino group; and R, R32 and R33 are independently alkyl wherein one of R18, R19, R30—R33 is a covalent bond that links the (b) group to a linker;
    • (iii) a compound of formula (c):
      Figure US20070134729A1-20070614-C00034

      wherein Rb is a covalent bond lining (c) to a linker;
    • (iv) a compound of formula (d):
      Figure US20070134729A1-20070614-C00035

      where Ra is:
      Figure US20070134729A1-20070614-C00036

      where:
  • R23 is a covalent bond that links (d) to a linker;
  • one of R24 and R25 is hydrogen, alkyl substituted alkyl, or aralkyl, and other is a covalent bond that hnks (d) to a linker; R26 is alkyl; or
    • (v) a compound of formula (e):
      Figure US20070134729A1-20070614-C00037

      wherein one of R21 and R22 is hydrogen and the other links (d) to a linker; and
  • X is selected from a compound of formula:.
    —Xa—Z—(Ya—Z)m—Xa
    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—, —OC(O)—, —C(O)NR—, —NRC(O)—, C(S), —C(S)O—, —C(S)NR—, —NRC(S)—, or a covalent bond where R is as defined below;
  • Z at each separate occurrence is selected from the group consisting of alkylene, substituted alkylene, cycloalkylene, substituted cylcoalkylene, alkenylene, substituted alkenylene, alkynylene, substituted alynylene, cycloalkenylene, substituted cycloalkenylene, arylene, heteroarylene, heterocyclene, or a covalent bond;
  • each Ya at each separate occurrence is selected from the group consisting of —O—, —C(O)—, —OC(O)—, —C(O)O—, —NR—, —S(O)n—, —C(O)NR′—,—NR′ C(O)—, —NR′C(O)NR′—, —NR′C(S)NR′—, —C(═NR′)—NR′—, —NR′—C(═NR′)—, —OC(O)—NR′—, —NR′—C(O)—O—, —N═C(Xa)—NR′—, —NR′—C(Xa)═N—,—P(O)(OR′)—O—, —O—P(O)(OR′)—, —S(O)nCR′R″—, —S(O)n—NR′—, —NR′—S(O)n—, —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 a second aspect, the invention relates to a method of treatment of mammals having a disease state that is treatable by antibacterial agents, comprising administering a therapeutically effective amount of a compound of Formula (I), or a mixture of compounds of Formula (I), thereto.
  • In a third aspect, the invention relates to a pharmaceutical composition comprising a therapeutically effective amount of one or more compounds of Formula (I) or a pharmaceutically acceptable salt thereof, admixed with at least one pharmaceutically acceptable excipient.
  • In a fourth aspect, this invention is directed to general synthetic methods for generating large libraries of diverse multimeric compounds which multimeric compounds are candidates for possessing multibinding properties. The diverse multimeric compound libraries provided by this invention are synthesized by combining a linker or linkers with a ligand or ligands to provide for a library of multimeric compounds wherein the linker and ligand each have 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 and polarization. The library of ligands is preferably selected to have diverse attachment points on the same ligand, different finctional groups at the same site of otherwise the same ligand, and the like.
  • In a fifth aspect, this invention is directed to libraries of diverse multimeric compounds which multimeric compounds are candidates for possessing multibinding properties. 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 targeting a receptor.
  • Accordingly, in one of its method aspects, this invention is directed to a method for identifying multimeric ligand compounds possessing multibinding properties 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 finctional groups react to formn a covalent linkage between said linker and at least two of said ligands; and
  • (d) assaying the multimeric ligand compounds produced in (c) above to identify multimneric ligand.compounds possessing multibinding properties.
  • In another of its method aspects, this invention is directed to a method for identifying multimeric ligand compounds possessing multibinding properties 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 flnctional 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 finctional 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 (c) above to identify multimeric ligand compounds possessing multibinding properties.
  • 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). Sequential addition is preferred when a mixture of different ligands is employed to ensure heterodimeric or multimeric compounds are prepared. Concurrent addition of the ligands occurs when at least a portion of the multimer compounds prepared are homomultimeric compounds.
  • The assay protocols recited in (d) can be conducted on the multimeric ligand compound library produced in (c) above, or preferably, each member of the library is isolated by preparative liquid chromatography mass spectrometry (LCMS).
  • In one of its composition aspects, this invention is directed to a library of multimeric ligand compounds which may possess multivalent properties 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 finctional 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.
  • In another of its composition aspects, this invention is directed to a library of multimeric ligand compounds which may possess multivalent properties 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 finctional 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 library of linkers employed in either the methods or the library aspects of this invention is 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. For example, in one embodiment, each of the linkers in the linker library may comprise linkers of different chain length and/or having different complementary reactive groups. Such linker lengths can preferably range from about 2 to 100 Å.
  • In another preferred embodiment, the ligand or mixture of ligands is selected to have reactive functionality at different sites on said ligands in order to provide for a range of orientations of said ligand on said multimeric ligand compounds. Such reactive functionality includes, by way of example, carboxylic acids, carboxylic acid halides, carboxyl esters, .amines, halides, pseudohalides, isocyanates, vinyl unsaturation, ketones, aldehydes, thiols, alcohols, anhydrides, boronates, and precursors thereof. It is understood, of course, that 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 other embodiments, the multimeric ligand compound is homomeric (i.e., each of the ligands is the same, although it may be attached at different points) or heteromeric (i.e., at least one of the ligands is different from the other ligands).
  • In addition to the combinatorial methods described herein, this invention provides for an iterative process for rationally evaluating what molecular constraints impart multibinding properties to a class of multimeric compounds or ligands targeting a receptor. Specifically, this method aspect is directed to a method for identifying multimeric ligand compounds possessing multibinding properties 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;
  • (c) repeating the process of (a) and (b) above until at least one multimeric compound is found to possess multibinding properties;
  • (d) evaluating what molecular constraints imparted 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 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.
  • Preferably, steps (e) and (f) are repeated at least two times, more preferably at from 2-50 times, even more preferably from 3 to 50 times, and still more preferably at least 5-50 times.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 illustrates examples of multibinding compounds comprising 2 ligands attached in different formats to a linker.
  • FIG. 2 illustrates examples of multibinding compounds comprising 3 ligands attached in different formats to a linker.
  • FIG. 3 illustrates examples of multibinding compounds comprising 4 ligands attached in different formats to a linker.
  • FIG. 4 illustrates examples of multibinding compounds comprising >4 ligands attached in different formats to a linker.
  • FIGS. 5, 6A, and 6B disclose some representative compounds of formula (a) and (b).
  • FIGS. 7-10 disclose examples of multibinding compounds comprising 2ligands attached in different formats.
  • FIGS. 11-23 illustrate synthesis of compounds of Formula (I).
  • DETAILED DESCRIPTION OF THE INVENTION Definitions
  • This invention is directed to multibinding compounds that are antibacterial agents and pharmaceutical compositions containing such compounds. When discussing such compounds, and compositions 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 1 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, nitro, guanidine, —C(═NRa)NHRb (where Ra and Rb are independently selected from hydrogen, alkyl, aryl, aralkyl, heteroaryl, or heteroaralkyl, —NHSO2NHRc (where Rc is hydrogen, alkyl, aryl, aralkyl, heteroaryl, or heteroaralkyl) —SO-alkyl, —SO-substituted alkyl, —SO-aryl,—SO-heteroaryl, —SO2-alkyl, —SO2-substituted alkyl, —SO2-aryl and 13 SO2-heteroaryl. This term is exemplified by groups such as hydroxymethyl, hydroxyethyl, hydroxypropyl, 2—aminoethyl, 3-aminopropyl, 2-methylaminoethyl, 3-dimethylaminopropyl, 2—sulfonamidoethyl, 2-carboxyethyl, and the like.
  • The term “al kylene” refers to a diradical of a 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 methylene (—CH2—), ethylene (—CH2CH2—), the propylene isomers (e.g., —CH2CH2CH2— and —CH(CH3)CH2—) and the like.
  • The term “substituted alkylene” refers to:
    • (a) 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 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, nitro, —SO-alkyl, —SO-substituted alkyl, —SO-aryl, —SO-heteroaryl, —SO2-alkyl, —SO2-substituted aLkyl, —SO2-aryl and —SO2-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;
    • (b) an alkylene group as defined above wherein one or more carbons atoms, is replaced by oxygen, sulfur, and —NR— where R is hydrogen, substituted alkyl, cycloalkyl, alkenyl cycloalkenyl, alkynyl, aryl, heteroaryl and heterocyclic.
  • The term “alkaryl” or “aralkyl” 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—, cycloaikyl-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-propoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, 1,2-dimethylbutoxy, and the like.
  • The term “substituted alkoxy” refers to the groups substituted alkyl-O—, substituted alkenyl-O—, substituted cycloalkyl-O—, substituted cycloalkenyl-O—, and substituted alkynyl-O— where substituted alkyl, substituted alkenyl, substituted cycloalkyl, substituted cycloalkenyl and substituted alkynyl are as defined herein.
  • 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. Preferred alkenyl groups include ethenyl (—CH═CH2), n-propenyl.(—CH2CH═CH2), iso-propenyl (—C(CH3)═CH2), 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 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, nitro, —SO-alkyl, —SO-substituted alkyl, —SO-aryl, —SO-heteroaryl, —SO2-aLkyl, —SO2-substituted alkyl, —SO2-aryl and —SO2-heteroaryl.
  • The term “alkenylene” refers to a diradical 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. This term is exemplified by groups such as ethenylene (—CH═CH—), the propenylene isomers (e.g., —CH2CH═CH—, —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, 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, nitro, —SO-alkyl, —SO-substituted alkyl, —SO-aryl, —SO-heteroaryl, —SO2-alkyl, —SO2-substituted alkyl, —SO2-aryl and —SO2-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, cycloalkenyl, substituted cycloalkenyl, aryl, heterocyclic or heteroaryl groups fused to the alkenylene 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 preferably from 1-6 sites of acetylene (triple bond) unsaturation. Preferred alkynyl groups include ethynyl (—C—CH), propargyl (—CH2C≡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 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, nitro, —SO-alkyl, —SO-substituted alkyl, —SO-aryl, —SO-heteroaryl, —SO2-alkyl, —SO2-substituted alkyl, —SO2-aryl, and —SO2-heteroaryl.
  • The term “alkynylene” refers to a diradical of an unsaturated hydrocarbon 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 acetylene (triple bond) unsaturation. Preferred alkynylene groups include ethynylene (—C≡C—), propargylene (—CH2C═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, 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, nitro, —SO-alkyl, —SO-substituted alkyl, —SO-aryl, —SO-heteroaryl, —SO2-alkyl, —SO2-substituted alkyl, —SO2-aryl and —SO2-heteroaryl
  • The term “acyl” refers to the groups HC(O)—, alkyl-C(O)—, substituted alkyl-C(O)—, alkenyl-C(O)—, substituted alkenyl-C(O)—, cycloalkyl-C(O)—, substituted cycloalkyl-C(O)—, cycloalkenyl-C(O)—, substituted cycloalkenyl-C(O)—, aryl-C(O)—, heteroaryl-C(O)— and heterocyclic-C(O)— where alkyl, substituted alkyl,.alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic are as defined herein.
  • 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 “sulfonylamino” refers to the group —NRSO2Ra where R is hydrogen, alkyl, substituted alkyl, aralkyl, or heteroaralkyl, and Ra is alkyl, substituted alkyl, amino, or substituted amino wherein alkyl, substituted alkyl, aralkyl, heteroaralkyl and substituted amino are as defined herein.
  • The term “aminoacyl” refers to the group —NRC(O)R where each R is independently hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, amino, substituted amino, aryl, heteroaryl, or heterocyclic wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, 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 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). The aryl group may optionally be fuised to a heterocyclic or cycloalkyl group. Preferred aryls.include 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 suhstituents, preferably 1 to 3 substituents, selected from the group consisting of acyloxy, 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, sulfonylamino, alkaryl, aryl, aryloxy, azido, carboxyl, carb6xylalkyl, cyano, halo, nitro, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, aminoacyloxy, oxyacylanino, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioheteroaryloxy, —SO-alkyl, —SO-substituted alkyl, —SO-aryl, —SO-heteroaryl,- —SO2-alkyl, —SO2-substituted alkyl, —SO2-aryl, —SO2-heteroaryl and trihalomethyl. Preferred aryl substituents include alkyl, alkoxy, halo, cyano, nitro, 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 “amino” refers to the group —NH2.
  • The term “substituted amino” refers to the group —NRR where each R is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, acyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, 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-substituted cycloalkyl”, “—C(O)O-alkenyl”, “-C(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, said cycloalkyl group may optionally be fused to an aryl or heteroaryl group. 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 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 alkyl, substituted alkyl, alkenyl, substituted alkenyl, 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, nitro, —SO-alkyl, 13 SO-substituted alkyl, —SO-aryl, —SO-heteroaryl, —SO2-alkyl, —SO2-substituted alkyl, —SO2-aryl and —SO2-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 unsaturation. Examples of suitable cycloalkenyl groups include, for instance, cyclobut-2-enyl, cyclopent-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 group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, 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, nitro, —SO-alkyl, —SO-substituted alkyl, —SO-aryl, —SO-heteroaryl, —SO2-alkyl, —SO2-substituted alkyl, —SO2-aryl and —SO2-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). The heteroaryl ring may optionally be fused to a cycloalkyl or heterocyclyl ring. Unless otherwise constrained by the definition for the heteroaryl substituent, such heteroaryl groups can be optionally substituted with 1 to 5 substituents, preferably 1 to 3 substituents, selected from the group consisting of acyloxy, 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, nitro, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, aminoacyloxy, oxyacylamino, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioheteroaryloxy, —SO-alkyl, —SO-substituted alkyl, —SO-aryl, —SO-heteroaryl, —SO2-alkyl, —SO2-substituted alkyl, —SO2-aryl, —SO2-heteroaryl and trihalomethyl. Preferred heteroaryl substituents include alkyl, alkoxy, halo, cyano, nitro, trihalomethyl, and thioalkoxy. Such heteroaryl groups can have a single ring (e.g., pyridyl or furyl) or multiple condensed rings (e.g., indolizinyl or benzothienyl). Preferred heteroaryls include pyridyl, pyrrolyl and furyl.
  • The term “heteroaryloxy” refers to the group heteroaryl-O—.
  • The term “heterocycle” or “heterocyclyl” refers to a monoradical saturated or 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 and further wherein one, two, or three of the ring carbon atoms may optionally be replaced with a carbonyl group (i.e., a keto group). The heterocycle group may optionally fused to an aryl or heteroaryl ring. Unless otherwise constrained by the definition for the heterocyclic substituent, such heterocyclic groups can be optionally substituted with 1 to 5, and preferably 1 to 3 substituents, selected from the group consisting of alkyl, acyloxy, alkyl, substituted alkyl, 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, nitro, —SO-alkyl, —SO-substituted alkyl, —SO-aryl, —SO-heteroaryl, —SO2-alkyl, —SO2-substituted alkyl, —SO2-aryl and —SO2-heteroaryl. Such heterocyclic groups can have a single ring or multiple condensed rings. Preferred heterocyclics include morpholino, piperidinyl, and the like.
  • Examples of heteroaryls and heterocycles include, but are not limited to, pyrrole, thiophene, furan imidazole, pyrazole, pyridine, pyrazine, pyrimidine, 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, pyrrolidine, piperidine, piperazine, indoline, morpholine, tetrahydrofuiranyl, tetrahydrothiophene, and the like as well as N-alkoxy-nitrogen containing heterocycles.
  • “Heterocycloamino” means a saturated, unsaturated, bridged or unbridged, monovalent cyclic group of 4 to 8 ring atoms, wherein at least one ring atom is N and optionally contains one or two additional ring heteroatoms selected from the group consisting of N, O , or S(O)n (where n is an integer from 0 to 2), the remaining ring atoms being C, where one or two C atoms may optionally be replaced by a carbonyl group. The heterocycloamino ring may be fused to a cycloalkyl, aryl or heteroaryl ring, and it may be optionally substituted with one or more substituents, preferably one or two substituents, selected from alkyl, substituted alkyl, cycloaLkyl, aryl, araiiyl, heteroaryl, heteroaralkyl, halo, cyano, acyl, amino, substituted amino, acylamino, —OR (where R is hydrogen, alkyl, alkenyl, cycloalkyl, acyl, aryl, heteroaryl, aralkyl, or heteroaralkyl), or —S(O)nR [where n is an integer from 0 to 2 and R is hydrogen (provided that n is 0), alkyl, alkenyl, cycloalkyl, amino, heterocyclo, aryl, heteroaryl, aralkyl, or heteroaralkyl]. More specifically the term heterocycloamino includes, but is not limited to, pyrrolidino, piperidino, morpholino, piperazino, indolino, quinuclidine, or thiomorpholino, and the derivatives thereof.
  • The term “heterocyclooxy” refers to the group heterocyclic-O—.
  • The term “thioheterocyclooxy” refers to the group heterocyclic-S—.
  • 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 and heterocyclic are as defined herein.
  • The term “spiro-attached cycloalkyl group” refers to a cycloalkyl group joined to another ring via one carbon atom common to both rings.
  • The term “thiol” refers to the group —SH.
  • The term “thioalkoxy” or “alkylthio” 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.
  • 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 and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto.
  • Pharnaceutically-acceptable base addition salts can be prepared from inorganic and organic bases. Salts derived from inorganic bases, include by way 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 arnines, trialkenyl amines, 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 amiines, disubstituted cycloalkenyl amine, trisubstituted cycloalkenyl amines, aryl amines, diaryl amiines, triaryl amines, heteroaryl amiines, diheteroaryl amines, triheteroaryl ainines, heterocyclic amnines, diheterocyclic amiines, 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, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, and the like. Also included are amiines where the two or three substituents, together with the amino nitrogen, form a heterocyclic or heteroaryl group. 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, 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.
  • 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, mtalic acid, malonic acid, succinic acid, maleic 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 “pharmaceutically-acceptable cation” refers to the cation of a 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 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 (See., T. W. Greene and P. G. H. Wuts, “Protective Groups in Organic Synthesis”, 2nd Ed.). 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, 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. 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), fluorenylmethoxy-carbonyl (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, 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 includes instances where said event or circumstance occurs and instances where it does not.
  • The term “ligand” or “ligands” as used herein denotes a compound that is a binding partner for penicillin binding proteins, a pencillinase enzyme, a cephalosporinase enzyme, a transpeptidase enzyme, a substrate of a transpeptidase enzyme, a beta-lactamase enzyme, a transglycoslase enzyme, or a transglycosylase enzyme substrate and is bound thereto by complementarity. The specific region or regions of the ligand that is (are) recognized by the penicillin binding proteins, a pencillinase enzyme, a cephalosporinase enzyme, a transpeptidase enzyme, a substrate of a transpeptidase enzyme, a beta-lactamase enzyme, a transglycoslase enzyme, or a transglycosylase enzyme substrate is designated as the “ligand domain”. A ligand may be either capable of binding to its target 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 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 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 usefuil in binding to penicillin binding proteins, a pencillinase enzyme, a cephalosporinase enzyme, a transpeptidase enzyme, a substrate of a transpeptidase enzyme, a beta-lactamase enzyme, a transglycoslase enzyme, or a transglycosylase enzyme substrate (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 penicillin binding proteins, a transglycoslase enzyme, or a taansglycosylase enzyme substrate 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 multivalency. The term “ligand” or “ligands” as used herein is intended to include the racemic forms of the ligands as well as individual enantiomers and diasteromers and non-racemic mixtures thereof.
  • The term “β-lactam antibiotic” refers to antibiotics, having a β-lactam ring core which can be depicted as follows:
    Figure US20070134729A1-20070614-C00038
  • The β-lactam antibiotics are classified into the penicillins, cephalosporins, carbapenems, oxacephems, carbacephems, and monobactams and include drugs such as Penicillin G, Penicillin V, Methicillin, Oxacillin, Cloxacillin, Dicloxacillin, Nafcillin, Ampicillin, Amoxicillin, Carbenicillin, Carbenicillin indanyl, Ticarcillin, Mezlocillin, Piperacillin Cephalothin, Cefazolin, Cephalexin, Cefadroxil, Cefamandole, Cefoxitin, Cefaclor, Cefuroxime, Cefuroxime axetil, Loracarbef, Cefonicid, Cefotetan, Ceforanide, Cefotaxime, Cefpodoxime proxetil, Ceftizoxime, Ceftriaxone, Cefoperazone, Ceftazidime, Cefepime Imipenem, Meropenem, Aztreonam, Ritipenem, L-695256, GV-143253, Sanifitrinem, Fropenem, Lactivicin, BO-2727, MEN-10700, Ro-48-8724, Cefosilis, SB-216477, S-4661, GG-326, BLA-857, PGE-8335534, PGE-542860, LB-10522, GV—129606, BO-2052A, CS-834, MK-826, YH-1226, YM-40220, MDL-63908, FCE25199, Panipenem, TOC-50, TOC-39, TOC-29, E-1101, Sulopenem, DU-6681, MC-02479, Temocillin, Carunonam, Ro-25-0534, SUN-A-0026, WS-1358A, Ro-25-1132, CGβ-57701,CGP -37697A, TMA-230, Syn-2190, Biapenem, CS-834, DWβ-204, DX-8739, CS-976, CKD-529, ER-35786, DZ-2640, 4-AAz, KR-21012, RO-25-0993, DA-1211, BMS-181139, J-11225, L-786392, DK-35C, Ro-25-6833, S-1090, E-1101, FK-518, DP-736, Cefditoren, LY-215891, R0-09-1428, Cefdaloxime, Cefoselis, KST-150185, Ro-09-1227, Cefclidin, Cefluprenam, Cefotiam, LB-10522, Cefcanel, BRL-57342, Cefprirome, YH-1226, Cefprozil, CKD-604, KST-150288, Cefcapene, Ro-24-8138, FK-312, Cefozopran, RU-59863, Ceftibuten, FR-193879, FK-041, Cefdinir, CP-6679, R0-63-9141, CFC-240, Cefpimizole, Cefminox, Cefetamet, CP-0467, PGE-7119699, R0-48-8391, AM-1817, AM-1732, MC-02002, BO-1341, BK-218, Ro-25-4835, RO-25-2016, YM-40220, Ro-23-9424, LY-206763, CR-240, YH-1266, MC-02331, Ro-44-3949, MC-02306, Ro-25-7103, BMS-180680. Preferred β-lactam antibiotics are Amoxicillin, Nafcillin, Cefadroxil, Ceftriaxone, Cefaclor, Aztreonam, Ceftazidime, Imipenem, Meropenem, Ritipenem, Ceftazidine, Pipericillin, Clauvlinic acid, Cefepime, Cefoxitin, Cefotaximne, Cefixime, Lefluzidine and derivatives thereof
  • The glycopeptide antibiotics are characterized by a multi-ring peptide core and at least one sugar attached at various sites, of which vancomycin is an important example. Examples of the glycopeptide class of ligands included in this definition may be found in “Glycopeptides Classification, Occurrence, and Discovery” by Rao, R. C. and Crandall, L. W., (Drugs and the Pharmaceutical Sciences” Vol. 63, edited by Ramakrishnan Nagarajan, published by Marcal Dekker, Inc.) which is hereby incorporated by reference. Disclosed are glycopeptides identified as Actaplanin, Actinodidin, Ardacin, Avoparcin, Azureomycm, A477, A35512, A40926, A41030, A42867, A47934, A80407, A82846, A83850, A84575, A84428, AB-65, Balhimycin, Chloroeremomycin, Chloroorientiein, Chloropolysporin, Decaplanin, N-demethylvancomycin., Eremomycin, Galacardin, Helvecardin. Izupeptin, Kibdelin, LL-AM374, Mannopeptin, MM45289, MM47756, MM47761, MM47921, MM47766, MM55260, MM55266, MM55270, MM56579, MM56598, OA-7653, Oreenticin, Parvodicin. Ristocetin, Ristomycin, Synrmonicin, Teicoplanin, UK-68597, UK-69542, UK-72051, Vancomycin, and the like. Another preferred class of ligands is the general class of glycopeptides disclosed above on which the sugar moiety is absent. For example removal of the disaccharide moiety appended to the phenol on vancomycin (as shown below as Formula II) by mild hydrolysis gives vancomycin aglycone. A further preferred class are glycopeptides that have been further appended with additional saccharide residues,.especially aminoglycosides, in a manner similar to vancosamine.
  • “Vancomycin” refers to the antibacterial compound whose structure is reproduced below as Formula Ia.
    Figure US20070134729A1-20070614-C00039
  • “Optional” or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. For example. “Optionally substituted glycopeptide” with respect to a compound of Formula (I) refers to a ligand as defined above in which those positions that are not linked to X may or may not be substituted by various groups as defined below. The term also includes those instances in which one amino acid of the basic core structure is replaced by another amino acid, for example as described in “Preparation and conformational analysis of vancomycin hexapeptide and aglucovancomycin hexapeptide”, by Booth, P. M., Williams, D. H., Univ. Chem. Lab., Cambridge, UK., J. Chem. Soc., Perkin Trans. I (1989), (12), 2335-9, and “The Edman degradation of vancomycin:preparation of vancomycin hexapeptide”, Booth, P. M., Stone, D. J. M, Williams, D. H., Univ. Chem. Lab., Cambridge, UK., J. Chem. Soc., Chem. Commun. (1987), (22), 1694-5.
  • “Optionally substituted vancomycin” with respect to the multibinding agents of the invention refers to vancomycin in which the hydroxy group at any position, the [R] position the carboxyl groups at the [C] position, or the amine groups at the [V].or [N] position that are not attached to the linker X may or may not be substituted by various groups. Such groups include: Ra where Ra at each occurrence is chosen from alkyl, alkyl optionally interrupted by 1-5 atoms-chosen from O, S, or —NRb— where Rb is alkyl, aryl, or heteroaryl, all of which are optionally.substituted, haloalkyl, alkenyl, alkynyl, alkylamino, alkylaminoalkyl, cycloalkyl, alkanoyl, aryl, heteroaryl, heterocyclic, additional saccharide residues, especially ammoglycosides, all of which are optionally substituted as defined above; and: NRWd in which Rc and Rd are independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkanoyl, aryl, heteroaryl, arylalkyl, or heteroarylalkyl, or Rc and Rd when taken together with the nitrogen atom to which they are attached represent a heterocyclic group, quarternary alkyl and aryl ammonium compounds, pyidinium ions, sulfonium ions, and the like, all of which are optionally substituted as defined above. An example of a preferred [C] substitution is dimethylaminopropylamino and glucosamino; and example of a preferred [V] substitution is alkyl, for example n-decyl, or alkylaminoalkyl, for example n-decylaminoethyl.
  • “Optionally substituted vancomycin aglycone” with respect to the multibinding agents of the invention refers to vancomycin aglycone in which the hydroxy group at any position, particularly the hydroxy group at the [O] position, the [R] position, the carboxy groups at the [C] position, or the amine group at the [N] position, that are not attached to the linker X may or may not be substituted by various groups —Ra as defined above.
  • “Transglycosylase enzyme substrate” as used herein denotes the molecular target of the transglycosylase enzyme. The substrate binds to the enzyme and eventually results in the synthesis of the bacterial cell wall. The action of this enzyme is inhibited by a ligand domain that binds to the enzyme itself and/or the enzyme substrate. A ligand such as vancomycin binds to this substrate and in effect “sequesters” the substrate to prevent its recognition by the enzyme and subsequent use in the construction of the bacterial cell wall. There is also a growing feeling that some glycopeptides or derivatives thereof may directly bind to and inhibit the transglycolase.
  • 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. In all cases, each ligand and linker in the multibinding compound is independently selected such that the multibinding compound includes both symmetric compounds (i.e., where each ligand as well as each linker is identical) and asymmetric compounds ((i.e., where at least one of the ligands is different from the other ligand(s) and/or at least one linker is different from the other linker(s)). Multibinding compounds provide a biological and/or 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, increased ability to kill cells such as fungal pathogens, cancer cells, etc., 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 “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
    Figure US20070134729A1-20070614-C00040

    when only one ligand is interacting with a ligand binding site. Examples of univalent interactions are depicted below.
  • 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 ligand binding sites 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 multibinding compound bearing three ligands versus a monovalent binding interaction, is shown below:
    Figure US20070134729A1-20070614-C00041
  • It should be understood that not all compounds that contain multiple copies of a ligand attached to a linker or to linkers 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 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 linker(s) in a specific manner in order to bring about the desired ligand-orienting result, and thus produce a multibinding event.
  • 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 ligand 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 multibinding agent produces an equivalent biological or therapeutic effect at a lower concentration than the aggregate unlinked ligand is indicative of enhanced potency.
  • 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 (i.e., the dissociation constants for each ligand-receptor complex) or, in cases where a biological effect is observed below the Y , the ratio of the respective EC50's (i.e., the concentrations that produce 50% of the maximum response for the ligand interacting with the two distinct ligand binding sites (receptors)).
  • The term “ligand binding site” denotes the site on a penicillin binding proteins, a transpeptidase enzyme, penicillinase enzyme, cephalosporinase enzyme, beta lactamase enzyme, a transpeptidase enzyme substrate, a transglycosylase enzyme and/or transglycosylase enzyme substrate 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 producing a unique biological effect, for example, agonism, antagonism, and modulatory effects, or it may maintain an ongoing biological event, and the like.
  • It should be recognized that the ligand binding sites of the enzyme or the receptor that participate in biological multivalent binding interactions are constrained to varying degrees by their intra- and inter-molecular associations. For example, ligand binding sites 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.
  • 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, 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:
  • (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;
  • (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.
  • 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 that is an antibacterial agent, and those disease states which have been found to be usefully treated by a specific multibinding compound of our invention.
  • 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 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’ refers to a group or groups that covalently attaches from 2 to 10 ligands (as identified above) in a manner that provides for a compound capable of multivalency. Among other features, the linker is a ligand-orienting entity that permits attachment of at least two copies of a ligand (which may be the same or different) thereto. In some cases, the linker may itself be biologically active. The term “linker” does not, however, extend to 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.
  • 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 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 of ligands to the ligand binding sites of an enzyme involved in cell wall biosynthesis and metabolism, a precursor used in the synthesis of the bacterial cell wall and/or the cell surface, whether such sites are located interiorly, both interiorly and on the periphery of the enzyme structure, or at any intermediate position thereof.
  • In the FIGS. 9, 10, 14-16, glycopeptides are depicted in a simplified form as a shaded box that shows only the carboxy terminus, labeled [C], the sugar amine terminus (e.g., vancosamine), labeled [V], and the “non-sugar” amino terminus, labeled [N] as follows:
    Figure US20070134729A1-20070614-C00042

    where R is hydrogen (as N-desmethylvancomycin) or methyl (as in vancomycin).
  • It can be seen by way of exemplification that one class of multivalent compounds that fall within the scope of the definition of Formula (I) include compounds wherein the glycopeptide ligand is connected by one or more linkers at the [C], [V], or [N] terminus.
  • Another class of multivalent compounds that fall within the scope of the definition of Formula (I) include compounds where the aglycone derivatives of glycopeptides. depicted as a triangle that shows only the carboxyl terminus, labeled [C], the aglycone hydroxy terminus labeled [9], and the “non-sugar” amino terminus, labeled [N] as follows:
    Figure US20070134729A1-20070614-C00043

    where R is hydrogen (as in N-desmethylvancomycin aglycone) or methyl (as in vancomycin aglycone) wherein the aglycone derivatives ligand is connected by one or more linkers at the [C], [V], or [N] terminus.
  • A third class of compounds falling within the scope of the invention are those in which the glycopeptides, or aglycone derivatives thereof, are linked via the [R] position. Reaction schemes that exemplify this linking strategy depict the ligands in a simplified form as above, i.e., as a shaded box in which the carboxyl terminus is labeled [C], the vancosamine amino terminus is labeled [V], and the “non-sugar” amino terminus is labeled [N], with the addition of the [R] position as a resorcinol derivative as shown below:
    Figure US20070134729A1-20070614-C00044

    where R is hydrogen or methyl.
  • PREFERRED EMBODIMENTS
  • While the broadest definition of this invention is set forth in the Summary of the Invention, certain compounds of Formula (I) are preferred.
  • (A) One preferred group of compounds is a multibinding compound of Formula (II):
    La—X—Lb  (II)
    wherein:
  • La is a beta lactam antibiotic is selected from the group consisting of:
    • (i) a compound of formula (a):
      Figure US20070134729A1-20070614-C00045

      wherein:
  • R is substituted alkyl, aryl, aralkyl, or heteroaryl wherein each of said substituent optionally links (a) to a linker via a covalent bond or R is a covalent bond that links (a) to a linker; and
  • R1 and R2 are, independently of each other, alkyl or at least one of R1 and R2 is a covalent bond linking (a) to a linker;
    • (ii) a compound of formula (b):
      Figure US20070134729A1-20070614-C00046

      wherein:
  • one of P and Q is O, S, or —CH2— and the other is —CH2—;
  • R3 is substituted alkyl, heteroarylalkyl, aralkyl, heterocyclylalkyl, or —C 6)═NOR7 (where R6 is aryl, heteroaryl, or substituted alkyl; and R7 is alkyl or substituted alkyl) wherein each of said substituent optionally links (b) to a linker or R3 is a covalent bond that links (b) to a linker; and
  • R4 is hydrogen, alkyl, alkenyl, substituted alkenylene, substituted alkyl, halo, heteroarylalkyl, heterocyclylalkyl, —SRa (where Ra is aryl, heteroaryl, heterocyclyl, or cycloalkyl) or —CH2SRa (where Ra is aryl, heteroaryl, heterocyclyl, or cycloalkyl) wherein each of said substituent optionally links (b) to a linker or R4 is a covalent bond that links (b) to a linker;
  • R5 is hydrogen, hydroxy, or alkoxy;
    • (iii) a compound of formula (c):
      Figure US20070134729A1-20070614-C00047

      wherein:
  • T is S or CH2;
  • R8a is alkyl;
  • W is O, S, —OCH2—, or CH2; and R8 is -(alkylene)-NHC(Rb=NH where Rb is a covalent bond linking (c) to a linker; or —W—R8 is a covalent bond that links (c) to a linker,
    • (iv) a compound of formula (d):
      Figure US20070134729A1-20070614-C00048

      wherein:
  • R9 and R9a are alkyl;
  • R10 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, halo, aryl, heteroaryl, heterocyclyl, aralkyl, heteroaralkyl, heterocyclylalkyl or —CH2SRa (where R1 is aryl, heteroaryl, heterocyclyl, or cycloalkyl) wherein each of said substituent optionally links (d) to a linker or at least one of R9 and R10 is a covalent bond that links (d) to a linker; or
  • R9 and R10 together with the carbon atoms to which they are attached form an aryl, heteroaryl, cycloalkyl, substituted cycloalkyl, or heterocyclyl ring of 4 to 7 ring atoms wherein one of the ring atoms optionally links (d) to a linker; or
    • (v) a compound of formula (e):
      Figure US20070134729A1-20070614-C00049

      wherein:
  • R11 is —SO3H or -(alkylene)-COOH;
  • R12 is alkyl, substituted alkyl, haloalkyl, alkoxy, aryl, aralkyl, heteroaryl, heteroaralkyl, cycloalkyl, substituted cycloalkyl, or heterocyclyl wherein each of said substituent optionally binds (e) to a linker or R12 is a covalent bond that links (e) to a linker; and
  • R13 is alkyl, acyl, or —COC(R14)═N—OR15 wherein R14 is aryl, heteroaryl which optionally links (e) to a linker, and R15 is -(alkylene)-COOR16 wherein R16 is hydrogen or optionally links (e) to a linker or R13 is a covalent bond that links (e) to a linker, preferably
  • La is selected from the group consisting of.
    • (i) a compound of formula (a):
      Figure US20070134729A1-20070614-C00050

      wherein:
  • R is:
    Figure US20070134729A1-20070614-C00051

    where:
  • R17 is a covalent bond that links the (a) group to a linker;
  • one of R18 and R19 is hydrogen and the other is a covalent bond that links the (a) group to a linker; and
  • R1 and k2 are methyl;
    • (ii) a compound of formula (b):
      Figure US20070134729A1-20070614-C00052

      where:
  • R3and R4are:
    R3 R4
    Figure US20070134729A1-20070614-C00053
    —CH2OCOCH3
    Figure US20070134729A1-20070614-C00054
    Figure US20070134729A1-20070614-C00055
    Figure US20070134729A1-20070614-C00056
    —CH3
    Figure US20070134729A1-20070614-C00057
    —CH3
    Figure US20070134729A1-20070614-C00058
    Figure US20070134729A1-20070614-C00059
    Figure US20070134729A1-20070614-C00060
    —CH2OCONHR19 —CH2OCOCH3
    Figure US20070134729A1-20070614-C00061
    —Cl
    Figure US20070134729A1-20070614-C00062
    —CH2OCONHR19
    Figure US20070134729A1-20070614-C00063
    Figure US20070134729A1-20070614-C00064
    Figure US20070134729A1-20070614-C00065
    —CH2OCOCH3, —CH2OCH3, H
    Figure US20070134729A1-20070614-C00066
    Figure US20070134729A1-20070614-C00067
    Figure US20070134729A1-20070614-C00068
    Figure US20070134729A1-20070614-C00069
    Figure US20070134729A1-20070614-C00070
    Figure US20070134729A1-20070614-C00071
    Figure US20070134729A1-20070614-C00072
    Figure US20070134729A1-20070614-C00073
    Figure US20070134729A1-20070614-C00074
    Figure US20070134729A1-20070614-C00075
    Figure US20070134729A1-20070614-C00076
    Figure US20070134729A1-20070614-C00077

    (Note:

    the R3 group in the left column is paired with the R4 in the right column)

    wherein:
  • n is 0 or 1; mis 1-5; Z is CH or N; Y is H or halo; R is alkyl; R17 is a covalent bond that links the (b) group to a linker; one of R18 and R19 is hydrogen or alkyl; R30 and R31 are, independently of each other, hydrogen or alkyl; or together with the nitrogen atom to which they are attached form a heterocycloamino group; and R, R32 and R33 are independently alkyl wherein one of R18 , R19, R30—R33 is a covalent bond that links the (b) group to a linker;
    • (iii) a compound of formula (c):
      Figure US20070134729A1-20070614-C00078

      wherein Rb is a covalent bond linking (c) to a linker;
    • (iv) a compound of formula (d):
      Figure US20070134729A1-20070614-C00079

      where Ra is:
      Figure US20070134729A1-20070614-C00080

      where:
  • R23 is a covalent bond that links (d) to a linker;
      • one of R24 and R25 is hydrogen, alkyl, substituted alkyl, or aralkyl, and other is a covalent bond that links (d) to a linker, R26 is alkyl; or
    • (v) a compound of formula (e):
      Figure US20070134729A1-20070614-C00081

      wherein one of R21 and R22 is hydrogen and the other links (d) to a linker;
  • Lb is an optionally substituted vancomycin which is linked to a linker via any hydroxyl group, carboxyl group or amino group; and
  • X is a linker and is selected from a compound of formula:
    —Xa—Z—(Ya—Z)m—Xa
    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—, —OC(O)—, —C(O)NR—, —NRC(O)—, C(S), —C(S)O—, —C(S)NR—, —NRC(S)—, or a covalent bond where R is as defined below;
  • Z 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;
  • each ya at each separate occurrence is selected from the group consisting of —O—, —C(O)—, —OC(O)—, —C(O)O—, —NR—, —S(O)n—, —C(O)NR′—, —NR′C(O)—, —NR′C(O)NR′—, —NR′C(S)NR′—, —C(═NR′)—NR′—, —NR′—C(═NR′)—, —OC(O)—NR′—, —NR′—C(O)—O—, —N═C(Xa)—NR′—,—NR′—C(XaY—N—,—P(O)(OR′)—O—, —O—P(O)(OR′)—, —S(O)nCR′R″—, —S(O)n—NR′—, —NR′—S(O)n—, —S—S—, and a covalent bond; where n is 0, 1 or 2; and R, R1 and R11 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; and pharmaceutically acceptable salts thereof provided that when Lb is vancomycin attached to a linker via the [C] terminus, then La cannot be cefalexin attached to the linker via acylation of its alpha amino group; and pharmaceutically acceptable salts thereof.
  • (B) Another more preferred group of compounds is a multibinding compound of Formula (III):
    Lc—X—Ld  (III)
    wherein:
  • ligands, Lc and Ld, are a beta lactam antibiotic and are independently selected from the group consisting of:
    • (i) a compound of formula (a):
      Figure US20070134729A1-20070614-C00082

      wherein:
  • R is substituted alkyl, aryl, aralkyl, or heteroaryl wherein each of said substituent optionally. links (a) to a linker via a covalent bond or R is a covalent bond that links (a) to a linker; and
  • R1 and R2 are, independently of each other, alkyl or at least one of R1 and R2 is a covalent bond linking (a) to a linker;
    • (ii) a compound of formula (b):
      Figure US20070134729A1-20070614-C00083

      wherein:
  • one of P and Q is O, S, or —CH2— and the other is —CH2—;
  • R3 is substituted alkyl, heteroarylalkyl, aralkyl, heterocyclylalkyl, or —C 6)═NOR7 (where R6 is aryl, heteroaryl, or substituted alkyl; and R7 is alkyl or substituted alkyl) wherein each of said substituient optionally links (b) to a linker or R3 is a covalent bond that links (b) to a linker; and
  • R4 is hydrogen, alkyl, alkenyl, substituted alkenylene, substituted alkyl, halo, heteroarylalkyl, heterocyclylalkyl, —SRa (where Ra is aryl, heteroaryl, heterocyclyl, or cycloalkyl) or —CH2SRa (where Ra is aryl, heteroaryl, heterocyclyl, or cycloalkyl) wherein each of said substituent optionally links (b) to a linker or R4 is a covalent bond that links (b) to a linker;
  • R5 is hydrogen, hydroxy, or alkoxy;
    • (iii) a compound of formula (c):
      Figure US20070134729A1-20070614-C00084

      wherein:
  • T is S or CH2;
  • R8 is alkyl;
  • W is O, S, —OCH2—, or CH2; and R8 is -(alkylene)-NHC(Rb)═NH where Rb is a covalent bond linking (c) to a linker; or —W—R8 is a covalent bond that links (c) to a linker;
    • (iv) a compound of formula (d):.
      Figure US20070134729A1-20070614-C00085

      wherein:
  • R9 and R9a are alkyl;
  • R10 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, halo, aryl, heteroaryl, heterocyclyl, aralkyl, heteroaralkyl, heterocyclylalkyl or —CH2SRa (where Ra is aryl, heteroaryl, heterocyclyl, or cycloalkyl) wherein each of said substituent optionally links (d) to a linker or at least one of R9 and R10 is a covalent bond that links (d) to a linker; or
  • R9 and R10 together with the carbon atoms to which they are attached form an aryl, heteroaryl, cycloalkyl, substituted cycloalkyl, or heterocyclyl ring of 4 to 7 ring atoms wherein one of the ring atoms optionally links (d) to a linker; or
    • (v) a compound of formula (e):
      Figure US20070134729A1-20070614-C00086

      wherein:
  • R11 is —SO3H or -(alkylene)-COOH;
  • R12 is alkyl, substituted alkyl, haloalkyl, alkoxy, aryl, aralkyl, heteroaryl, heteroaralkyl, cycloalkyl, substituted cycloalkyl, or heterocyclyl wherein each of said substituent optionally binds (e) to a linker or R12 is a covalent bond that links (e) to a linker; and
  • R13 is alkyl, acyl, or —COC(R14)═N—OR15 wherein R14 is aryl, heteroaryl which optionally links (e) to a linker, and R15 is -(alkylene)-COOR16 wherein R16 is hydrogen or optionally links (e) to a linker or R13 is a covalent bond that links (e) to a linker, preferably
  • Lc and Ld are independently selected from the group consisting of:
    • (i) a compound of formula (a):
      Figure US20070134729A1-20070614-C00087

      wherein:
  • R is:
    Figure US20070134729A1-20070614-C00088

    where:
  • R17 is a covalent bond that. links the (a) group to a linker, one of R18 and R19 is hydrogen and the other is a covalent bond that links the (a) group to a linker;
    • (ii) a compound of formula (b):
      Figure US20070134729A1-20070614-C00089

      where:
  • R3 and R4 are:
    R3 R4
    Figure US20070134729A1-20070614-C00090
    —CH2OCOCH3
    Figure US20070134729A1-20070614-C00091
    Figure US20070134729A1-20070614-C00092
    Figure US20070134729A1-20070614-C00093
    —CH3
    Figure US20070134729A1-20070614-C00094
    —CH3
    Figure US20070134729A1-20070614-C00095
    Figure US20070134729A1-20070614-C00096
    Figure US20070134729A1-20070614-C00097
    —CH2OCONHR19 —CH2OCOCH3
    Figure US20070134729A1-20070614-C00098
    —Cl
    Figure US20070134729A1-20070614-C00099
    —CH2OCONHR19
    Figure US20070134729A1-20070614-C00100
    Figure US20070134729A1-20070614-C00101
    Figure US20070134729A1-20070614-C00102
    —CH2OCOCH3, —CH2OCH3, H
    Figure US20070134729A1-20070614-C00103
    Figure US20070134729A1-20070614-C00104
    Figure US20070134729A1-20070614-C00105
    Figure US20070134729A1-20070614-C00106
    Figure US20070134729A1-20070614-C00107
    Figure US20070134729A1-20070614-C00108
    Figure US20070134729A1-20070614-C00109
    Figure US20070134729A1-20070614-C00110
    Figure US20070134729A1-20070614-C00111
    Figure US20070134729A1-20070614-C00112
    Figure US20070134729A1-20070614-C00113
    Figure US20070134729A1-20070614-C00114

    (Note:

    the R3 group in the left column is paired with the R4 in the right column)

    wherein:
  • n is 0 or 1; m is 1-5; Z is CH or N; Y is H or halo; R is alkyl;
  • R17 is a covalent bond that links the (b) group to a linker; one of R18 and R19 is hydrogen or alkyl; R30 and R31 are, independently of each other, hydrogen or alkyl; or together with the nitrogen atom to which they are attached form a heterocycloamino group; and R, R32 and R33 are independently alkyl wherein one of R18, R19, R30—R33 is a covalent bond that links the (b) group to a linker;
    • (iii) a compound of formula (c):
      Figure US20070134729A1-20070614-C00115
    • wherein Rb is a covalent bond linking (c) to a linker;
    • (iv) a compound of (c) formula (d):
      Figure US20070134729A1-20070614-C00116

      where Ra is:
      Figure US20070134729A1-20070614-C00117

      where:
  • R23 is a covalent bond that liniks (d) to a linker;
      • one of R24 and R25 is hydrogen, alkyl, substituted alkyl, or aralkyl, and other is a covalent bond that links (d) to a linker; R26 is alkyl; or
    • (v) a compound of formula (e):
      Figure US20070134729A1-20070614-C00118

      wherein one of R21 and R22 is hydrogen and the other links (d) to a linker; and
  • X is a linker is selected from a compound of formula:
    —Xa—Z—(Ya—Z)m—Xa
    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—, —OC(O)—, —C(O)NR—, —NRC(O)—, C(S), —C(S)O—, —C(S)NR—, —NRC(S)—, or a covalent bond where R is as defined below;
  • Z at each separate occurrence is selected from the group consisting of alkylene, substituted alkylene, cycloalkylene, substituted cylcoallylene, alkenylene, substituted alkenylene, alkynylene, substituted alkynylene, cycloalkenylene, substituted cycloalkenylene, arylene, heteroarylene, heterocyclene, or a covalent bond;
  • each Ya at each separate occurrence is selected from the group consisting of —O—, —C(O)—, —OC(O)—, —C(O)O—, —NR—, —S(O)n—, —C(O)NR′—, —NR′C(O), —NR′C(O)NR′—, —NR′C(S)NR′—, —C(═NR′)—NR′—, —NR′—C(═NR′)—, —OC(O)—NR′—, —NR′—C(O)—O—, —N═C(Xa)—NR′—, —NR′—C(Xa)═N—, —P(O)(OR′)—O—, —O—P(O)(OR′)—,—S(O)nCR′R″—,—S(O) n—NR′—, —NR′—S(O)n—, —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; and pharmaceutically acceptable salts thereof.
  • Within the above more preferred groups, an even more preferred group of compounds is that wherein:
  • La, Lc, and Ld are independently selected from the group consisting of:
    Figure US20070134729A1-20070614-C00119
    Figure US20070134729A1-20070614-C00120

    Lb is selected from the group consisting of:
    Figure US20070134729A1-20070614-C00121

    wherein the atom carrying the bond with the dashed line indicates the point of attachment of the ligand to the linker; and
  • the linker is selected from the group consisting of:
    Figure US20070134729A1-20070614-C00122
    Figure US20070134729A1-20070614-C00123
    Figure US20070134729A1-20070614-C00124
    Figure US20070134729A1-20070614-C00125
    Figure US20070134729A1-20070614-C00126
    Figure US20070134729A1-20070614-C00127
    Figure US20070134729A1-20070614-C00128
    Figure US20070134729A1-20070614-C00129
    Figure US20070134729A1-20070614-C00130

    LINKERS DERIVED FROM AMINOALDEHYDES
    Figure US20070134729A1-20070614-C00131

    Linkers Derived from Aminoacids
    Figure US20070134729A1-20070614-C00132
  • Representative compounds of the invention are shown in the table below:
  • (I) Compounds of Formula (III) wherein the ligands are selected from a compound of formula (b) and are linked to a linker, X, via the R3 group and where P, Q. R4, and R5 and are as defined below are:
    Figure US20070134729A1-20070614-C00133
    Cpd
    No Linker X
    1
    Figure US20070134729A1-20070614-C00134
    2
    Figure US20070134729A1-20070614-C00135
    3
    Figure US20070134729A1-20070614-C00136
    4
    Figure US20070134729A1-20070614-C00137
    5
    Figure US20070134729A1-20070614-C00138
    6
    Figure US20070134729A1-20070614-C00139
    7
    Figure US20070134729A1-20070614-C00140
    8
    Figure US20070134729A1-20070614-C00141
    9
    Figure US20070134729A1-20070614-C00142
    10
    Figure US20070134729A1-20070614-C00143
    11
    Figure US20070134729A1-20070614-C00144
    12
    Figure US20070134729A1-20070614-C00145
    13
    Figure US20070134729A1-20070614-C00146
    14
    Figure US20070134729A1-20070614-C00147
    15
    Figure US20070134729A1-20070614-C00148
    16
    Figure US20070134729A1-20070614-C00149
    17
    Figure US20070134729A1-20070614-C00150
    18
    Figure US20070134729A1-20070614-C00151
    19
    Figure US20070134729A1-20070614-C00152
    20
    Figure US20070134729A1-20070614-C00153
    21
    Figure US20070134729A1-20070614-C00154
    22
    Figure US20070134729A1-20070614-C00155
    Cpd
    No P Q R4 R5
    1 CH3 H —CH2 S
    2 CH3 H —CH2 S
    3 CH3 H —CH2 S
    4 CH3 H —CH2 S
    5 CH3 H —CH2 S
    6 CH3 H —CH2 S
    7 CH3 H —CH2 S
    8 CH3 H —CH2 S
    9 CH3 H —CH2 S
    10 CH3 H —CH2 S
    11 CH3 H —CH2 S
    12 CH3 H —CH2 S
    13 CH3 H —CH2 S
    14 CH3 H —CH2 S
    15 —CH2-1-methyl- H —CH2 S
    1H-tetrazol-5-
    ylsulfanyl
    16 —CH2-1-methyl- H —CH2 S
    1H-tetrazol-5-
    ylsulfanyl
    17 —CH2-1-methyl- H —CH2 S
    1H-tetrazol-5-
    ylsulfanyl
    18 —CH2-1-methyl- H —CH2 S
    1H-tetrazol-5-
    ylsulfanyl
    19 —CH2-1-methyl- H —CH2 S
    1H-tetrazol-5-
    ylsulfanyl
    20 —CH2-1-methyl- H —CH2 S
    1H-tetrazol-5-
    ylsulfanyl
    21 —CH2-1-methyl- H —CH2 S
    1H-tetrazol-5-
    ylsulfanyl
    22 —CH2-1-methyl- H —CH2 S
    1H-tetrazol-5-
    ylsulfanyl
  • Compounds of Formula (III) wherein the ligands are selected from a compound of formula (1,) and are linked to a linker, X, via the R 4 group and where P, Q, R3, and R5 and are as defined below are:
    Figure US20070134729A1-20070614-C00156
    Cpd No Linker X P Q R3 R5
    1
    Figure US20070134729A1-20070614-C00157
    (2-aminothiazol-4-yl)- methoxyiminomethyl H —CH2 S

    III. Other compounds of the invention are:
    Figure US20070134729A1-20070614-C00158
    Figure US20070134729A1-20070614-C00159
  • General Synthetic Scheme
  • Compounds of this invention can be made by the methods depicted in the reaction schemes shown below.
  • The starting materials and reagents used in preparing these compounds are either available from commercial suppliers such as Aldrich Chemical Co., (Milwaukee, Wis., USA), Bachem (Torrance, Calif., USA), Emka-Chemie, or Sigma (St. Louis, Mo., USA) or are prepared by methods known to those skilled in the art following procedures set forth in references such as Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-15 (John Wiley and Sons, 1991); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplementals (Elsevier Science Publishers, 1989), Organic-Reactions, Volumes 140 (John Wiley and Sons, 1991), March's Advanced Organic Chemistry, (John Wiley and Sons, 4th Edition), and Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989).
  • The starting materials and the intermediates of the reaction may be isolated and purified if desired using conventional techniques, including but not limited to filtration, distillation, crystallization, chromatography, and the like. Such materials may be characterized using conventional means, including physical constants and spectral data.
  • Furthermore, 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.
  • These schemes are merely illustrative of some methods by which the compounds of this invention can be synthesized, and various modifications to these schemes can be made and will be suggested to one skilled in the art having referred to this disclosure.
  • Preparation of a multibinding compound of Formlula (I)
  • In general, a multibinding compound of Formula (I) can be prepared as illustrated and described in Scheme A below.
  • A multibinding compound of Formula (I) can be prepared by covalently attaching the ligands, L, to a linker, X, as shown in Scheme A below.
    Figure US20070134729A1-20070614-C00160
  • In method (a), a multibinding compound of Formula (I) is prepared in one step, by covalently attaching the ligands, L, to a linker, X, where FG1 and FG2 represent a functional group such as halo, pseudohalides, boronates, amino, hydroxy, thio, aldehyde, ketone, carboxy, carboxy derivatives such as acid halide, ester, amido, and the like. This method is preferred for preparing compounds of Formula (I) where both the ligands are identical.
  • In method (b), the compounds of Formula (I) are prepared in a stepwise manner by covalently attaching one equivalent of a ligand, L1, with a ligand X where FG1 and FG2 represent a functional group as defined above, and FG2PG is a protected finctional group to give an intermediate of formula (II). Deprotection of the second functional group on the ligand, followed by reaction with a ligand L2, which may be same or different than ligand L1, then provides a compound of Formula (I). This method is suitable for preparing compounds of Formula (I) where the ligands are the non-identical.
  • The ligands are covalently attached to the linker using conventional chemical techniques providing for covalent linkage of the ligand to the linker. 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 as shown in Table I below.
    TABLE I
    Representative Complementary Binding Chemistries
    First Reactive Group Second Reactive Group Linkage
    carboxyl amine amide
    sulfonyl halide amine sulfonamide
    hydroxyl alkyl/aryl halide ether
    hydroxyl isocyanate urethane
    amine epoxide β-hydroxyamine
    amine alkyl/aryl halide alkylamine
    hydroxyl carboxyl ester
    amine aldehyde/NaCNBH3 amine
    hydroxylamine sulfonyl halide sulfonamide
    aldehyde amine/NaCHBH3 amine
    aldehyde amine/NaCHBH3 amine
    amine isocyanate urea
  • By way of example, reaction between a carboxylic acid of either the linker or the β-lactam and a primary or secondary amine of the β-lactam or the linker in the presence of suitable, well-known activating agents such as dicyclohexylcarbodiimide, results in formation of an amide bond covalently linking the β-lactam to the linker, reaction between an amine group of either the linker or the β-lactam and a sulfonylbhalide of the β-lactarn or the linker, in the presence of a base such as triethylamine, pyridine, an the like results in formation of a sulfonamide bond covalently linking the β-lactam to the linker, and reaction between an alcohol or phenol group of either the linker or the β-lactam and an alkyl or aryl halide of the β-lactam or the β-lactarn in the presence of a base such as triethylamine, pyridine, and the like, results in formation of an ether bond covalently linking the β-lactam to the linker.
  • Any-compound which is an antibacterial agent can be used as a ligand in this invention. Typically, a compound selected for use as a ligand will have at least one functional group, such as an amino, 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.
  • Linkers can be attached to different positions on the ligand molecule to achieve different orientations of the ligand domains, and thereby facilitate multivalency. While a number of positions on the ligands are synthetically practical for linking, it is:preferred to preserve those ligand substructures which are most important for ligand-receptor binding.
  • It will be apparent to one skilled in the art that the above chemistries are not limited to preparing multibindingbinding compounds of Formula (I) and can be used to prepare tri-, tetra-, etc., multibinding compounds of Formula (I).
  • The linker is attached to the ligand at a position that retains ligand 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-activity relationships (SAR) of the ligand and/or 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 linker), the univalent linker-ligand conjugate may be tested for retention of activity in the relevant assay.
  • 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 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 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 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 multiple bonds (alkenyl, alkenylene, alikyyl 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 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 (H2N(CH2)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”.
  • Different frameworks can be designed to provide preferred orientations of the ligands. Such frameworks may be represented by using an array of dots (as shown below) wherein each dot may potentially be an atom, such as C, O, N, S, P, H, F, Cl, Br, and F or the dot may alternatively indicate the absence of an atom at that position. To facilitate the understanding of the framework structure, the framework is illustrated as a two dimensional array in the following diagram, although clearly the framework is a three dimensional array in practice:
    Figure US20070134729A1-20070614-C00161
  • Each dot is either an atom, chosen from carbon, hydrogen, oxygen, nitrogen, sulfur, phosphorus, or halogen, or the dot represents a point in space (i.e., an absence of an atom). As is apparent to the skilled artisan, only certain atoms on the grid have the ability to act as an attachment point for the ligands, namely, C, O, N, S and P.
  • Atoms can be connected to each other via bonds (single, double or triple bonds with acceptable resonance and tautomeric forms), with regard to the usual constraints of chemical bonding. Ligands may be attached to the framework via single, double or triple bonds (with chemically acceptable tautomeric and resonance forms). Multiple ligand groups (2 to 10) can be attached to the framework such that the minimal, shortest path distance between adjacent ligand groups does not exceed 100 atoms. Preferably, the linker connections to the ligand is selected such that the maximum spatial distance between two adjacent ligands is no more than 100 Å.
  • An example of a linker as presented by the grid is shown below for a biphenyl construct.
    Figure US20070134729A1-20070614-C00162
  • Nodes (1,2), (2,0), (4,4), (5,2), (4,0), (6,2), (7,4), (9,4), (10,2), (9,0), (7,0) all represent carbon atoms. Node (10,0) represents a chlorine atom. All other nodes (or dots) are points in space (i.e., represent an absence of atoms).
  • Nodes (1,2) and (9,4) are attachment points. Hydrogen atoms are affixed to nodes (2,4), (4,4), (4,0), (2,0), (7,4), (10,2) and (7,0). Nodes (5,2) and (6,2) are connected single bond.
  • The carbon atoms present are connected by either a single or double bonds, taking into consideration the principle of resonance and/or tautomerism.
  • 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 SNN PNN
    CNO NNO ONO SNO PNO
    CNS NNS ONS SNS PNS
    CNP NNP ONP SNP PNP
    COC NOC OOC SOC POC
    COO NON OON SON PON
    COC NOO OOO SOO POO
    COP NOP OOS SOS POS
    OOP SOP POP
    CSC NSC
    CSN NSN OSC SSC PSC
    CSO NSO OSN SSN PSN
    CSS NSS OSO SSO PSO
    CSP NSP OSS SSS PSS
    OSP SSP PSP
    CPC NPC
    CPN NPN OPC SPC PPC
    CPO NPO OPN SPN PPN
    CPS NPS OPO SPO PPO
    CPP NPP OPS SPS PPS
    OPP SPP PPP
  • 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 Edition, Wiley-Interscience, New York, N.Y. (1992). These arrangements are described in the grid of dots shown in the scheme above. 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.
  • Examples. of molecular structures in which the above bonding patterns could be employed as components of the linker are shown below.
    Figure US20070134729A1-20070614-C00163

    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. 1 illustrates a usefuil 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. 1 , di splay vectors around similar central core structures such as a phenyl structure (Panel A) and a cyclohexane structure (Panel B) 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. 2) and compounds of higher valency (FIGS. 3 and 4).
  • 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 flexibility, antigenicity, stability, and the like. For example, the introduction of one or more poly(ethylene 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 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 use ancillary groups such as, for example, small repeating units of ethylene glycols, alcohols, polyols (e.g., glycerin, glycerol propoxylate, saccharides, including mono-, oligosaccharides, etc.), carboxylates (e.g., small repeating units of glutamic acid, acrylic acid, etc.), arnines (e.g., tetraethylenepentamine), and the like) to enhance the water solubility and/or hydrophilicity of the multibinding 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 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 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 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. Hydrophilic characteristics derive from the presence of phosphate, carboxylic, sulfate, amino, sulfhydryl, nitro 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 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, dipalmitoylphosphatidylcholine, dioleoylphosphatidylcholirne, 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 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 linker and the ancillary group(s) 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 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 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, 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.
  • 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.
  • 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 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, poly(ethylene glycol).
  • As explained above, the multibinding compounds described herein comprise 2-10 ligands attached to a linker-that attaches the ligands in such a manner that they are presented to the enzyme 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 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 which ligands are attached. Thus, it should be recognized that the ligands can 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 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. 1 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 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 (L)3X, 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 FIGS. 2 and 3 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
    in a branched array, e.g.,
    Figure US20070134729A1-20070614-C00164

    (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.,
    Figure US20070134729A1-20070614-C00165

    where X and L are as defined herein. Alternatively, it could be represented as an 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. 4 where, as before, the 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 multi-ring linker (e.g., biphenyl) could accommodate a larger number of ligands.
  • Certain of the above described compounds may alternatively be represented as cyclic chains of the form:
    Figure US20070134729A1-20070614-C00166

    and variants thereof
  • All of the above variations are intended to be within the scope of the invention defined by the Formula (L)p(X)q.
  • 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 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.
  • Combinatorial Libraries
  • The methods described above lend themselves to combinatorial approaches for identifying multimeric compounds which possess multibinding 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 multibinding compound with its target(s) 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: (I) the identity of ligand(s), (2) the orientation of ligands, (3) the valency of the 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 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:
  • Selection of ligand(s):
  • 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. 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, log P, 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.
  • Orientation: Selection of Ligand Attachment Points and Linking Chemistry
  • Several points are chosen on each ligand 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 receptor(s) in multiple relative orientations, an important multibinding design parameter. The only requirement for choosing attachment points is that attaching to at least 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 protease inhibitor bound to its target allows one to identify one or more sites where linker attachment will not preclude the enzyme:inhibitor interaction. Alternatively, evaluation of ligand/target binding by nuclear magnetic resonance will permit the identification of sites non-essential for ligand/target binding. See, for example, Fesik, et al., U.S. Pat. No. 5,891,643. When such structural information is not available, utilization of structure-activity relationships (SAR) for ligands will suggest positions where 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 activity. This selection derives from, for example, heterobivalent interactions within the context of a single target molecule. For example, consider a receptor antagonist ligand bound to its target receptor, 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 receptor molecule at sites proximal to the antagonist binding site, which include elements of the receptor that are not part of the formal antagonist binding site and/or elements of the matrix surrounding the receptor such as the membrane. Here, the most favorable orientation for interaction of the second ligand molecule with the receptor/matrix may be achieved by attaching it to the linker at a position which abrogates activity of the ligand at the formal antagonist binding site. Another way to consider 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 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. For example, a 5HT4 receptor antagonist and a bladder-selective muscarinic M3 antagonist may be joined to a linker through attachment points which do not abrogate the binding affinity of the monomeric ligands for their respective receptor sites. The dimeric compound may achieve enhanced affinity for both receptors due to favorable interactions between the 5HT4 ligand and elements of the M3 receptor proximal to the formal M3 antagonist binding site and between the M3 ligand and elements of the 5HT4 receptor proximal to the formal 5HT4 antagonist binding site. Thus, the dimeric compound may be more potent and selective antagonist of overactive bladder and a superior therapy for urinary urge incontinence.
  • 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, stable and intrinsically inocuous under typical chemical and physiological conditions, and compatible with a large number of available linkers. Amide bonds, ethers, amines, carbamates, ureas, and sulfonamides are but a few examples of preferred linkages.
  • Linkers: Spanning Relevant Multibinding Parameters Through Selection of Valency, Linker Length, Linker Geometry, Rigidity, Physical Properties, and Chemical Functional Groups
  • 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:
  • 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 Lengh
  • 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, typically enzymes and soluble receptor targets. In other instances where high-resolution structural information is not available (such as 7TM G-protein coupled receptors), 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 Å, with more preferred linker distances of 3-12 Å. In situations where two binding sites reside on separate (e.g., protein) target sites, preferred linker distances are 20-100 Å, with more preferred distances of 30-70 Å.
  • Linker 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 cds- or trans-arrangements at the 1,1- vs. 1,2- vs. 1,3- vs. 1,4- positions around a cyclohexane core or in cis- or trans-arrangements at a 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.
  • Linker Physical Properties:
  • 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.
  • Linker Chemical Functional Groups:
  • 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.
  • Combinatorial Synthesis
  • 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 (B 1, B2, B3) joined in all possible combinations provide for at least 15 possible combinations of multibinding compounds:
    A1—A1 A1-A2 A1-B1 A1-B2 A1-B3 A2—A2 A2-B1 A2-B2
    A2-B3 B1—B1 B1-B2 B1-B3 B2—B2 B2-B3 B3—B3
  • When each of these combinations is joined by 10 different linkers, a library of 150 candidate multibinding compounds results.
  • Given the combinatorial nature of the library, common chemistries are preferably used to join the reactive functionalities on the ligands with complementary reactive functionalities on the linkers. The library therefore lends 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, for example, chromatographic methods (e.g., HPLC).
  • Analysis of Array by Biochemical, Analytical, Pharmacological, and Computational Methods:
  • Various methods are used to characterize the properties and activities of the candidate multibinding compounds in the library to determine which compounds possess multibinding properties. Physical constants such as solubility under various solvent conditions and logD/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 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 determined. Pharmacological data, including oral absorption, 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.
  • 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 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. Pat. 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 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 Jul. 11, 1998. Such methods couple frontal affinity 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.
  • Follow-up Synthesis and Analysis of Additional Array(s):
  • 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 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 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.
  • To further elaborate upon this procedure, suitable divalent linkers include, by way of example only, those derived from dicarboxylic acids, disulfonylhalides, dialdehydes, diketones, dihalides, diisocyanates,diamines, diols, mixtures of carboxylic acids, sulfonylhalides, aldehydes, ketones, halides, isocyanates, amines and diols. In each case, the carboxylic acid, sulfonylhalide, 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:
  • Complementary Binding Chemistries
  • First Reactive Group Second Reactive Group Linkage
    carboxyl amine amide
    sulfonyl halide amine sulfonamide
    hydroxyl alkyl/aryl halide ether
    hydroxyl isocyanate urethane
    amine epoxide β-hydroxyamine
    amine alkyl/aryl halide alkylamine
    hydroxyl carboxyl ester
    amine aldehyde/NaCNBH3 amine
    hydroxylamine sulfonyl halide sulfonamide
    aldehyde amine/NaCHBH3 amine
    aldehyde amine/NaCHBH3 amine
    amine isocynate urea
  • The following table illustrates, by way of examples, starting materials (identified as X-1 through X-418) that can be used to prepare linkers incorporated in the multibinding compounds of this invention utilizing the chemistry described above. For example, 1,10-decanedicarboxylic acid, X1, can be reacted with 2 equivalents of a ligand carrying an amino group in the presence of a coupling reagent such as DCC to provide a multibinding compound of formula (I) wherein the ligands are linked via 1,10-decanediamido linking group.
    Figure US20070134729A1-20070614-C00167
    Figure US20070134729A1-20070614-C00168
    Figure US20070134729A1-20070614-C00169
    Figure US20070134729A1-20070614-C00170
    Figure US20070134729A1-20070614-C00171
    Figure US20070134729A1-20070614-C00172
    Figure US20070134729A1-20070614-C00173
    Figure US20070134729A1-20070614-C00174
    Figure US20070134729A1-20070614-C00175
    Figure US20070134729A1-20070614-C00176
    Figure US20070134729A1-20070614-C00177
    Figure US20070134729A1-20070614-C00178
    Figure US20070134729A1-20070614-C00179
    Figure US20070134729A1-20070614-C00180
    Figure US20070134729A1-20070614-C00181
    Figure US20070134729A1-20070614-C00182
    Figure US20070134729A1-20070614-C00183
    Figure US20070134729A1-20070614-C00184
    Figure US20070134729A1-20070614-C00185
    Figure US20070134729A1-20070614-C00186
    Figure US20070134729A1-20070614-C00187
    Figure US20070134729A1-20070614-C00188
    Figure US20070134729A1-20070614-C00189
    Figure US20070134729A1-20070614-C00190
    Figure US20070134729A1-20070614-C00191
    Figure US20070134729A1-20070614-C00192
    Figure US20070134729A1-20070614-C00193
    Figure US20070134729A1-20070614-C00194
  • For a multibinding compounds of the Invention, beta lactam antibiotic ligands represented as L1, for use in this invention include, by way of example, L1-1 through L1-5, the ligands L1-1 through L1-5 having been selected from the compounds of formula (a)-(e) disclosed in the Summary of the invention: compound (a) (L1-1), compound (b) (L1-2), compound (c) (L1-3), compound (d) (L1-4) and compound (e) (L1-5).
  • The glycopeptide ligands represented as L2 for use in this invention include, by way of example, L2-1 through L2-2: L2 -1 being an optionally substituted vancomycin and L2-2 being an aglycone derivative of an optionally substituted vancomycin.
  • Combinations of-ligands (L1 and L2) and linkers (X) per this invention include, by way example only, heterodimers wherein a first ligand, L1, selected from L1-1 through L1-5 above, and a second ligand, L2, and a linker, X, are selected from the following:
    L2-1/X-1- L2-1/X-2- L2-1/X-3- L2-1/X-4- L2-1/X-5- L2-1/X-6-
    L2-1/X-7- L2-1/X-8- L2-1/X-9- L2-1/X-10- L2-1/X-11- L2-1/X-12-
    L2-1/X-13- L2-1/X-14- L2-1/X-15- Lz-1/X-16- L2-1/X-17- L2-1/X-18-
    L2-1/X-19- L2-1/X-20- L2-1/X-21- L2-1/X-22- L2-1/X-23- L2-1/X-24-
    L2-1/X-25- L2-1/X-26- L2-1/X-27- L2-1/X-28- L2-1/X-29- L2-1/X-30-
    L2-1/X-31- L2-1/X-32- L2-1/X-33- L2-1/X-34- L2-1/X-35- L2-1/X-36-
    L2-1/X-37- L2-1/X-38- L2-1/X-39- L2-1/X-40- L2-1/X-41- L2-1/X-42-
    L2-1/X-43- L2-1/X-44- L2-1/X-45- L2-1/X-46- L2-1/X-47- L2-1/X-48-
    L2-1/X-49- L2-1/X-50- L2-1/X-51- L2-1/X-52- L2-1/X-53- L2-1/X-54-
    L2-1/X-55- L2-1/X-56- L2-1/X-57- L2-1/X-58- L2-1/X-59- L2-1/X-60-
    L2-1/X-61- L2-1/X-62- L2-1/X-63- L2-1/X-64- L2-1/X-65- L2-1/X-66-
    L2-1/X-67- L2-1/X-68- L2-1/X-69- L2-1/X-70- L2-1/X-71- L2-1/X-72-
    L2-1/X-73- L2-1/X-74- L2-1/X-75- L2-1/X-76- L2-1/X-77- L2-1/X-78-
    L2-1/X-79- L2-1/X-80- L2-1/X-81- L2-1/X-82- L2-1/X-83- L2-1/X-84-
    L2-1/X-85- L2-1/X-86- L2-1/X-87- L2-1/X-88- L2-1/X-89- L2-1/X-90-
    L2-1/X-91- L2-1/X-92- L2-1/X-93- L2-1/X-94- L2-1/X-95- L2-1/X-96-
    L2-1/X-97- L2-1/X-98- L2-1/X-99- L2-1/X-100- L2-1/X-101- L2-1/X-102-
    L2-1/X-103- L2-1/X-104- L2-1/X-105- L2-1/X-106- L2-1/X-107- L2-1/X-108-
    L2-1/X-109- L2-1/X-110- L2-1/X-111- L2-1/X-112- L2-1/X-113- L2-1/X-114-
    L2-1/X-115- L2-1/X-116- L2-1/X-117- L2-1/X-118- L2-1/X-119- L2-1/X-120-
    L2-1/X-121- L2-1/X-122- L2-1/X-123- L2-1/X-124- L2-1/X-125- L2-1/X-126-
    L2-1/X-127- L2-1/X-128- L2-1/X-129- L2-1/X-130- L2-1/X-131- L2-1/X-132-
    L2-1/X-133- L2-1/X-134- L2-1/X-135- L2-1/X-136- L2-1/X-137- L2-1/X-138-
    L2-1/X-139- L2-1/X-140- L2-1/X-141- L2-1/X-142- L2-1/X-143- L2-1/X-144-
    L2-1/X-145- L2-1/X-146- L2-1/X-147- L2-1/X-148- L2-1/X-149- L2-1/X-150-
    L2-1/X-151- L2-1/X-152- L2-1/X-153- L2-1/X-154- L2-1/X-155- L2-1/X-156-
    L2-1/X-157- L2-1/X-158- L2-1/X-159- L2-1/X-160- L2-1/X-161- L2-1/X-162-
    L2-1/X-163- L2-1/X-164- L2-1/X-165- L2-1/X-166- L2-1/X-167- L2-1/X-168-
    L2-1/X-169- L2-1/X-170- L2-1/X-171- L2-1/X-172-
    L2-1/X-173- L2-1/X-174- L2-1/X-175- L2-1/X-176- L2-1/X-177- L2-1/X-178-
    L2-1/X-179- L2-1/X-180- L2-1/X-181- L2-1/X-182- L2-1/X-183- L2-1/X-184-
    L2-1/X-185- L2-1/X-186- L2-1/X-187- L2-1/X-188- L2-1/X-189- L2-1/X-190-
    L2-1/X-191- L2-1/X-192- L2-1/X-193- L2-1/X-194- L2-1/X-195- L2-1/X-196-
    L2-1/X-197- L2-1/X-198- L2-1/X-199- L2-1/X-200- L2-1/X-201- L2-1/X-202-
    L2-1/X-203- L2-1/X-204- L2-1/X-205- L2-1/X-206- L2-1/X-207- L2-1/X-208-
    L2-1/X-209- L2-1/X-210- L2-1/X-211- L2-1/X-212- L2-1/X-213- L2-1/X-214-
    L2-1/X-215- L2-1/X-216- L2-1/X-217- L2-1/X-218- L2-1/X-219- L2-1/X-220-
    L2-1/X-221- L2-1/X-222- L2-1/X-223- L2-1/X-224- L2-1/X-225- L2-1/X-226-
    L2-1/X-227- L2-1/X-228- L2-1/X-229- L2-1/X-230- L2-1/X-231- L2-1/X-232-
    L2-1/X-233- L2-1/X-234- L2-1/X-235- L2-1/X-236- L2-1/X-237- L2-1/X-238-
    L2-1/X-239- L2-1/X-240- L2-1/X-241- L2-1/X-242- L2-1/X-243- L2-1/X-244-
    L2-1/X-245- L2-1/X-246- L2-1/X-247- L2-1/X-248- L2-1/X-249- L2-1/X-250-
    L2-1/X-251- L2-1/X-252- L2-1/X-253- L2-1/X-254- L2-1/X-255- L2-1/X-256-
    L2-1/X-257- L2-1/X-258- L2-1/X-259- L2-1/X-260- L2-1/X-261- L2-1/X-262-
    L2-1/X-263- L2-1/X-264- L2-1/X-265- L2-1/X-266- L2-1/X-267- L2-1/X-268-
    L2-1/X-269- L2-1/X-270- L2-1/X-271- L2-1/X-272- L2-1/X-273- L2-1/X-274-
    L2-1/X-275- L2-1/X-276- L2-1/X-277- L2-1/X-278- L2-1/X-279- L2-1/X-280-
    L2-1/X-281- L2-1/X-282- L2-1/X-283- L2-1/X-284- L2-1/X-285- L2-1/X-286-
    L2-1/X-287- L2-1/X-288- L2-1/X-289- L2-1/X-290- L2-1/X-291- L2-1/X-292-
    L2-1/X-293- L2-1/X-294- L2-1/X-295- L2-1/X-296- L2-1/X-297- L2-1/X-298-
    L2-1/X-299- L2-1/X-300- L2-1/X-301- L2-1/X-302- L2-1/X-303- L2-1/X-304-
    L2-1/X-305- L2-1/X-306- L2-1/X-307- L2-1/X-308- L2-1/X-309- L2-1/X-310-
    L2-1/X-311- L2-1/X-312- L2-1/X-313- L2-1/X-314- L2-1/X-315- L2-1/X-316-
    L2-1/X-317- L2-1/X-318- L2-1/X-319- L2-1/X-320- L2-1/X-321- L2-1/X-322-
    L2-1/X-323- L2-1/X-324- L2-1/X-325- L2-1/X-326- L2-1/X-327- L2-1/X-328-
    L2-1/X-329- L2-1/X-330- L2-1/X-331- L2-1/X-332- L2-1/X-333- L2-1/X-334-
    L2-1/X-335- L2-1/X-336- L2-1/X-337- L2-1/X-338- L2-1/X-339- L2-1/X-340-
    L2-1/X-341- L2-1/X-342- L2-1/X-343- L2-1/X-344- L2-1/X-345- L2-1/X-346-
    L2-1/X-347- L2-1/X-348- L2-1/X-349- L2-1/X-350- L2-1/X-351- L2-1/X-352-
    L2-1/X-353- L2-1/X-354- L2-1/X-355- L2-1/X-356- L2-1/X-357- L2-1/X-358-
    L2-1/X-359- L2-1/X-360- L2-1/X-361- L2-1/X-362- L2-1/X-363- L2-1/X-364-
    L2-1/X-365- L2-1/X-366- L2-1/X-367- L2-1/X-368- L2-1/X-369- L2-1/X-370-
    L2-1/X-371- L2-1/X-372- L2-1/X-373- L2-1/X-374- L2-1/X-375- L2-1/X-376-
    L2-1/X-377- L2-1/X-378- L2-1/X-379- L2-1/X-380- L2-1/X-381- L2-1/X-382-
    L2-1/X-383- L2-1/X-384- L2-1/X-385- L2-1/X-386- L2-1/X-387- L2-1/X-388-
    L2-1/X-389- L2-1/X-390- L2-1/X-391- L2-1/X-392- L2-1/X-393- L2-1/X-394-
    L2-1/X-395- L2-1/X-396- L2-1/X-397- L2-1/X-398- L2-1/X-399- L2-1/X-400-
    L2-1/X-401- L2-1/X-402- L2-1/X-403- L2-1/X-404- L2-1/X-405- L2-1/X-406-
    L2-1/X-407- L2-1/X-408- L2-1/X-409- L2-1/X-410- L2-1/X-411- L2-1/X-412-
    L2-1/X-413- L2-1/X-414- L2-1/X-415- L2-1/X-416- L2-1/X-417- L2-1/X-418-

    and so on, substituting L2-2.
  • Utility, Testing, and Administration Utility
  • The compounds of the invention, and their pharmaceutically acceptable salts, are useful in medical treatments and exhibit biological activity, including antibacterial activity, which can be demonstrated in the tests described in the Examples. The antibacterial activity of the instant compounds may be determined by testing in standardized in vitro dilution tests for minimum inhibitory concentration (MICs). Such tests are well known to those skilled in the art, and are referenced and described in the fourth edition of “Antibiotics in Laboratory Medicine”, by Victor Lorian, M. D., published by Williams and Wilkins, which is hereby incorporated by reference. Using such standard microbiological procedures, the compounds of this invention will be found to exhibit activity against gram-positive and gram-negative bacteria such as Staphylococcus aureus, Escherichia coli and Pseudomonas aeruginosa at test levels.
  • The compounds of the present invention are useful in the treatment in mammals of bacterial infections, by both gram-positive and gram-negative bacteria. The compounds may be administered to the mammals in the form of a pharmaceutical composition comprising the compounds of the invention admixed with a pharmaceutically acceptable excipient.
  • Pharmaceutical Formulations
  • 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, transdermnal, subcutaneous, intravenous, intramuscular, and intranasal. These compounds are effective as injectable intranasal and oral compositions. Such compositions are prepared in a manner well known in the pharmaceutical art and comprise at least one active compound.
  • 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 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), 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 30 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 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).
  • 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.
  • 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 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 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 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.
  • EXAMPLES
  • The following preparations and examples are given to enable those skilled in the art to more clearly understand and to practice the present invention. They should not be considered as limiting the scope of the invention, but merely as being illustrative and representative thereof.
  • In the examples below, the following abbreviations have the following meanings. Unless otherwise stated, all temperatures are in degrees Celsius. If an abbreviation is not defined, it has its generally accepted meaning.
  • Å=Angstroms
  • cm=centimeter
  • DCC=dicyclohexyl carbodiimide
  • DMF=N,N-dimethylformamide
  • DMSO=dimethylsulfoxide
  • g=gram
  • HPLC=high performance liquid chromatography
  • mg=milligram
  • min=minute
  • mL=milliliter
  • mm=millimeter
  • mmol=millimol
  • N=normal
  • THF=tetrahydroftiran
  • μL=microliters
  • μm=microns
  • Synthetic Examples Example 1 Synthesis of Amoxicillin Homodimer (Following FIG. 11)
  • Step 1
  • A slurry of (D)-4-hydroxyphenyl glycine 1 (10 mmol) in methanol (100 mL) is stirred with cooling in an ice bath. Thionyl chloride (11 mmol) is added dropwise over the course of 15 minutes. After addition is complete, the mixture is allowed to stir in the cooling bath for an additional 2 hours. The mixture is then concentrated to dryness to afford crude (D)-4-hydroxyphenyl glycine methyl ester hydrochloride. This material is dissolved and stirred in 100 mL dimethylformamide and treated sequentially with diisopropylethyl amine (22 mmol) followed by allyl 1-benzotriazolyl carbonate (11 mmol). After stirring 1 hour at room temperature, volatiles are removed under reduced pressure and the residue is fractionated by silica gel chromatography using ethyl acetate/hexane eluent to afford Aloc-protected methyl ester 2.
  • A solution of compound 2 (6.0 mmol) and tetraethylene glycol (3.0 mmol) in anhydrous tetrahydrofuran (25 mL) is stirred at room temperature under nitrogen and then treated sequentially with triphenylphosphine (9.0 mmol) and diethylazodicarboxylate (6.6 mmol). The reaction is stirred for 4 hours and then concentrated under reduced pressure. The crude is fractionated by silica gel chromatography using ethyl acetate/hexane eluent to afford 3.
  • Step 3
  • A solution of compound 3 (2.0 mmol) in 20 mL methanol is treated with a solution of lithium hydroxide (5 mmol) in 2 mL water. The reaction is stirred at room temperature for 2 hours and then poured into 100 mL 1 N sodium hydrogen sulfate solution and extracted with ethyl acetate. The organic extract is dried over anhydrous sodium sulfate, filtered, and then concentrated to dryness under vacuum to afford crude diacid 4 which is used without further purification.
  • Step 4
  • Diacid 4 is dissolved in 10 mL anhydrous dimethylformamide and treated sequentially with hydroxybenzotriazole (5.0 mmol), diisopropylethyl amine (4.0 mmol) and PyBOP (4.0 mmol). After stirring for 15 minutes at room temperature, the activated diacid is treated with (+)-6-aminopenicillanic acid 5 (4.0 mmol) and the coupling reaction is stirred overnight at room temperature. Volatiles are removed under vacuum and the crude is fractionated by reverse-phase HPLC using a linear gradient of acetonitrile in water (both buffered with 0.1% trifluoracetic acid) to afford 6 after lyopholization of the appropriate fractions.
  • Step 5
  • Diacid 6 (1.0 mmol) is dissolved in 10 mL anhydrous tetrahydrofuran and stirred under nitrogen at room temperature and treated sequentially with pyrrolidine (3.0 mmol) and tetrakis(triphenylphosphine)palladium[0] (0.15 mmol). After 2 hours, the mixture is evaporated to dryness and then fractionated by reverse-phase HPLC using a linear gradient of acetonitrile in water (both buffered with 0.1% trifluoracetic acid) to afford the desired amoxicillin dimer 7 after lyopholization of the appropriate fractions.
  • Example 2 Synthesis of Imipenem Homodimer (Following FIG. 12)
  • Step 1
  • 4,4′-Dipiperidine hydrochloride (10 mmol) is dissolved in water (100 mL), stirred at room temperature, and treated sequentially with triethylamine (40 mmol) and 2-iminothiolane hydrochloride (20 mmol). After two hours the reaction mixture is frozen and lyopholized and the diamidine dithiol 8 is recovered as the dihydrochloride after crystallization from HCl/diethyl ether.
  • Step 2
  • Compound 9 (4.0 mmol) is generated in acetonitrile (20 mL) as described (Salzmann et al. J Am. Chem. Soc. 1980, 102, 6163 and Lelillo et al. Tetrahedron Lett. 1980, 21, 2783). This is then treated with compound 8 (2.0 mmol) and diisopropylethyl amine (9.0 mmol) and the reaction is stirred at 0 C for 1 hour. The PNB-protected adduct precipitates from the reaction mixture and is isolated by filtration. This material is then dissolved in a mixture of tetrahydrofuran and water buffered to pH 7.0 with morpholinopropane sulfonic acid, treated with 10% palladium on carbon (200 mg) and subjected to 40 psi H2 for 4 hours. The mixture is filtered through a pad of celite to remove catalyst and chromatographed at 4° C. on a column of Dowex 50×4 (Na+cycle, 200-400 mesh) resin eluted with deionized water. The desired compound 10 is recovered upon lyopholization of the appropriate fractions.
  • Example 3 Synthesis of Imipenem Homodimer (Following FIG. 13)
  • Thienamycin 11 (2.0 numol) is dissolved in aqueous buffer (morpholinopropane sulfonic acid, pH 8.2) and stirred in an ice/water bath. Dimethyloctanediimidate dihydrochloride 12 (1.0 mmol) is added as a solid and the reaction is stirred one hour in the cooling bath and then two hours at room temperature. The mixture is then chromatographed at 4° C. on a column of Dowex 50×4 (Na+cycle, 200-400 mesh) resin eluted with deionized water. The desired compound 13 is recovered upon lyopholization of the appropriate fractions.
  • Example 4 Synthesis of Vancomycin-Amoxicillin Heterodimer (Following FIG. 14)
  • Method A
  • Step 1
  • A slurry of (D)-4-hydroxyphenyl glycine 1 (10 mmol) in methanol (100 mL) is stirred with cooling in an ice bath. Thionyl chloride (11 mmol) is added dropwise over the course of 15 minutes. After addition is complete, the mixture is allowed to stir in the cooling bath for an additional 2 hours. The mixture is then concentrated to dryness to afford crude (D)-4-hydroxyphenyl glycine methyl ester hydrochloride. This material is dissolved and stirred in 100 mL dimethylformamide and treated sequentially with diisopropylethyl amine (22 mmol) followed by allyl 1-benzotriazolyl carbonate (11 mmol). After stirring I hour at room temperature, volatiles are removed under reduced pressure and the residue is fractionated by silica gel chromatography using ethyl acetate/hexane eluent to afford Aloc-protected (D)-4-hydroxyphenyl glycine methyl ester. The ester (7.0 mmol) is dissolved in methanol (40 mL, stirred at room temperature, and treated with a solution of lithium hydroxide (8.0 mmol) in 20 mL water. The reaction is stirred at room temperature for 2 hours and then poured into 100 mL 1 N sodium hydrogen sulfate solution and extracted with ethyl acetate. The organic extract is dried over anhydrous sodium sulfate, filtered, and then concentrated under reduced pressure. The crude is fractionated via chromatography on silica gel using methanol/methylene chloride/trifluoracetic acid eluent to afford N-Aloc (D)-4-hydroxyphenyl glycine 14.
  • Step 2
  • Compound 14 (5.0 mmol) is dissolved in anhydrous dimethylformamide (20 mL), stirred at room temperature, and treated sequentially with hydroxybenzotriazole (5.0 mmol), diisopropylethyl amine (5.0 mmol) and PyBOP (5.0 mmol). After stirring for 15 minutes at room temperature, the activated acid is treated with (+)-6-aminopenicillanic acid (5.0 mmol) and the coupling reaction is stirred overnight at room temperature. The mixture is then treated with allyl bromide (5.0 mmol) and stirred for an additional 24 hours. Volatiles are removed under vacuum and the crude is fractionated by silica gel chromatography using methanol/methylene chloride eluent to afford N-Aloc (D)-4-hydroxyphenyl glycine allyl ester 16.
  • Step 3
  • Compound 16 (1.0 mmol) is dissolved in anhydrous dimethylformamide (5.0 mL), stirred in an ice/water bath, and treated sequentially with N,N-dimethylaminopyridine (0.1 mmol) and carbonyldiimidazole (1.0 mmol). The ice bath is removed and the reaction mixture is allowed to warm to room temperature. The imidazolide 16 thus produced is used without further manipulation in the coupling reactions described below.
  • Vancomycin-2-aminoethanamide 18 (compound 18 prepared as described in Example 5 below, 1.0 mmol) is dissolved in 5.0 mL anhydrous dimethylformamide, stirred at room temperature, and treated sequentially with diisopropylethyl amine (4.0 mmol) and the solution of the imidazolide 16 (prepared in Step 3 above). After 2 hours, volatiles are removed under vacuum and the residue is triturated with acetonitrile. The solid is then redissolved in 10 mL 1:1 anhydrous tetrahydrofliran:anhydrous dimethylformamide, stirred under nitrogen at room temperature, and treated sequentially with pyrrolidine (3.0 mmol) and tetrakis(triphenylphosphine)palladium[0] (0.25 mmol). After 2 hours, the mixture is concentrated under vacuum and the residue is dissolved in 0.1% aqueous trifluoroacetic acid and fractionated by reverse-phase HPLC using a linear gradient of acetonitrile in water (both buffered with 0.1% trifluoracetic acid) to afford the desired compound 19 upon lyopholization of the appropriate fractions.
  • Method B
  • Step 1
  • Vancomycin hydrochloride 17 (10 mmol) is slurried in 100 mL 1:1 methanol:anhydrous dimethylformamide, stirred at room temperature, and treated sequentially with diisopropylethyl amine (20 mmol) and Fmoc glycinal (prepared as described by Salvi et al. Tetrahedron Lett. 1994, 35, 1181-1184). After 2 hours the reaction mixture is cooled in an ice water bath and treated further with sodium cyanoborohydride (4.0 mmol) and trifluoroacetic acid (30 mmol). After 2 additional hours the crude product is precipitated by dropwise addition to a ten-fold volume of acetonitrile, and then fractionated by reverse-phase HPLC using a linear gradient of acetonitrile in water (both buffered with 0.1% trifluoracetic acid) to afford the adducts reductively alkylated on the N-methylamino terminus 20 and on the N′-amino group of the vancosamine residue 21.
  • Compounds 20 and 21 (2.0 mmol each) are separately dissolved in anhydrous dimethylformamide (10 mL), stirred at room temperature and treated with excess piperidine (1.0 mL). After one hour the crude products are precipitated by dropwise addition to 50 mL acetonitrile with vigorous stirring. The crude products are fractionated by reverse-phase HPLC using a linear gradient of acetonitrile in water (both buffered with 0.1% trifluoracetic acid) to afford the N-aminoethyl adduct 22 and the the N′-aminoethyl adduct 23 upon lyopholization of the appropriate fractions.
  • Compounds 22 and 23 are subsequently elaborated to desired heterobivalent compounds 24 and 25, respectively by following step 4 described for the conversion of compound 18 to compound 19.
  • Example 5 Synthesis of Vancomycin-Imipenem Heterodimer (Following FIG. 15)
  • Step 1
  • Vancomycin hydrochloride 17 (10 mmol) is) is dissolved in water (100 mL), stirred at room temperature, and treated sequentially with triethylamine (40 mmol) and 2-iminothiolane hydrochloride (10 mmol). After two hours the reaction mixture is fractionated by reverse-phase HPLC using a linear gradient of acetonitrile in water (both buffered with 0. 1% trifluoracetic acid) to afford the imine adducts modified on the N-methylamino terminus compound 26 and on the N′-amino group of the vancosamine residue compound 27.
  • Compound 28 (2.0 mmol) is generated in acetonitrile (10 mL) as described (Salzmann et al. J Am. Chem. Soc. 1980, 102, 6163 and Lelillo et al. Tetrahedron Lett. 1980, 21, 2783). This is then treated with a solution of compound 26 (2.0 mmol) and diisopropylethyl amine (11 mmol) in 10 mL anhydrous dimethylformamide and the reaction is stirred at 0 C for 1 hour. After removal of volatiles under vacuum, the crude product is dissolved in a mixture of tetrahydrofuran and water buffered to pH 7.0 with morpholinopropane sulfonic acid, treated with 10% platinum oxide (20 mg) and subjected to 40 psi H2 for 4 hours. The mixture is filtered through a pad of celite to remove catalyst and chromatographed at 4° C. on a column of Dowex 50×4 (Na+cycle, 200-400 mesh) resin eluted with deionized water. The desired compound 29 is recovered upon lyopholization of the appropriate fractions.
  • In a like manner, using adduct 27 in place of 26, compound 30 is prepared.
  • Example 6 Synthesis of Vancomycin-Imipenem Heterodimer (Following FIG. 16)
  • Step 1
  • Vancomycin hydrochloride 17 (10 mmol) is dissolved in 100 mL 1:1 anhydrous dimethylsulfoxide:dimethylformamide, stirred at room temperature, and treated sequentially with ethylenediamine (20 mmol), hydroxybenzotriazole (10 rnmol), and PyBOP (10 mmol). After two hours, the crude product is precipitated by dropwise addition to 1 vigorously stirred acetonitrile, and then fractionated by reverse-phase HPLC using a linear gradient of acetonitrile in water (both buffered with 0.1% trifluoracetic acid) to afford compound 31 after lyopholization of the appropriate fractions.
  • Step 2
  • Compound 31 (5.0 mmol) is) is dissolved in water (50 mL), stirred at room temperature, and treated sequentially with triethylamine (20 mmol) and 2-iminothiolane hydrochloride (5.0 mmol). After two hours the reaction mixture is fractionated by reverse-phase HPLC using a linear gradient of acetonitrile in water (both buffered with 0. 1% trifluoracetic acid) to afford compound 32 after lyopholization of the appropriate fractions.
  • Step 3
  • Compound 28 (2.0 nmmol) is generated in acetonitrile (10 mnL) as described (Salzmann et al. J Am. Chem. Soc. 1980, 102, 6163 and Lelillo et al. Tetrahedron Lett. 1980, 21, 2783). This is then treated with a solution of compound 32 (2.0 mmol) and diisopropylethyl amine (11 mmol) in 10 mL anhydrous dimethylformamide and the reaction is stirred at 0° C. for 1 hour. After removal of volatiles under vacuum, the crude product is dissolved in a mixture of tetrahydrofuran and water buffered to pH 7.0 with morpholinopropane sulfonic acid, treated with 10% platinum oxide (20 mg) and subjected to 40 psi H2 for 4 hours. The mixture is filtered through a pad of celite to remove catalyst and chromatographed at 4° C. on a column of Dowex 50×4 (Na+cycle, 200-400 mesh) resin eluted with deionized water. The desired compound 33 is recovered upon lyopholization of the appropriate fractions.
  • Example 7 Synthesis of Cephaclor Dimer (Following FIG. 17)
  • Step 1
  • A solution of 10 mmols of Cephaclor 34 (commercially available) in methanol (10 mL) is treated to pH 6 with acetic acid. 1,3,5-Trioxane (4 mmols) is then added followed by sodium cyanoborohydride (6 mmols). When HPLC indicates that the reaction is complete, it is quenched with aqueous acetic acid (keeping the pH 6-6.5) and the solvent removed in vacuo. The crude product is purified by HPLC to afford a compound 35.
  • Step 2
  • A mixture of 35 (4 mmols) in DMF (10 mL) is treated with 2 mmols of 1,8-dibromooctane and the reaction kept at 40° C. until HPLC indicates completion. The solvent removed in vacuo and the crude product is purified by HPLC to afford the desired compound 36.
  • In a similar manner, compound 42 may be prepared in a similar manner from the PNB ester of commercially available Amoxicillin.
  • Example 8 Synthesis of Cephaclor Dimer (Following FIG. 18)
  • A mixture of 35 in (4 mmols) in THF (10 mL) with N-ethyldiisopropylamine (4 mmols) is treated with sebacoyl chloride (2 mmols) and the reaction kept at room temperature until HPLC indicates completion. The solvent removed in vacuo and the crude product is purified by HPLC to afford the desired compound 37.
  • Example 9 Synthesis of Cephaclor-Ampicillin Heterodimer (Following FIG. 19)
  • Step 1
  • A mixture of compound 35 (4 mmols) and compound 38 in DMF (10 mL) is treated with 1,8-dibromooctane (4 mnmols) and the reaction kept at 40° C. until HPLC indicates completion. The solvent removed in vacuo and the crude product is purified by HPLC to afford the desired compound 39.
  • Example 10 Synthesis of Carbapenem-Amoxicillin Heterodimer (Following FIG. 20)
  • A mixture of carbapenem 40 (4 mmols) and 1 ,8-dibromooctane (10 mmols) in DMF (5 mL) is kept at 40° C. until HPLC indicates completion. The solvent removed in vacuo and the crude product is purified by HPLC to afford compound 41.
  • A solution of of 42 (5 mmols) in anhydrous DMF (5 mL) with N-ethyldiisopropylamine (5 mmols) is treated with chlorotrimethylsilane (5 mmols) followed by compound 41 and the reaction stirred at 40° C. until HPLC indicates completion. After addition of water (2 mL) and removal of volatiles under vacuum, the crude product is dissolved in a mixture of THF and water buffered to pH 7.0 with morpholinopropane sulfonic acid, treated with 10% palladium of carbon (200 mg) and subjected to 40 psi H2 for four hours. The mixture is filtered and purified by HPLC to afford the desired compound 43.
  • Example 11 Synthesis of Carbapenem-Amoxicillin Heterodimer (Following FIG. 21)
  • Step 1
  • A solution of compound 44 (10 nmmols) in THF (10 mL) is treated with Boc anhydride (11 mmols) and after lhr. the volatiles are removed under vacuum to afford intermediate 45.
  • Step 2
  • A solution of compound 46 (2 mmols) in acetonitrile ((10 mL) is treated with a solution of intermediate 45 (2 mmols) and N-ethyldiisopropylamine (II mmols) in anhydrous DMF (10 mL) and the reaction stirred at 0° C. for 1 hour. The solvents are removed in vacuo and the crude product is purified by HPLC to afford compound 47.
  • Step 3
  • A solution of commercially available Cefoclor 48 (20 mmols) in THF (25 mL) is treated with Boc anhydride (22 mmols). After 1 hour of p-nitrobenzyl alcohol (22 mmols) is added followed by dicyclohexylcarbodiimide (22 mmols). When complete as indicated by HPLC, trifluoroacetic acid (1 mL) is added and when removal of the Boc group is complete the reaction is filtered and the solvent removed. The residue is purified by chromatography to afford compound 49.
  • Step 4
  • A solution of intermediate 49 (4 mmols) and N-Boc-8-aminooctanoic acid (4 mmols) in anhydrous DMF (10 mL) is cooled under N2 with stirring in an ice-water bath. To the stirred solution is added 1-hydroxybenzotriazole (7 mmols) followed by 1-(3-dimethylaminopropyl)-3-ethylcarbodiinide hydrochloride (5.5 mmols). The cooling bath is removed and the reaction followed by TLC. When complete, the mixture is partitioned between water and ethyl acetate and the aqueous phase back extracted with ethyl acetate. The combined organic extracts are washed with water followed by sat sodium carbonate, dried over sodium sulfate and the solvent removed in vacuo. The crude product is purified by chromatography to afford compound 50.
  • Step 5
  • A solution of compound 50 (4 rnmols) in methylene chloride (5 mL) is treated with trifluoroacetic acid (0.5 mL) and when Boc removal is complete, washed with aqueous sodium bicarbonate and water, dried over sodium sulfate and the solvent removed. The product is dissolved in anhydrous DMF (10 mL) and compound 47 (4 mmols) is added. The solution is cooled under N2 with stirring in an ice-water bath. To the stirred solution is added 1-30 hydroxybenzotriazole (7 mmols) followed by 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (5.5 mmols). The cooling bath is removed and the reaction followed by TLC. When the reaction is complete, the mixture is partitioned between water and ethyl acetate and the aqueous phase back extracted with ethyl acetate. The combined organic extracts are washed with water followed by sat. sodium bicarbonate, dried over sodium sulfate and the solvent removed in vacuo. The crude product is purified by chromatography and dissolved in a mixture of THF and water buffered to pH 7.0 with morpholinopropane sulfonic acid. Add 10% platinum oxide (20 mg) and subject the reaction mixture to 40 psi H2 for four hours. The reaction mixture is filtered and purified by chromatography. The product is dissolved in a mixture of THF (5 mL) and trifluoroacetic acid (0.5 mL). After the reaction is complete volatiles are removed under vacuum and the crude product is purified by HPLC to afford the desired compound 51.
  • Example 12 Synthesis of Ceftazidime Homodimer (Following FIG. 22)
  • Step 1
  • To chloromethylcephalosporonic acid chloride 52 (32.0 g, 74 mmol) in a 1:1 mixture of acetonitrile and dimethylformamide (300 mL) was added compound 53 (36.9, 156 mmol). After 4 h, the crude product was precipitated by diluting the solution with ether (800 mL). The solid was filtered and purified by reverse phase HPLC to give compound 54.
  • Step 2
  • A mixture of compound 54 (7.1 g, 9.5 mmol) and anisole (3 mL) in trifluoroacetic acid (30 mL) was stirred for 20 min. The desired product 55 was precipitated out by diluting the solution with ether (600 mL), then filtered and dried.
  • Step 3
  • To a solution of hexadecanedioic acid (12 mg, 0.05 rnmol) in dimethylformamide (0.3 mL) was added HATU (40 mg, 0.11 mmol). After 1 h, compound 55 (75 mg, 0.10 mmol) was added and stirring was continued. After 4 h, 0.5% aqueous trifluoroacetic acid (0.5 mL) was added and the reaction mixture was purified by reverse phase HPLC to give the desired compound 56.
  • Example 13 Synthesis of Cefoperazone Homodimer (Following FIG. 23)
  • Step 1
  • To a solution of cefoperazone sodium salt (10.0 g) in water (100 mL) was added 6 N hydrochloric acid until the pH of the solution was approximately 2. The white precipitates were filtered off to give compound 57.
  • Step 2
  • Compound 57 (1.1 g, 1.71 mmol) and sodium bicarbonate (0.16 g, 1.88 mmol) were combined and then a solution of p-methoxybenzyl bromide (0.51 g, 2.56 mmol) in dimethylformamide/dioxane (5 ml/3 ml) was added. The reaction mixture was allowed to stir overnight and then concentrated. Ethyl acetate was added and the organic layer was washed with saturated sodium bicarbonate, brine, dried over magnesium sulfate and filtered. The filtrate was concentrated to give compound 58.
  • Step 3
  • To a solution of compound 58 (21.8 mg, 28.5 mmol) in acetonitrile (145 mL) was added diisopropylethylamine (5.96 mL, 34.2 mmol), followed by p-nitrophenyl chloroformate (5.8 g, 34.2 mmol). After I h, the reaction mixture was concentrated and the crude product 59 was used in the next step without further purification.
  • Step 4
  • To a solution of compound 59 (25.0 g, 26.9 mmol) in dichloromethane (120 mL) was added a 1:1 mixture of trifluoroacetic acid and anisole (80 mL). After 1 h, the reaction mixture was concentrated and the residue was dissolved in a minimum amount of acetonitrile and cold ether was added. The desired product 60 was isolated as a yellow solid.
  • Step 5
  • To a solution of compound 60 (0.162 g, 0.20 mmol) in dimethylformamide A(0.20 mL) was added diisopropylethylamine (0.034 mL, 0.20 mL) and 1,9-diaminononane (16 mg, 0.10 mmol) and the reaction mixture was stirred at room temperature. After 4 h, the reaction mixture was diluted with dimethylformamide (0.80 mL) and the desired product 61 was isolate using preparative HPLC.
  • Formulation Examples Example 1
  • Hard gelatin capsules containing the following ingredients are prepared:
    Quantity
    Ingredient (mg/capsule)
    Active Ingredient 30.0
    Starch 305.0
    Magnesium stearate 5.0
  • The above ingredients are mixed and filled into hard gelatin capsules in 340 mg quantities.
  • Example 2
  • A tablet Formula is prepared using the ingredients below:
    Quantity
    Ingredient (mg/tablet)
    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.
  • Example 3
  • A dry powder inhaler formulation is prepared containing the following components:
    Ingredient Weight %
    Active Ingredient 5
    Lactose 95
  • The active ingredient is mixed with the lactose and the mixture is added to a dry powder inhaling appliance.
  • Example 4
  • Tablets, each containing 30 mg of active ingredient, are prepared as follows:
    Quantity
    Ingredient (mg/tablet)
    Active Ingredient 30.0 mg 
    Starch 45.0 mg 
    Micro-crystalline cellulose 35.0 mg 
    Polyvinylpyrrolidone 4.0 mg
    (as 10% solution in sterile water)
    Sodium carboxymethyl starch 4.5 mg
    Magnesium stearate 0.5 mg
    Talc 1.0 mg
    Total 120 mg 
  • 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 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.
  • Example 5
  • Capsules, each containing 40 mg of medicament are made as follows:
    Quantity
    Ingredient (mg/capsule)
    Active Ingredient  40.0 mg
    Starch 109.0 mg
    Magnesium stearate  1.0 mg
    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 quantities.
  • Example 6
  • Suppositories, each containing 25 mg of active ingredient are made as follows:
    Ingredient Amount
    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.
  • Example 7
  • Suspensions, each containing 50 mg of medicament per 5.0 mL dose are made as follows:
    Ingredient Amount
    Active Ingredient 50.0 mg
    Xanthan gum 4.0 mg
    Sodium carboxymethyl cellulose (11%)
    Microcrystalline cellulose (89%) 50.0 mg
    Sucrose 1.75 g
    Sodium benzoate 10.0 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.
  • Example 8
  • A formulation may be prepared as follows:
    Quantity
    Ingredient (mg/capsule)
    Active Ingredient  15.0 mg
    Starch 407.0 mg
    Magnesium stearate  3.0 mg
    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.
  • Examlple 9
  • A formulation may be prepared as follows:
    Ingredient Quantity
    Active Ingredient 5.0 mg
    Corn Oil 1.0 mL
  • Example 10
  • A topical formulation may be prepared as follows:
    Ingredient Quantity
    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. Pat. No. 5,023,252, issued Jun. 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, edited by E. W. Martin (Mack Publishing Company, 18th ed., 1990).
  • Biological Examples Example 1 Deternination of Antibacterial Activity In Vitro Determination of Antibacterial Activity
  • β-lactam resistant bacteria are obtained and phenotyped based on their sensitivity. Minimal inhibitory concentrations (MICs) are measured in a microdilution broth procedure under NCCLS guidelines. The compounds are serially diluted into Mueller-Hinton broth in 96-well microtiter plates. Overnight cultures of bacterial strains are diluted based on absorbance at 600 nm so that the final concentration in each well was 5×105 cfu/ml. Plates are returned to a 35° C. incubator. The following day (or 24 hours in the case of Enterococci strains), MICs are determined by visual inspection of the plates.
  • Bacterial strains which may be tested in this model include, but are not limited to, those found in Table I and Table II below. Growth conditions may be modified as necessary for each particular strain. Growing conditions and growth media for the strains listed in Table I and Table II are known in the art.
  • Determination of Kill Time
  • Experiments to determine the time required to kill the bacteria are conducted as described in Lorian. These experiments are conducted with both staphylococcus and enterococcus strains.
  • Briefly, several colonies are selected from an agar plate and grown at 35° C. under constant agitation until a turbidity of approximately 1.5×108 CFU/ml is achieved. The sample is diluted to about 6×106 CFU/ml and incubated at 35° C. under constant agitation. At various times, aliquots are removed and five ten-fold serial dilutions are performed. The pour plate method is used to determine the number of colony forming units (CFUs).
  • In Vivo Determination of Antibacterial Activity
  • Acute Tolerability Studies in Mice
  • In these studies, the compounds of Formula (I) are administered either intravenously or subcutaneously and observed for 5-15 minutes. If there are no adverse effects, the dose is increased in a second group of mice. This dose incrementation continues until mortality occurs, or the dose is maximized. Generally, dosing begins at 20 mg/kg and increases by 20 mg/kg each time until the maximum tolerated dose (MTD) is achieved.
  • Bioavailability Studies in Mice
  • Mice are administered the compound of Formula (I) either intravenously or subcutaneously at a therapeutic dose (in general, approximately 50 mg/kg). Groups of animals are placed in metabolic cages so that urine and feces may be collected for analysis. Groups of animals (n=3) are sacrificed at various times (10 min, 1 hour and 4 hours). Blood is collected by cardiac puncture and the following organs are harvested: lung, liver, heart, brain, kidney, and spleen. Tissues were weighed and prepared for HPLC analysis. HPLC analysis on the tissue homogenates and fluids is used to determine the concentration of the compound of Formula (I). Metabolic products resulting from changes to the compound of Formula (I) are also determined.
  • Mouse Septecemia Model
  • In this model, an appropriately virulent strain of bacteria (most commonly S. aureus, or E. faecalis or E. faecium) is administered intraperitoneally to mice (N=5 to 10 mice per group). The bacteria was combined with hog gastric mucin to enhance virulence. The dose of bacteria (normally 105-107) is that which is sufficient to induce mortality in all of the mice over a three day period. One hour after the bacteria is administered, the compound of Formula (I) is administered in a single dose, either IV or subcutaneously. Each dose is administered to groups of 5 to 10 mice, at doses that typically range from a maximum of about 20 mg/kg to a minimum of less than 1 mg/kg. A positive control (normally β-lactam with β-lactam sensitive strains) is administered in each experiment. The dose at which approximately 50% of the animals are saved is calculated from the results.
  • Neutropenic Thigh Model
  • In this model, antibacterial activity of the compound of Formula (I) is evaluated against an appropriately virulent strain of bacteria (most commonly S. aureus sensitive or resistant to β-lactams). Mice are initially rendered neutropenic by administration of cyclophosphamide at 200 mg/kg on day 0 and day 2. On day 4, they are infected in the left anterior thigh by an IM injection of a single dose of bacteria. The mice are administered the compound of Formula (I) one hour after the administration of bacteria. At various later times (normally 1, 2.5, 4 and 24 hours) the mice are sacrificed (3 per time point). The thigh is excised, homogenized and the number of CFUs (colony forming units) is determined by plating. Blood is also plated to determine the CFUs in the blood.
  • Pharmnacokinetic Studies
  • The rate at which the compound of Formula (I) is removed from the blood can be determined in either rats or mice. In rats, the test animals are cannulated in the jugular vein. A compound of Formula (I) is administered via tail vein injection, and at various time points (normally 5, 15, 30, 60 minutes and 2, 4, 6 and 24 hours) blood is withdrawn from the cannula. In mice, a compound of Formula (I) is also administered via tail vein injection, and at various time points. Blood is normally obtained by cardiac puncture. The concentration of the remaining compound of Formula (I) is determined by HPLC.
  • The foregoing invention has been described in some detail by way of illustration and example, for purposes of clarity and understanding. It will be obvious to one of skill in the art that changes and modifications may be practiced within the scope of the appended claims. Therefore, it is to be understood that the above description is intended to be illustrative and not restrictive. The scope of the invention should, therefore, be determined not with reference to the above description, but should instead be determined with reference to the following appended claims, along with the full scope of equivalents to which such claims are entitled.
  • All patents, patent applications and publications cited in this application are hereby incorporated by reference in their entirety for all purposes to the same extent as if each individual patent, patent application or publication were so individually denoted.

Claims (2)

1. A multibinding compound which comprises from 2-10 ligands covalently connected by a linker or linkers wherein each of said ligands comprises a ligand domain capable of binding to penicillin binding proteins, a transpeptidase enzyme, a substrate of a transpeptidase enzyme, a beta-lactamase enzyme, a pencillinase enzyme, a cephalosporinase enzyme, a transglycoslase enzyme, or a transglycosylase enzyme substrate provided that:
(i) all the ligands in a multibinding compound of Formula (I) cannot be either a beta lactam antibiotic, an optionally substituted glycopeptide antibiotic, or an aglycone derivative of an optionally substituted glycopeptide antibiotic;
(ii) when p is 2 and q is 1 then at least one of the ligands is a beta lactam antibiotic; and
(iii) when p is 2, q is 1, and one of the ligands is vancomycin attached via the [C], then the other cannot be cefalexin attached to the linker via acylation of its alpha amino group.
2-40. (canceled)
US11/603,287 1998-06-08 2006-11-21 Novel antibacterial agents Abandoned US20070134729A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/603,287 US20070134729A1 (en) 1998-06-08 2006-11-21 Novel antibacterial agents

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US8844898P 1998-06-08 1998-06-08
US9307298P 1998-07-16 1998-07-16
US31719899A 1999-05-24 1999-05-24
US45792699A 1999-12-08 1999-12-08
US11/603,287 US20070134729A1 (en) 1998-06-08 2006-11-21 Novel antibacterial agents

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US45792699A Continuation 1998-06-08 1999-12-08

Publications (1)

Publication Number Publication Date
US20070134729A1 true US20070134729A1 (en) 2007-06-14

Family

ID=26778665

Family Applications (6)

Application Number Title Priority Date Filing Date
US09/325,662 Expired - Lifetime US6288234B1 (en) 1998-06-08 1999-06-04 Multibinding inhibitors of microsomal triglyceride transferase protein
US09/327,899 Expired - Lifetime US6566509B1 (en) 1998-06-08 1999-06-08 Multivalent macrolide antibiotics
US09/760,827 Abandoned US20020028943A1 (en) 1998-06-08 2001-01-17 Multibinding inhibitors of microsomal triglyceride transferase protein
US10/161,279 Abandoned US20040023290A1 (en) 1998-06-08 2002-06-03 Novel therapeutic agents that modulate enzymatic processes
US10/330,381 Expired - Lifetime US7179794B2 (en) 1998-06-08 2002-12-27 Multivalent macrolide antibiotics
US11/603,287 Abandoned US20070134729A1 (en) 1998-06-08 2006-11-21 Novel antibacterial agents

Family Applications Before (5)

Application Number Title Priority Date Filing Date
US09/325,662 Expired - Lifetime US6288234B1 (en) 1998-06-08 1999-06-04 Multibinding inhibitors of microsomal triglyceride transferase protein
US09/327,899 Expired - Lifetime US6566509B1 (en) 1998-06-08 1999-06-08 Multivalent macrolide antibiotics
US09/760,827 Abandoned US20020028943A1 (en) 1998-06-08 2001-01-17 Multibinding inhibitors of microsomal triglyceride transferase protein
US10/161,279 Abandoned US20040023290A1 (en) 1998-06-08 2002-06-03 Novel therapeutic agents that modulate enzymatic processes
US10/330,381 Expired - Lifetime US7179794B2 (en) 1998-06-08 2002-12-27 Multivalent macrolide antibiotics

Country Status (17)

Country Link
US (6) US6288234B1 (en)
EP (11) EP1085846A2 (en)
JP (3) JP2004512255A (en)
AR (3) AR018631A1 (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)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080039374A1 (en) * 2001-10-12 2008-02-14 Fatheree Paul R Cross-lined glycopeptide-cephalosporin antibiotics
US20080069870A1 (en) * 2005-04-12 2008-03-20 Elan Corporation Pic Controlled Release Compositions Comprising a Cephalosporin for the Treatment of a Bacterial Infection
US20080193544A1 (en) * 2005-03-16 2008-08-14 Nycomed Gmbh Taste Masked Dosage Form Containing Roflumilast
US8431154B2 (en) 2002-02-20 2013-04-30 Takeda Gmbh Oral dosage form containing a PDE 4 inhibitor as an active ingredient and polyvinylpyrrolidone as excipient
US8536206B2 (en) 2003-03-08 2013-09-17 Takeda Gmbh Process for the preparation of roflumilast
CN103755728A (en) * 2013-12-24 2014-04-30 深圳华润九新药业有限公司 Cefazolin derivative and preparation method thereof as well as oral antibiotic preparation
WO2015022335A1 (en) * 2013-08-12 2015-02-19 Katholieke Universiteit Leuven Vancomycin analogs
WO2015114666A3 (en) * 2014-01-29 2015-11-05 Vyome Biosciences Pvt. Ltd. Treatments for resistant acne
US11071769B2 (en) * 2017-05-22 2021-07-27 Insmed Incorporated Lipo-glycopeptide cleavable derivatives and uses thereof

Families Citing this family (148)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2347027T3 (en) * 1998-05-22 2010-10-22 The Board Of Trustees Of The Leland Stanford Junior University BIFUNCTIONAL MOLECULES AND THERAPIES BASED ON THE SAME.
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
EP1257269B1 (en) * 2000-02-24 2004-11-03 Kopran Research Laboratories Limited Orally administrable acid stable anti-ulcer benzimidazole derivatives
WO2001098540A2 (en) * 2000-06-22 2001-12-27 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
EP1461323B1 (en) * 2001-06-12 2013-10-09 Wellstat Therapeutics Corporation Compounds for the treatment of metabolic disorders
RS50712B (en) 2001-06-28 2010-06-30 Pfizer Products Inc. Triamide substituted indoles, benzofuranes and benzothiophenes as inhibitors of microsomal triglyceride transfer protein (mpt) and/or apolipoprotein b (apo b) secretion
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
AU2002329785A1 (en) * 2001-08-17 2003-03-03 The Trustees Of Princeton University Bifunctional glycopeptides antibiotics and combinatorial libraries thereof
MXPA04002303A (en) 2001-09-11 2004-06-29 Astrazeneca Ab Oxazolidinone and/or isoxazoline as antibacterial agents.
CA2463039A1 (en) 2001-09-26 2003-04-03 Bayer Pharmaceuticals Corporation 1,6-naphthyridine derivatives as antidiabetics
AUPR879601A0 (en) * 2001-11-09 2001-12-06 Biota Scientific Management Pty Ltd Novel chemical compounds and their use
AU2002357366A1 (en) * 2001-12-20 2003-07-09 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
AU2003215245A1 (en) 2002-02-15 2003-09-09 Sympore Gmbh Conjugates of biologically active compounds, methods for their preparation and use, formulation and pharmaceutical applications thereof
AU2003211113B2 (en) * 2002-02-15 2007-08-09 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
MXPA04002602A (en) * 2002-02-28 2004-08-11 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
US6878686B2 (en) 2002-05-24 2005-04-12 Theravance, Inc. Cross-linked glycopeptide-cephalosporin antibiotics
JP4530851B2 (en) * 2002-07-08 2010-08-25 グラクソスミスクライン・イストラジヴァッキ・センタル・ザグレブ・ドルズバ・ゼー・オメイェノ・オドゴヴォルノスティオ Novel compounds and compositions as carriers for steroid / nonsteroidal anti-inflammatory active molecules, anti-neoplastic active molecules and antiviral active molecules
PL374646A1 (en) * 2002-07-08 2005-10-31 Pliva-Istrazivacki Institutt D.O.O. Novel nonsteroidal anti-inflammatory substances, compositions and methods for their use
WO2004007444A2 (en) 2002-07-11 2004-01-22 Vicuron Pharmaceuticals, Inc. N-hydroxyamide derivatives possessing antibacterial activity
CN1315859C (en) 2002-08-15 2007-05-16 维库罗恩医药品公司 Lincomycin derivatives possessing antibacterial activity
US7199105B2 (en) 2002-08-15 2007-04-03 Vicuron Pharmaceuticals, Inc. Lincomycin derivatives possessing antibacterial activity
TW200420573A (en) * 2002-09-26 2004-10-16 Rib X Pharmaceuticals Inc Bifunctional heterocyclic compounds and methods of making and using same
EP1627045A2 (en) * 2003-02-28 2006-02-22 Plexxikon, Inc. Pyk2 crystal structure and uses
MXPA05009430A (en) * 2003-03-05 2006-04-07 Rib X Pharmaceuticals Inc Bifunctional heterocyclic compounds and methods of making and using the same.
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
CA2521621C (en) * 2003-04-15 2011-11-01 Wellstat Therapeutics Corporation Compounds for the treatment of metabolic disorders
US7271252B2 (en) * 2003-04-22 2007-09-18 Roche Diagnostics Operations, Inc. Reagents for detecting efavirenz
NZ543789A (en) * 2003-04-30 2008-03-28 Wellstat Therapeutics Corp Compounds for the treatment of metabolic disorders
WO2004113350A1 (en) 2003-05-23 2004-12-29 Theravance, Inc. Cross-linked glycopeptide - cephalosporin antibiotics
US7256177B2 (en) 2003-06-17 2007-08-14 Vicuron Pharmaceuticals, Inc. Lincomycin derivatives possessing antibacterial activity
US7199106B2 (en) 2003-06-17 2007-04-03 Vicuron Pharmaceuticals, Inc. Lincomycin derivatives possessing antimicrobial 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
US7348338B2 (en) * 2003-07-17 2008-03-25 Plexxikon, Inc. PPAR active compounds
RU2356889C2 (en) * 2003-07-17 2009-05-27 Плекссикон, Инк. Compounds, which are active towards receptors, activated by peroxisome proliferator
MXPA06002139A (en) * 2003-08-29 2006-05-31 Dynogen Pharmaceuticals Inc Compositions useful for treating gastrointestinal motility disorders.
WO2005021486A1 (en) * 2003-08-29 2005-03-10 Japan Tobacco Inc. Ester derivative and medicinal use thereof
US20050164300A1 (en) * 2003-09-15 2005-07-28 Plexxikon, Inc. Molecular scaffolds for kinase ligand development
EP1682563A1 (en) * 2003-10-30 2006-07-26 Rib-X Pharmaceuticals, Inc. Bifunctional macrolide heterocyclic compounds and mehtods of making and using the same
EP1699808A1 (en) * 2003-11-18 2006-09-13 Rib-X Pharmaceuticals, Inc. Bifunctional macrolide heterocyclic compounds and methods of making and using the same
US20070066641A1 (en) * 2003-12-19 2007-03-22 Prabha Ibrahim Compounds and methods for development of RET modulators
EP1696920B8 (en) * 2003-12-19 2015-05-06 Plexxikon Inc. Compounds and methods for development of ret modulators
AU2005210594A1 (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
EP1723159B1 (en) 2004-02-27 2019-06-12 Melinta Therapeutics, Inc. Macrocyclic compounds and methods of making and using the same
JP2008503446A (en) 2004-05-06 2008-02-07 プレキシコン,インコーポレーテッド PDE4B inhibitor and use thereof
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
WO2006027711A2 (en) * 2004-08-26 2006-03-16 Nicholas Piramal India Limited Prodrugs and codrugs containing bio- cleavable disulfide linkers
TW200616604A (en) 2004-08-26 2006-06-01 Nicholas Piramal India Ltd Nitric oxide releasing prodrugs containing bio-cleavable linker
CA2583428A1 (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
BRPI0516772A (en) * 2004-10-25 2008-09-23 Japan Tobacco Inc solid formulation with improved solubility and stability and method for producing said formulation
EP1819673A2 (en) * 2004-11-30 2007-08-22 Plexxikon, Inc. Indole derivatives for use as ppar active compounds
MX2007006777A (en) 2004-12-06 2007-08-06 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
CA2608733A1 (en) * 2005-05-17 2007-02-01 Plexxikon, Inc. Pyrrol (2,3-b) pyridine derivatives protein kinase inhibitors
CA2609783A1 (en) * 2005-05-27 2006-12-07 Pfizer Products Inc. Combination of a cannabinoid-1- receptor-antagonist and a microsomal triglyceride transfer protein inhibitor for treating obesity or mainataining weight loss
EP3088400A1 (en) * 2005-06-22 2016-11-02 Plexxikon Inc. Pyrrolo[2,3-b]pyridine derivatives as protein kinase inhibitors
JP5395432B2 (en) 2005-08-24 2014-01-22 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
EP1934237A2 (en) 2005-08-24 2008-06-25 Rib-X Pharmaceuticals, Inc. Triazole compounds and methods of making and using the same
NZ567162A (en) * 2005-09-07 2011-06-30 Plexxikon Inc 1, 3-disubstituted indole derivatives for use as PPAR modulators
DK1928438T3 (en) 2005-09-26 2013-04-22 Avigen Inc Use of ibudilast for the treatment of drug dependence
US7622256B2 (en) 2006-05-31 2009-11-24 Avigen, Inc. Method for selecting compounds that modulate MIF-induced expression of ICAM-1 and/or VCAM-1
EP2094241A4 (en) * 2006-11-14 2013-04-17 Ariad Pharma Inc 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
MX2009006688A (en) 2006-12-21 2009-06-30 Plexxikon Inc Compounds and methods for kinase modulation, and indications therefor.
PE20121126A1 (en) * 2006-12-21 2012-08-24 Plexxikon Inc PIRROLO [2,3-B] PYRIDINES COMPOUNDS AS KINASE MODULATORS
ES2392116T3 (en) * 2007-01-30 2012-12-04 Avigen, Inc. Procedures for the treatment of acute and subchronic pain
US8889632B2 (en) 2007-01-31 2014-11-18 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
CN101730708B (en) 2007-03-28 2013-09-18 哈佛大学校长及研究员协会 Stitched 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
EP2205600A1 (en) 2007-10-25 2010-07-14 Achaogen, Inc. CARBACEPHEM ß -LACTAM ANTIBIOTICS
CA2723358A1 (en) * 2008-05-05 2009-11-12 Allison B. Reiss Method for improving cardiovascular risk profile of cox inhibitors
AU2009248057B2 (en) * 2008-05-13 2013-02-21 Genmedica Therapeutics Sl Salicylate conjugates useful 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
CA2755072A1 (en) * 2009-03-16 2010-09-23 Genmedica Therapeutics Sl Combination therapies for treating metabolic disorders
EA031116B1 (en) * 2009-04-03 2018-11-30 Ф. Хоффманн-Ля Рош Аг PROPANE-1-SULFONIC ACID {3-[5-(4-CHLORO-PHENYL)-1H-PYRROLO[2,3-b]PYRIDINE-3-CARBONYL]-2,4-DIFLUORO-PHENYL}AMIDE CRYSTALLINE POLYMORPH FORMS
RU2624506C2 (en) 2009-04-29 2017-07-04 АМАРИН КОРПОРЕЙШН ПиЭлСи Pharmaceutical compositions containing epa and cardiovascular agents and their application methods
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
MX2012005284A (en) 2009-11-06 2012-06-28 Plexxikon Inc Compounds and methods for kinase modulation, and indications therefor.
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
CA2807685C (en) 2010-08-13 2020-10-06 Aileron Therapeutics, Inc. P53 derived peptidomimetic macrocycle
EP2605650A4 (en) 2010-08-18 2014-05-21 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
PT2672967T (en) 2011-02-07 2018-12-07 Plexxikon Inc Compounds and methods for kinase modulation, and indications therefor
TWI558702B (en) 2011-02-21 2016-11-21 普雷辛肯公司 Solid forms of a pharmaceutically active substance
WO2012154879A2 (en) 2011-05-09 2012-11-15 Van Andel Research Institute Autophagy inhibitors
EP2716642B1 (en) * 2011-05-30 2016-07-20 Astellas Pharma Inc. Imidazopyridine compound
WO2014081405A2 (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
EA028342B1 (en) 2011-09-09 2017-11-30 Мерк Шарп И Доум Корп. Methods for treating pneumonia
US9096684B2 (en) 2011-10-18 2015-08-04 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
CN115813902A (en) 2012-01-20 2023-03-21 德玛公司 Use of substituted hexitols for the treatment of malignant tumors
JP6450192B2 (en) 2012-02-15 2019-01-09 エイルロン セラピューティクス,インコーポレイテッド Triazole-bridged and thioether-bridged peptidomimetic macrocycles
WO2013123266A1 (en) 2012-02-15 2013-08-22 Aileron Therapeutics, Inc. 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
US9604919B2 (en) 2012-11-01 2017-03-28 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
WO2014084312A1 (en) * 2012-11-30 2014-06-05 アステラス製薬株式会社 Imidazopyridine compound
US8883857B2 (en) 2012-12-07 2014-11-11 Baylor College Of Medicine Small molecule xanthine oxidase inhibitors and methods of use
US20140275000A1 (en) 2013-03-15 2014-09-18 Cubist Pharmaceuticals, Inc. Ceftolozane pharmaceutical compositions
WO2014144295A1 (en) 2013-03-15 2014-09-18 Cubist Pharmaceuticals, Inc. Ceftolozane antibiotic compositions
US9872906B2 (en) 2013-03-15 2018-01-23 Merck Sharp & Dohme Corp. Ceftolozane antibiotic compositions
WO2014168986A1 (en) 2013-04-08 2014-10-16 Brown Dennis M Therapeutic benefit of suboptimally administered chemical compounds
WO2015013581A1 (en) 2013-07-26 2015-01-29 Update Pharma Inc. Combinatorial methods 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
WO2015035376A2 (en) 2013-09-09 2015-03-12 Calixa Therapeutics, Inc. Treating infections with ceftolozane/tazobactam in subjects having impaired renal function
US8906898B1 (en) 2013-09-27 2014-12-09 Calixa Therapeutics, Inc. Solid forms of ceftolozane
MX2016013027A (en) 2014-04-04 2017-05-23 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
AU2015320549A1 (en) 2014-09-24 2017-04-13 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
SG11201702175YA (en) 2014-09-24 2017-04-27 Aileron Therapeutics Inc Peptidomimetic macrocycles and formulations thereof
EP3215490A1 (en) * 2014-11-05 2017-09-13 Hetero Research Foundation Process for the preparation of lomitapide
BR112017019738A2 (en) 2015-03-20 2018-05-29 Aileron Therapeutics Inc peptidomimetic macrocycles and their uses
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
EP3347372A4 (en) 2015-09-10 2019-09-04 Aileron Therapeutics, Inc. Peptidomimetic macrocycles as modulators of mcl-1
CN108348510B (en) 2015-09-17 2022-01-04 圣母大学 Benzylamine-containing heterocyclic compounds and compositions useful against mycobacterial infections
SG11201804037XA (en) * 2015-11-16 2018-06-28 Evonik Roehm Gmbh Injection solution comprising a non-nucleoside reverse-transcriptase inhibitor and poly(lactide-co-glycolide)
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
US11642365B2 (en) * 2017-09-22 2023-05-09 John Mansell Compositions and methods for treatment of sepsis-related disorders
BR112023004249A2 (en) * 2020-09-15 2023-04-04 Mitsubishi Tanabe Pharma Corp TRIAZINE COMPOUND SALT, CRYSTALLINE FORM AND PRODUCTION METHOD

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5693791A (en) * 1995-04-11 1997-12-02 Truett; William L. Antibiotics and process for preparation
US6437119B1 (en) * 1998-05-07 2002-08-20 William Lawrence Truett Compounds formed from two or three antibiotics and their processes of preparation

Family Cites Families (45)

* 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
WO1992005802A1 (en) * 1990-09-28 1992-04-16 Neorx Corporation Polymeric carriers for release of covalently linked agents
US5312837A (en) * 1991-01-29 1994-05-17 Genelabs Technologies, Inc. Method of treating viral infections with aryl macrocyclic compounds
US5770358A (en) 1991-09-18 1998-06-23 Affymax Technologies N.V. Tagged synthetic oligomer libraries
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
MX9605366A (en) * 1994-05-07 1997-12-31 Astra Ab Bicyclic amidine derivatives 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
WO1996040640A1 (en) 1995-06-07 1996-12-19 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
AU2332497A (en) * 1996-03-19 1997-10-10 Salk Institute For Biological Studies, The (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
US5885983A (en) 1996-05-10 1999-03-23 Bristol-Myers Squibb Company Inhibitors of microsomal triglyceride transfer protein and method
US5827875A (en) 1996-05-10 1998-10-27 Bristol-Myers Squibb Company Inhibitors of microsomal triglyceride transfer protein and method
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
JP2002513379A (en) 1996-07-24 2002-05-08 ブリストル―マイヤーズ・スクイブ・カンパニー Treatment of tumors with high LDL requirements using MTP inhibitors
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
YU23499A (en) 1996-11-27 2001-07-10 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
JP2001508049A (en) * 1996-12-18 2001-06-19 イーライ・リリー・アンド・カンパニー Combined production method of condensed pyrimidine library
WO1998027979A1 (en) 1996-12-20 1998-07-02 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
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
CA2276467A1 (en) 1997-01-17 1998-07-23 Bristol-Myers Squibb Company Method for treating atherosclerosis with an mpt inhibitor and cholesterol lowering drugs
WO1998031225A1 (en) 1997-01-17 1998-07-23 Bristol-Myers Squibb Company A method of inhibiting or treating phytosterolemia with an mtp inhibitor
CA2240325A1 (en) 1998-03-27 1998-11-14 Synsorb Biotech, Inc. Methods for screening compound libraries
US20020131972A1 (en) * 1998-05-21 2002-09-19 Daniel Sem Multi-partite ligands and methods of identifying and using same

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5693791A (en) * 1995-04-11 1997-12-02 Truett; William L. Antibiotics and process for preparation
US6437119B1 (en) * 1998-05-07 2002-08-20 William Lawrence Truett Compounds formed from two or three antibiotics and their processes of preparation

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7601690B2 (en) 2001-10-12 2009-10-13 Theravance, Inc. Cross-linked glycopeptide-cephalosporin antibiotics
US20080039374A1 (en) * 2001-10-12 2008-02-14 Fatheree Paul R Cross-lined glycopeptide-cephalosporin antibiotics
US8044195B2 (en) 2001-10-12 2011-10-25 Theravance, Inc. Cross-linked glycopeptide-cephalosporin antibiotics
US20080108558A1 (en) * 2001-10-12 2008-05-08 Fatheree Paul R Cross-linked glycopeptide-cephalosporin antibiotics
US8557978B2 (en) 2001-10-12 2013-10-15 Theravance, Inc. Cross-linked glycopeptide-cephalosporin antibiotics
US20080194464A1 (en) * 2001-10-12 2008-08-14 Fatheree Paul R Cross-linked glycopeptide-cephalosporin antibiotics
US20080039611A1 (en) * 2001-10-12 2008-02-14 Fatheree Paul R Cross-linked glycopeptide-cephalosporin antibiotics
US7612037B2 (en) 2001-10-12 2009-11-03 Theravance, Inc. Cross-linked glycopeptide-cephalosporin antibiotics
US7713931B2 (en) 2001-10-12 2010-05-11 Theravance, Inc. Cross-linked glycopeptide-cephalosporin antibiotics
US7655621B2 (en) 2001-10-12 2010-02-02 Theravance, Inc. Cross-linked glycopeptide-cephalosporin antibiotics
US7649080B2 (en) 2001-10-12 2010-01-19 Theravance, Inc. Cross-linked glycopeptide-cephalosporin antibiotics
US20100197569A1 (en) * 2001-10-12 2010-08-05 Theravance, Inc. Cross-linked glycopeptide-cephalosporin antibiotics
US9468598B2 (en) 2002-02-20 2016-10-18 Astrazeneca Ab Oral dosage form containing a PDE 4 inhibitor as an active ingredient and polyvinylpyrrolidon as excipient
US8431154B2 (en) 2002-02-20 2013-04-30 Takeda Gmbh Oral dosage form containing a PDE 4 inhibitor as an active ingredient and polyvinylpyrrolidone as excipient
US8536206B2 (en) 2003-03-08 2013-09-17 Takeda Gmbh Process for the preparation of roflumilast
US8604064B2 (en) 2003-03-10 2013-12-10 Takeda Gmbh Process for the preparation of roflumilast
US8618142B2 (en) 2003-03-10 2013-12-31 Takeda Gmbh Process for the preparation of roflumilast
US8663694B2 (en) 2005-03-16 2014-03-04 Takeda Gmbh Taste masked dosage form containing roflumilast
US20080193544A1 (en) * 2005-03-16 2008-08-14 Nycomed Gmbh Taste Masked Dosage Form Containing Roflumilast
US20080069870A1 (en) * 2005-04-12 2008-03-20 Elan Corporation Pic Controlled Release Compositions Comprising a Cephalosporin for the Treatment of a Bacterial Infection
WO2015022335A1 (en) * 2013-08-12 2015-02-19 Katholieke Universiteit Leuven Vancomycin analogs
CN103755728A (en) * 2013-12-24 2014-04-30 深圳华润九新药业有限公司 Cefazolin derivative and preparation method thereof as well as oral antibiotic preparation
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
WO2015114666A3 (en) * 2014-01-29 2015-11-05 Vyome Biosciences Pvt. Ltd. Treatments for resistant acne
US10071103B2 (en) 2014-01-29 2018-09-11 Vyome Biosciennces Pvt. Ltd. Treatments for resistant acne
US11045479B2 (en) 2014-01-29 2021-06-29 Vyome Therapeutics Limited Treatments for resistant acne
US11071769B2 (en) * 2017-05-22 2021-07-27 Insmed Incorporated Lipo-glycopeptide cleavable derivatives and uses thereof
US11857597B2 (en) 2017-05-22 2024-01-02 Insmed Incorporated Lipo-glycopeptide cleavable derivatives and uses thereof

Also Published As

Publication number Publication date
WO1999064051A1 (en) 1999-12-16
WO1999063929A3 (en) 2000-02-03
WO1999064048A9 (en) 2000-07-20
AU4818099A (en) 1999-12-30
CA2319474A1 (en) 1999-12-16
WO1999063988A1 (en) 1999-12-16
AU4426399A (en) 1999-12-30
AU4674599A (en) 1999-12-30
EP1085861A1 (en) 2001-03-28
AU4543899A (en) 1999-12-30
US6288234B1 (en) 2001-09-11
ATE322910T1 (en) 2006-04-15
WO1999063940A3 (en) 2001-06-07
AU4426699A (en) 1999-12-30
WO1999063940A2 (en) 1999-12-16
EP0989857A4 (en) 2001-01-17
WO1999064033A9 (en) 2000-07-27
WO1999064054A1 (en) 1999-12-16
DK1005356T3 (en) 2006-08-14
CA2320241A1 (en) 1999-12-16
AR019635A1 (en) 2002-02-27
SG106036A1 (en) 2004-09-30
US7179794B2 (en) 2007-02-20
DE69930818D1 (en) 2006-05-24
WO1999063937A3 (en) 2000-03-02
WO1999064047A9 (en) 2000-03-16
EP0989857A1 (en) 2000-04-05
JP2002517465A (en) 2002-06-18
AU4430599A (en) 1999-12-30
EP1003541A1 (en) 2000-05-31
ES2263274T3 (en) 2006-12-01
AR018631A1 (en) 2001-11-28
CA2321166A1 (en) 1999-12-16
WO1999064037A1 (en) 1999-12-16
CA2316999A1 (en) 1999-12-16
SG80631A1 (en) 2001-05-22
CY1105087T1 (en) 2009-11-04
WO1999064049A1 (en) 1999-12-16
NZ505979A (en) 2003-09-26
JP2002517422A (en) 2002-06-18
EP1085846A2 (en) 2001-03-28
AU4551899A (en) 1999-12-30
HK1028737A1 (en) 2001-03-02
EP1083919A1 (en) 2001-03-21
WO1999064054A9 (en) 2000-03-02
EP1124528A1 (en) 2001-08-22
EP1005356A4 (en) 2001-08-22
CA2319120A1 (en) 1999-12-16
AU4551799A (en) 1999-12-30
CA2321190A1 (en) 1999-12-16
CA2321162A1 (en) 1999-12-16
EP1083920A1 (en) 2001-03-21
CA2321274A1 (en) 1999-12-16
WO1999063937A2 (en) 1999-12-16
US20020028943A1 (en) 2002-03-07
EP1085893A1 (en) 2001-03-28
EP1005356B1 (en) 2006-04-12
WO1999064047A1 (en) 1999-12-16
WO1999063929A9 (en) 2000-03-16
WO1999064033A1 (en) 1999-12-16
CA2319495A1 (en) 1999-12-16
DE69930818T2 (en) 2006-12-07
EP1143991A2 (en) 2001-10-17
AU4551699A (en) 1999-12-30
AU764401B2 (en) 2003-08-21
CA2319477A1 (en) 1999-12-16
TWI239959B (en) 2005-09-21
WO1999063929A2 (en) 1999-12-16
EP1005356A1 (en) 2000-06-07
WO1999063937A9 (en) 2001-06-28
WO1999064049A9 (en) 2001-01-18
PT1005356E (en) 2006-07-31
JP2004512255A (en) 2004-04-22
AU4677399A (en) 1999-12-30
CA2319080C (en) 2011-05-31
WO1999063988A9 (en) 2000-05-04
US20040023290A1 (en) 2004-02-05
WO1999064047A8 (en) 2001-03-08
CA2319080A1 (en) 1999-12-16
SG90053A1 (en) 2002-07-23
EP1085891A1 (en) 2001-03-28
AR019634A1 (en) 2002-02-27
AU4677199A (en) 1999-12-30
US20030176670A1 (en) 2003-09-18
WO1999064048A1 (en) 1999-12-16
US6566509B1 (en) 2003-05-20

Similar Documents

Publication Publication Date Title
US20070134729A1 (en) Novel antibacterial agents
US7101964B2 (en) Intermediate for preparing glycopeptide derivatives
US20070154948A1 (en) Novel antibacterial agents
US6518242B1 (en) Derivatives of glycopeptide antibacterial agents
CA2318394A1 (en) Novel antibacterial agents
US20030008812A1 (en) Glycopeptide derivatives
US6420560B1 (en) H1—histamine receptor antagonists

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: THERAVANCE BIOPHARMA ANTIBIOTICS IP, LLC, CALIFORN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:THERAVANCE, INC.;REEL/FRAME:033162/0550

Effective date: 20140601