US20030130349A1 - Inhibitors of alpha 4beta1 mediated cell adhesion - Google Patents

Inhibitors of alpha 4beta1 mediated cell adhesion Download PDF

Info

Publication number
US20030130349A1
US20030130349A1 US10/193,137 US19313702A US2003130349A1 US 20030130349 A1 US20030130349 A1 US 20030130349A1 US 19313702 A US19313702 A US 19313702A US 2003130349 A1 US2003130349 A1 US 2003130349A1
Authority
US
United States
Prior art keywords
group
mmol
alkyl
alkylene
formula
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US10/193,137
Other versions
US6596752B1 (en
Inventor
Thomas Lobl
Bradley Teegarden
Alexander Polinsky
Gilbert Rishton
Masafumi Yamagishi
Steven Tanis
Jed Fisher
Edward Thomas
Robert Chrusciel
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.)
Tanabe Seiyaku Co Ltd
Original Assignee
Tanabe Seiyaku Co Ltd
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 Tanabe Seiyaku Co Ltd filed Critical Tanabe Seiyaku Co Ltd
Priority to US10/193,137 priority Critical patent/US6596752B1/en
Publication of US20030130349A1 publication Critical patent/US20030130349A1/en
Application granted granted Critical
Publication of US6596752B1 publication Critical patent/US6596752B1/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/46Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino or carboxyl groups bound to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/57Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of rings other than six-membered aromatic rings
    • C07C233/60Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of rings other than 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 singly-bound oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/57Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of rings other than six-membered aromatic rings
    • C07C233/63Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of rings other than 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 carboxyl groups
    • 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
    • C07C233/81Carboxylic 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 carboxyl groups
    • C07C233/82Carboxylic 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 carboxyl groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom
    • C07C233/83Carboxylic 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 carboxyl groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic 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
    • C07C233/81Carboxylic 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 carboxyl groups
    • C07C233/82Carboxylic 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 carboxyl groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom
    • C07C233/87Carboxylic 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 carboxyl groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom of a carbon skeleton containing six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/40Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of rings other than six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/42Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/44Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton with carbon atoms of carboxamide groups and singly-bound oxygen atoms bound to carbon atoms of the same non-condensed six-membered aromatic ring
    • C07C235/58Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton with carbon atoms of carboxamide groups and singly-bound oxygen atoms bound to carbon atoms of the same non-condensed six-membered aromatic ring with carbon atoms of carboxamide groups and singly-bound oxygen atoms, bound in ortho-position to carbon atoms of the same non-condensed six-membered aromatic ring
    • C07C235/64Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton with carbon atoms of carboxamide groups and singly-bound oxygen atoms bound to carbon atoms of the same non-condensed six-membered aromatic ring with carbon atoms of carboxamide groups and singly-bound oxygen atoms, bound in ortho-position to carbon atoms of the same non-condensed six-membered aromatic ring having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/70Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups and doubly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/84Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups and doubly-bound oxygen atoms bound to the same carbon skeleton with the carbon atom of at least one of the carboxamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C237/22Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton having nitrogen atoms of amino groups bound to the carbon skeleton of the acid part, further acylated
    • 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
    • C07C237/28Carboxylic 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 non-condensed six-membered aromatic ring of the carbon skeleton
    • C07C237/40Carboxylic 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 non-condensed six-membered aromatic ring of the carbon skeleton having the nitrogen atom of the carboxamide group bound to a carbon atom of a six-membered aromatic ring
    • 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
    • C07C237/28Carboxylic 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 non-condensed six-membered aromatic ring of the carbon skeleton
    • C07C237/42Carboxylic 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 non-condensed six-membered aromatic ring of the carbon skeleton having nitrogen atoms of amino groups bound to the carbon skeleton of the acid part, further acylated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C255/00Carboxylic acid nitriles
    • C07C255/01Carboxylic acid nitriles having cyano groups bound to acyclic carbon atoms
    • C07C255/24Carboxylic acid nitriles having cyano groups bound to acyclic carbon atoms containing cyano groups and singly-bound nitrogen atoms, not being further bound to other hetero atoms, bound to the same saturated acyclic carbon skeleton
    • C07C255/29Carboxylic acid nitriles having cyano groups bound to acyclic carbon atoms containing cyano groups and singly-bound nitrogen atoms, not being further bound to other hetero atoms, bound to the same saturated acyclic carbon skeleton containing cyano groups and acylated amino groups bound to the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C255/00Carboxylic acid nitriles
    • C07C255/45Carboxylic acid nitriles having cyano groups bound to carbon atoms of rings other than six-membered aromatic rings
    • C07C255/46Carboxylic acid nitriles having cyano groups bound to carbon atoms of rings other than six-membered aromatic rings to carbon atoms of non-condensed rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C259/00Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups
    • C07C259/04Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups without replacement of the other oxygen atom of the carboxyl group, e.g. hydroxamic acids
    • C07C259/08Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups without replacement of the other oxygen atom of the carboxyl group, e.g. hydroxamic acids having carbon atoms of hydroxamic groups bound to carbon atoms of rings other than six-membered aromatic rings
    • 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
    • C07C271/22Esters 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 carboxyl groups
    • 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
    • C07C271/26Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atom of at least one of the carbamate groups bound to a carbon atom of a six-membered aromatic ring
    • C07C271/28Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atom of at least one of the carbamate groups bound to a carbon atom of a six-membered aromatic ring to a carbon atom of a non-condensed six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C275/00Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups
    • C07C275/26Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of urea groups bound to carbon atoms of rings other than six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C275/00Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups
    • C07C275/28Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of urea groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton
    • C07C275/42Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of urea groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton being further substituted by carboxyl groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C311/00Amides of sulfonic acids, i.e. compounds having singly-bound oxygen atoms of sulfo groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/15Sulfonamides having sulfur atoms of sulfonamide groups bound to carbon atoms of six-membered aromatic rings
    • C07C311/21Sulfonamides having sulfur atoms of sulfonamide groups bound to carbon atoms of six-membered aromatic rings having the nitrogen atom of at least one of the sulfonamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C311/00Amides of sulfonic acids, i.e. compounds having singly-bound oxygen atoms of sulfo groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/50Compounds containing any of the groups, X being a hetero atom, Y being any atom
    • C07C311/52Y being a hetero atom
    • C07C311/64X and Y being nitrogen atoms, e.g. N-sulfonylguanidine
    • 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
    • C07D207/04Heterocyclic 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 no double bonds between ring members or between ring members and non-ring members
    • C07D207/10Heterocyclic 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 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
    • C07D207/16Carbon 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
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/18Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D209/20Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals substituted additionally by nitrogen atoms, e.g. tryptophane
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/62Oxygen or sulfur atoms
    • C07D213/63One oxygen atom
    • C07D213/64One oxygen atom attached in position 2 or 6
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • C07D213/82Amides; Imides in position 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/22Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the nitrogen-containing ring
    • C07D217/26Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • 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
    • C07D233/64Heterocyclic 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 substituted hydrocarbon radicals attached to ring carbon atoms, e.g. histidine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/16Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms
    • C07D295/18Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms by radicals derived from carboxylic acids, or sulfur or nitrogen analogues thereof
    • C07D295/182Radicals derived from carboxylic acids
    • C07D295/185Radicals derived from carboxylic acids from aliphatic carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/16Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms
    • C07D295/18Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms by radicals derived from carboxylic acids, or sulfur or nitrogen analogues thereof
    • C07D295/182Radicals derived from carboxylic acids
    • C07D295/192Radicals derived from carboxylic acids from aromatic carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/56Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three 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
    • C07D307/68Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/06Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to the ring carbon atoms
    • C07D333/24Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals

Definitions

  • the present invention relates to small molecules that are potent inhibitors of ⁇ 4 ⁇ 1 mediated adhesion to either VCAM or CS-1 and which could be useful for the treatment of inflammatory diseases.
  • the extracellular matrix is the major component of connective tissue which provides structural integrity, and promotes cell migration and differentiation. As part of these functions, extracellular matrix molecules such as fibronectin, collagen, laminin, von Willebrand factor, thrombospondin, fibrinogen, and tenascin have been shown to support adhesion of cells in vitro. This adhesive interaction is critical for a number of biological processes including hemostasis, thrombosis, wound healing, tumor metastasis, immunity and inflammation.
  • Fibronectin is the prototype ECM molecule.
  • the major cell attachment site in the fibronectin molecule has been reproduced synthetically with the amino acid sequence arginine-glycine-aspartic acid, or RGD using single letter nomenclature.
  • Peptides containing the RGD sequence which either inhibit or promote cell adhesion have been described (U.S. Pat. Nos.
  • the most active cell-binding site within this alternatively spliced region is composed of 25 amino acids where the carboxy terminus contains the sequence EILDVPST
  • the amino acid sequence EILDVPST forms a recognition motif on FN for cell surface receptors.
  • the receptors which recognize these sites on FN belong to a gene superfamily called integrins which consist of heterodimeric complexes of non-covalently associated alpha and beta subunits.
  • a common ⁇ subunit combines with unique a subunits to form an adhesion receptor of defined specificity.
  • 8 ⁇ subunits have been identified which can dimerize with 16 distinct ⁇ subunits forming 22 distinct integrins.
  • the ⁇ 1 subfamily also known as the VLA family (Very Late Activation Antigens), binds to ECM molecules such as FN, collagen and laminin.
  • VLA family Very Late Activation Antigens
  • Leukocyte interaction with FN at the two spatially separate binding domains is mediated by two distinct integrins.
  • the RGD site is recognized by the Integrin ⁇ 5 ⁇ 1
  • EILDV is recognized by ⁇ 4 ⁇ 1 (Pytela et al., Cell 40:191 (1985); Wayner et al., J. Cell Biol. 109:1321 (1989); Guan et al, Cell 60:53 (1990)).
  • Vascular endothelial cells form the interface between blood and tissues and control the passage of leukocytes as well as plasma fluid into tissues.
  • a variety of signals generated at the site of inflammation can activate both endothelial cells as well as circulating leukocytes so that they become more adhesive to one another. Following this initial adhesion the leukocytes migrate into the tissues to perform host defense functions.
  • adhesion molecules have been identified which are involved in leukocyte-endothelial interactions.
  • ⁇ 4 ⁇ 1 interacts with a cytokine inducible protein on endothelial cells termed vascular cell adhesion molecule (VCAM).
  • VCAM vascular cell adhesion molecule
  • ⁇ 2 integrins include CD11a/CD18, CD11b/CD18, and CD11c/CD18.
  • the ⁇ 7 subunit associates with ⁇ 4 to form a unique ⁇ 4 ⁇ 7 heterodimer which binds to FN, to VCAM, and to Mucosal Addressin Cell Adhesion Molecule-1 (MAdCAM) (Ruegg et al, J. Cell.Biol. 117:179 (1992); Andrew et al., J. Immunol. 153:3847 (1994); Briskin et al., Nature 363:461 (1993); Shyjan et al, J. Immunol. 156:2851 (1996)).
  • MAdCAM Mucosal Addressin Cell Adhesion Molecule-1
  • ⁇ 4 integrins are widely expressed on different cell types including hematopoietic progenitors, lymphocytes, natural killer cells, monocytes, eosinophils, basophils, and mast cells (Helmer, M. E., Annu. Rev. Immunol. 8:365 (1990)).
  • Other molecules on endothelial cells which bind to the leukocytes include ICAM-1, ICAM-2, E-selectin and P-selectin (Carlos and Harlan, Immunol. Rev.
  • ⁇ 4 ⁇ 1 plays a critical role in the pathogenesis of a variety of diseases.
  • Monoclonal antibodies directed against ⁇ 4 have been tested in a variety of disease models.
  • Anti- ⁇ 4 antibodies block adhesion of lymphocytes to synovial endothelial cells; this adhesion plays a potential role in rheumatoid arthritis (van Dinther-Janssen et al, J. Immunol. 147:4207 (1991).
  • ⁇ 4 has also been implicated with respect to rheumatoid arthritis in separate studies (Laffon et al, J. Clin. Invest.
  • Anti- ⁇ 4 monoclonal antibodies also inhibit insulitis and delay the onset of diabetes in the non-obese diabetic mouse (Baron et al, J. Clin. Invest. 93:1700 (1994); Yang et al, Proc. Natl. Acad. Sci. USA 90:10494 (1993); Burkly et al, Diabetes 43:529 (1994)).
  • ⁇ 4 is also implicated in atherosclerosis due to its endothelial expression during atherogenesis (Cybuisky et al, Science 251:788 (1991)). The migration of leukocytes to an inflammatory site can also be blocked by anti- ⁇ 4 antibodies.
  • inhibitors of leukocyte endothelial adhesion may block the costimulatory signals mediated by integrins and thus inhibit overproduction of inflammatory cytokines.
  • the peptides GRDGSP or EILDV were tested against contact hypersensitivity response. The contact hypersensitivity response was found to be blocked by GRDGSP or EILDV suggesting that both ⁇ 4 ⁇ 1 , and ⁇ 5 ⁇ 1 are involved in this inflammatory response.
  • ⁇ 4 ⁇ 1 -mediated conditions include the inflammatory disorders rheumatoid arthritis, allergic disorders, asthma, spontaneous chronic colitis, insulitis, contact hypersensitivity response, atherosclerosis and autoimmune encephalomyelitis.
  • inflammatory disorders rheumatoid arthritis, allergic disorders, asthma, spontaneous chronic colitis, insulitis, contact hypersensitivity response, atherosclerosis and autoimmune encephalomyelitis.
  • small molecules that are potent inhibitors of ⁇ 4 ⁇ 1 mediated adhesion to either VCAM-1 or CS-1 may be used as a form of treatment in numerous inflammatory diseases.
  • these inflammatory conditions could be expanded to include adult respiratory distress syndrome, AIDS, cardiovascular diseases, thrombosis or harmful platelet aggregation, reocclusion following thrombolys's, allograft rejection, reperfusion injury, psoriasis, eczema, contact dermatitis and other skin inflammatory diseases, osteoporosis, osteoarthritis, atherosclerosis, neoplastic diseases including metastasis of neoplastic or cancerous growth, wound healing enhancement, treatment of certain eye diseases such as detaching retina, Type I diabetes, multiple sclerosis, systemic lupus erythematosus (SLE), inflammatory and immunoinflammatory conditions including ophthalmic inflammatory conditions and inflammatory bowel diseases, ulcerative colitis, regional enteritis and other autoimmune diseases. Accordingly, a compound which could inhibit these conditions is desirable.
  • n is an integer of 0 or 1
  • R 1 is a hydrogen atom or a methyl group
  • R 2 can be selected from the following: a —CN group; a —COOH group; a —(C 1-6 alkylene)OH group, preferably a —(C 1-3 alkylene)OH group; a CH 2 O(C 1-6 alkyl) group, preferably a —CH 2 O(C 1-3 alkyl) group; a —(C 1-3 alkylene)COOH group, preferably a —(C 1-13 alkylene)COOH group; a —CH 2 O(C 1-6 alkylene)O(C 1-6 alkyl) group, preferably a —CH 2 O(C 1-3 alkylene)O(C 6 alkyl) group or a —CH 2 O(C 1-6 alkylene)O(C 1-3 alkyl) group, more preferably a —CH 2 O(C 1-3 alkyl)
  • a —CONHOCH 2 Ph group a —CONH(C 1-6 alkylene)CN group, preferably a —CONH(C 1-3 alkylene)CN group; a —COO(C 1-6 alkyl) group, preferably a —COO(C 1-3 alkyl) group; a —CH 2 O(C 1-6 alkylene)CONH 2 group, preferably a —CH 2 O(C 1-3 alkylene)CONH 2 group; a —CONH(C 1-6 alkylene)CONH 2 group, preferably a —CONH(C 1-3 alkylene)CONH 2 group; a —CONHOH group; a —NHCOOCH 2 Ph group; or a group selected from the following formula:
  • the C 1-6 alkylene is preferably C 1-3 aklylene
  • the C 2-7 alkenylene is preferably C 2-4 alkenylene
  • the C 1-6 alkyl is preferably C 1-3 alkyl
  • the C 3-7 cycloalkyl is preferably C 3-6 cycloalkyl.
  • R 3 can be a hydrogen atom or a methyl group
  • X can be a methylene group or a —CO— group
  • R 4 can selected from the following: a hydrogen atom; or a C 1-6 alkyl group, preferably a C 1-3 alkyl group.
  • R 5 can be a group selected from the following: a —COOH group or an ester or an amide thereof; a —(C 1-6 alkylene)COOH group, preferably a —(C 1-3 alkylene)COOH group, or an ester or an amide thereof; a —(C 1-7 alkylene)O(C 1-6 alkyl) group, preferably a —(C 1-4 alkylene)O(C 1-6 alkyl) group or a —(C 1-7 alkylene)O(C 1-3 alkyl) group, more preferably a —(C 1-4 alkylene)O(C 1-3 alkyl) group; a —(C 1-7 alkylene)OH group, preferably a —(C 1-4 alkylene)OH group; a —COO(C 1-6 alkyl) group, preferably a —COO(C 1-3 alkyl) group; a —CONH(C
  • R 6 can be a substituted or unsubstituted monocyclic or bicyclic aryl group, a substituted or unsubstituted monocyclic or bicyclic heteroaryl group, a substituted or unsubstituted monocyclic or bicyclic arylcarbonylamino-C 1-6 alkyl group, a substituted or unsubstituted monocyclic or bicyclic aliphatic heterocyclic carbonyl group, a 9- fluorenylmethyloxycarbonylamino —C 1-6 alkyl group, a 3-tosylguanidino-C 1-6 alkyl group;
  • R 1 and R 3 must be different and when R 2 or R 6 is a —COOH group or contains a —COOH group, then a pharmaceutically acceptable ester or a pharmaceutically acceptable amide thereof are included and also with the proviso that [1S-[1, (R*) 3 ⁇ ]]- ⁇ -[[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]amino]-4-(2,6-dichlorobenzoyl)- ⁇ -oxo-1-pyrazinebutanoic acid methyl ester or [1S-[1 ⁇ , (R*),3 ⁇ ]]- ⁇ -[[(3-Carboxy-2,2,3-trimethylcyclopentyl) carbonyl]amino]-4-(2,6-dichlorobenzoyl)- ⁇ -oxo-1-pyrazinebutanoic acid methyl ester are excluded.
  • an aryl group or aryl moiety in the arylcabonylamino group is a 5- or 6-membered aromatic hydrocarbon ring; and including any bicyclic group in which any of the above ring is fused to another above ring; and substituted by zero (0) to three (3) substituents.
  • aryl can include phenyl, a C 1-6 alkoxyphenyl group and naphthyl group. Each of these moieties may be substituted as appropriate.
  • a heteroaryl is a 5- or 6-membered partially saturated or unsaturated ring containing from one (1) to four (4) heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur; and including any bicyclic group in which any of the above heterocyclic rings is fused to a benzene ring, C 3 -C 8 cycloalkyl, or another heterocycle; and if chemically feasible, the nitrogen and sulfur atoms may be in the oxidized forms; and substituted by zero (0) to three (3) substituents.
  • heteroaryl can include 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, 3-pyridazinyl, 4-pyridazinyl, 3-pyrazinyl, 2-quinolyl, 3-quinolyl, 1-isoquinolyl, 3-isoquinolyl, 4-isoquinolyl, 2-quinazolinyl, 4-quinazolinyl, 2-quinoxalinyl, 1-phthalazinyl, 2-imidazolyl, 4-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 3-pyrazolyl, 4-pyrazolyl, 5-pyrazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-indolinyl, 3-indolyl, 3-indazo
  • an aliphatic heterocyclic moiety in aliphatic heterocyclic carbonyl group is a 5- or 6-membered saturated ring containing from one (1) to four (4) heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur; and including any bicyclic group in which any of the above heterocyclic rings is fused to a benzene ring, C 3 -C 8 cycloalkyl, or another heterocycle; and if chemically feasible, the nitrogen and sulfur atoms may be in the oxidized forms; and substituted by zero (0) to three (3) substituents.
  • Examples of aliphatic heterocyclic can include piperazinyl group, pyrrolidinyl group, piperidyl group, homopiperidyl group, thiomorpholino group, and morpholino group. Each of these moieties may be substituted as appropriate.
  • C 1-6 alkyl represents an alkyl group having 1 to 6 carbon atoms. This group may be straight or branched. Illustrative but non-limiting examples of a C 1-6 alkyl group are methyl, ethyl, n-propyl, isopropyl, n-butyl, iso-butyl, tert-butyl, sec-butyl, n-pentyl, isopentyl and n-hexyl. It is understood that this type of nomenclature extends to terms such as “C 1-6 methoxy” and therefore encompasses both straight and branced methoxy groups having 1 to 6 carbon atoms.
  • the desired compounds of the present invention have preferred steric configurations. Accordingly, a preferred steric configuration is represented by compounds of the formula [I-1]:
  • n, R 1 through R 6 and X are as defined above.
  • R 5 can be selected from the following: a —COOH group; a —(C 1-6 alkylene)COOH group, preferably a —(C 1-3 alkylene)COOH group; a —(C 1-7 alkylene)O(C 1-6 alkyl) group, preferably a —(C 1-4 alkylene)O(C 1-6 alkyl) group or a —(C 1-7 alkylene)O(C 1-3 alkyl) group, more preferably a —(C 1-4 alkylene)O(C 1-3 alkyl) group; a —(C 1-7 alkylene)OH group, preferably a —(C 1-4 alkylene)OH group; a —COO(C 1-6 alkyl) group, preferably a —COO(C 1-3 alkyl) group; a —CONH(C 1-6 alkyl) group
  • R 6 can be selected from the following formula:
  • R 7 which occurs one or more times and which may be the same or different in each occurrence, may be selected from the following: a —H group; a —OH group; a —NO 2 group; a —NH 2 group; a —C 1-5 alkyl group, preferably a —C 1-3 alkyl group; a —F group; a —Cl group; a —Br group; a —I group; a —COOH group; a —COO(C 1-6 alkyl) group, preferably a —COO(C 1-3 alkyl) group; a —O(C 1 -C 8 alkyl) group, preferably a —O(C 1-4 alkyl) group; a CONH(C 1-6 alkylene)COOH group, preferably a —CONH(C 1-3 alkylene)COOH group; a —OCH 2 (C 3-7 cycloalkyl)
  • R 8 which occurs one or more times and which may be the same or different in each occurrence, may be selected from the following: a —H group; an —OH group; a —NH 2 group; a —NO 2 group; a —C 1-7 alkyl group, preferably a —C 1-4 alkyl group; a —F group; a —Cl group; a —Br group; a —I group; a —CF 3 group; a phenyl group, or a —O(C 1-6 alkyl) group, preferably a —O(C 1-3 alkyl) group.
  • R 9 may be selected from the following: a —H group; a —Cl alkyl group, preferably a —C 1-3 alkyl group; a —C 3-7 cycloalkyl group, preferably a —C 3-6 cycloalkyl group; a —(C 1-6 alkylene)aryl group, preferably a —(C 1-3 alkylene)aryl group; an aryl group; or a group selected from the following formula:
  • R 10 which occurs one or more times and which may be the same or different in each occurrence, may be selected from the following: a —H group; a —F group; a —Cl group; a —Br group; an —I group; a —NO 2 group; a —C 1-6 alkyl group, preferably a —C 1-3 alkyl group; or a —O(C 1-6 alkyl) group, preferably a —O(C 1-3 alkyl) group.
  • R 11 may be selected from the following:
  • R 12 which occurs one or more times and which may be the same or different in each occurrence, may be selected from the following: a —H group; a —CF 3 group; a —OCH 3 group; a —F group; a —Br group; a —Cl group; or an —I group;
  • R 9 is other than hydrogen.
  • R 6 is selected from the following:
  • Y is selected from either a hydrogen atom or a chlorine atom.
  • R 2 is selected from the following: a —COOH group or an ester or an amide thereof; a —CONHCH 2 COOH group; a —CONHOCH 2 Ph group; or a —CONHCH 2 CONH 2 group.
  • R 1 is a —CH 3 group
  • R 2 is a —COOH group
  • a —CONHCH 2 COOH group a CONHOCH 2 Ph group or a —CONHCH 2 CONH 2 group
  • R 3 and R 4 are hydrogen atoms.
  • X is —CO—
  • R 5 is —COOH
  • n is 1, and
  • R 6 is represented by the following formula
  • R 8 occurs 2 or 3 times and is a chlorine atom.
  • R 1 may be a hydrogen atom or a methyl group.
  • R 2 may be selected from the following: a —CN group; a —COOH group; a —CONH 2 group; a —CONHOH group; a —CON(CH 3 ) 2 group; a —CH 2 OCH 2 COOH group; a —CH ⁇ CHCOOH group; a —CONHCH 2 COOH group; a —CONH(CH 2 ) 2 COOH group; a CONHCH 2 CONH 2 group; a —CONH(CH 2 ) 2 CN group; a group selected from the following:
  • R 3 may be a hydrogen atom or a methyl group and R 5 may be a —COOH group or a COOMe group.
  • R 6 may be selected from the following:
  • Z 1 may be selected from the following: a —O— group; a —NHCO— group; a —NHCH 2 — group, a —OCH 2 — group; a —CONH-group; a —NHSO 2 — group; a —NHCOCH 2 — group; or a —N(CH 3 )CH 2 — group.
  • R 13 may be selected from the following: a —H group; a -iBuO group; a —CH 3 group; a i-Bu group; or a group selected from the following:
  • R 14 which occurs one or more times and which may be the same or different in each occurence, may be selected from the following: a —H group; a —F group; a —Cl group; a —Br group; an —I group; a —CH 3 group; a —OCH 3 group; a —CF 3 group; a —NO, group; a —NH 2 group; or a -n-C 7 H 15 group.
  • R 15 may be selected from the following: a —H group; a —OH group; a —NO 2 group; or a group selected from the following:
  • R 16 may be selected from the following
  • R 17 which occurs one or more times and which may be the same of different in each occurrence, may be selected from the following: a —H group; a —Cl group; a —OCH 3 group; or a —CF 3 group, provided that R 1 and R 3 must be different.
  • R 1 is a hydrogen atom or a methyl group
  • R 2 is selected from the following: a —CN group; a —COOH group; a —COOMe group; a —CONH 2 group; a —CONHOH group; a —CON(CH 3 ) 2 group; a —CH 2 OCH 2 COOH group; a —CH ⁇ CHCOOH group; a —CONHCH 2 COOH group; a —CONH(CH 2 ) 2 COOH group; a —CONHCH 2 CONH 2 group; a —CONH(CH 2 ) 2 CN group; or a group selected from the following:
  • R 3 is a hydrogen atom or a methyl group.
  • R 5 is a —COOH group or a —COOMe group and R 6 is selected from the following:
  • Z 1 may be selected from the following: a —O— group; a —NHCO— group; a —NHCH 2 — group; a —OCH 2 — group; a —CONH— group; a —NHSO 2 — group; or a —NHCOCH 2 — group.
  • R 13 may be selected from the following: a —H group; a -iBuO group; a -i-Bu group; or a group selected from the following:
  • R 14 which occurs one or more times and which may be the same or different in each occurence, may be selected from the following: a —H group; a —F group; a —Cl group; a —Br group; an —I group; a —CH 3 group; a —OCH 3 group; a —CF 3 group; a —NO 2 group; a —NH, group; or a -n-C 7 H 15 group.
  • R 15 may be selected from the following: a —H group; a —OH group; a —NO 2 group; or a group selected from the following:
  • R 16 may be selected from the following:
  • R 17 which occurs one or more times and which may be the same of different in each occurrence, is a chlorine atom, provided that R 1 and R 3 must be different.
  • R 1 is methyl group and R 2 may be selected from the following: a —CN group; a —COOH group; a —CONH 2 group; a —CONHOH group; a —CH 2 OCH 2 COOH group; a —CH ⁇ CHCOOH group; a —CONHCH 2 COOH group; a —CONH(CH 2 ) 2 COOH group; a —CONHCH 2 CONH 2 group; a —CONH(CH 2 ) 2 CN group; or a group selected from the following:
  • R 3 is a hydrogen atom
  • R 5 is a —COOH group or a —COOMe group
  • R 6 may be selected from the following:
  • Z 1 may be selected from the following: a —NHCO— group; a —OCH 2 -group; a —NHCH 2 — group; a —CONH— group; or a —NHSO 2 -group.
  • R 13 may be selected from the following:
  • R 14 which occurs one or more times and which may be the same or different in each occurence, may be selected from the following: a —F group; a —Cl group; a —Br group; an —I group; a —CH 3 group; a —OCH 3 group; a —CF 3 group; or a —NO 2 group.
  • R 15 may be a —H group or a —OH group.
  • R 1 is a hydrogen atom or a methyl group and R 2 may be selected from the following: a —CN group; a —COOH group; a —CONH 2 group; a —CONHOH group; a —CONHOCH 3 group; a —CH 2 OCH 2 COOH group; a —CH ⁇ CHCOOH group; a —CONHCH 2 COOH group; a —CONH(CH 2 ) 2 COOH group; a —CONHCH 2 CONH 2 group; a —CONH(CH 2 ) 2 CN group; or a group selected from the following:
  • R 3 is a hydrogen atom or a methyl group
  • R 5 is a —COOH group or a —COOMe group
  • R 6 may be selected from the following:
  • Z 1 may be selected from the following: a —O— group; a —NHCO— group; a —NHCH 2 — group; a —OCH 2 — group; a —CONH— group; or a —NHSO 2 — group.
  • R 13 may be selected from the following: a —H group; a -iBuO group; or a group selected from the following:
  • R 14 which occurs one or more times and which may be the same or different in each occurence, may be selected from the following: a —H group; a —OH; a —F group; a —Cl group; a —Br group; an —I group; a —CH 3 group; a —OCH 3 group; a —CF 3 group; a —NO 2 group; or a —NH 2 group.
  • R 15 may be selected from the following: a —H group; a —OH group; a —NO 2 group; or a group selected from the following:
  • R 16 may be selected from the following:
  • R 17 which occurs one or more times and which may be the same or different in each occurrence, may be a —Cl group or a —CF 3 group, provided that R 1 and R 3 must be different.
  • R 1 may be a hydrogen atom or methyl group
  • R 2 may be selected from the following: a —CN group; a —COOH group; a —CONH 2 group; a —CONHOH group; a —CH 2 OCH 2 COOH group; a —CH ⁇ CHCOOH group; a —CONHCH 2 COOH group; a —CONH(CH 21 ) 2 COOH group; a —CONHCH 2 CONH 2 group; a —CONH(CH 2 ) 2 CN group; or a group selected from the following:
  • R 3 may be a hydrogen atom or a methyl group.
  • R 1 is —COOH group or a —COOMe group and R 6 may be selected from the following:
  • Z 1 may be selected from the following: a —O— group; a —NHCO— group; a —NHCH 2 — group; a —OCH 2 — group; a —CONH— group; or a —NHSO 2 — group.
  • R 13 may be a —H group or a group selected from the following:
  • R 14 which occurs one or more times and which may be the same or different in each occurence, may be selected from the following: a —H group; a —F group; a —Cl group; a —Br group; an —I group; a —CH 3 group; a —OCH 3 group; a —CF 3 group; a —NO 2 group; or a —NH 2 group.
  • R 15 may be selected from the following: a —H group; a —OH group, a —NO 2 group; or a group selected from the following:
  • R 16 may be selected from the following:
  • R 17 which occurs one or more times and which may be the same or different in each occurrence, may be a —H group or a —Cl group, provided that R 1 and R 3 must be different.
  • R 1 is a methyl group
  • R 2 may be selected from the following: a —CN group; a —COOH group; a —CONH 2 group; a —CONHOH group; a —CH 2 OCH 2 COOH group; a —CH ⁇ CHCOOH group; a —CONHCH 2 COOH group; a —CONH(CH 2 ) 2 COOH group; a —CONH(CH 2 ) 2 CN group; a —CONHCH 2 CONH 2 group; or a group selected from the following:
  • R 3 is a hydrogen atom
  • R 5 is a —COOH group or a —COOMe group
  • R 6 may be selected from the following:
  • Z 1 may be selected from the following: a —NHCO— group; a —NHCH 2 — group; a —NAcCH 2 — group; a —OCH 2 — group; or a —CONH-group.
  • R 13 may be selected from the following:
  • R 14 which occurs one or more times and which may be the same or different in each occurence, may be selected from the following: a —F group; a —Cl group; a —Br group; an —I group; a —OCH 3 group; a —CF 3 group; or a —NO 2 group.
  • R 1 is a —H group or a —NO 2 group.
  • Preferred compounds according to formula [I] may be selected from the group consisting of (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)-carbonyl]-1-[(3,4-dichlorophenyl)methyl]-L-histidine, (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine, (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine, (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-[(2,6-dichlorophenyl)methyl]-3-nitro
  • the desired compounds of the present invention may exist in the form of optical isomers based on asymmetric carbon atoms thereof, and the present invention also includes these optical isomers and mixtures thereof.
  • the steric configuration of a bond need not be fixed.
  • a bond may be of any acceptable configuration.
  • a compound may be a mixture with several different configurations of the same bond.
  • the desired compounds of the present invention may be used in the form of an ester or amide thereof.
  • a C 1-6 alkyl ester there may be mentioned a C 2-7 alkenyl ester, a C 2-7 alkynyl ester, a C 2-7 alkanoyloxy-C 1-6 alkyl ester, an aryl-C 1-6 alkyl ester or an aryl ester.
  • the amide thereof there may be mentioned an, amide (—CONH 2 ), a mono or di N—C 1-6 alkyl amide, an N—C 3-8 cycloalkyl amide, an N-aryl amide or an N-aryl-C 1-6 alkyl amide.
  • the desired compound of the present invention may be clinically used either in a free form or in the form of pharmaceutically acceptable salts thereof.
  • Pharmaceutically acceptable salts include acid-addition salts with inorganic acid or organic acid (e.g., hydrochloride, sulfate, nitrate, hydrobromide, methanesulfonate, p-toluenesulfonate, acetate), salt with inorganic base, organic base or amino acid (e.g., triethylamine salt, a salt with lysine, an alkali metal salt, an alkali earth metal salt and the like).
  • the compound may also be formulated into a pharmaceutical composition comprising a therapeutically effective amount of the compound as defined above and a pharmaceutically acceptable carrier or diluent.
  • the compound can also be used for treating or preventing ⁇ 4 ⁇ 1 adhesion mediated conditions in a mammal such as a human.
  • This method may comprise administering to a mammal or a human patient an effective amount of the compound or composition as explained above.
  • This method can be used to treat such inflammatory conditions as rheumatoid arthritis, asthma, allergy conditions, adult respiratory distress syndrome, AIDS, cardiovascular diseases, thrombosis or harmful platelet aggregation, reocclusion following thrombolysis, allograft rejection, reperfusion injury, psoriasis, eczema, contact dermatitis and other skin inflammatory diseases, osteoporosis, osteoarthritis, atherosclerosis, neoplastic diseases including metastasis of neoplastic or cancerous growth, wound healing enhancement, treatment of certain eye diseases such as detaching retina, Type I diabetes, multiple sclerosis, systemic lupus erythematosus (SLE), inflammatory and immunoinflammatory conditions including ophthalmic inflammatory conditions and inflammatory bowel diseases, ulcerative colitis, atherosclerosis, regional enteritis and other autoimmune diseases.
  • inflammatory conditions as rheumatoid arthritis, asthma, allergy conditions, adult respiratory distress syndrome, AIDS, cardiovascular diseases,
  • the desired compound of the present invention or pharmaceutically acceptable salts thereof may be administered either orally or parenterally, and it may be used as a suitable pharmaceutical preparation, for example, a tablet, a granule, a capsule, a powder, an injection, and an inhalation by a conventional process.
  • the dose of the desired compound of the present invention or a pharmaceutically acceptable salt thereof varies depending on an administration method, age, body weight, and state of a patient, but, in general, the daily dose is preferably about 0.1 to 100 mg/kg/day, particularly preferably 1 to 100 mg/kg/day.
  • the compound of the present invention be administered in the form of an Aerosol.
  • routes of administration include intravenous, oral, intramuscular, and subcutaneous.
  • compositions containing the compounds of the present invention can be prepared to provide for an excellent means for administering in aerosol form for inhalation therapy. Accordingly, the present invention will provide for self-propelling compositions containing the compounds of the present invention.
  • Propellants employed should be non-toxic and have a vapor pressure suitable for the conditions under which administration occurs.
  • These propellants can be fluorinated or fluorochlorinated lower saturated aliphatic hydrocarbons.
  • the preferred propellants of this type are the halogenated alkanes containing not more than two carbon atoms and at least one fluorine atom. Illustrative of these are trichloromonofluoromethane, dichlorodifluoromethane, monochlorotrifluoromethane, dichloromonofluoromethane and 1,2-dichloro-1,1,2,2-tetrafluoroethane. These compounds are available from E. I. duPont de Nemours and Company under the trade name “Freon”. These propellants may be employed singularly or in admixture.
  • an organic solvent may also be employed.
  • the organic solvent must be non-toxic and without undesirable effects on inhalation in the amount present in the aerosol produced.
  • the solvent should be substantially anhydrous, completely miscible with the propellant or mixture of propellants employed and have a suitable boiling point. Examples of such solvents included non-toxic aliphatic alcohols such as ethanol; ethers such as ethyl ether and vinyl ether; ketones such as acetone; and suitable halogenated lower alkanes.
  • the composition may also optionally contain a non-toxic hygroscopic glycol.
  • the glycol must be substantially miscible with the organic solvent and the propellant employed. Satisfactory glycols include propylene glycol, triethylene glycol, glycerol, butylene glycol and hexylene glycol.
  • the compounds of formula (I) can be formulated into pharmaceutical compositions.
  • the particular disease in question, its severity, as well as the age, sex, weight, and condition of the subject to be treated, must be taken into consideration and this perusal is to be determined by the skill of the attendant physician.
  • a suitable daily dose of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for a mammalian subject suffering from, or likely to suffer from, any condition as described hereinbefore is 0.1 mg to 100 mg of the compound of formula I, per kilogram body weight of the mammalian subject.
  • the dose may be in the range of 0.5 to 500 mg of the compound per kilogram body weight, the most preferred dosage being 0.5 to 50 mg/kg of mammal body weight administered two to three times daily.
  • a suitable dose may be in the range of 0.1 ⁇ g to 100 ⁇ g of the compound per kilogram, typically about 0.1 ⁇ g/kg.
  • a suitable dose of a compound of Formula (I), or a physiologically acceptable salt thereof may be as specified in the preceding paragraph, but most preferably is from 1 mg to 10 mg of the compound per kilogram, the most preferred dosage being from 1 mg to 5 mg/kg of mammal body weight, for example, from 1 to 2 mg/kg.
  • a unit dosage of an orally administrable composition encompassed by the present invention contains less than about 1.0 g of a formula (I) compound.
  • formulation, both for human and veterinary use, of the present invention may be presented to the mammal by inhalation.
  • the dose may be in the range of 0.5 to 500 mg of the compound, per kg body weight.
  • the most preferred dosage being 0.5 to 50 mg/kg of mammal body weight administered two to three times daily.
  • the compounds and compositions of the present invention can be administered to patients suffering from a condition listed herein in an amount which is effective to fully or partially alleviate undesired symptoms of the condition.
  • the symptoms may be caused by inappropriate cell adhesion mediated by 0401 integrins.
  • Such inappropriate cell adhesion would typically be expected to occur as a result of increased VCAM-1 and/or CS-1 expression on the surface of endothelial cells.
  • Increased VCAM-1 and/or CS-1 expression can be due to a normal inflammation response or due to abnormal inflammatory states.
  • an effective dose of a compound of the invention may reduce the increased cell adhesion due to increased VCAM-1 expression by endothelial cells.
  • Reducing the adhesion observed in the disease state by 50% can be considered an effective reduction in adhesion. More preferably, a reduction in adhesion by 90%, is achieved. Most preferably adhesion mediated by VCAM-1/U40, and/or CS-1 interaction is abolished by an effective dose. Clinically, in some instances, effect of the compound can be observed or a decrease in white cell infiltration into tissues or a site of injury. To achieve a therapeutic effect, then, the compounds or compositions of the present invention are administered to provide a dose effective to reduce or eliminate inappropriate cell adhesion or to alleviate undesired symptoms.
  • an active ingredient While it is possible for an active ingredient to be administered alone, it is preferable to present it as a pharmaceutical formulation comprising a compound of Formula (I) and a pharmaceutically acceptable carrier thereof. Such formulations constitute a further feature of the present invention.
  • the formulations, both for human and veterinary medical use, of the present invention comprise an active ingredient of Formula (I), in association with a pharmaceutically acceptable carrier thereof and optionally other therapeutic ingredient (s), which are generally known to be effective in treating the disease or condition encountered.
  • the carrier(s) must be “acceptable” in the sense of being compatible with the other ingredients of the formulations and not deleterious to the recipient thereof.
  • the formulations include those in a form suitable for oral, pulmonary, ophthalmic, rectal, parenteral (including subcutaneous, intramuscular, and intravenous), intra-articular, topical, nasal inhalation (e.g., with an aerosol) or buccal administration.
  • parenteral including subcutaneous, intramuscular, and intravenous
  • intra-articular including subcutaneous, intramuscular, and intravenous
  • nasal inhalation e.g., with an aerosol
  • buccal administration Such formulation are understood to include long-acting formulations known in the art.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. All methods may include the step of bringing the active ingredient into association with the carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired form.
  • Formulations of the present invention suitable for oral administration may be in the form of discrete units such as capsules, cachets, tablets, or lozenges, each containing a predetermined amount of the active ingredient in the form of a powder or granules; in the form of a solution or suspension in an aqueous liquid.
  • Formulations for other uses could involve a nonaqueous liquid; in the form of an oil-in-water emulsion or a water-in-oil emulsion; in the form of an aerosol; or in the form of a cream or ointment or impregnated into a transdermal patch for use in administering the active ingredient transdermally, to a patient in need thereof.
  • the active ingredient of the present inventive compositions may also be administered to a patient in need thereof in the form of a bolus, electuary, or paste.
  • the desired compound [I] can be prepared by the following methods:
  • R 5a is a group of the formula: —COOR 22 , —(C 1-6 alkylene)COOR 22 , C 1-7 alkylene)O(C 1-6 alkyl), —(C 1-7 alkylene) OH, —COO(C 1-6 alkyl)-CONH(C 1-6 alkyl) or —CONH 2 ,
  • R 22 is a protecting group for the carboxyl group, and the other symbols are the same as defined above.
  • R 22 can be selected from a conventional protecting group for a carboxyl group, for example, a C 1-6 alkyl group, a C 2-7 alkenyl group, a C 2-7 alkynyl group, a C 2-7 alkanoyloxy-C 1-6 alkyl group, an aryl-C 1-6 alkyl group (e.g., benzyl group) or an aryl group (e.g., phenyl group) and the like.
  • a conventional protecting group for a carboxyl group for example, a C 1-6 alkyl group, a C 2-7 alkenyl group, a C 2-7 alkynyl group, a C 2-7 alkanoyloxy-C 1-6 alkyl group, an aryl-C 1-6 alkyl group (e.g., benzyl group) or an aryl group (e.g., phenyl group) and the like.
  • a salt of the compound [III-a] includes, for example, salt with an inorganic acid (e.g., hydrochloride, sulfate) and salt with an inorganic base (e.g., an alkali metal salt such as sodium salt or potassium salt, an alkali earth metal salt such as magnesium salt or calcium salt).
  • an inorganic acid e.g., hydrochloride, sulfate
  • an inorganic base e.g., an alkali metal salt such as sodium salt or potassium salt, an alkali earth metal salt such as magnesium salt or calcium salt.
  • the reaction of the compound [II] and the compound [III-a] or a salt thereof is carried out in the presence of a base in a suitable solvent or without a solvent.
  • the base can be selected from an organic base (e.g., DIEA, DMAP, Et 3 N, DBU), an alkali metal hydride (e.g., NaH, LiH), an alkali metal carbonate (e.g., Na 2 CO 3 , Na 2 KO 3 ) an alkali metal hydrogen carbonate (e.g., NaHCO 3 , KHCO 3 ), an alkali metal amide (e.g., NaNH 2 ), an alkali metal alkoxide (e.g., NaOMe, KOMe), an alkyl-alkali metal (n-BuLi, t-BuLi), an alkali metal hydroxide (e.g., NaOH, KOH), an alkali earth metal hydroxide (e.g., Ba(OH) 2 ), and
  • the solvent can be selected from any one which does not disturb the reaction, for example, DMF, THF, benzene, toluene, DMSO, CH 3 CN or a mixture thereof.
  • the reaction is preferably carried out at a temperature from 0° C. to 100° C., more preferably at a temperature from 400% to 80° C.
  • the removal of said protecting group from the products can be carried out by a conventional method, which is selected according to the types of the protecting groups to be removed, for example, hydrolysis, acid treatment, catalytic reduction, and the like.
  • a more preferred method than method A is:
  • R 2a is a group of the formula: —CN, —COOR 23 , —COOH, —(C 1-6 alkylene)OH, —CH 2 O(C 1-6 alkyl), —(C 1-6 alkylene)COOH, —(C 1-6 alkylene)COOR 23 , —CH 2 C(C 1-6 alkylene)O(C 1-6 alkyl), —CH 2 O(C 1-6 alkylene) COOH, —CH 2 O(C 1-6 alkylene) COOR 23 , —(C 2-7 alkenylene) COOH, —(C 2-7 alkenylene) COOR 23 , —CO(C 1-6 alkylene)COOH, —CO(C 1-6 alkylene)COOR 23 , —CO(C 2-7 alkenylene)COOH, —CO(C 1-6 alkylene)COOR 23 , —CO(C 2-7 alkenylene)COOH, —CO(C 1-6 alkylene)CO
  • R is a group of the formula: —CN, —COOR 23 , —(C 1-6 alkylene) OH, —CH 2 O(C 1-6 alkyl), —(C 1-6 alkylene) COOR 23 , —CH 2 O(C 1-6 alkylene)O(C 1-6 alkyl), —CH 2 O(C 1-6 alkylene)COOR 23 , —(C 2-7 alkenylene) COOR 23 , —CO(C 1-6 alkylene)COOR 23 , —CO(C 2-7 alkenylene) COOR 23 , —CO(C 1-6 alkylene)O(C 1-6 alkyl), —CO(C 1-6 alkylene)CO(C 1-6 alkyl), —CONH(C 1-6 alkyl), —CONHO(C 1-6 alkyl), —CONH(C 1-6 alkylene)COOR 23 , —CONH(C 3-7 cycloalkyl), —CONH 2
  • R 23 is a protecting group for the carboxyl group, and the other symbols are the same as defined above.
  • R 2 is a group of the formula: —CN, —COOR 23 , —COOH, —(C 1-6 alkylene) OH, —CH 2 O(C 1-6 alkyl), —(C 1-6 alkylene)COOH, —(C 1-6 alkylene)COOR 23 , —CH 2 O(16 alkylene)O(C 1-16 alkyl), —CH 2 O(C 1-6 alkylene)COOH, —CH 2 O(C 1-16 alkylene)COOR 23 , —(C 2-7 alkenylene)COOH, —(C 2-7 alkenylene)COOR 23 , —CO(C 1-16 alkylene)COOH, —CO(C 1-16 alkylene)COOR 23 , —CO(C 2-7 alkenylene)COOH, —CO(C 1-27 alkenylene)COOR 23 , —CO(C 1-6 alkylene)O(C 1-6 alkyl), —CO
  • X is a group of the formula: —CO—, i.e., the compound of the formula-[I-c]:
  • R 22 and R 23 are the same or different conventional protecting group for a carboxyl group, for example, a C 1-6 alkyl group, a C 2-7 alkenyl group, a C 2-7 alkynyl group, a C 2-7 alkanoyloxy-C 1-6 alkyl group, an aryl-C 1-6 alkyl group (e.g., benzyl group) or an aryl group (e.g., phenyl group) and the like.
  • a carboxyl group for example, a C 1-6 alkyl group, a C 2-7 alkenyl group, a C 2-7 alkynyl group, a C 2-7 alkanoyloxy-C 1-6 alkyl group, an aryl-C 1-6 alkyl group (e.g., benzyl group) or an aryl group (e.g., phenyl group) and the like.
  • a salt of the compound [III-a] and/or [IV] includes, for example, salt with an inorganic acid (e.g., hydrochloride, sulfate) and salt with an inorganic base (e.g., an alkali metal salt such as sodium, potassium and calcium, an alkali earth metal salt such as barium).
  • an inorganic acid e.g., hydrochloride, sulfate
  • an inorganic base e.g., an alkali metal salt such as sodium, potassium and calcium, an alkali earth metal salt such as barium.
  • the condensation reaction of the compound [IV] or a salt thereof with the compound [III-a] or a salt thereof is carried out in the presence of a condensing reagent in a suitable solvent or without a solvent.
  • the condensing reagent can be selected from any one which can be used for a conventional peptide synthesis, for example, BOP-Cl, BOP reagent, DCC and WSCI.
  • the solvent can be selected from any one which does not disturb the condensation reaction, for example, CH 2 Cl 2 , DMF or a mixture thereof.
  • the reaction is preferably carried out at a room temperature.
  • the removal of said protecting group from the products can be carried out by a conventional method, which is selected according to the types of the protecting groups to be removed, for example, hydrolysis, acid treatment, catalytic reduction, and the like.
  • a salt of the compound [V] and/or [III-b] includes, for example, salt with an inorganic acid (e.g., hydrochloride, sulfate) and salt with an inorganic base (e.g., an alkali metal salt, an alkali earth metal salt).
  • an inorganic acid e.g., hydrochloride, sulfate
  • an inorganic base e.g., an alkali metal salt, an alkali earth metal salt
  • the reductive alkylation of the compound [V] or a salt thereof with the compound [III-b] or a salt thereof is carried out by a conventional method in the presence of a reducing agent in a suitable solvent or without a solvent.
  • the reducing agent is preferably sodium borohydride, sodium cyanoborohydride, and the like.
  • the solvent can be selected from any one which does not disturb the reaction, for example, alkanol such as methanol, alkanoic acid such as AcOH, THF or a mixture thereof.
  • the reaction is preferably carried out at a temperature from 0° C. to a room temperature.
  • reaction of the compound [I-f] or a salt thereof and the compound [VII] is carried out in the presence of an acid acceptor in a suitable solvent or without a solvent.
  • the acid acceptor and the solvent can be selected from the base or the solvent used in Method A.
  • the reaction is preferably carried out at room temperature.
  • the removal of said protecting group from the products can be carried out by a conventional method, which is selected according to the types of the protecting groups to be removed, for example, hydrolysis, acid treatment, catalytic reduction, and the like.
  • the desired compound [I] of the present invention can be converted to each other.
  • Such conversion of the present compound [I] into the other compound [I] may be carried out by selecting one of the following procedures from (a) to (e) according to the type of substituent thereof, and if desired, followed by removing the protecting group for the carboxyl group by a conventional method.
  • the compound [I] wherein R 6 is an amino-substituted aryl group can be prepared by the reduction of the compound [I] wherein the corresponding R 6 is an aralklyoxycarbonyl amino group- or nitro-substituted aryl group.
  • the reduction can be, for example, a, catalytic reduction using a palladium catalyst such as palladium on an activated carbon, a platinum catalyst such as platinum oxide, and the like.
  • the catalytic reduction is preferably carried out at a room temperature.
  • R 2 is a group of the formula: —CONH 2 , —CONH(C 1-6 alkyl), —CONHO(C 1-6 alkyl), —CONH(C 1-6 alkylene)COOR 23 , —CONH(C 3-7 cycloalkyl), —CONH(C 1-6 alkylene) CONH 2 , —CONHOCH 2 Ph, —CONH(C 1-6 alkylene)CN, CONHOH,
  • [0180] can be prepared by reacting the compound [I] wherein the corresponding R 2 is a group of the formula: —COOH with a substituted or unsubstituted amine selected from a group of the formula: NH 3 , NH 2 (C 1-6 alkyl), NH 2 O(C 1-6 alkyl), NH 2 (C 1-6 alkylene)COOR 23 , NH 2 (C 3-7 cycloalkyl), NH 2 (C 1-6 alkylene)CONH 2 , NH 2 OH, NH 2 OCH 2 Ph, NH 2 (C 1-6 alkylene)CN,
  • R 23 is defined as above, in the presence of a condensing reagent (e.g., BOP reagent) which can be used for a conventional peptide synthesis, and removing the protecting group for the carboxyl group, if desired.
  • a condensing reagent e.g., BOP reagent
  • the reaction is preferably carried out at a temperature from 0° C. to a room temperature.
  • the compound [I] wherein R 4 is a C 1-6 alkyl group can be prepared by reacting the compound [I] wherein the corresponding R 4 is a hydrogen atom with a C 1-6 alkyl halide (e.g., methyl iodide, butyl iodide) in the presence of metal hydride (e.g., NaH).
  • a C 1-6 alkyl halide e.g., methyl iodide, butyl iodide
  • metal hydride e.g., NaH
  • R 6 is a C 2-6 alkanoylamino-, C 3-7 cycloalkylcarbonylamino-, aryl C 2-7 alkanoylamino-, arylcarbonylamino-, C 1-5 alkyloxycarbonylamino-, C 3-7 cycloalkyloxycarbonylamino-, aryl C 1-6 alkyloxycarbonylamino-, arylureido, or arylsulfonylamino-substituted aryl group can be prepared by reacting the compound [I] wherein the corresponding R 6 is an amino aryl group or a (C 1-6 alkyl)-amino-substituted aryl group with a C 2-16 alkanoic acid, an anhydride of C 2-6 alkanoic acid, C 2-6 alkanoyl halide, C 3-7 cycloalkcanecarboxylic acid, anhydride of
  • R 24 is a protecting group for the amino group, and the other symbols are the same as defined above.
  • [0190] can be prepared by condensing the compound [I] wherein the corresponding R 2 is a group of the formula: —COOH with a group of the formula [VIII]:
  • R 24 can be selected from a conventional protecting group for an amino group, for example, tert-butoxy-carbonyl grop (BOC), benzyloxycarbonyl group (Cbz) and the like.
  • BOC tert-butoxy-carbonyl grop
  • Cbz benzyloxycarbonyl group
  • the solvent used for the Procedures (a) to (e) may be selected from any one which does not disturb the procedures, for example, THF, methanol, DMF, CH 2 Cl, or a mixture thereof.
  • the compound [II] may be prepared by reacting a compound of the formula [VI]:
  • C 1-6 alkanoyl halide e.g., AcCl
  • C 1-6 alkanoic anhydride e.g., Ac 2 O
  • the compound [III-a] may be prepared by a conventional method, which is selected according to the types of the substituents, for example, by the following schemes:
  • R is (1) a substituted or unsubstituted monocyclic or bicyclic aryl group or (2) a substituted or unsubstituted monocyclic or bicyclic heteroaryl group,
  • Hal is a halogen atom and other symbols are the same as defined above.
  • the compound [IV] may be prepared as shown in various locations of the present application, for example, in Schemes 7, 8, 9, 10 and 11.
  • Example 12 (525 mg, 95%) as a pale yellow foam: 1 H NMR (300 MHz, Acetone-d 6 ), (major isomer) ⁇ 7.4-7.5 (m, 3H), 7.23 (d, 2H), 6.99 (d, 2H), 5.30 (d, 1H), 5.28 (s, 2H), 4.6-4.7 (m, 1H), 3.13 (dd, 1H), 2.97 (dd, 1H), 2.81 (t, 1H), 2.5-2.6 (m, 1H), 2.0-2.1 (m, 1H), 1.6-1.7 (m, 1H), 1.3-1.4 (m, 1H), 1.27 (s, 3H), 1.19 (s, 3H), 0.80 (s, 3H); 13 C NMR (75 MHz, Acetone-d 6 ), (major isomer) ⁇ 177.79, 173.99, 172.94, 158.53, 137
  • Benzyl bromide is eluted through neutral alumina (10 mL in a 30 mL sintered glass funnel) to give a colorless liquid (15 mL, 126 mmol) which is added to a stirred solution of (1R,3S)-camphoric acid 1-(1,1-dimethylethyl) ester (15-D) (30 g, 117 mmol), N,N-diisopropylethyl amine (24 mL, 138 mmol), and acetonitrile (90 ml). After seven days, the mixture is filtered to give a white solid (diisopropylethyl amine hydrobromide) and a yellow liquid which is placed in the freezer. After two days, the mixture is filtered (two 50 mL diethyl ether rinses) to give a white solid (24 g, 59% yield).
  • Example 181 is isolated by suction filtration (with water washes) to give Example 181 as a white solid 0.4 g, 58% yield)
  • the mixture is allowed to stir for 48 hours as the ice melts and the solution warms to room temperature
  • the solution is cast into methylene chloride (0.25L) and 1N aq. NaOH (0.25L).
  • the organic phase is separated and washed in order with 1N aq. HCl (0.25L), water (5 ⁇ 0.25L), and brine (0.25L).
  • the organic phase is dried and concentrated in vacuo to give the crude amide as an ivory powder.
  • Benzyl ester 9-C (0.3 g, 0.83 mmol) was dissolved in absolute EtOH (10 mL). The solution was treated with a catalytic amount of 10% Pd/C (0.2 g) and hydrogenated at 20 psi for 1 h on a Parr hydrogenation apparatus. The suspension was filtered through a Celite cake and washed cake with EtOH.
  • Alcohol 10-B (1.0 g, 4.8 mmol), in Et 2 O (20 mL), was treated with boron trifluoride dimethyl etherate (0.1 equiv, 0.48 mmol, 0.06 mL) and an excess amount of CH 2 N 2 Et 2 O. The reaction mixture was stirred overnight. The mixture was filtered and solvent was removed on rotary evaporator.
  • Ester 10-C (0.37 g, 1.7 mmol) was dissolved in THF (40 mL) and treated with LiOH.H 2 O (10 equiv, 17 mmol, 0.71 g) in H 2 O (20 mL), MeOH (10 mL) and H 2 O 2 (10 mL). The mixture was heated at reflux for about 8 h. Solvent was removed via a rotary evaporator. Dissolved residue in H 2 O (50 mL), lowered pH to 5 with hydrochloric acid, extracted aqueous portion with EtOAc (3 ⁇ 25 mL) followed by CHCl 3 (3 ⁇ 25 mL).
  • Residue was treated with acidic H 2 O (30 mL) and extracted with CHCl 3 (3 ⁇ 15 mL) Combined organic layers, washed with saturated NaHCO 3 (20 mL), dried (MgSO 4 ) and solvent was removed under reduced pressure. Crude product was purified by silica gel (20 mm ⁇ 16.5 cm) flash chromatography using 10%-15% EtOAc/hexanes as eluent. Fractions containing the compound were combined and solvent was removed under reduced pressure.
  • Ester 10-F (0.27 g, 0.5 mmol) was treated with LiOH.H 2 O (10 equiv, 5.0 mmol, 0.21 g) in H 2 O (8 mL). The reaction mixture was stirred for 1 h. THF (3 mL) was added to complete dissolution of starting material and MeOH (3 mL) was added to convert reaction mixture to homogeneous solution. The volume was reduced under vacuum. H 2 O (40 mL) was added and the mixture was acidified with HCl.
  • reaction mixture is evaporated to dryness, mixed with toluene (200 mL), and shaken with cold 0.5N aq. HCl (50 mL) followed by water (4 ⁇ 50 mL). The organic layer is then evaporated to dryness in vacuo to give an off-white solid which is recrystallized from ethyl acetate to give a white solid (11-F-1) (0.59 g, 58% yield).
  • Example 182 is accomplished in vacuo to give Example 182 as a white solid (0.25 g, 51% yield).
  • Ester 12-B (0.7 g, 3.6 mmol) was dissolved in THF (20 mL) and treated with LiOH.H 2 O (10 equiv, 1.5 g, 35.8 mmol) in H 2 O (20 mL) and MeOH (10 mL) After 2 h, the solvent was removed in vacuo.
  • Example 58 (866 mg, 95%) as a colorless foam: 1 H NMR (300 MHz, CDCl 3 ), ⁇ 7.56 (2H), 7.33 (2H), 7.08 (2H), 5.84 (1H), 4.8-4.9 (1H), 3.74 (3H), 3.0-3.2 (3H), 2.4-2.6 (2H), 2.0-2.2 (1H), 1.6-1.8 (1H), 1.43 (9H), 1.21 (3H), 1.14 (3H), 0.76 (3H) 13 C NMR (75 MHz, CDCl 3 ), ⁇ 174.92, 172.55, 172.00, 161.81, 136.29, 135.91, 134.46, 133.00, 132.69, 129.75, 128.08, 120.51, 80.19, 56.60, 54.33, 53.04, 52.35, 46.32, 37.14, 32.26, 27.99
  • Example 58 (810 mg, 1.27 mmol) was dissolved in CH 2 Cl 2 (5 mL) containing TFA (5 mL). This reaction mixture was stirred for 6 h at RT. Evaporation of the solvent under reduced pressure provided a crude oil.
  • Example 92 (703 mg, 95 O) as a colorless foam: 1 H NMR (300 MHz, Acetone-d 6 ), ⁇ 9.68 (1H), 7.66 (2H), 7.44 (2H), 7.17 (2H), 6.93 (1H), 4.88 (1H), 3.72 (3H), 3.17 (1H), 3.05 (1H), 2.79 (1H), 2.5-2.6 (1H), 2.1-2.2 (1H), 1.7-1.8 (1H), 1.4-1.5 (1H), 1.25 (3H), 1.21 (3H), 0.84 (3H); 13 C NMR (75 MHz, Acetone-d 6 ), ⁇ 176.92, 172.14, 171.14, 160.87, 136.16, 134.58, 134.48, 132.16, 132.12, 128.93, 127.08, 119.23, 59.24, 55.25, 52.60
  • Example 92 (684 mg, 1.17 mmol) was dissolved in H 2 O (8 mL) containing LiOH (127 mg, 5.27 mmol). After 6 h at RT the mixture was acidified with 3 N HCl (3 mL) and the precipitate filtered and washed with cold H 2 O (3 mL).
  • Example 93 (547 mg, 82%) as a colorless solid: 1 H NMR (300 MHz, Acetone-d 6 ), ⁇ 9.96 (1H), 7.70 (2H), 7.58 (2H), 7.30 (2H), 4.8-4.9 (1H), 3.23 (1H), 3.07 (1H), 2.89 (1H), 2.5-2.6 (1H), 2.0-2.2 (1H), 1.6-1.7 (1H), 1.4-1.5 (1H), 1.26 (3H), 1.19 (3H), 0.81 (3H); 13 C NMR (75 MHz, Acetone-d 6 ), ⁇ 178.52, 174.10, 173.62, 162.22, 138.12, 136.08, 134.47, 133.69, 130.77, 128.90, 120.54, 56.92, 54.56, 53.89, 47.24, 37.48, 33.37, 23.52, 23.04, 22.31, 21.73; ESMS (m/z) 569 ([M ⁇ H] ⁇ ).
  • Example 101 (605 mg, 1.15 mmol) was dissolved in H 2 O (5 mL) containing LiOH (138 mg, 5.76 mmol). This was stirred for 4 hours. The solution thus obtained was acidified with 2 N HCl (10 mL) and the precipitate filtered. The filter cake was washed with cold H 2 O (3 mL) and then dried under high vacuum to provide Example 102 (539 mg, 92%) as a white amorphous powder:
  • the combined aqueous phases were washed with pentane (0.35L) and the pH of the aq. layer was adjusted to ca. 4 with 1N aq. HCl.
  • the aqueous layer was extracted with ether (4 ⁇ 0.35L) and the combined organic phases were concentrated in vacuo to give a colorless oil which slowly solidified to afford 96 g (90%) of 10-A as a white crystalline solid.
  • An analytical sample can be obtained by recrystallizing 10-A from ether-pentane (1:1) to provide 10-A as clear hexagonal plates.
  • the bottle was surrounded by aluminum foil, dry ice was added to cool the bottle and contents, and the bottle was removed from the shaker when the pressure reading was ca. 0 psi.
  • the isobutylene was condensed from the reaction vessel via a cold finger condenser over ca. 2 hours.
  • the resulting thin oil was dissolved in pentane (0.5L), the organic phase was washed with water (2 ⁇ 0.25L), 0.5N aq. NaOH (2 ⁇ 50 mL), and water (2 ⁇ 0.25L).
  • the organic phase was concentrated in vacuo to provide 15-C as a clear colorless oil which slowly solidified at room temperature. Recrystallization from petroleum ether gave 15-C as a fine white crystalline solid (26.86 g, 89%).
  • Boc-piperazine (1.9 gm, 10.4 mmol) was dissolved in THF (10 mL) and CH 2 Cl 2 (1 mL) containing DIEA (5.6 mL, 32.2 mmol). To this solution 2,6-dichlorobenzoyl chloride (1.6 mL, 11.4 mmol) was added at 0° C. After stirring the reaction for 1 hour at 0° C., 1N HCl (30 mL) was added. The mixture was extracted with CH 2 Cl 2 (2 ⁇ 20 mL) and the combined organics dried (Na 2 SO 4 ), filtered and the solvent removed in vacuo.
  • Example 176 (0.18 g, 0.287 mmol) was dissolved in CH 2 Cl 2 (1.0 mL) and TFA (1.0 mL). After stirring at room temperature for 1 hr the solvent was evaporated and the resultant gum triturated with ether. The solid was washed with ether and dried under vacuum to provide [1S-[1 ⁇ (R*),3 ⁇ ]]- ⁇ -[[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]amino]-4-(2,6-dichlorobenzoyl)- ⁇ -oxo-1-piperazinebutanoic acid methyl ester as a colorless solid (0.14-g, 87%). ESMS (m/z) 570 (MH + ).
  • Example 178 (0.19 g, 0.303 mmol) was dissolved in CH 2 Cl 2 (1.0 mL) and TFA (1.0 mL). After stirring at room temperature for 1 hr the solvent was evaporated and the resultant gum triturated with ether. The solid was washed with ether and dried under vacuum to provide [1S-[1 ⁇ (R*),3 ⁇ ]]- ⁇ -[[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonylamino]-4-[2,6-dichlorobenzoyl)- ⁇ -oxo-1-piperazinebutanoic acid methyl ester as a colorless solid (98 mg, 58%). ESMS (m/z) 570 (MH + ).
  • the ester 19-C-4 (0.258 g., 0.4 mmol) is hydrolyzed in a manner similar to that described above for Example 212.
  • the reaction mixture is then brought to pH 7, filtered and the filtrate transferred to a C-18 reversed phase HPLC column and eluted with a 0-10% acetonitrile/0.01 aqueous Na 2 CO 3 gradient. Evaporation is accomplished in vacuo to give the target compound as a white solid (0.2 g, 76% yield).
  • Example 214 As a white solid (0.43 g, 85% yield).
  • Example 215 as a white solid (0.4 g, 74% yield).
  • methyl trifluoromethanesulfonate (3.5 mL, 30.9 mmol). After an additional 12 hours of stirring, the reaction mixture is acidified with trifluoroacetic acid (50 mL, 649 mmol), and is allowed to stir for another 16 hours. The mixture is then diluted with toluene (200 mL), shaken with water (4 ⁇ 120 mL), and evaporated to dryness, giving 20-B as a pale brown solid (2.35 g, 10.97 mmol, 40% yield).
  • camphoric acid methyl ester 20-B (0.35 g, 1.63 mmol), EDC (0.35 g, 1.83 mmol), HOBT (0.25 g, 1.85 mmol), 4-dimethylaminopyridine (0.05 g, 0.41 mmol), in methylene chloride (10 mL) is stirred together in a 25 mL, 2-neck flask cooled in an ice water bath.
  • the mixture is evaporated to dryness, giving a colorless oil which is mixed with THF (100 mL) and water (50 mL) and then shaken sequentially with water (2 ⁇ 50 mL), aq. HCl (0.5N, 4 ⁇ 30 mL), satd. aq. NaHCO 3 (2 ⁇ 50 mL), and water (2 ⁇ 50 mL).
  • the organic layer is then evaporated to dryness, giving a colorless oil (0.91 g) which is chromatographed on silica gel with 10% methanol/chloroform to give 21-C as a white, foamy solid (0.57 g, 70% yield).
  • Example 218 (0.14 g, 0.25 mmol) in methanol (8 mL) is treated with a solution of LiOH—H 2 O (0.053 g, 1.27 mmol) in water (4 mL) over 15 minutes. The mixture is allowed to stir for 1.5 hours at room temperature, then the solvent is removed in vacuo. The residue is dissolved in water (25 mL), the pH is, adjusted to ca. 4 with 1N aq. HCl, and the mixture is extracted with ethyl acetate (3 ⁇ 25 mL). The combined organic extracts are dried (MgSO 4 ), and concentrated in vacuo to give the crude material as a sticky solid.
  • Boc-Tryptophan-O-methyl ester 24-A (636 mg, 2.00 mmol, 1 eq) was dissolved in dry DMF. To this solution NaH (88 mg, 2.20 mmol, 1.1 eq) was added with evolution of H 2 . To this mixture benzyl bromide (285 ⁇ L, 2.40 mmol, 1.2 eq) was added and the reaction stirred for 3 hours at room temperature. The reaction was quenched with brine (15 mL) and extracted with Et 2 O (3 ⁇ 15 mL). The combined organics were dried (Na 2 SO 4 ), filtered and the solvent removed in vacuo. The residue was purified by column chromatography (SiO 2 , Hexanes to 30% EtOAc/Hexanes gradient elution) to provide 426 mg (52 W) of the benzyl indole 24-B.
  • Example 196 was produced as described in Example 2.
  • Example 190 The preparation of Example 190 is as follows. Boc Tyr(2,6-dichlorobenzyl)-OH (25-A) (1.31 gm, 2.97 mmol) was dissolved in THF (5 mL) and cooled to ⁇ 78° C. under dry N 2 . BH 3 THF (5.9 mL, 5.9 mmol, 1N) was added and the reaction warmed slowly to room temperature with stirring for 3 hours. The reaction was cooled to 0° C. and quenched with H 2 O (1 mL) and warmed to room temperature. After the addition of 1 N HCl (25 mL), the mixture was extracted with EtOAc (3 ⁇ 25 ml) and the combined organic phases were dried over Na 2 SO 4 .
  • Example 56 (0.27 gm, 0.62 mmol) was dissolved in CH 2 Cl 2 (10 mL) and 2,6-dichlorophenylisocyanate (0.18 gm, 0.94 mmol) and DIEA (327 ⁇ L, 1.86 mmol) was added and the reaction stirred overnight. After the addition of 1 N HCl (20 mL), the mixture was extracted with EtOAc (3 ⁇ 25 mL) and the combined organic phases were dried over Na 2 SO 4 .
  • Example 227 (310 mg, 82%): ESMS (m/z) 620 (MH + ).
  • Example 227 (250 mg, 0.40 mmol) was dissolved in CH 2 Cl 2 (1.5 mL) and TFA (1.5 mL). After 1 h, the solvent was removed and the residue triturated with Et 2 O (3 ⁇ 5 mL) to form a gum. The residue was purified by column chromatography (SiO 2 , gradient elution: 100% hexanes 25% acetone/hexanes) to provide Example 228 (170 mg, 73%): ESMS (m/z) 564 (MH + ).
  • Example 228 (130 mg, 0.23 mmol) was dissolved in THF/CH 3 OH (5 mL/mL, respectively) and LiOH (22 mg, 0.53 mmol) was added in H 2 O (1 mL). After 2 h the solvent was evaporated and the residue dissolved in H 2 O (3 mL). The solution was precipitated with the addition of 1 N HCl (2 mL). The solvent was collected by vacuum filtration and washed with cold H 2 O (2 ⁇ 2 mL). The solid material was then thoroughly dried under high vacuum to afford Example 229 (80 mg, 64%) as a white solid: ESMS (m/z) 550 (MH + ).
  • Example 54 (387 mg, 0.704 mmol) and morpholine (0.14 mL, 1.55 mmol) were dissolved in CH 2 Cl 2 (15 mL). This solution was treated with BOP-Cl (215 mg, 0.845 mmol) and stirred under dry N 2 at room temperature. After 18 h the reaction was treated with 1 N HCl (10 mL) and extracted with CH 2 Cl 2 (3 ⁇ 15 mL). The combined organics were dried over Na 2 SO 4 , filtered and the solvent evaporated.
  • Example 226 160 mg, 37%) as a colorless oil: ESMS (m/z) 618 (M+Na) + .
  • Example 226 The methyl ester (Example 226) (160 mg, 0.258 mmol), was dissolved in THF (5 mL) and LiOH (12 mg, 0.52 mmol) was added in H 2 O (5 mL). After 4 h 1 N HCl (3 mL) was added and the precipitate collected by vacuum filtration washing with cooled H 2 O (3 ⁇ 3 mL) The product was thoroughly dried under high vacuum to provide Example 225 (148 mg, 95%) as an amorphous powder: ESMS (m/z) 602 (M ⁇ H).
  • N-[(1,1-Dimethylethoxy)carbonyl]-4-nitro-L-phenylalanine methyl ester (31-B) (C 15 H 20 N 2 O 6 )
  • a solution of N-Boc-4-nitrophenylalanine (31-A) (25.2 g, 81.28 mmol) and DMAP (0.82 g, 6.7 mmol) in dry DMF is cooled to 0° C. under Ar, and treated with MeOH (7.55 mL, 186 mmol) and DCC (18.975 g, 91.04 mmol).
  • the reaction mixture is stirred overnight at room temperature and filtered.
  • the filtrate is washed with satd NaHCO 3 and brine.
  • This CHCl 3 solution is extracted with H 2 O (2 ⁇ 300 mL), aqueous 1 M Na 2 S 2 O 3 .5H 2 O (300 mL), and H 2 O (3 ⁇ 300 mL); and then is dried (Na 2 SO 4 ), filtered, and concentrated to give a yellow oil.
  • the PdCl 2 [P(Ph) 3 ] 2 catalyst (0.41 g) is added, folowed immediately by an O 2 -free solution of (34-C) (2.46 g, 5.94 mmol) in dry 1:1 THF/CH 3 C(O)N(Me) 2 (17.8 mL).
  • the resulting reaction mixture is stirred at 65 ⁇ 5° C. under Ar for 5 h. It is quenched with satd aqueous NH 4 Cl (150 mL).
  • the resulting mixture is extracted with EtOAc (3 ⁇ 300 mL). The combined EtOAc portions are washed with brine (300 mL), dried (Na 2 SO 4 ), filtered and concentrated to a yellow-green colored oil.
  • the CH 2 Cl 2 mixture is washed with H 2 O (3 ⁇ 200 mL), 0.5 M aqueous HCl (2 ⁇ 200 mL), satd aqueous NaHCO 3 (2 ⁇ 200 mL), and H 2 O (2 ⁇ 200 mL).
  • the combined aqueous washes are extracted with one portion of CH 2 Cl 2 (200 mL).
  • the combined CH 2 Cl 2 portions are dried (Na 2 SO 4 ), filtered, and concnetrated to give a yellow-colored oil.
  • N 2 was bubbled through a mixture of N-Boc-4-iodo-L-phenylalanine functionalized Wang resin (35-B) (500 mg, ca. 0.3 mmol), PPh 3 (105 mg, 0.4 mmol), 2,4,6-trichloroaniline (490 mg, 2.5 mmol) and DIEA (1.74 mL, 10 mmol) in NMP (10 mL) for 10 min.
  • Pd 2 dba 3 (92 mg, 0.1 mmol) was added and the reaction mixture was placed under a CO atmosphere and heated (bath temp. 70° C.) for 18 h.
  • Example 220 was prepared as described in Schemes 35 and 2 starting from 4-aminobiphenyl and 35-B: physical properties as follows: 1 H NMR (300 MHz, CD 3 OD) ⁇ 7.87 (1H), 7.76 (2H), 7.62 (4H), 7.44 (4H), 7.30 (1H), 4.79 (1H), 3.30 (1H), 3.01 (1H), 2.72 (1H), 2.54 (1H), 2.02 (1H), 1.62 (1H), 1.58 (1H), 1.28 (3H), 1.20 (3H), 0.78 (3H); 13 C NMR (75 MHz, CD 3 OD) ⁇ 179.75, 175.49, 174.98, 168.82, 143.38, 142.03, 139.42, 138.75, 134.74, 130.70, 130.01, 128.86, 128.38, 128.31, 127.87, 122.68, 57.56, 54.69, 47.81, 38.43, 33.87, 23.94, 23.25, 22
  • Example 221 was prepared as described in Schemes 35 and 2 starting from 4-chloroaniline and 35-B: physical properties as follows: 1 H NMR (300 MHz, CD 3 OD) ⁇ 7.85 (2H), 7.68 (2H), 7.40 (2H), 7.36 (2H), 4.82 (1H), 3.29 (1H), 3.05 (1H), 2.81 (1H), 2.54 (1H), 1.96 (1H), 1.70 (1H), 1,24 (1H), 0.91 (3H), 0.86 (3H), 0.77 (3H); 13 C NMR (75 MHz, CD 3 OD) ⁇ 179.74, 175.55, 174.65, 168.73, 143.39, 138.93, 134.-5, 130.66, 130.55, 128.86, 123.68, 57.54, 54.62, 47.81, 38.33, 35.92, 33.86, 30.30, 29.00, 26.34, 23.21, 22.51, 21.98, 21.16; HRMS (FAB) calcd for
  • Example 222 was prepared as described in Schemes 35 and 2 starting from 2-(triflouromethyl)aniline and 35-B. Physical properties as follows: 1 H NMR (300 MHz, CD 3 OD) ⁇ 7.86 (2H), 7.66 (4H), 7.40 (2H), 4.78 (1H), 3.30 (1H), 3.10 (1H), 2.72 (1H), 2.49 (1H), 1.98 (1H), 1.62 (1H), 1.48 (1H), 1.28 (3H), 1.20 (3H), 0.78 (3H); 13 C NMR (75 MHz, CD 3 OD) ⁇ 179.70, 175.77, 173.54, 169.59, 143.34, 136.74, 124.18, 133.85, 131.74, 130.73, 128.89, 128.30, 127.13, 123.51, 57.56, 55.05, 62.54, 57.56, 55.05, 54.58, 50.06, 47.80, 38.21, 33.84, 23.89, 23.
  • Example 224 was prepared as described in Schemes 35 and 2 starting from methyl L-leucinate and 35-B: Physical properties as follows: 1 H NMR (300 MHz, CD 3 OD) ⁇ 7.78 (2H), 7.34 (2H), 4.87 (1H), 4.72 (1H), 3.30 (1H), 3.02 (1H), 2.82 (1H), 2.68 (1H), 1.98 (1H), 1.74 (5H), 1.23 (3H), 0.97 (9H), 0.77 (3H); 13 C NMR (75 MHz, CD 3 OD) ⁇ 179.73, 176.33, 175.23, 174.66, 170.35, 143.05, 133.87, 130.52, 128.72, 62.41, 57.53, 54.63, 52.77, 47.79, 41.51, 38.30, 26.42, 23.90, 23.56, 23.18, 22.51, 21.93; MS (FAB) m/z (rel.
  • R 36-1 and R 36-2 are independently defined as —CH 3 , t-Bu, or —CH 2 C 6 H 5 and relative configuration is depicted by bold and dotted lines.
  • Scheme 36 teaches a general method (etherification, epimerization and ester deprotection) for the preparation of selectively protected camphoric acid isomers 36-B, 36-D, 36-G and 36-F.
  • R 37-1 and R 37-2 are independently defined as —H or —CH 3 and R 37-3 and R 37-4 are independently defined as —H, —CH 3 , t-Bu, or —CH 2 C 6 H 5 .
  • Scheme 37 teaches methods for the coupling of camphoric acid monoester isomers to isomers of 37-B and ester deprotection for the preparation of isomers of Example 54. 37-C-1 through 37-C-15.
  • the precipitate is filtered washing with water to obtain (1S-cis)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (1.1265 g.
  • the precipitate is filtered washing with water to obtain (1S-trans)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine (692.2 mg).
  • Physical properties as follows: m.p. 145-150° C.; 1 H NMR (300 MHz, DMSO-d 6 ).
  • the reaction mixture is concentrated partially (to remove most of the EtOH), basified to pH 12 with 1 N NaOH, and extracted twice with CH 2 Cl 2 .
  • the combined CH 2 Cl 2 extracts are discarded.
  • the aqueous solution is acidified with concentrated HCl to pH 3, and then is extracted with CH 2 Cl 2 .
  • the oil is dissolved in MeOH (1.0 mL) and then treated with H 2 O (0.65 mL) and 1.00 M LiOH 0.35 mL). After 16 h a second portion of 1.00 M LiOH is added and the hydrolysis is allowed to proceed for an additional 4 h.
  • the solution was diluted with H 2 O and the pH adjusted to ca. 8-9.
  • the neutralized solution is diluted with MeOH and then concentrated.
  • the aqueous concentrate is diluted with additional H 2 O, basified to pH 13 (1N NaOH), and extracted with Et 2 O. The Et 2 O extract is discarded.
  • the aqueous phase is acidified to pH 2 (conc. HCl) and is extracted with EtOAc.
  • Example 253 (93 mg) as a colorless oil: 1 H NMR (CD 3 OD) ⁇ 7.61 (2H), 7.55-7.44 (3H), 7.25 (2H), 4.69-4.78 (1H), 3.23 (1H), 3.00 (1H), 2.80-2.70 (1H), 2.60-2.46 (1H), 2.10-1.94 (1H), 1.78-1.61 (1H), 1.52-1.39 (1H), 1.36 (9H); 1.27 (3H), 1.23 (3H), 0.81 (3H); MS (+ESI) m/z 557, 555.
  • DIEA (0.65 g) was added dropwise to a mixture of (1R)camphoric anhydride (0.18 g) and O-benzyl-L-tyrosine methyl ester hydrochloride (0.33 g) in DMF (2 ml) at 0° C.
  • the mixture was stirred at 40° C. for 15 hr, cooled, diluted with AcOEt, and acidified with 1N HCl to pH 5.
  • the organic layer was washed with H 2 O, brine, dried over Na 2 SO 4 , and the solvent was removed in vacuo.
  • DIEA (4.48 g) was added to a mixture of 1-(3,4-dichlorobenzyl)-L-histidine methyl ester (5.56 g) and (1R)-camphoric anhydride (2.53 g) in DMF (50 ml). The mixture was stirred at 40° C. for 17 hr, cooled, diluted with H 2 O, acidified with 5% HCl to pH 5, and extracted with CHCl 3 . The extract was washed with H 2 O, brine, and dried over NaSO 4 .
  • Benzyl (S)-2-amino-3-(2-naphthyl) propionate tosylate (0.20 g) was partitioned between AcOH and sat. NaHCO 3 . The organic layer was washed with H 2 O, brine, dried over Na 2 SO 4 , and the solvent was removed in vacuo. DMF (5 ml) and (1R)-camphoric anhydride (0.20 g) were added to the residue. The mixture was stirred at 30-40° C. for 17 hr, cooled, and poured into H 2 O. The resulting mixture was extracted with AcOH. The extract was washed with H 2 O, brine, dried over Na 2 SO 4 , and the solvent was removed in vacuo.
  • Examples 7 through 51 were prepared in a similar manner as described in Examples 1-6, and are shown in Tables 1, 2, and 3. TABLE 1 Examples 7 through 20: Ex. physicochemical No. * R 0 R 15 Z 1 R 13 R 18 property 7 S H H single t-BuO— H M.P.: 98—100° C. bond MS(m/z) 420 (MH + ) 8 S CH 3 H —OCH 2 — H gum MS(m/z): 518 (MH + ) 9 S H H —OCH 2 — H M.P.: 92-93° C.
  • Example 39 is an isomer of Example 12 and is therefore synthesized in a manner similar to that of Example 12.
  • HCl gas was bubbled through a solution of N-(tert-butoxycarbonyl)-4-(2,6-dichlorobenzamido)-L-phenylalanine (800 mg) in MeOH (15 ml) for 5 minutes and the mixture was stirred for 3 hr. at room temperature. Excess HCl was removed by bubbling N 2 through the mixture and the solvent was removed in vacuo. The residue was washed with ether and dried.
  • Examples 60-153 were prepared in a similar manner, as described in Examples 52-59, and are shown in Table 4. TABLE 4 Examples 60 through 153: physico- Ex. chemical No. R 19 R 0 R 15 Z 1 R 13 property 60 H CH 3 single bond OH gum MS (m/z): 565 (MH + ) 61 H H single bond OH M.P.: 168-171° C. MS (m/z): 549 ([M ⁇ H] ⁇ ) 62 H CH 3 NO 2 —OCH 2 — gum MS (m/z): 581 (MH + ) 63 H H NO 2 —OCH 2 — M.P.: 92-94° C.
  • BOP Reagent (674 mg) was added to a mixture of (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine methyl ester (743 mg) and aq. NH 4 OH (0.5 ml) in THF (10 ml). The mixture was stirred for 24 hr. and sat. LiCl (15 ml) was added. The resulting mixture was extracted with AcOEt and the extract was dried over Na 2 SO 4 .
  • Examples 156-166 were prepared in a similar manner, as described in Examples 154 and 155 and are shown in Table 5. TABLE 5 Examples 156 through 166: Ex. physicochemical No. R 19 R 20 R 0 R 6 property 156 H H M.P.: 85-87° C.
  • Example 167 was prepared from O-benzyl-L-tyrosine methyl ester hydrochloride and (1R,3S)-3-(tert-butoxycarbonyl)-1,2,2-trimethylcyclopentanecarboxylic acid in a similar manner as described in Example 52.
  • Example 168 was prepared in a similar manner as described in Example 53. Physicochemical property: gum; MS (m/z): 468 (MH + ); MP 191-192° C. (d).
  • Example 169 was prepared in a similar manner as described in Example 54, MS (m/z): 454 (MH + ).
  • NaCNBH 3 (104 mg) was added to a mixture of O-[(2,6-dichlorophenyl)methyl]-L-tyrosine methyl ester (490 mg), (1R,cis)-3-formyl-1,2,2-trimethylcyclopentanecarboxylic acid, (1R,5S)-4-hydroxy-1,8,8-trimethyl-3-oxabicyclo[3,2,1]-octane-2-one (153 mg), AcOH (0.5 ml), and MeOH (25 ml) under argon. The mixture was stirred for 72 hr. at room temperature. The solvent was removed in vacuo and 10% HCl (20 ml) was added. The resulting mixture was stirred for 2 hr.

Abstract

The present invention relates to small molecules according to the formula [I]:
Figure US20030130349A1-20030710-C00001
which are potent inhibitors of α4β1 mediated adhesion to either VCAM or CS-1 and which can be used for treating or preventing α4β1 adhesion mediated conditions in a mammal such as a human.

Description

    BACKGROUND OF THE INVENTION
  • 1. Field of the Invention [0001]
  • The present invention relates to small molecules that are potent inhibitors of β[0002] 4β1 mediated adhesion to either VCAM or CS-1 and which could be useful for the treatment of inflammatory diseases.
  • 2. Description of Related Art [0003]
  • The extracellular matrix (ECM) is the major component of connective tissue which provides structural integrity, and promotes cell migration and differentiation. As part of these functions, extracellular matrix molecules such as fibronectin, collagen, laminin, von Willebrand factor, thrombospondin, fibrinogen, and tenascin have been shown to support adhesion of cells in vitro. This adhesive interaction is critical for a number of biological processes including hemostasis, thrombosis, wound healing, tumor metastasis, immunity and inflammation. [0004]
  • Fibronectin (FN) is the prototype ECM molecule. The major cell attachment site in the fibronectin molecule has been reproduced synthetically with the amino acid sequence arginine-glycine-aspartic acid, or RGD using single letter nomenclature. Peptides containing the RGD sequence which either inhibit or promote cell adhesion have been described (U.S. Pat. Nos. 4,589,881; 4,661,111; 4,517,686; 4,683,291; 4,578,079; 4,614,517; and 4,792,525) Changes in the peptide as small as the exchange of alanine for glycine or glutamic acid for aspartic acid, which constitute the addition of a single methyl or methylene group to the tripeptide, eliminates these activities (Pierschbacher et al., [0005] Proc. Natl. Acad. Sci. USA 81:5985 (1984)). Recently, a second FN cell binding domain has been identified within the alternatively spliced region of the A chain of the molecule, known as the connecting segment 1 (CS-1). The most active cell-binding site within this alternatively spliced region is composed of 25 amino acids where the carboxy terminus contains the sequence EILDVPST The amino acid sequence EILDVPST forms a recognition motif on FN for cell surface receptors. (Wayner et al., J. Cell Biol. 109:1321 (1989); Guan et al., Cell 60:53 (1990)).
  • The receptors which recognize these sites on FN belong to a gene superfamily called integrins which consist of heterodimeric complexes of non-covalently associated alpha and beta subunits. A common β subunit combines with unique a subunits to form an adhesion receptor of defined specificity. To date, 8 β subunits have been identified which can dimerize with 16 distinct α subunits forming 22 distinct integrins. The β1 subfamily, also known as the VLA family (Very Late Activation Antigens), binds to ECM molecules such as FN, collagen and laminin. For reviews, see, Hynes, [0006] Cell 48:549 (1987); Hemler, Annu. Rev. Immunol. 8:365 (1990). Leukocyte interaction with FN at the two spatially separate binding domains is mediated by two distinct integrins. The RGD site is recognized by the Integrin α5β1, while, EILDV is recognized by α4β1 (Pytela et al., Cell 40:191 (1985); Wayner et al., J. Cell Biol. 109:1321 (1989); Guan et al, Cell 60:53 (1990)).
  • Vascular endothelial cells form the interface between blood and tissues and control the passage of leukocytes as well as plasma fluid into tissues. A variety of signals generated at the site of inflammation can activate both endothelial cells as well as circulating leukocytes so that they become more adhesive to one another. Following this initial adhesion the leukocytes migrate into the tissues to perform host defense functions. Several adhesion molecules have been identified which are involved in leukocyte-endothelial interactions. [0007]
  • In the β[0008] 1 subfamily, in addition to binding to fibronectin, α4β1 interacts with a cytokine inducible protein on endothelial cells termed vascular cell adhesion molecule (VCAM). Further involved in the leukocyte-endothelial adhesion process is the β2 integrin subfamily. β2 integrins include CD11a/CD18, CD11b/CD18, and CD11c/CD18. In addition, the β7 subunit associates with α4 to form a unique α4β7 heterodimer which binds to FN, to VCAM, and to Mucosal Addressin Cell Adhesion Molecule-1 (MAdCAM) (Ruegg et al, J. Cell.Biol. 117:179 (1992); Andrew et al., J. Immunol. 153:3847 (1994); Briskin et al., Nature 363:461 (1993); Shyjan et al, J. Immunol. 156:2851 (1996)). α4 integrins are widely expressed on different cell types including hematopoietic progenitors, lymphocytes, natural killer cells, monocytes, eosinophils, basophils, and mast cells (Helmer, M. E., Annu. Rev. Immunol. 8:365 (1990)). Other molecules on endothelial cells which bind to the leukocytes include ICAM-1, ICAM-2, E-selectin and P-selectin (Carlos and Harlan, Immunol. Rev. 114:1 (1990); Osborn, L., Cell 62:3 (1990); Springer T., Nature 346:425 (1990); Geng et al., Nature 347:757 (1990); Stoolman, Cell 56:907 (1989)).
  • A number of in vitro and in vivo studies indicate that α[0009] 4β1 plays a critical role in the pathogenesis of a variety of diseases. Monoclonal antibodies directed against α4 have been tested in a variety of disease models. Anti-α4 antibodies block adhesion of lymphocytes to synovial endothelial cells; this adhesion plays a potential role in rheumatoid arthritis (van Dinther-Janssen et al, J. Immunol. 147:4207 (1991). α4 has also been implicated with respect to rheumatoid arthritis in separate studies (Laffon et al, J. Clin. Invest. 88:546 (1991); Morales-Ducret et al, J. Immunol. 149:1424 (1992)). A significant number of studies have evaluated the role of α4 in allergy and asthma. For example, monoclonal antibodies to α4 block adhesion of basophils and eosinophils to cytokine activated endothelial cells (Walsh et al, J. Immunol. 146:3419 (1991); Bochner et al, J. Exp. Med. 173:1553 (1991)) Monoclonal antibodies to α4 were also effective in several lung antigen challenge models (Abraham et al, J. Clin. Invest. 93:776 (1994), Weg et al, J. Exp. Med. 177:561 (1993)). The cotton-top tamarin, which experiences spontaneous chronic colitis, showed a significant attenuation of their colitis when anti-α4 antibody was administered (Podolsky et al, J. Clin. Invest. 92:372 (1993); Bell et al, J. Immunol. 151:4790 (1993)). In a rat and mouse model, autoimmune encephalomyelitis was blocked by anti-α4 antibody (Yednock et al, Nature 356:63 (1992); Baron et al, J. Exp. Med. 177:57 (1993)) Anti-α4 monoclonal antibodies also inhibit insulitis and delay the onset of diabetes in the non-obese diabetic mouse (Baron et al, J. Clin. Invest. 93:1700 (1994); Yang et al, Proc. Natl. Acad. Sci. USA 90:10494 (1993); Burkly et al, Diabetes 43:529 (1994)). α4 is also implicated in atherosclerosis due to its endothelial expression during atherogenesis (Cybuisky et al, Science 251:788 (1991)). The migration of leukocytes to an inflammatory site can also be blocked by anti-α4 antibodies. In addition to the blocking of migration, inhibitors of leukocyte endothelial adhesion may block the costimulatory signals mediated by integrins and thus inhibit overproduction of inflammatory cytokines. In a separate set of experiments not using anti-α4 antibodies, the peptides GRDGSP or EILDV were tested against contact hypersensitivity response. The contact hypersensitivity response was found to be blocked by GRDGSP or EILDV suggesting that both α4β1, and α5β1 are involved in this inflammatory response.
  • Other ailments which may involve α[0010] 4β1-mediated conditions include the inflammatory disorders rheumatoid arthritis, allergic disorders, asthma, spontaneous chronic colitis, insulitis, contact hypersensitivity response, atherosclerosis and autoimmune encephalomyelitis., These studies illustrate that small molecules that are potent inhibitors of α4β1 mediated adhesion to either VCAM-1 or CS-1 may be used as a form of treatment in numerous inflammatory diseases. However, these inflammatory conditions could be expanded to include adult respiratory distress syndrome, AIDS, cardiovascular diseases, thrombosis or harmful platelet aggregation, reocclusion following thrombolys's, allograft rejection, reperfusion injury, psoriasis, eczema, contact dermatitis and other skin inflammatory diseases, osteoporosis, osteoarthritis, atherosclerosis, neoplastic diseases including metastasis of neoplastic or cancerous growth, wound healing enhancement, treatment of certain eye diseases such as detaching retina, Type I diabetes, multiple sclerosis, systemic lupus erythematosus (SLE), inflammatory and immunoinflammatory conditions including ophthalmic inflammatory conditions and inflammatory bowel diseases, ulcerative colitis, regional enteritis and other autoimmune diseases. Accordingly, a compound which could inhibit these conditions is desirable.
  • SUMMARY OF THE INVENTION
  • The present invention is directed to a compound of the formula [I]: [0011]
    Figure US20030130349A1-20030710-C00002
  • In the above formula [I], n is an integer of 0 or 1, R[0012] 1 is a hydrogen atom or a methyl group, and R2 can be selected from the following: a —CN group; a —COOH group; a —(C1-6alkylene)OH group, preferably a —(C1-3alkylene)OH group; a CH2O(C1-6 alkyl) group, preferably a —CH2O(C1-3 alkyl) group; a —(C1-3 alkylene)COOH group, preferably a —(C1-13 alkylene)COOH group; a —CH2O(C1-6 alkylene)O(C1-6 alkyl) group, preferably a —CH2O(C1-3 alkylene)O(C6 alkyl) group or a —CH2O(C1-6 alkylene)O(C1-3 alkyl) group, more preferably a —CH2O(C1-3 alkylene)O(C1-3 alkyl) group; a —CH2O(C1-6 alkylene)COOH group, preferably a —CH2O(C1-3 alkylene)COOH group; a —(C2-7 alkenylene)COOH group, preferably a —(C2-4 alkenylene)COOH group; a —CO(C1-6 alkylene)COOH group, preferably a —CO(C1-3 alkylene)COOH group; a —CO(C2-7 alkenylene)COOH group, preferably a —CO(C2-4 alkenylene)COOH group; a —CO(C1-6 alkylene)O(C6 alkyl) group, preferably a —CO(C1-3 alkylene)O(C1-6 alkyl) group or a —CO(C1-6 alkylene)O(C1-3 alkyl) group, more preferably a —CO(C1-3 alkylene)O(C1-3 alkyl) group; a —CO(C1-6 alkylene)CO(C1-6 alkyl) group, preferably a —CO(C1-3 alkylene)CO(C1-6 alkyl) group or a —CO(C1-6 alkylene)CO(C1-3 alkyl) group, more preferably a —CO(C1-3 alkylene)CO(C1-3 alkyl) group; a —CONH(C1-6 alkyl) group, preferably a —CONH(C1-3 alkyl) group; a —CONHO(C1-6 alkyl) group, preferably a —CONHO(C1-3 alkyl) group; a —CONH(C1-6 alkylene) COOH group, preferably a —CONH(C1-3 alkylene)COOH group, a —CONH2 group; a —CONH(C3-7 cycloalkyl) group, preferably a —CON (C3-6 cycloalkyl) group; a group as follows:
    Figure US20030130349A1-20030710-C00003
  • a —CONHOCH[0013] 2Ph group; a —CONH(C1-6 alkylene)CN group, preferably a —CONH(C1-3 alkylene)CN group; a —COO(C1-6 alkyl) group, preferably a —COO(C1-3 alkyl) group; a —CH2O(C1-6 alkylene)CONH2 group, preferably a —CH2O(C1-3 alkylene)CONH2 group; a —CONH(C1-6 alkylene)CONH2 group, preferably a —CONH(C1-3 alkylene)CONH2 group; a —CONHOH group; a —NHCOOCH2Ph group; or a group selected from the following formula:
    Figure US20030130349A1-20030710-C00004
  • In the above Formula (I), in R[0014] 2, the C1-6 alkylene is preferably C1-3 aklylene, the C2-7 alkenylene is preferably C2-4 alkenylene, the C1-6 alkyl is preferably C1-3 alkyl and the C3-7 cycloalkyl is preferably C3-6 cycloalkyl.
  • In the above formula [I], R[0015] 3 can be a hydrogen atom or a methyl group, X can be a methylene group or a —CO— group, and R4 can selected from the following: a hydrogen atom; or a C1-6 alkyl group, preferably a C1-3 alkyl group.
  • In the above formula [I], R[0016] 5 can be a group selected from the following: a —COOH group or an ester or an amide thereof; a —(C1-6 alkylene)COOH group, preferably a —(C1-3 alkylene)COOH group, or an ester or an amide thereof; a —(C1-7 alkylene)O(C1-6 alkyl) group, preferably a —(C1-4 alkylene)O(C1-6 alkyl) group or a —(C1-7 alkylene)O(C1-3 alkyl) group, more preferably a —(C1-4 alkylene)O(C1-3 alkyl) group; a —(C1-7 alkylene)OH group, preferably a —(C1-4 alkylene)OH group; a —COO(C1-6 alkyl) group, preferably a —COO(C1-3 alkyl) group; a —CONH(C1-6 alkyl) group, preferably a —CONH(C1-3 alkyl) group; or a —CONH2 group.
  • In the above formula [I], R[0017] 6 can be a substituted or unsubstituted monocyclic or bicyclic aryl group, a substituted or unsubstituted monocyclic or bicyclic heteroaryl group, a substituted or unsubstituted monocyclic or bicyclic arylcarbonylamino-C1-6 alkyl group, a substituted or unsubstituted monocyclic or bicyclic aliphatic heterocyclic carbonyl group, a 9- fluorenylmethyloxycarbonylamino —C1-6 alkyl group, a 3-tosylguanidino-C1-6 alkyl group;
    Figure US20030130349A1-20030710-C00005
  • provided that R[0018] 1 and R3 must be different and when R2 or R6 is a —COOH group or contains a —COOH group, then a pharmaceutically acceptable ester or a pharmaceutically acceptable amide thereof are included and also with the proviso that [1S-[1, (R*) 3α]]-α-[[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]amino]-4-(2,6-dichlorobenzoyl)-γ-oxo-1-pyrazinebutanoic acid methyl ester or [1S-[1α, (R*),3α]]-β-[[(3-Carboxy-2,2,3-trimethylcyclopentyl) carbonyl]amino]-4-(2,6-dichlorobenzoyl)-γ-oxo-1-pyrazinebutanoic acid methyl ester are excluded.
  • In the above formula [I], in R[0019] 6, an aryl group or aryl moiety in the arylcabonylamino group is a 5- or 6-membered aromatic hydrocarbon ring; and including any bicyclic group in which any of the above ring is fused to another above ring; and substituted by zero (0) to three (3) substituents.
  • Examples of aryl can include phenyl, a C[0020] 1-6 alkoxyphenyl group and naphthyl group. Each of these moieties may be substituted as appropriate.
  • In R[0021] 6, a heteroaryl is a 5- or 6-membered partially saturated or unsaturated ring containing from one (1) to four (4) heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur; and including any bicyclic group in which any of the above heterocyclic rings is fused to a benzene ring, C3-C8 cycloalkyl, or another heterocycle; and if chemically feasible, the nitrogen and sulfur atoms may be in the oxidized forms; and substituted by zero (0) to three (3) substituents.
  • Examples of heteroaryl can include 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, 3-pyridazinyl, 4-pyridazinyl, 3-pyrazinyl, 2-quinolyl, 3-quinolyl, 1-isoquinolyl, 3-isoquinolyl, 4-isoquinolyl, 2-quinazolinyl, 4-quinazolinyl, 2-quinoxalinyl, 1-phthalazinyl, 2-imidazolyl, 4-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 3-pyrazolyl, 4-pyrazolyl, 5-pyrazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-indolinyl, 3-indolyl, 3-indazolyl, 2-benzoazolyl, 2-benzothiazolyl, 2-benzimidazolyl, 2-benzofuryl, 3-benzofuryl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-pyrrolyl, 3-pyrrolyl, 1,2,4-oxadiazolyl-3-yl, 1,2,4-oxadiazol-5-yl, 1,2,4-thiadiazol-3-yl, 1,2,4-thiadiazol-5-yl, 1,2,4-triazol-3-yl, 1,2,4-triazol-5-yl, 1,2,3,4-tetrazol-5-yl, 5-oxazolyl, 1-pyrrolyl, 1-pyrazolyl, 1,2,3-triazol-1-yl, 1,2,4-triazol-1-yl, 1-tetrazolyl, 1-indolinyl, 1-indazolyl, 2-isoindolyl, 1-purinyl, 3-isothiazolyl, 4-isothiazolyl and 5-isothiazolyl. Each of these moieties may be substituted as appropriate. [0022]
  • In R[0023] 6, an aliphatic heterocyclic moiety in aliphatic heterocyclic carbonyl group is a 5- or 6-membered saturated ring containing from one (1) to four (4) heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur; and including any bicyclic group in which any of the above heterocyclic rings is fused to a benzene ring, C3-C8 cycloalkyl, or another heterocycle; and if chemically feasible, the nitrogen and sulfur atoms may be in the oxidized forms; and substituted by zero (0) to three (3) substituents.
  • Examples of aliphatic heterocyclic can include piperazinyl group, pyrrolidinyl group, piperidyl group, homopiperidyl group, thiomorpholino group, and morpholino group. Each of these moieties may be substituted as appropriate. [0024]
  • According to the present invention, the term “C[0025] 1-6 alkyl” represents an alkyl group having 1 to 6 carbon atoms. This group may be straight or branched. Illustrative but non-limiting examples of a C1-6 alkyl group are methyl, ethyl, n-propyl, isopropyl, n-butyl, iso-butyl, tert-butyl, sec-butyl, n-pentyl, isopentyl and n-hexyl. It is understood that this type of nomenclature extends to terms such as “C1-6 methoxy” and therefore encompasses both straight and branced methoxy groups having 1 to 6 carbon atoms.
  • Also, in the above formula [I] with all substituents as described above, a pharmaceutically acceptable salt thereof is included. [0026]
  • The desired compounds of the present invention have preferred steric configurations. Accordingly, a preferred steric configuration is represented by compounds of the formula [I-1]: [0027]
    Figure US20030130349A1-20030710-C00006
  • wherein n, R[0028] 1 through R6 and X are as defined above.
  • A more preferred steric configuration is represented by compounds according to the formula [I-2]: [0029]
    Figure US20030130349A1-20030710-C00007
  • wherein n, R[0030] 1 through R4, R6 and X are as defined above and R5 can be selected from the following: a —COOH group; a —(C1-6 alkylene)COOH group, preferably a —(C1-3 alkylene)COOH group; a —(C1-7 alkylene)O(C1-6 alkyl) group, preferably a —(C1-4 alkylene)O(C1-6 alkyl) group or a —(C1-7 alkylene)O(C1-3 alkyl) group, more preferably a —(C1-4 alkylene)O(C1-3 alkyl) group; a —(C1-7 alkylene)OH group, preferably a —(C1-4 alkylene)OH group; a —COO(C1-6 alkyl) group, preferably a —COO(C1-3 alkyl) group; a —CONH(C1-6 alkyl) group, preferably a —CONH(C1-3 alkyl) group or a —CONH2 group.
  • In a preferred embodiment of the present invention, R[0031] 6 can be selected from the following formula:
    Figure US20030130349A1-20030710-C00008
  • In the above, R[0032] 7, which occurs one or more times and which may be the same or different in each occurrence, may be selected from the following: a —H group; a —OH group; a —NO2 group; a —NH2 group; a —C1-5 alkyl group, preferably a —C1-3 alkyl group; a —F group; a —Cl group; a —Br group; a —I group; a —COOH group; a —COO(C1-6 alkyl) group, preferably a —COO(C1-3 alkyl) group; a —O(C1-C8 alkyl) group, preferably a —O(C1-4 alkyl) group; a CONH(C1-6 alkylene)COOH group, preferably a —CONH(C1-3 alkylene)COOH group; a —OCH2(C3-7 cycloalkyl) group, preferably a —OCH2(C3-6 cycloalkyl) group; or a substituent selected from the following formula:
    Figure US20030130349A1-20030710-C00009
  • In the above, R[0033] 8, which occurs one or more times and which may be the same or different in each occurrence, may be selected from the following: a —H group; an —OH group; a —NH2 group; a —NO2 group; a —C1-7 alkyl group, preferably a —C1-4 alkyl group; a —F group; a —Cl group; a —Br group; a —I group; a —CF3 group; a phenyl group, or a —O(C1-6 alkyl) group, preferably a —O(C1-3 alkyl) group.
  • In the above, R[0034] 9 may be selected from the following: a —H group; a —Cl alkyl group, preferably a —C1-3 alkyl group; a —C3-7 cycloalkyl group, preferably a —C3-6 cycloalkyl group; a —(C1-6alkylene)aryl group, preferably a —(C1-3 alkylene)aryl group; an aryl group; or a group selected from the following formula:
    Figure US20030130349A1-20030710-C00010
  • In the above, R[0035] 10, which occurs one or more times and which may be the same or different in each occurrence, may be selected from the following: a —H group; a —F group; a —Cl group; a —Br group; an —I group; a —NO2 group; a —C1-6 alkyl group, preferably a —C1-3 alkyl group; or a —O(C1-6 alkyl) group, preferably a —O(C1-3 alkyl) group.
  • In the above, R[0036] 11 may be selected from the following:
    Figure US20030130349A1-20030710-C00011
  • In the above, R[0037] 12, which occurs one or more times and which may be the same or different in each occurrence, may be selected from the following: a —H group; a —CF3 group; a —OCH3 group; a —F group; a —Br group; a —Cl group; or an —I group;
  • The above embodiments carrying the proviso that when R[0038] 7 is the formula:
    Figure US20030130349A1-20030710-C00012
  • then R[0039] 9 is other than hydrogen.
  • In a more preferred embodiment of the present invention, R[0040] 6 is selected from the following:
    Figure US20030130349A1-20030710-C00013
  • In the above, Y is selected from either a hydrogen atom or a chlorine atom. [0041]
  • In a more preferred embodiment of the present invention, R[0042] 2 is selected from the following: a —COOH group or an ester or an amide thereof; a —CONHCH2COOH group; a —CONHOCH2Ph group; or a —CONHCH2CONH2 group.
  • In another preferred embodiment of the present invention, R[0043] 1 is a —CH3 group, and R2 is a —COOH group; a —CONHCH2COOH group; a CONHOCH2Ph group or a —CONHCH2CONH2 group, and R3 and R4 are hydrogen atoms. Also, X is —CO—, R5 is —COOH, n is 1, and
  • R[0044] 6 is represented by the following formula
    Figure US20030130349A1-20030710-C00014
  • wherein R[0045] 7 is
    Figure US20030130349A1-20030710-C00015
  • and R[0046] 8 occurs 2 or 3 times and is a chlorine atom.
  • Other compounds within the scope of the present invention are compounds of the formula [I-3]: [0047]
    Figure US20030130349A1-20030710-C00016
  • In the above formula [I-3], R[0048] 1 may be a hydrogen atom or a methyl group. Also in the above formula [I-3], R2 may be selected from the following: a —CN group; a —COOH group; a —CONH2 group; a —CONHOH group; a —CON(CH3)2 group; a —CH2OCH2COOH group; a —CH═CHCOOH group; a —CONHCH2COOH group; a —CONH(CH2)2COOH group; a CONHCH2CONH2 group; a —CONH(CH2)2CN group; a group selected from the following:
    Figure US20030130349A1-20030710-C00017
  • In the above formula [I-3], R[0049] 3 may be a hydrogen atom or a methyl group and R5 may be a —COOH group or a COOMe group.
  • In the above formula [I-3], R[0050] 6 may be selected from the following:
    Figure US20030130349A1-20030710-C00018
  • In the above, Z[0051] 1 may be selected from the following: a —O— group; a —NHCO— group; a —NHCH2— group, a —OCH2— group; a —CONH-group; a —NHSO2— group; a —NHCOCH2— group; or a —N(CH3)CH2— group.
  • In the above, R[0052] 13 may be selected from the following: a —H group; a -iBuO group; a —CH3 group; a i-Bu group; or a group selected from the following:
    Figure US20030130349A1-20030710-C00019
  • In the above, R[0053] 14, which occurs one or more times and which may be the same or different in each occurence, may be selected from the following: a —H group; a —F group; a —Cl group; a —Br group; an —I group; a —CH3 group; a —OCH3 group; a —CF3 group; a —NO, group; a —NH2 group; or a -n-C7H15 group.
  • In the above, R[0054] 15 may be selected from the following: a —H group; a —OH group; a —NO2 group; or a group selected from the following:
    Figure US20030130349A1-20030710-C00020
  • In the above, R[0055] 16 may be selected from the following
    Figure US20030130349A1-20030710-C00021
  • In the above, R[0056] 17, which occurs one or more times and which may be the same of different in each occurrence, may be selected from the following: a —H group; a —Cl group; a —OCH3 group; or a —CF3 group, provided that R1 and R3 must be different.
  • In a another embodiment of the compounds according to formula [I-3], R[0057] 1 is a hydrogen atom or a methyl group, and R2 is selected from the following: a —CN group; a —COOH group; a —COOMe group; a —CONH2 group; a —CONHOH group; a —CON(CH3)2 group; a —CH2OCH2COOH group; a —CH═CHCOOH group; a —CONHCH2COOH group; a —CONH(CH2)2COOH group; a —CONHCH2CONH2 group; a —CONH(CH2)2CN group; or a group selected from the following:
    Figure US20030130349A1-20030710-C00022
  • and R[0058] 3 is a hydrogen atom or a methyl group.
  • In the another embodiment of formula [I-3], R[0059] 5 is a —COOH group or a —COOMe group and R6 is selected from the following:
    Figure US20030130349A1-20030710-C00023
  • In the above embodiment of formula [I-3], Z[0060] 1 may be selected from the following: a —O— group; a —NHCO— group; a —NHCH2— group; a —OCH2— group; a —CONH— group; a —NHSO2— group; or a —NHCOCH2— group.
  • In the above embodiment of formula [I-3], R[0061] 13 may be selected from the following: a —H group; a -iBuO group; a -i-Bu group; or a group selected from the following:
    Figure US20030130349A1-20030710-C00024
  • In the above embodiment of formula [I-3], R[0062] 14, which occurs one or more times and which may be the same or different in each occurence, may be selected from the following: a —H group; a —F group; a —Cl group; a —Br group; an —I group; a —CH3 group; a —OCH3 group; a —CF3 group; a —NO2 group; a —NH, group; or a -n-C7H15 group.
  • In the above embodiment of formula [I-3], R[0063] 15 may be selected from the following: a —H group; a —OH group; a —NO2 group; or a group selected from the following:
    Figure US20030130349A1-20030710-C00025
  • In the above embodiment of formula [I-3], R[0064] 16 may be selected from the following:
    Figure US20030130349A1-20030710-C00026
  • In the above embodiment of formula [I-3] R[0065] 17, which occurs one or more times and which may be the same of different in each occurrence, is a chlorine atom, provided that R1 and R3 must be different.
  • In another embodiment of formula [I-3], R[0066] 1 is methyl group and R2 may be selected from the following: a —CN group; a —COOH group; a —CONH2 group; a —CONHOH group; a —CH2OCH2COOH group; a —CH═CHCOOH group; a —CONHCH2COOH group; a —CONH(CH2)2COOH group; a —CONHCH2CONH2 group; a —CONH(CH2)2CN group; or a group selected from the following:
    Figure US20030130349A1-20030710-C00027
  • In the above, R[0067] 3 is a hydrogen atom, R5 is a —COOH group or a —COOMe group and R6 may be selected from the following:
    Figure US20030130349A1-20030710-C00028
  • In the above embodiment of formula [I-3], Z[0068] 1 may be selected from the following: a —NHCO— group; a —OCH2-group; a —NHCH2— group; a —CONH— group; or a —NHSO2-group.
  • In the above embodiment of formula [I-3], R[0069] 13 may be selected from the following:
    Figure US20030130349A1-20030710-C00029
  • In another embodiment of formula [I-3], R[0070] 14, which occurs one or more times and which may be the same or different in each occurence, may be selected from the following: a —F group; a —Cl group; a —Br group; an —I group; a —CH3 group; a —OCH3 group; a —CF3 group; or a —NO2 group.
  • In another embodiment of formula [I-3], R[0071] 15 may be a —H group or a —OH group.
  • In a another embodiment of formula [I-3], R[0072] 1 is a hydrogen atom or a methyl group and R2 may be selected from the following: a —CN group; a —COOH group; a —CONH2 group; a —CONHOH group; a —CONHOCH3 group; a —CH2OCH2COOH group; a —CH═CHCOOH group; a —CONHCH2COOH group; a —CONH(CH2)2COOH group; a —CONHCH2CONH2 group; a —CONH(CH2)2CN group; or a group selected from the following:
    Figure US20030130349A1-20030710-C00030
  • In another embodiment of formula [I-3], R[0073] 3 is a hydrogen atom or a methyl group, R5 is a —COOH group or a —COOMe group, and R6 may be selected from the following:
    Figure US20030130349A1-20030710-C00031
  • In another embodiment of formula [I-3], Z[0074] 1 may be selected from the following: a —O— group; a —NHCO— group; a —NHCH2— group; a —OCH2— group; a —CONH— group; or a —NHSO2— group.
  • In another embodiment of formula [I-3], R[0075] 13, may be selected from the following: a —H group; a -iBuO group; or a group selected from the following:
    Figure US20030130349A1-20030710-C00032
  • In another embodiment of formula [I-3], R[0076] 14, which occurs one or more times and which may be the same or different in each occurence, may be selected from the following: a —H group; a —OH; a —F group; a —Cl group; a —Br group; an —I group; a —CH3 group; a —OCH3 group; a —CF3 group; a —NO2 group; or a —NH2 group.
  • In another embodiment of formula [I-3], R[0077] 15 may be selected from the following: a —H group; a —OH group; a —NO2 group; or a group selected from the following:
    Figure US20030130349A1-20030710-C00033
  • In another embodiment of formula [I-3], R[0078] 16 may be selected from the following:
    Figure US20030130349A1-20030710-C00034
  • In another embodiment of formula [I-3], R[0079] 17, which occurs one or more times and which may be the same or different in each occurrence, may be a —Cl group or a —CF3 group, provided that R1 and R3 must be different.
  • In another embodiment of formula [I-3], R[0080] 1 may be a hydrogen atom or methyl group, R2 may be selected from the following: a —CN group; a —COOH group; a —CONH2 group; a —CONHOH group; a —CH2OCH2COOH group; a —CH═CHCOOH group; a —CONHCH2COOH group; a —CONH(CH21)2COOH group; a —CONHCH2CONH2 group; a —CONH(CH2)2CN group; or a group selected from the following:
    Figure US20030130349A1-20030710-C00035
  • and R[0081] 3 may be a hydrogen atom or a methyl group.
  • In another embodiment of formula [I-3], R[0082] 1 is —COOH group or a —COOMe group and R6 may be selected from the following:
    Figure US20030130349A1-20030710-C00036
  • In another embodiment of formula [I-3], Z[0083] 1 may be selected from the following: a —O— group; a —NHCO— group; a —NHCH2— group; a —OCH2— group; a —CONH— group; or a —NHSO2— group.
  • In another embodiment of formula [I-3], R[0084] 13 may be a —H group or a group selected from the following:
    Figure US20030130349A1-20030710-C00037
  • In another embodiment of formula [I-3], R[0085] 14, which occurs one or more times and which may be the same or different in each occurence, may be selected from the following: a —H group; a —F group; a —Cl group; a —Br group; an —I group; a —CH3 group; a —OCH3 group; a —CF3 group; a —NO2 group; or a —NH2 group.
  • In another embodiment of formula [I-3], R[0086] 15 may be selected from the following: a —H group; a —OH group, a —NO2 group; or a group selected from the following:
    Figure US20030130349A1-20030710-C00038
  • In another embodiment of formula [I-3], R[0087] 16 may be selected from the following:
    Figure US20030130349A1-20030710-C00039
  • In another embodiment of formula [I-3], R[0088] 17, which occurs one or more times and which may be the same or different in each occurrence, may be a —H group or a —Cl group, provided that R1 and R3 must be different.
  • In another embodiment of formula [I-3], R[0089] 1 is a methyl group, and R2 may be selected from the following: a —CN group; a —COOH group; a —CONH2 group; a —CONHOH group; a —CH2OCH2COOH group; a —CH═CHCOOH group; a —CONHCH2COOH group; a —CONH(CH2)2COOH group; a —CONH(CH2)2CN group; a —CONHCH2CONH2 group; or a group selected from the following:
    Figure US20030130349A1-20030710-C00040
  • In another embodiment of formula [I-3], R[0090] 3 is a hydrogen atom, R5 is a —COOH group or a —COOMe group, and R6 may be selected from the following:
    Figure US20030130349A1-20030710-C00041
  • In another embodiment of formula [I-3], Z[0091] 1 may be selected from the following: a —NHCO— group; a —NHCH2— group; a —NAcCH2— group; a —OCH2— group; or a —CONH-group.
  • In another embodiment of formula [I-3], R[0092] 13 may be selected from the following:
    Figure US20030130349A1-20030710-C00042
  • In another embodiment of formula [I-3], R[0093] 14, which occurs one or more times and which may be the same or different in each occurence, may be selected from the following: a —F group; a —Cl group; a —Br group; an —I group; a —OCH3 group; a —CF3 group; or a —NO2 group.
  • In another embodiment of formula [I-3], R[0094] 1 is a —H group or a —NO2 group.
  • Preferred compounds according to formula [I] may be selected from the group consisting of (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)-carbonyl]-1-[(3,4-dichlorophenyl)methyl]-L-histidine, (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine, (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine, (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-[(2,6-dichlorophenyl)methyl]-3-nitro-L-tyrosine, (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[[(2,4,6-trichlorophenyl)carbonyl]-amino]-L-phenylalanine, (1S-cis)-N-[[3-[[(2-Amino-2-oxoethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine, [0095]
  • (1S-cis)-N-[[3-[[(Carboxymethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine, and (1S-cis)-N-[(3-Cyano-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine. [0096]
  • The desired compounds of the present invention may exist in the form of optical isomers based on asymmetric carbon atoms thereof, and the present invention also includes these optical isomers and mixtures thereof. [0097]
  • In an embodiment of the present invention, the steric configuration of a bond need not be fixed. A bond may be of any acceptable configuration. Further, a compound may be a mixture with several different configurations of the same bond. [0098]
  • The desired compounds of the present invention may be used in the form of an ester or amide thereof. As the ester thereof, there may be mentioned a C[0099] 1-6 alkyl ester, a C2-7 alkenyl ester, a C2-7 alkynyl ester, a C2-7 alkanoyloxy-C1-6 alkyl ester, an aryl-C1-6 alkyl ester or an aryl ester. As the amide thereof, there may be mentioned an, amide (—CONH2), a mono or di N—C1-6 alkyl amide, an N—C3-8 cycloalkyl amide, an N-aryl amide or an N-aryl-C1-6 alkyl amide.
  • The desired compound of the present invention may be clinically used either in a free form or in the form of pharmaceutically acceptable salts thereof. Pharmaceutically acceptable salts include acid-addition salts with inorganic acid or organic acid (e.g., hydrochloride, sulfate, nitrate, hydrobromide, methanesulfonate, p-toluenesulfonate, acetate), salt with inorganic base, organic base or amino acid (e.g., triethylamine salt, a salt with lysine, an alkali metal salt, an alkali earth metal salt and the like). [0100]
  • The compound may also be formulated into a pharmaceutical composition comprising a therapeutically effective amount of the compound as defined above and a pharmaceutically acceptable carrier or diluent. [0101]
  • The compound can also be used for treating or preventing α[0102] 4β1 adhesion mediated conditions in a mammal such as a human. This method may comprise administering to a mammal or a human patient an effective amount of the compound or composition as explained above.
  • This method can be used to treat such inflammatory conditions as rheumatoid arthritis, asthma, allergy conditions, adult respiratory distress syndrome, AIDS, cardiovascular diseases, thrombosis or harmful platelet aggregation, reocclusion following thrombolysis, allograft rejection, reperfusion injury, psoriasis, eczema, contact dermatitis and other skin inflammatory diseases, osteoporosis, osteoarthritis, atherosclerosis, neoplastic diseases including metastasis of neoplastic or cancerous growth, wound healing enhancement, treatment of certain eye diseases such as detaching retina, Type I diabetes, multiple sclerosis, systemic lupus erythematosus (SLE), inflammatory and immunoinflammatory conditions including ophthalmic inflammatory conditions and inflammatory bowel diseases, ulcerative colitis, atherosclerosis, regional enteritis and other autoimmune diseases. [0103]
  • The desired compound of the present invention or pharmaceutically acceptable salts thereof may be administered either orally or parenterally, and it may be used as a suitable pharmaceutical preparation, for example, a tablet, a granule, a capsule, a powder, an injection, and an inhalation by a conventional process. [0104]
  • The dose of the desired compound of the present invention or a pharmaceutically acceptable salt thereof varies depending on an administration method, age, body weight, and state of a patient, but, in general, the daily dose is preferably about 0.1 to 100 mg/kg/day, particularly preferably 1 to 100 mg/kg/day. [0105]
  • Preferred Routes of Administration for Asthma: [0106]
  • It is preferred that the compound of the present invention be administered in the form of an Aerosol. However, other routes of administration include intravenous, oral, intramuscular, and subcutaneous. [0107]
  • In the case of aerosol administration, compositions containing the compounds of the present invention can be prepared to provide for an excellent means for administering in aerosol form for inhalation therapy. Accordingly, the present invention will provide for self-propelling compositions containing the compounds of the present invention. [0108]
  • Propellants employed should be non-toxic and have a vapor pressure suitable for the conditions under which administration occurs. These propellants can be fluorinated or fluorochlorinated lower saturated aliphatic hydrocarbons. The preferred propellants of this type are the halogenated alkanes containing not more than two carbon atoms and at least one fluorine atom. Illustrative of these are trichloromonofluoromethane, dichlorodifluoromethane, monochlorotrifluoromethane, dichloromonofluoromethane and 1,2-dichloro-1,1,2,2-tetrafluoroethane. These compounds are available from E. I. duPont de Nemours and Company under the trade name “Freon”. These propellants may be employed singularly or in admixture. [0109]
  • In addition to the propellant, an organic solvent may also be employed. The organic solvent must be non-toxic and without undesirable effects on inhalation in the amount present in the aerosol produced. In addition, the solvent should be substantially anhydrous, completely miscible with the propellant or mixture of propellants employed and have a suitable boiling point. Examples of such solvents included non-toxic aliphatic alcohols such as ethanol; ethers such as ethyl ether and vinyl ether; ketones such as acetone; and suitable halogenated lower alkanes. [0110]
  • In addition to the organic solvent, the composition may also optionally contain a non-toxic hygroscopic glycol. The glycol must be substantially miscible with the organic solvent and the propellant employed. Satisfactory glycols include propylene glycol, triethylene glycol, glycerol, butylene glycol and hexylene glycol. [0111]
  • The above indicated methods of admistration and formulation of aerosol compositions should not be viewed as limiting. The compounds of the present invention can be formulated in anyway deemed suitable to one of ordinary skill in the art so as to obtain the desired effects. [0112]
  • Pharmaceutical Compositions [0113]
  • As indicated previously, the compounds of formula (I) can be formulated into pharmaceutical compositions. In determining when a compound of formula (I) is indicated for the treatment of a given disease, the particular disease in question, its severity, as well as the age, sex, weight, and condition of the subject to be treated, must be taken into consideration and this perusal is to be determined by the skill of the attendant physician. [0114]
  • For medical use, the amount of a compound of Formula (I) required to achieve a therapeutic effect will, of course, vary both with the particular compound, the route of administration, the patient under treatment, and the particular disorder or disease being treated. A suitable daily dose of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for a mammalian subject suffering from, or likely to suffer from, any condition as described hereinbefore is 0.1 mg to 100 mg of the compound of formula I, per kilogram body weight of the mammalian subject. In the case of systematic administration, the dose may be in the range of 0.5 to 500 mg of the compound per kilogram body weight, the most preferred dosage being 0.5 to 50 mg/kg of mammal body weight administered two to three times daily. In the case of topical administration, e.g., to the skin or eye, a suitable dose may be in the range of 0.1 μg to 100 μg of the compound per kilogram, typically about 0.1 μg/kg. [0115]
  • In the case of oral dosing, a suitable dose of a compound of Formula (I), or a physiologically acceptable salt thereof, may be as specified in the preceding paragraph, but most preferably is from 1 mg to 10 mg of the compound per kilogram, the most preferred dosage being from 1 mg to 5 mg/kg of mammal body weight, for example, from 1 to 2 mg/kg. Most preferably, a unit dosage of an orally administrable composition encompassed by the present invention contains less than about 1.0 g of a formula (I) compound. [0116]
  • It is understood that formulation, both for human and veterinary use, of the present invention may be presented to the mammal by inhalation. To achieve therapeutic effect, the dose may be in the range of 0.5 to 500 mg of the compound, per kg body weight. The most preferred dosage being 0.5 to 50 mg/kg of mammal body weight administered two to three times daily. [0117]
  • It is understood that the ordinarily skilled physician or veterinarian will readily determine and prescribe the effective amount of a compound of Formula I to prevent or arrest the progress of the condition for which treatment is administered. In so proceeding, the physician or veterinarian could employ relatively low doses at first, subsequently increasing the dose until a maximum response is obtained. [0118]
  • The compounds and compositions of the present invention can be administered to patients suffering from a condition listed herein in an amount which is effective to fully or partially alleviate undesired symptoms of the condition. The symptoms may be caused by inappropriate cell adhesion mediated by 0401 integrins. Such inappropriate cell adhesion would typically be expected to occur as a result of increased VCAM-1 and/or CS-1 expression on the surface of endothelial cells. Increased VCAM-1 and/or CS-1 expression can be due to a normal inflammation response or due to abnormal inflammatory states. In either case, an effective dose of a compound of the invention may reduce the increased cell adhesion due to increased VCAM-1 expression by endothelial cells. Reducing the adhesion observed in the disease state by 50% can be considered an effective reduction in adhesion. More preferably, a reduction in adhesion by 90%, is achieved. Most preferably adhesion mediated by VCAM-1/U40, and/or CS-1 interaction is abolished by an effective dose. Clinically, in some instances, effect of the compound can be observed or a decrease in white cell infiltration into tissues or a site of injury. To achieve a therapeutic effect, then, the compounds or compositions of the present invention are administered to provide a dose effective to reduce or eliminate inappropriate cell adhesion or to alleviate undesired symptoms. [0119]
  • While it is possible for an active ingredient to be administered alone, it is preferable to present it as a pharmaceutical formulation comprising a compound of Formula (I) and a pharmaceutically acceptable carrier thereof. Such formulations constitute a further feature of the present invention. [0120]
  • The formulations, both for human and veterinary medical use, of the present invention comprise an active ingredient of Formula (I), in association with a pharmaceutically acceptable carrier thereof and optionally other therapeutic ingredient (s), which are generally known to be effective in treating the disease or condition encountered. The carrier(s) must be “acceptable” in the sense of being compatible with the other ingredients of the formulations and not deleterious to the recipient thereof. [0121]
  • The formulations include those in a form suitable for oral, pulmonary, ophthalmic, rectal, parenteral (including subcutaneous, intramuscular, and intravenous), intra-articular, topical, nasal inhalation (e.g., with an aerosol) or buccal administration. Such formulation are understood to include long-acting formulations known in the art. [0122]
  • The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. All methods may include the step of bringing the active ingredient into association with the carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired form. [0123]
  • Formulations of the present invention suitable for oral administration may be in the form of discrete units such as capsules, cachets, tablets, or lozenges, each containing a predetermined amount of the active ingredient in the form of a powder or granules; in the form of a solution or suspension in an aqueous liquid. Formulations for other uses could involve a nonaqueous liquid; in the form of an oil-in-water emulsion or a water-in-oil emulsion; in the form of an aerosol; or in the form of a cream or ointment or impregnated into a transdermal patch for use in administering the active ingredient transdermally, to a patient in need thereof. The active ingredient of the present inventive compositions may also be administered to a patient in need thereof in the form of a bolus, electuary, or paste. [0124]
  • The practitioner is referred to “Remington: The Science and Practice of Pharmacy,” 19th Edition, c. 1995 by the Philadelphia College of Pharmacy and Science, as a comprehensive tome on pharmaceutical preparations. [0125]
    Abbreviations
    Ac2O: Acetic anhydride
    AcOEt: Ethyl acetate
    BCECF-AM: 2′,7′-bis-(2-carboxyethyl)-5-(and 6-)
    carboxyfluorescein acetoxymethyl ester
    BOP-Cl: Bis (2-oxo-3-oxazolidinyl) phosphinic
    chloride
    BOP Reagent: Benzotriazol-1-yloxy-tris
    (dimethylamino)-phosphonium
    hexafluorophosphate
    DMEM: Dulbecco's Minimal Eagle's Media
    DMF: Dimethyl formamide
    DIEA: Diisopropylethylamine
    EDC: 1-(3-Dimethylaminopropyl)-3-
    ethylcarbodiimide hydrochloride
    Et: Ethyl
    EtOH: Ethanol
    HATU: N-[(Dimethylamino)-1H-1,2,3-triazolo[4,5-
    b]-pyridin-1-ylmethylene]-N-
    methylmethanaminium hexafluorophosphate
    N-oxide
    HBSS: Hank's Balanced Salt Solution
    HBTU: O-Benzotriazol-1-yl-N,N,N′,N′-
    tetramethyluronium hexafluorophosphate
    HOBT: 1-Hydroxybenzotriazole
    HSA: Human serum albumin
    LDA: Lithium diisopropylamide
    Me: Methyl
    meq: milliequivalent
    MeOH: Methanol
    n-Bu: n-Butyl
    NMP: 1-Methyl-2-pyrrolidinone
    PBS: Phosphate buffered saline
    Pd-C: Palladium on charcoal
    Ph: Phenyl
    SPDP: 3-(2-pyridyldithio) propionic acid N-
    hydroxysuccinimide ester
    t-Bu: t-butyl
    THF: Tetrahydrofuran
    TFA: Trifluoroacetic acid
  • According to the present invention, the desired compound [I] can be prepared by the following methods: [0126]
    Figure US20030130349A1-20030710-C00043
  • wherein R[0127] 5a is a group of the formula: —COOR22, —(C1-6 alkylene)COOR22, C1-7 alkylene)O(C1-6 alkyl), —(C1-7alkylene) OH, —COO(C1-6 alkyl)-CONH(C1-6 alkyl) or —CONH2,
  • R[0128] 22 is a protecting group for the carboxyl group, and the other symbols are the same as defined above.
  • Method A: [0129]
  • The compound of the formula [I] wherein R[0130] 2 is a group of the formula: —COOH and X is a group of the formula: —CO—, an ester thereof, an amide thereof or a pharmaceutically acceptable salt thereof, i.e., the compound of the formula [I-a]:
    Figure US20030130349A1-20030710-C00044
  • wherein the symbols are the same as defined above, an ester thereof, an amide thereof or a pharmaceutically acceptable salt thereof may be prepared by [0131]
  • (1) reacting a compound of the formula [II]: [0132]
    Figure US20030130349A1-20030710-C00045
  • wherein the symbols are the same as defined above, with a compound of the formula [III-a]: [0133]
    Figure US20030130349A1-20030710-C00046
  • wherein the symbols are the same as defined above, or a salt thereof, [0134]
  • (2) removing the protecting group for the carboxyl group, if desired, and [0135]
  • (3) converting the resulting compound into an ester thereof, an amide thereof or a pharmaceutically acceptable salt thereof by a conventional method, if further desired. [0136]
  • R[0137] 22 can be selected from a conventional protecting group for a carboxyl group, for example, a C1-6 alkyl group, a C2-7 alkenyl group, a C2-7 alkynyl group, a C2-7 alkanoyloxy-C1-6 alkyl group, an aryl-C1-6 alkyl group (e.g., benzyl group) or an aryl group (e.g., phenyl group) and the like.
  • A salt of the compound [III-a] includes, for example, salt with an inorganic acid (e.g., hydrochloride, sulfate) and salt with an inorganic base (e.g., an alkali metal salt such as sodium salt or potassium salt, an alkali earth metal salt such as magnesium salt or calcium salt). [0138]
  • The reaction of the compound [II] and the compound [III-a] or a salt thereof is carried out in the presence of a base in a suitable solvent or without a solvent. The base can be selected from an organic base (e.g., DIEA, DMAP, Et[0139] 3N, DBU), an alkali metal hydride (e.g., NaH, LiH), an alkali metal carbonate (e.g., Na2CO3, Na2KO3) an alkali metal hydrogen carbonate (e.g., NaHCO3, KHCO3), an alkali metal amide (e.g., NaNH2), an alkali metal alkoxide (e.g., NaOMe, KOMe), an alkyl-alkali metal (n-BuLi, t-BuLi), an alkali metal hydroxide (e.g., NaOH, KOH), an alkali earth metal hydroxide (e.g., Ba(OH)2), and the like. The solvent can be selected from any one which does not disturb the reaction, for example, DMF, THF, benzene, toluene, DMSO, CH3CN or a mixture thereof. The reaction is preferably carried out at a temperature from 0° C. to 100° C., more preferably at a temperature from 400% to 80° C.
  • The removal of said protecting group from the products can be carried out by a conventional method, which is selected according to the types of the protecting groups to be removed, for example, hydrolysis, acid treatment, catalytic reduction, and the like. [0140]
  • A more preferred method than method A is: [0141]
    Figure US20030130349A1-20030710-C00047
  • R[0142] 2a is a group of the formula: —CN, —COOR23, —COOH, —(C1-6 alkylene)OH, —CH2O(C1-6 alkyl), —(C1-6 alkylene)COOH, —(C1-6 alkylene)COOR23, —CH2C(C1-6 alkylene)O(C1-6 alkyl), —CH2O(C1-6 alkylene) COOH, —CH2O(C1-6 alkylene) COOR23, —(C2-7 alkenylene) COOH, —(C2-7 alkenylene) COOR23, —CO(C1-6 alkylene)COOH, —CO(C1-6 alkylene)COOR23, —CO(C2-7 alkenylene)COOH, —CO(C2-7 alkenylene)COOR23, —CO(C1-6 alkylene)O(C1-6 alkyl), —CO(C1-6 alkylene)CO(C1-6 alkyl), —CONH(C1-6 alkyl), —CONHO(C1-6 alkyl), —CONH(C1-6 alkylene) COOH, —CONH(C1-6 alkylene)COOR23, —CONH(C3-7 cycloalkyl), —CONH2, —CONH(C1-6 alkylene)CONH2, —CONHOH, —NHCO2CH2Ph, —CONHOCH2Ph, —CONH(C1-6alkylene) CN, —COO(C1-6alkyl), —CH2O(C1-6alkylene) CONH2,
    Figure US20030130349A1-20030710-C00048
  • R is a group of the formula: —CN, —COOR[0143] 23, —(C1-6 alkylene) OH, —CH2O(C1-6 alkyl), —(C1-6 alkylene) COOR23, —CH2O(C1-6 alkylene)O(C1-6 alkyl), —CH2O(C1-6 alkylene)COOR23, —(C2-7 alkenylene) COOR23, —CO(C1-6 alkylene)COOR23, —CO(C2-7 alkenylene) COOR23, —CO(C1-6 alkylene)O(C1-6 alkyl), —CO(C1-6 alkylene)CO(C1-6 alkyl), —CONH(C1-6 alkyl), —CONHO(C1-6 alkyl), —CONH(C1-6 alkylene)COOR23, —CONH(C3-7 cycloalkyl), —CONH2, —CONH(C1-6 alkylene) CONH2, —CONHOH, —NHCO2CH2Ph, —CONHOCH2Ph, —CONH(C1-6alkylene) CN, —COO(C1-6alkyl) —CH2O(C1-6alkylene) CONH2,
    Figure US20030130349A1-20030710-C00049
  • R[0144] 23 is a protecting group for the carboxyl group, and the other symbols are the same as defined above.
  • Method B: [0145]
  • The compound of the formula [II, an ester thereof, an amide thereof or a pharmaceutically acceptable salt thereof, [0146]
  • wherein R[0147] 2 is a group of the formula: —CN, —COOR23, —COOH, —(C1-6 alkylene) OH, —CH2O(C1-6 alkyl), —(C1-6 alkylene)COOH, —(C1-6 alkylene)COOR23, —CH2O(16 alkylene)O(C1-16 alkyl), —CH2O(C1-6 alkylene)COOH, —CH2O(C1-16 alkylene)COOR23, —(C2-7 alkenylene)COOH, —(C2-7 alkenylene)COOR23, —CO(C1-16 alkylene)COOH, —CO(C1-16 alkylene)COOR23, —CO(C2-7 alkenylene)COOH, —CO(C1-27 alkenylene)COOR23, —CO(C1-6 alkylene)O(C1-6 alkyl), —CO(C1-6 alkylene)CO(C1-6 alkyl), —CONH(C1-6 alkyl), —CONHO(C1-6 alkyl), —CONH(C1-16 alkylene) COOH, —CONH(C1-16 alkylene)COOR23, —CONH(C3-7 cycloalkyl), —CONH2, —CONH(C1-6 alkylene)CONH2, —CONHOH, —NHCO2CH2Ph, CONHOCH2Ph, —CONH(C1-6alkylene) CN, —COO(C1-6alkyl), —CH2O(C1-6alkylene) CONH2 or
    Figure US20030130349A1-20030710-C00050
  • X is a group of the formula: —CO—, i.e., the compound of the formula-[I-c]: [0148]
    Figure US20030130349A1-20030710-C00051
  • wherein [0149]
  • the symbols are the same as defined above, may be prepared by [0150]
  • (1) condensing a compound of the formula [IV]: [0151]
    Figure US20030130349A1-20030710-C00052
  • wherein the symbols are the same as defined above, or a salt thereof, with a compound of the formula [III-a]: [0152]
    Figure US20030130349A1-20030710-C00053
  • wherein the symbols are the same as defined above, or a salt thereof, [0153]
  • (2) removing the protecting group for the carboxyl group and hydroxyl group, if desired, and [0154]
  • (3) converting the resulting compound into an ester thereof, an amide thereof or a pharmaceutically acceptable salt thereof by a conventional method, if further desired. [0155]
  • R[0156] 22 and R23 are the same or different conventional protecting group for a carboxyl group, for example, a C1-6 alkyl group, a C2-7 alkenyl group, a C2-7 alkynyl group, a C2-7 alkanoyloxy-C1-6 alkyl group, an aryl-C1-6 alkyl group (e.g., benzyl group) or an aryl group (e.g., phenyl group) and the like.
  • A salt of the compound [III-a] and/or [IV] includes, for example, salt with an inorganic acid (e.g., hydrochloride, sulfate) and salt with an inorganic base (e.g., an alkali metal salt such as sodium, potassium and calcium, an alkali earth metal salt such as barium). [0157]
  • The condensation reaction of the compound [IV] or a salt thereof with the compound [III-a] or a salt thereof is carried out in the presence of a condensing reagent in a suitable solvent or without a solvent. The condensing reagent can be selected from any one which can be used for a conventional peptide synthesis, for example, BOP-Cl, BOP reagent, DCC and WSCI. [0158]
  • The solvent can be selected from any one which does not disturb the condensation reaction, for example, CH[0159] 2Cl2, DMF or a mixture thereof. The reaction is preferably carried out at a room temperature.
  • The removal of said protecting group from the products can be carried out by a conventional method, which is selected according to the types of the protecting groups to be removed, for example, hydrolysis, acid treatment, catalytic reduction, and the like. [0160]
    Figure US20030130349A1-20030710-C00054
  • wherein the symbols are the same as defined above. [0161]
  • Method C: [0162]
  • The compound of the formula [I] wherein X is a methylene group, an ester thereof, an amide thereof or a pharmaceutically acceptable salt thereof, i.e., the compound of the formula [I-e]: [0163]
    Figure US20030130349A1-20030710-C00055
  • wherein [0164]
  • the symbols are the same as defined above, an ester thereof, an amide thereof or a pharmaceutically acceptable salt thereof, may be prepared by [0165]
  • (1) reacting a compound of the formula [V]: [0166]
    Figure US20030130349A1-20030710-C00056
  • wherein the symbols are the same as defined above, or a salt thereof in the presence of a reducing agent with a compound of the formula [III-b]: [0167]
    Figure US20030130349A1-20030710-C00057
  • wherein the symbols are the same as defined above, [0168]
  • (2) removing the protecting group for the carboxyl group, if desired, and [0169]
  • (3) converting the resulting compound into an ester thereof, an amide thereof or a pharmaceutically acceptable salt thereof by a conventional method, if further desired. [0170]
  • A salt of the compound [V] and/or [III-b] includes, for example, salt with an inorganic acid (e.g., hydrochloride, sulfate) and salt with an inorganic base (e.g., an alkali metal salt, an alkali earth metal salt). [0171]
  • The reductive alkylation of the compound [V] or a salt thereof with the compound [III-b] or a salt thereof is carried out by a conventional method in the presence of a reducing agent in a suitable solvent or without a solvent. The reducing agent is preferably sodium borohydride, sodium cyanoborohydride, and the like. The solvent can be selected from any one which does not disturb the reaction, for example, alkanol such as methanol, alkanoic acid such as AcOH, THF or a mixture thereof. The reaction is preferably carried out at a temperature from 0° C. to a room temperature. [0172]
  • The reaction of the compound [I-f] or a salt thereof and the compound [VII] is carried out in the presence of an acid acceptor in a suitable solvent or without a solvent. The acid acceptor and the solvent can be selected from the base or the solvent used in Method A. The reaction is preferably carried out at room temperature. [0173]
  • The removal of said protecting group from the products can be carried out by a conventional method, which is selected according to the types of the protecting groups to be removed, for example, hydrolysis, acid treatment, catalytic reduction, and the like. [0174]
  • The desired compound [I] of the present invention can be converted to each other. Such conversion of the present compound [I] into the other compound [I] may be carried out by selecting one of the following procedures from (a) to (e) according to the type of substituent thereof, and if desired, followed by removing the protecting group for the carboxyl group by a conventional method. [0175]
  • Procedure (a): [0176]
  • The compound [I] wherein R[0177] 6 is an amino-substituted aryl group can be prepared by the reduction of the compound [I] wherein the corresponding R6 is an aralklyoxycarbonyl amino group- or nitro-substituted aryl group. The reduction can be, for example, a, catalytic reduction using a palladium catalyst such as palladium on an activated carbon, a platinum catalyst such as platinum oxide, and the like. The catalytic reduction is preferably carried out at a room temperature.
  • Procedure (b): [0178]
  • The compound [I] wherein R[0179] 2 is a group of the formula: —CONH2, —CONH(C1-6 alkyl), —CONHO(C1-6 alkyl), —CONH(C1-6 alkylene)COOR23, —CONH(C3-7 cycloalkyl), —CONH(C1-6 alkylene) CONH2, —CONHOCH2Ph, —CONH(C1-6 alkylene)CN, CONHOH,
    Figure US20030130349A1-20030710-C00058
  • can be prepared by reacting the compound [I] wherein the corresponding R[0180] 2 is a group of the formula: —COOH with a substituted or unsubstituted amine selected from a group of the formula: NH3, NH2(C1-6 alkyl), NH2O(C1-6 alkyl), NH2(C1-6 alkylene)COOR23, NH2(C3-7 cycloalkyl), NH2(C1-6 alkylene)CONH2, NH2OH, NH2OCH2Ph, NH2 (C1-6 alkylene)CN,
    Figure US20030130349A1-20030710-C00059
  • wherein R[0181] 23 is defined as above, in the presence of a condensing reagent (e.g., BOP reagent) which can be used for a conventional peptide synthesis, and removing the protecting group for the carboxyl group, if desired. The reaction is preferably carried out at a temperature from 0° C. to a room temperature.
  • Procedure (c): [0182]
  • The compound [I] wherein R[0183] 4 is a C1-6 alkyl group can be prepared by reacting the compound [I] wherein the corresponding R4 is a hydrogen atom with a C1-6 alkyl halide (e.g., methyl iodide, butyl iodide) in the presence of metal hydride (e.g., NaH). The reaction is preferably carried out at a temperature from 0° C. to room temperature.
  • Procedure (d) [0184]
  • The compound [I] wherein R[0185] 6 is a C2-6 alkanoylamino-, C3-7 cycloalkylcarbonylamino-, aryl C2-7 alkanoylamino-, arylcarbonylamino-, C1-5 alkyloxycarbonylamino-, C3-7 cycloalkyloxycarbonylamino-, aryl C1-6 alkyloxycarbonylamino-, arylureido, or arylsulfonylamino-substituted aryl group can be prepared by reacting the compound [I] wherein the corresponding R6 is an amino aryl group or a (C1-6 alkyl)-amino-substituted aryl group with a C2-16 alkanoic acid, an anhydride of C2-6 alkanoic acid, C2-6alkanoyl halide, C3-7 cycloalkcanecarboxylic acid, anhydride of C3-7 cycloalkane-carboxylic acid, C3-7 cycloalkanoyl halide, aryl C2-7 alkanoic acid, anhydride of aryl C2-7 alkanoic acid, aryl C2-7 alkanoyl halide, arylcarboxylic acid, anhydride of arylcarboxylic acid, arylcarbonyl halide, C1-5 alkyl halogenoformate, arylisocyanate, or arylsulfonyl halide in the presence or absence of an acid acceptor (e.g., DIEA) and in the presence or absence of a condensing reagent (e.g., BOP-Cl) which can be used for a conventional peptide synthesis. The reaction is preferably carried out at a temperature from one to a room temperature.
  • Procedure (e): [0186]
  • {circle over (P)}=a resin which is used in a conventional solid phase peptide synthesis [0187]
    Figure US20030130349A1-20030710-C00060
  • wherein R[0188] 24 is a protecting group for the amino group, and the other symbols are the same as defined above.
  • The compound of the formula [I-1]: [0189]
    Figure US20030130349A1-20030710-C00061
  • can be prepared by condensing the compound [I] wherein the corresponding R[0190] 2 is a group of the formula: —COOH with a group of the formula [VIII]:
    Figure US20030130349A1-20030710-C00062
  • wherein the symbols are the same as defined above, by a conventional solid phase peptide synthesis method also known as Merrifield method (Journal of American Chemical Society 85, 2149-2154 (1963)), followed by the deprotection of amino group and carboxyl group by a conventional method. [0191]
  • R[0192] 24 can be selected from a conventional protecting group for an amino group, for example, tert-butoxy-carbonyl grop (BOC), benzyloxycarbonyl group (Cbz) and the like.
  • The solvent used for the Procedures (a) to (e) may be selected from any one which does not disturb the procedures, for example, THF, methanol, DMF, CH[0193] 2Cl, or a mixture thereof.
  • General Description for Synthesis of Intermediates [0194]
  • The compound [II] may be prepared by reacting a compound of the formula [VI]: [0195]
    Figure US20030130349A1-20030710-C00063
  • wherein the symbols are the same as defined above, in the presence of C[0196] 1-6 alkanoyl halide (e.g., AcCl) and/or C1-6 alkanoic anhydride (e.g., Ac2O).
  • The compound [III-a] may be prepared by a conventional method, which is selected according to the types of the substituents, for example, by the following schemes: [0197]
    Figure US20030130349A1-20030710-C00064
    Figure US20030130349A1-20030710-C00065
    Figure US20030130349A1-20030710-C00066
  • wherein R is (1) a substituted or unsubstituted monocyclic or bicyclic aryl group or (2) a substituted or unsubstituted monocyclic or bicyclic heteroaryl group, Hal is a halogen atom and other symbols are the same as defined above. [0198]
  • The compound [IV] may be prepared as shown in various locations of the present application, for example, in Schemes 7, 8, 9, 10 and 11. [0199]
  • The desired compound [I] of the present invention may also be prepared by the methods as shown in the following Schemes. [0200]
    Figure US20030130349A1-20030710-C00067
  • Commercially available (L)-p-nitroPhe-OH (5a-A) (50.6 g, 240.6 mmol) was dissolved in MeOH (250 mL) and dry HCl was bubbled through the solution for 45 minutes at 0° C. The mixture was refluxed for 15 minutes and allowed to stand overnight. The HCl salt precipitated and the solid material was collected by filtration and washed with Et[0201] 2O (3×50 mL). The solid methyl ester (5a-B) thus obtained was pale yellow (55.3 g, 88%): mp=215-218° C. (d).
  • The HCl salt of (L)-p-nitroPhe-OMe (5a-B) (5.2 g, 19.8 mmol) was dissolved in THF (30 mL) containing DIEA (10.3 mL, 59.4 mmol) To this solution was added (1R-cis)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic acid 1-(1,1-dimethylethyl) ester (15-D) (5.1 g, 19.8 mmol) and BOP reagent (10.6 g, 23.9 mmol) and the solution was stirred under dry N[0202] 2 for 72 hours. Work-up of the coupling reaction was performed by the addition of 1N HCl (60 mL) and extraction with EtOAc (2×20 mL). The combined organic phase was washed with saturated NaHCO3 (20 mL), then saturated LiCl (15 mL) and dried over Na2SO4. The solution was filtered, solvent evaporated and the residue chromatographed (SiO2, gradient elution: 100% hexanes→50% EtOAc/hexanes) to provide the fully protected intermediate (1S-cis)-N-[3-{(1,1-dimethylethoxy)-2,2,3-trimethylcyclopentyl]carbonyl]-4-nitro-L-phenylalanine methyl ester (5a-C) (7.1 g, 77%):
  • [0203] 1H NMR (300 MHz, CDCl3), δ 8.14 (2H), 7.35 (2H). 6.18 (1H), 4.99 (1H), 3.76 (3H), 3.34 (1H), 3.20 (1H), 2.62 (1H), 2.5-2.6 (2H), 2.1-2.2 (1H), 1.6-1.8 (1H), 1.4-1.5 (1H), 1.45 (9H), 1.25 (3H), 1.16 (3H), 0.81 (3H); 13C NMR (75 MHz, CDCl3), δ 174.57, 172.44, 171.30, 146.69, 144.01, 129.87, 123.31, 79.89, 56.35, 53.98, 52.52, 52.24, 46.10, 37.38, 32.08, 27.72, 22.64, 22.27, 21.62, 20.40; ESMS (m/z) 463 (MH+).
  • The above compound, (5a-C) (2.7 g, 5.77 mmol), was dissolved in MeOH (40 mL) and degassed with N[0204] 2. To this solution was added 10% Pd-C (250 mg) and H2 gas was bubbled through the resultant slurry for 15 minutes and the reaction was stirred an additional 3 hours under an atmosphere of H2. The mixture was filtered through celite and the celite washed with CH3OH. The solvent was evaporated to afford Example 56 (5a-D) (2.49 g, 100):
  • [0205] 1H NMR (300 MHz, CDCl3), δ 6.88 (2H), 6.64 (2H), 5.71 (1H), 4.82 (1H), 3.72 (3H), 3.0-3.1 (2H), 2.5-2.6 (2H), 2.1-2.2 (1H), 1.6-1.8 (1H), 1.4-1.5 (1H), 1.43 (9H) 1.21 (3H), 1.14 (3H), 0.80 (3H); ESMS (m/z) 433 (MH+).
    Figure US20030130349A1-20030710-C00068
  • Commercially available Boc-(L)-Phe(4-N-Cbz)-OH (5b-A) (6.2 g, 14.9 mmol) was dissolved in MeOH (20 mL) and dry HCl was bubbled through the solution for 10 minutes. This mixture was stirred for 1 hour. The solvent was evaporated and the solid material (5b-B) thus obtained was washed with cold Et[0206] 2O (3×20 mL). This solid material was dissolved in THF (25 mL) containing DIEA (7.8 mL, 44.8 mmol). To this solution was added (15-D) (4.2 g, 16.4 mmol) and BOP reagent (7.9 g, 17.9 mmol) and the solution was stirred under dry N2 overnight. Work-up of the coupling reaction was performed by the addition of 1N HCl (60 mL) and extraction with EtOAc (2×50 mL). The combined organic phase was washed with saturated LiCl (35 mL) and dried over Na2SO4. The solution was filtered, solvent evaporated and the residue chromatographed (SiO2, gradient elution: 100% hexanes→500 EtOAc/hexanes) to provide the fully protected intermediate 4-benzyloxycarbonylamino-N-[[(1S,3R)-3-(tert-butoxycarbonyl)-2,2,3-trimethylcyclopentyl]carbonyl]-L-phenylalanine methyl ester (5b-C) (6.5 g, 77%) 1H NMR (300 MHz, CDCl3), δ 7.3-7.4 (6H), 7.17 (1H), 7.01 (2H), 5.80 (1H), 5.17 (2H), 4.86 (1H), 3.70 (3H) 3.06 (2H), 2.5-2.6 (2H) 2.1-2.2 (1H), 1.6-1.8 (1H), 1.4-1.5 (1H), 1.43 (9H) 1.21 (3H), 1.13 (3H), 0.80 (3H); 13C NMR (75 MHz, CDCl3) δ 174.88, 172.43, 172.06, 153.32, 136.98, 135.95, 130.55, 129.58, 128.46, 128.19, 128.15, 118.71, 80.05, 66.80, 56.54, 54.26, 52.98, 52.17, 46.25, 36.94, 32.21, 27.92, 22.81, 22.32, 21.82, 20.46; ESMS (m/z) 567 (MH+).
  • The above compound, (5b-C) (5.74 g, 10.13 mmol), was dissolved in MeOH/THF (4:1, 50 mL) and degassed with N[0207] 2. To this solution was added 10% Pd-C (500 mg) and H2 gas was bubbled through the resultant slurry for 1 hour. The reaction was stirred an additional 3 hours under an atmosphere of H2. The mixture was filtered though celite and the celite washed with CH3OH. The solvent was evaporated to afford Example 56 (5a-D) (4.38 g, 100%):
  • [0208] 1H NMR (300 MHz, CDCl3), δ 6.90 (2H), 6.68 (2H), 5.73 (1H), 4.82 (1H), 3.72 (3H), 3.0-3.1 (2H), 2.5-2.6 (2H), 2.1-2.2 (1H), 1.6-1.8 (1H), 1.4-1.5 (1H), 1.43 (9H), 1.21 (3H), 1.14 (3H), 0.80 (3H); ESMS (m/z) 433 (MH+).
    Figure US20030130349A1-20030710-C00069
  • (1R)-Camphoric anhydride (243 mg, 1.33 mmol) was dissolved in THF (10 mL) containing DIEA (1.2 mL, 6.67 mmol). To this solution O-2,6-dichlorobenzyl-L-tyrosine methyl ester (6-A) (618 mg, 1.58 mmol) was added and the solution stirred at 45° C. for 1 h. The reaction was cooled to room temperature and 1 N HCl (20 mL) was added. This was extracted with EtOAc (2×20 mL) and the combined organics were dried (Na[0209] 2SO4), filtered, and the solvent removed in vacuo. The residue was chromatographed (SiO2, 10% MeOH in CH2Cl2) to provide (6-B) (668 mg, 93%) as a colorless oil: 1H NMR (300 MHz, CDCl3), (major isomer) δ 7.34 (d, 2H), 7.23 (dd, 1H), 7.04 (d, 2H), 6.9-7.0 (m, 2H), 5.85 (d, 1H), 5.23 (s, 2H), 4.88 (q, 1H), 3.73 (s, 3H), 3.0-3.2 (m, 2H), 2.5-2.6 (m, 2H), 2.2-2.3 (m, 1H), 1.7-1.8 (m, 1H), 1.4-1.5 (m, 1H), 1.23 (s, 3H), 1.22 (s, 3H), 0.84 (s, 3H); ESMS (m/z) 536 (MH+).
  • (6-B) (570 mg, 1.06 mmol) was dissolved in THF (2 mL). To this solution LiOH (89 mg, 3.72 mmol) was added in H[0210] 2O (2 mL) and the mixture stirred for 12 h at RT. The reaction was acidified with 1 N HCl (10 mL) and then extracted with EtOAc (2×20 mL). The combined organics were dried (Na2SO4), filtered, and the solvent removed in vacuo to provide Example 12 (6-C) (525 mg, 95%) as a pale yellow foam: 1H NMR (300 MHz, Acetone-d6), (major isomer) δ 7.4-7.5 (m, 3H), 7.23 (d, 2H), 6.99 (d, 2H), 5.30 (d, 1H), 5.28 (s, 2H), 4.6-4.7 (m, 1H), 3.13 (dd, 1H), 2.97 (dd, 1H), 2.81 (t, 1H), 2.5-2.6 (m, 1H), 2.0-2.1 (m, 1H), 1.6-1.7 (m, 1H), 1.3-1.4 (m, 1H), 1.27 (s, 3H), 1.19 (s, 3H), 0.80 (s, 3H); 13C NMR (75 MHz, Acetone-d6), (major isomer) δ 177.79, 173.99, 172.94, 158.53, 137.39, 133.18, 132.01, 131.23, 129.52, 115.24, 65.78, 56.75, 54.47, 53.67, 46.91, 37.19, 33.30, 23.44, 23.01, 22.38, 21.64; ESMS (m/z) 520 (M−H).
    Figure US20030130349A1-20030710-C00070
  • Preparation 7-A Scheme 7, 7-A Stereochemistry=1S-cis (Intermediate for Examples 181, 185, and 188)
  • (1R,3S)-Camphoric Acid 1-(1,1-Dimethylethyl)-3-phenylmethyl ester (C[0211] 21H30O4)
  • Benzyl bromide is eluted through neutral alumina (10 mL in a 30 mL sintered glass funnel) to give a colorless liquid (15 mL, 126 mmol) which is added to a stirred solution of (1R,3S)-camphoric acid 1-(1,1-dimethylethyl) ester (15-D) (30 g, 117 mmol), N,N-diisopropylethyl amine (24 mL, 138 mmol), and acetonitrile (90 ml). After seven days, the mixture is filtered to give a white solid (diisopropylethyl amine hydrobromide) and a yellow liquid which is placed in the freezer. After two days, the mixture is filtered (two 50 mL diethyl ether rinses) to give a white solid (24 g, 59% yield). [0212]
  • [0213] 1H NMR: (300 MHz, CDCl3): δ 7.37-7.29 (5H), 5.15 (1H), 5.09 (1H), 2.85-2.79 (1H), 2.54-2.46 (1H), 2.24-2.15 (1H), 1.86-1.74 (1H), 1.49-1.36 (1H), 1.43 (9H), 1.23 (,3H), 1.15 (3H), 0.78 (3H); IR (nujol) 1737, 1724, 1717, 1346, 1272, 1259, 1219, 1210, 1162, 1124, 1116, 1084, 852, 737, 696 cm−1; MS (FAB) m/z (rel. intensity) 347 (M+H, 35), 348 (8), 347 (35), 292 (8), 291 (43), 273 (12), 109 (13), 92 (9), 91 (99), 57 (30), 41 (9); HRMS (FAB) calcd for C21H30O4+H+ 347.2222, found 347.2232; Anal. Calcd for C21H30O4: C, 72.80; H, 8.73; Found: C, 72.79; H, 8.90.
  • (1R,3S)-Camphoric Acid 3-phenylmethyl ester (7-A) (C[0214] 17H22O4)
  • To (1R,3S)-camphoric Acid 1-(1,1-Dimethylethyl)-3-phenylmethyl ester (24 g, 69 mmol) is added trifluoroacetic acid (15 mL). After stirring for two days, the solution is evaporated in vacuo to give a pale yellow oil which is dissolved in toluene (250 mL) and shaken with water (6×100 mL). Evaporation of the toluene gave a colorless oil which slowly crystallizes to give 7-A as an oily, white solid (16.4 g, 81% yield). [0215]
  • [0216] 1H-NMR: (300 MHz, CDCl3) δ 7.38-7.16 (5H), 5.17 (1H), 5.11 (1H), 2.87 (1H), 2.60-2.49 (1H) 2.30-2.21 (1H), 1.91-1.80 (1H) 11.57-1.48 (1H), 1.27 (3H), 1.25 (3H), 0.84 (3H); IR (liq.) 3067, 3034, 2972, 2888, 1732, 1696, 1457, 1378, 1285, 1231, 1212, 1166, 1124, 752, 698 cm−1; MS (FAB) m/z (rel. intensity) 291 (M+H, 44), 391 (5), 292 (8), 291 (44), 273 (6), 245 (4), 155 (3), 109 (10), 92 (9), 91 (99), 41 (3); HRMS (FAB) m/z calcd for C17H22O4+H+ 291.1596, found 291.1603; Anal. Calcd for C17H22O4: C, 70.32; H, 7.64; Found: C, 70.21; H, 7.89.
  • Preparation 7-C-1 Scheme 7, 7-C: wherein R7-1=H Scereochemistry=1S-cis
  • (1R-cis)-N-[[3-[(phenylmethoxy)carbonyl]-1,2,2-trimethylcyclopentyl]carbonyl]glycine (1,1-dimethylethyl) ester (7-C-1) (C[0217] 23H33NO5)
  • To (1R,3S)camphoric acid 3-phenylmethyl ester 7-A (0.736 g, 2.53 mm) in dry DMF (5 mL) is added diisopropylethyl amine (3 mL, 17.2 mmol) and HATU (1.05 g, 2.76 mmol). Thirty minutes later, 7-B (R[0218] 7-1=H) hydrochloride (0.855 g, 5.10 mm) is added. After overnight stirring, the mixture is evaporated to dryness (in vacuo/N2 flow) and then mixed with toluene (50 mL) and THF (50 mL) and washed with water (2×50 mL), 1N HCl (50 mL), and water (4×50 mL). The organic layer is then evaporated to dryness, giving 7-C(R7-1=H) as an off-white solid (0.9 g, 90%)
  • [0219] 1H NMR (300 MHz, CDCl3) δ 7.37-7.15 (5H) 6.05 (1H), 5.17-5.08 (2H), 3.99-3.82 (2H), 2.89-2.83 (1H), 2.51-2.40 (1H), 2.29-2.22 (1H), 1.94-1.80 (1H), 1.59-1.51 (1H), 1.47 (9H), 1.31 (3H), 1.21 (3H), 0.78 (3H). MS (ES+) m/z 404.1 (parent).
  • Preparation 7-D Scheme 7, 7-D: wherein R7-1=H Stereochemistry=1S-cis
  • (1R-cis)-N-[[3-carboxy-1,2,2-trimethylcyclopentyl]carbonyl]glycine (1,1-dimethylethyl) ester (7-D-1) (C[0220] 16H27NO5)
  • Ester 7-C (R[0221] 7-1=H, 0.97 g, 2.4 mmol) in THF (12 mL) and ethanol (6 mL) is shaken with 10% Pd/C (0.115 g) in a Parr bottle under an H2 (38 psi) atmosphere. After 8 hours, the bottle is removed from the shaker and filtered through Celite (with 3×30 mL ethanol rinses). The filtrate is evaporated to dryness, giving 7-D (R7-1=H) as a thick, colorless oil (0.6 g, 79% yield).
  • [0222] 1H NMR (300 MHz, CDCl3) δ 6.13 (1H), 4.0-3.84 (2H) 2.86-2.80 (1H), 2.49-2.39 (1H), 2.27-2.17 (1H), 1.94-1.80 (1H), 1.60-1.52 (1H), 1.46 (9H), 1.34 (3H), 1.22 (3H), 0.88 (3H); IR (liq.) 3385, 2976, 2940, 2887, 1732, 1644, 1528, 1478, 1459, 1405, 1394, 1369, 1277, 1227, 1158 cm−1; MS (FAB) m/z (rel. intensity) 314 (M+H, 99), 315 (18), 314 (99), 258 (51), 109 (22), 95 (9), 76 (20), 69 (12), 57 (28), 55 (12), 41 (14). HRMS (FAB) m/z calcd for C16H27NO5+H1 314.1967, found 314.1974; Anal. Calcd for C16H27NO5 C, 61.32; H, 8.68; N, 4.47; Found: C, 61.70; H, 8.86; N, 4.14; Melt Solvate: 3.906 Ethanol.
  • Preparation 7-F-1 Scheme 7, 7-F: wherein R7-1=H, R4=H, R5=CO2CH3 R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl Stereochemistry=(1S-cis)-L
  • (1S-cis)-N-[[3-[[(1,1-dimethylethoxycarbonylmethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester (7-F-1) (C[0223] 33H41Cl2N3O7)
  • (1R-cis)-N-[[3-carboxy-1,2,2-trimethylcyclopentyl]carbonyl]glycine (1,1-dimethylethyl) ester 7-D (R[0224] 7-1=H, 0.356 g, 1.14 mmol) is dissolved in methylene chloride (6 mL), under N2 in a round bottom flask, and is cooled in an ice water bath. To this stirred solution is added N,N-diisopropylethylamine (1 mL, 5.7 mmol), EDC (0.242 g, 1.26 mmol), HOBt (0.181 g, 1.34 mmol), and 4-N,N-dimethylaminopyridine (0.016 g, 0.13 mmol) followed 30 minutes later by 4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester hydrochloride 7-E-1.HCl (7E: R4=H, R5=CO2CH3, R6=4-[(2,6-dichlorobenzoyl)amino]phenyl, Stereochemistry=S) (0.487 g, 1.2 mmol). After two days of stirring (the ice bath is allowed to melt), the reaction mixture is evaporated to dryness and then partitioned between THF (100 mL), diethyl ether (50 mL), and water (50 mL). The organic layer is washed with water (3×50 mL), aqueous HCl (0.5N, cold, 3×30 mL), aqueous sodium bicarbonate (1×50 mL), water (3×30 mL, to pH 7), and then evaporated to dryness, giving 7-F (R7-1=H, R4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl,
  • Stereochemistry=(1S-cis)-L) as a white solid (0.63 g, 80% yield). [0225]
  • [0226] 1H NMR (300 MHz, CDCl3) δ 7.57 (2H), 7.38-7.26 (3H) 7.10 (2H), 6.07 (1H), 5.80 (1H), 4.89 (1H), 3.97-3.74 (2H), 3.21-3.05 (2H), 2.58-2.52 (1H), 2.45-2.35 (1H), 2.30-2.18 (1H), 1.90-1.75 (1H), 1.58-1.50 (1H), 1.46 (9H), 1.30 (3H), 1.20 (3H), 0.79 (3H); IR (nujol) 1739, 1668, 1643, 1609, 1560, 1538, 1516, 1431, 1414, 1327, 1288, 1256, 1235, 1195, 1161 cm−1; MS (FAB) m/z (rel. intensity) 662 (M+H, 84), 664 (58), 663 (39), 662 (84), 533 (39), 531 (55), 240 (58), 194 (37), 173 (41), 109 (99), 57 (44); HRMS (FA3) m/z calcd for C33H41Cl2N3O7+H+ 662.2399, found 662.2410.
  • Preparation 7-G-1 Scheme 7, 7-G: wherein R7-1=H, R4=H, R5=CO2CH3 R6=4-[(2,6-Dichlorobenzoyl)amino]-phenyl Stereochemistry=(1S-cis)-L
  • (1S-cis)-N-[[3-[[(Carboxymethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester (7-G-1) (C[0227] 29H33Cl2N3O7)
  • (1S-cis)-N-[[3-[[(1,1-dimethylethyloxycarbonylmethyl)damino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester 7-F (R[0228] 7-1=H, R4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorobenzoyl)amino]-phenyl, Stereochemistry=(1S-cis)-L, 0.802 g, 1.21 mmol) is stirred overnight in trifluoroacetic acid (3 mL). The solution is then diluted with toluene (5 mL) and evaporated to dryness in vacuo to give an off white solid which was recrystallized from chloroform/diethyl ether to give 7-G (R7-1=H, R4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorobenzoyl),amino]-phenyl, Stereochemistry (1S-cis)-L as a white solid (0.7 g, 90% yield).
  • [0229] 1H-NMR: (300 MHz, DMSO-d6): δ 10.68 (1H), 7.92 (2H), 7.58-7.44 (3H), 7.19 (2H), 4.53-4.45 (1H), 3.75-3.62 (2H), 3.58 (3H), 3.03-2.85 (2H), 2.68-2.62 (1H), 2.40-2.28 (1H), 1.98-1.81 (1H), 1.70-1.50 (1H), 1.30-1.25 (1H), 1.17 (3H), 1.09 (3H).
  • Preparation of Example 181 Scheme 7, 7-G: wherein R7=H R4=H, R5=CO2H, R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl, Stereochemistry=(1S-cis)-L
  • (1S-cis)-N-[[3-[[(Carboxymethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (Example 181) (C[0230] 28H31Cl2N3O7)
  • (1S-cis)-N-[[3-[[(Carboxymethyl)amino]carbonyl]-2,2,3-trimethylcyclopentenyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester. (7-G-1) (0.7 g, 1.15 mmol) is dissolved in methanol (12 mL). To this is added a mixture of LiOH H[0231] 2O (0.243 g, 5.8 mmol), aqueous H2O2 (30%, 2 mL), and H2O (2 mL). After overnight stirring, the reaction mixture is diluted with water (50 mL), and evaporated (room temperature, in vacuo/N2 flow) until the methanol is gone. The aqueous solution is then transferred to a separatory funnel and shaken with diethyl ether (2×20 mL). The aqueous layer is then evaporated, to remove residual diethyl ether, and cooled in an ice water bath. The stirred solution is then brought to pH 3-4 using aqueous HCl (1N). The resultant precipitate is isolated by suction filtration (with water washes) to give Example 181 as a white solid 0.4 g, 58% yield)
  • [0232] 1H-NMR: (300 MHz, DMSO-d6): 612.45 (1H), 10.6 (1H) 7.74 (2H), 7.57-7.44 (3H), 7.20 (2H), 4.48-4.40 (1H), 3.65 (2H), 2.94 (2H), 2.64 (1H), 2.35 (1H), 1.90 (1H), 1.58 (1H), 1.29 (1H), 1.18 (3H), 1.08 (3H), 0.60 (3H); IR (nujol) 3124, 3088, 3078, 1738, 1666, 1628, 1612, 1588, 1563, 1552, 1521, 1429, 1334, 1197, 1170 cm−1; MS (FAB) m/z (rel. intensity) 592 (M+H, 99), 595 (20), 594 (69), 593 (41), 592 (99), 519 (25), 517 (38), 240 (55), 175 (23), 173 (33), 109 (64); HRMS (FAB) m/z calcd for C28H31Cl2N3O7+H+ 592.1617, found 592.1606; Anal. Calcd for C28H31Cl2N3O7: C, 56.76; H, 5.27; N, 7.09; Found: C, 54.92; H, 5.41; N, 6.91; KF Water: 3.05% H2O.
  • Preparation 7F-2 Scheme 7, 7-F: wherein wherein R7-1=H, R4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorophenyl)methoxy]-phenyl Stereochemistry=(1S-cis)-L
  • [1S-cis]-O-[((2,6-Dichlorophenyl)methyl)]-N-[[3-[[(1,1-diemethylethoxy)carbonylmethyl)amino]-carbonyl]-2,2,3-trimethylcyclopentyl)carbonyl]-L-tyrosine methyl ester (7-F-2) (C[0233] 33H42Cl2N2O7) is prepared from 7-D (R7-1=H and O-[(2,6-dichlorophenyl)methyl]-L-tyrosine methyl ester 7E-2 (7-E: R4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorophenyl)methoxy]phenyl, Stereochemistry=S) as taught by Scheme 7.
  • [0234] 1H NMR (CDCl3) δ 7.39-6.94 (7H), 6.09 (1H), 5.78 (1H), 5.25 (2H), 4.87 (1H), 3.90 (2H), 3.74 (3H), 3.09 (2H), 2.59-2.20 (3H), 1.80 (1H), 1.56 (1H), 1.47 (9H), 1.30 (3H), 1.26 (3H), 0.81 (3H); IR (mull) 3327, 1762, 1741, 1664, 1637, 1538, 1512, 1440, 1241, 1229, 1206, 1198, 1174, 1156, 1022 cm−1; MS (FAB) m/z (rel. intensity) 649 (M+H, 50), 651 (34), 649 (50), 518 (21), 296 (23), 240 (44), 194 (28), 161 (26), 159 (41), 109 (99), 57 (37)
  • Preparation 7-G-2 Scheme 7, 7-G: wherein R7-1=H, R4=H, R1=CO2CH3, R6=4-[(2,6-Dichlorophenyl)methoxy]phenyl Stereochemistry=(1S-cis)-L
  • [1S-cis]-O-[((2,6-Dichlorophenyl)methyl)]-N-[[3-[[(Carboxymethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-L-tyrosine methyl ester (7-G-2) (C[0235] 29H34Cl2N2O7) is prepared from 7-P-2 as taught by Scheme 7.
  • [0236] 1H NMR (CDCl3) δ 8.14 (1H), 7.37-6.93 (7H), 6.51 (1H), 6.02 (1H), 5.24 (2H), 4.85 (1H), 4.02 (2H), 3.73 (3H), 3.09 (2H), 2.57 (1H), 2.41 (1H), 2.25 (1H), 1.84 (1H), 1.56 (1H), 1.26 (3H), 1.20 (3H), 0.79 (3H); MS (ES+) m/z 592.9.
  • Preparation of Example 185 Scheme 7, 7-G: wherein R7-1=H, R4=H, R5=CO2H, R6=4-[(2,6-Dichlorophenyl)methoxy]phenyl, Stereochemistry=(1S-cis)-L
  • (1S-cis)-N-[[3-[(Carboxymethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine (Example 185) (C[0237] 28H32Cl2N2O7) is prepared from 7-G-2 (R7-1=H, R4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorophenyl)methoxy]-phenyl, Stereochemistry=S) as taught by Scheme 7.
  • [0238] 1H NMR (300 MHz, DMSO-d6) 7.8-6.9 (9H), 5.18 (2H), 4.42 (1H), 3.8-3.6 (2H), 3.02-2.82 (2H), 2.65 (1H), 2.38 (1H), 1.91 (1H), 1.58 (11H), 1.30 (11H), 1.19 (3H), 1.10 (3H), 0.61 (3H); IR (nujol) 3409, 1733, 1645, 1612, 1585, 1564, 1511, 1439, 1297, 1239, 1197, 1179, 1018, 786, 770 cm−1; MS (FAB) m/z (rel. intensity) 579 (M+H, 99), 582 (22), 581 (67), 580 (44), 579 (99), 578 (21), 240 (34), 161 (21), 159 (34), 109 (46), 91 (37); HRMS (FAB) m/z calcd for C28H32Cl2N2O7+H+ 579.1664, found 579.1667.
  • Preparation 7-C-2 Scheme 7, 7-C: wherein R7-1=CH3 Stereochemistry=[1S-[1α,3α(R*)]]
  • [1S-[1α,3α(R*)]]-3-[[[1-(1,1-Dimethylethoxycarbonyl)ethyl]amino]carbonyl]-2,2,3-trimethylcyclopentanecarboxylic acid [phenyl(methyl)] ester (7-C-2) (C[0239] 24H35NO5) is prepared from 7-A and 7-B. (R7-1=CH3, Stereochemistry=s) as taught by Scheme 7.
  • [0240] 1H NMR (300 MHz, CDCl3) δ 7.37-7.16 (5H), 6.18 (1H), 5.12 (2H), 4.41 (1H), 2.84 (1H), 2.46 (1H), 2.28 (1H), 1.87 (1H), 1.52 (1H), 1.51 (9H), 1.35 (3H), 1.28 (3H), 1.20 (3H), 0.78 (3H). MS (ES+) m/z 455.1
  • Preparation 7-D-2 Scheme 7, 7-D: wherein wherein R7-1=CH3 Stereochemistry=[1S-[1α,3α(R*)]]
  • [1S-[1α,3α(R*)]]-3-[[[1-(1,1-Dimethylethoxycarbonyl)ethyl]amino]carbonyl]-2,2,3-trimethylcyclopentanecarboxylic acid (7-D-2) (C[0241] 17H29NO5) is prepared from 7-C-2 as taught by Scheme 7.
  • [0242] 1H NMR (300 MHz, CDCl3) δ 6.28 (1H), 4.44 (1H), 2.86 (1H) 2.43 (1H), 2.23 (1H), 1.93 (1H), 1.56 (1H), 1.47 (9H), 1.36 (3H), 1.31 (3H), 1.22 (3H), 0.90 (3H); MS (ES−) m/z 326.1.
  • Preparation 7-F-3 Scheme 7, 7-F: wherein R7-1=CH3, R4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorophenyl)methoxy]phenyl Stereochemistry=[1S-[1α,3α(R*)]]
  • [1S-[1α,3α(R*)]]-O-[(2,6-Dichlorophenyl)methyl]-N-[[3-[[[1-(1,1-dimethylethoxycarbonyl)ethyl]amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-L-tyrosine methyl ester (7-F-3) (C[0243] 34H44Cl2N2O7) is prepared from 7-D-2 and 7-E-2 (7E: R4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorophenyl)methoxy]phenyl) as taught by Scheme 7.
  • [0244] 1H NMR (300 MHz, CDCl3) δ 7.38-6.93 (7H), 6.20 (1H), 5.77 (1H), 5.25 (2H), 4.87 (1H), 4.42 (1H), 3.73 (3H), 3.09 (2H), 2.44 (2H), 1.76 (2H), 1.52 (1H), 1.46 (9H), 1.35 (3H), 1.27 (3H), 1.20 (3H), 0.80 (3H). IR (nujol) 1739, 1654, 1612, 1585, 1565, 1511, 1439, 1344, 1300, 1240, 1198, 1177, 1154, 1017, 768 cm−1. MS (FAB) m/z (rel. intensity) 663 (M+H, 82), 665 (60), 664 (37), 663 (82), 518 (27), 254 (51), 208 (24), 161 (28), 159 (45), 109 (99), 57 (33).
  • Preparation 7-G-3 Scheme 7, 7-G: wherein R7-1=CH3, R4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorophenyl)methoxy]phenyl Stereochemistry=[-[1α,3α(R*)-L
  • [1S-[1α,3α(R*)]]-O-[(2,6-Dichlorophenyl)methyl]-N-[[3-[[(1-Carboxyethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-L-tyrosine methyl ester (7-G-3) (C[0245] 30H36Cl2N2O7) is prepared from 7F-3 as taught by Scheme 7.
  • [0246] 1H NMR (300 MHz, CDCl3) δ 10.55 (1H), 7.38-6.94 (7H) 6.47 (1H), 6.12 (1H), 5.25 (2H), 4.87 (2H), 4.55 (1H), 3.75 (3H), 3.10 (2H), 2.60 (1H), 2.41 (1H), 2.25 (1H), 1.87 (1H), 1.58 (1H), 1.46 (3H), 1.24 (3H), 1.21 (3H), 0.78 (3H); MS (ES+) m/z 606.8.
  • Preparation of Example 188 Scheme 7, 7-G: wherein R7-1=CH3, R4=H, R5=CO2H, R6=4-[(2,6-Dichlorophenyl)methoxy]phenyl, Stereochemistry=[1S-[1α,3α(R*)]
  • [1S-[1α,3α(R)]]-N-[[3-[(1-Carboxyethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-O-[(2,6-dichlorophenyl) methyl]L-tyrosine (C[0247] 29H34Cl2N2O7) is prepared from 7-G-3 as taught by Scheme 7.
  • [0248] 1H NMR (300 MHz, DMSO-d6 7.71 (1H), 7.54-7.43 (3H), 7.28 (1H), 7.16 (2H), 6.93 (2H), 5.16 (2H), 4.40 (1H), 4.16 (1H), 3.02-2.80 (2H), 2.63 (1H), 2.35 (1H), 1.86 (1H), 1.54 (1H), 1.35-1.23 (4H), 1,14 (3H), 1.08 (3H), 0.59 (3H); IR (nujol) 3427, 3031, 1731, 1645, 1612, 1585, 1565, 1512, 1439, 1297, 1239, 1230, 1197, 1179, 1017 cm−1; MS (FAB) m/z (rel. intensity) 593 (M+H, 99), 596 (22), 595 (69), 594 (43), 593 (99), 592 (17), 504 (22), 254 (63), 161 (44), 159 (40), 109 (72).
    Figure US20030130349A1-20030710-C00071
  • Preparation 8-C-1 Scheme 8, 8-C: wherein: R8-1=H, R8-2H Stereochemistry=1S-cis (1S-cis)-N-[1-[(phenylmethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]glycine amide (8-C-1) (C19H26N2O4)
  • A solution of (1R-cis)-[3-(phenylmethoxy)carbonyl]-1,2,2-trimethylcyclopentanecarboxylic acid (7-A) (1 g, 3.44 mmol) in dry DMF is cooled in an ice water bath and diisopropylethylamine (1.77 g, 13.76 mmol, 2.39 mL), HATU (1.35 g, 3.55 mmol), and 8-B (R[0249] 8-1=H, R8-2=H) (0.38 g, 3.44 mmol) are added in order. The mixture is allowed to stir for 48 hours as the ice melts and the solution warms to room temperature The solution is cast into methylene chloride (0.25L) and 1N aq. NaOH (0.25L). The organic phase is separated and washed in order with 1N aq. HCl (0.25L), water (5×0.25L), and brine (0.25L). The organic phase is dried and concentrated in vacuo to give the crude amide as an ivory powder. The crude amide is recrystallized from hexanes-chloroform to furnish the target amide 8-C (R8-1=H, R8-2=H) as a fine, free flowing, white powder.
  • MP: 163-164° C.; [0250] 1H-NMR (300 MHz, CDCl3): δ 7.25-7.40 (5H) 6.36 (1H), 6.08 (1H), 5.33 (3H), 5.15 (1H), 5.10 (1H), 3.93 (2H), 2.85 (1H), 2.43 (1H), 2.27 (1H), 1.40-1.65 (2H), 1.30 (3H), 1.21 (3H), 0.76 (3H); IR (nujol) 3383, 3361, 3184, 2924, 1766, 1710, 1684, 1626, 1527, 1402, 1253, 1166, 753 cm−1; MS (EI) m/z (rel intensity) 346 (M+, 4), 329 (2), 273 (3), 255 (3), 239 (3), 211 (6), 153 (11), 109 (17), 91 (base); Anal. calcd for C19H26N2O4: C, 65.88; H, 7.56; N, 8.09; Found: C, 65.94, H, 7.65; N, 8.09.
  • Preparation 8-D-1 Scheme 8, 8-D: wherein: R8-1=H, R8-2=H Stereochemistry=1S-cis
  • (1R-cis)-N-[[3-carboxy-1,2,2-trimethylcyclopentyl]-carbonyl]glycine amide (8-D-1) (C[0251] 12H20N2O4).
  • A solution of 8-C (R[0252] 8-1=H, R8-2=H) (3.46 g, 10 mmol) in THF (225 mL), containing 10% Pd/C (1.14 g), is hydrogenated under 50 psi of hydrogen for 12 hours. The catalyst is removed by filtration through a cake of Celite, the filter cake is rinsed with THF (100 mL), and the combined filtrates are concentrated in vacuo to give the crude product as a white foam. The crude material is recrystallized from hexanes-THF to afford the target compound 8-D (R8-1=H, R8-2=H) (2.4 g, 94%) as fine white needles.
  • MP: 86-87° C.; [0253] 1H-NMR (300 MHz, DMSO-d6): δ 12.00 (1H) 7.28 (1H), 7.13 (1H), 6.92 (1H), 3.56 (2H), 2.67 (1H), 2.34 (1H), 1.97 (1H) 1.72 (1H), 1.36 (1H), 1.19 (3H), 1.09 (3H), 0.68 (3H); IR (nujol) 3496, 3391, 3189, 2924, 1729, 1705, 1686, 1623, 1519, 1401, 1280, 1245, 1200, 665 cm−1; MS (EI) m/z (rel intensity) 238 (2), 221 (6), 195 (base) 138 (26), 109 (81), 95 (67); Anal. calcd for C12H2N2O4: C, 56.24; H, 7.86; N, 10.93; Found: C, 55.90; H, 8.05; N, 10.50.
  • Preparation 8-F-1 Scheme 8, 8-F: wherein R8-1=H, R8-2=H, R4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl, Stereochemistry=(1S-cis)-L
  • (1S-cis)-N-[[3-[[(2-Amino-2-oxoethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester (C[0254] 29H34Cl2N4O6)
  • To 8-D (R[0255] 8-1=H, R8-2=H)) (1.03 g, 4 mmol) in dry DMF (25 mL), cooled in a an ice-water bath, is added in order, diisopropylethylamine (2.07 g, 16 mmol, 2.8 mL), 4-(2,6-dichlorobenzamido)-L-phenylalanine methyl ester hydrochloride 7-E-1.HCl (7-E: R4=H, R5=CO2CH3, R6=4-[(2,6-dichlorobenzoyl)amino]phenyl) (1.48 g, 4 mmol), and HATU (1.49 g, 4.2 mmol). The mixture is stirred for 48 hours as the ice melts and the mixture warms to room temperature. The solution is cast into ethyl acetate (1L) and this solution is washed successively with 1N aq. HCl (1L), 1N aq. NaOH (1L), water (4×1L), and brine (1L). The organic phase is seperated, dried (Na2SO4), and concentrated in vacuo to give the desired product 8-F (R8-1=H, R8-2=H, R4=H, R5=C2CH3, R6=4-[(2,6-dichlorobenzoyl)amino]phenyl) (1.12 g, 42%) as a fine, white powder.
  • MP: 232-233° C.; [0256] 1H-NMR (300 MHz, DMSO-d6): δ 10.75 (1H), 7.94 (1H), 7.45-7.70 (5H), 7.35 (1H), 7.19 (2H), 7.13 (1H), 6.92 (1H), 4.81 (1H), 3.57 (3H), 3.55 (2H), 2.92 (2H), 2.64 (1H), 2.34 (1H), 1.88 (1H), 1.62 (1H), 1.30 (1H), 1.16 (3H), 1.07 (3H), 0.58 (3H); IR (nujol): 3344, 3251, 3194, 3126, 3072, 2924, 1743, 1699, 1669, 1652, 1623, 1528, 1432, 1328, 799 cm−1; MS (FAB): m/z (rel. intensity) 605 (M+2H), base), 531 (28), 503 (2), 367 (7), 349 (17), 256 (10), 239 (66), 194 (31), 173 (37), 137 (12), 109 (83); Anal. calcd for C29H34Cl2N4O6.0.3H2O: C, 57.02; H, 5.71; N, 9.17; Found: C, 57.01; H, 5.86; N, 8.89.
  • Preparation of Example 180 Scheme 8, 8-G: wherein R8-1=H, R8-2=H, R4=H, R5=CO2H, R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl, Stereochemistry=(1S-cis)-L
  • (1S-cis)-N-[[3-[[(2-Amino-2-oxoethyl)amino]-carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (Example 180) (C[0257] 28H32Cl2N4O6)
  • To 8-F (R[0258] 8-1=H, R8-2=H, R4=H, R5=CO2CH3, R6 4-[(2,6-dichlorobenzoyl)amino]-phenyl) (1.05 g, 1.7 mmol), dissolved in methanol (30 mL), is added a solution of LiOH2H2O (0.32 g, 7.65 mmol) in water (10 mL), dropwise over 15 minutes. The mixture stirs for 18 hours at room temperature and the pH is then adjusted to ca. 7 by the careful addition of 1N aq. HCl. The majority of the methanol, is removed in vacuo and the pH of the resulting solution is adjusted to ca. 2 with 1N aq. HCl. The resulting flocculent white precipitate is isolated by filtration and dried. The solid is crushed and washed with water (2×10 mL) and dried in vacuo at 50° C. to give 0.97 g (97%) of 8-G (R8-1=H, R8-2, R4=H, R5=CO2H, R6=4-[(2,6-dichlorobenzoyl)amino]-phenyl) as a white, powdery solid.
  • MP: 203-205° C.; [0259] 1H NMR (300 MHz, DMSO-d6): 12.51 (1H) 10.70 (1H), 7.75 (1H), 7.45-7.57 (3H), 7.33 (1H), 7.20 (2H), 7.11 (1H), 6.92 (1H), 4.43 (1H), 3.63 (1H), 3.47 (2H), 3.30 (2H), 3.01 (1H), 2.84 (1H), 2.31 (1H), 1.87 (1H), 1.55 (1H), 1.31 (1H), 1.17 (3H), 1.08 (3H), 0.59 (3H); IR (nujoll): 3511, 3325, 3128, 3082, 2868, 1722, 1697, 1664, 1614, 1555, 1537, 1417, 1337, 1246, 799 cm−1; MS (FAB) m/z (rel. intensity) 591 (M+H, base), 517 (32), 335 (26), 239 (32), 173 (39), 109 (63), 57 (80); HRMS (FAB) m/z calcd for C28H32Cl2N4O6+H+ 591.1777, found 591.1747.
  • Preparation 8-F-2 Scheme 8, 8-F: wherein R8-1=H, R8-2=H, R4=H, R5=CO2CH31 R6=4-[(2,6-Dichlorophenyl)methoxy]phenyl Stereochemistry=(1S-cis)-L
  • (1S-cis)-N-[[3-[[(2-Amino-2-oxoethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine methyl ester (8-F-2) (C[0260] 29H35Cl2N3O6) is prepared from 8-D (R8-1=H, R8-2=H) and 7-E-2 (R4=H, R5=CO2CH3, R6=4-[(2,6-dichlorophenyl)methoxy]phenyl) as taught by Scheme 8.
  • [0261] 1H NMR (CDCl3) δ 7.37-6.93 (7H), 6.54 (1H), 6.40 (TH), 5.82 (1H), 5.60 (1H), 524 (2H), 4.85 (1H), 3.93 (2H), 3.73 (3H), 3.09 (2H), 2.54 (1H), 2.40 (1H), 2.23 (1H), 1.78 (1H), 1.52 (1H), 1.27 (3H), 1.20 (3H), 0.78 (3H); MS (ES+) m/z 591.9.
  • Preparation of Example 187 Scheme 8, 8-G: wherein R8-1=H, R8-2=H, R4=H, R5=CO2H, R6=4-[(2,6-Dichlorophenyl)methoxy]phenyl Stereochemistry=(1S-cis)-L
  • (1S-cis)-N-[[3-[[(2-amino-2-oxoethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine (Example 187) (C[0262] 28H33Cl2N3O6) is prepared from 8-F (R8-1=H, R8-2=H, R4=H, R5=CO2CH3, R6=4-[(2,6-dichlorophenyl)methoxy]phenyl) as taught by Scheme 8.
  • [0263] 1H NMR (300 MHz, DMSO-d6) 7.74 (2H), 7.55-7.40 (4H) 7.15 (3H), 6.94 (3H), 5.16 (2H), 4.41 (1H), 3.75-3.48 (2H), 3.1-2.8 (2H), 2.63 (1H), 2.33 (1H), 1.87 (1H), 1.54 (1H), 1.32 (1H), 1.17 (3H), 1.08 (3H), 0.58 (3H); MS (FA) m/z (rel. intensity) 578 (M+H, 99), 581 (30), 580 (72), 579 (57), 578 (99), 577 (19), 504 (17) 322 (18), 239 (35), 161 (29), 159 (34); HRMS (FAB) m/z calcd for C28H33Cl2N3O6+H+ 578.1824, found 578.1836.
    Figure US20030130349A1-20030710-C00072
  • Preparation of Example 183 Scheme 9, 9-G: wherein R4=H, R6=4-[(2,6-dichlorobenzoyl)amino]phenyl Stereochemistry=(1S-cis)-L
  • (1S-cis)-N-[[3-(carboxymethoxymethyl)-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (Example 183) 7-A (1.15 g, 3.96 mmol) was dissolved in dry THF (5 mL). The reaction flask was immersed in a −15° C. bath (ethylene glycol/CO[0264] 2), then diborane-tetrahydrofuran (1M, 1 equiv, 3.96 mmol, 3.96 mL) was added slowly. The reaction solution was stirred at −15° C. under N2 during the day, and was equilibrated to room temperature overnight The reaction solution was treated with potassium carbonate (1.2 g) in H2O (25 mL). The THF layer was separated and the aqueous phase was extracted with EtOAc (3×20 mL). The organic layers were combined, washed with saturated NaCl (20 mL) and dried (MgSO4). Solvent was removed in vacuo from the mixture to yield 0.83 g (76%) of 9-B. The crude product was chromatographed, initially using CH2Cl2, as eluent followed by CH2Cl2\MeOH (56) Recovered desired product 9-B (0.76 g, 69%) from column: 1H NMR (CDCl3) δ 0.80 (3H), 1.00 (3H), 1.11 (3H), 1.32-1.47 (1H), 1.62-1.91 (2H), 2.09-2.22 (1H), 2.86 (1H), 3.48-3.59 (2H), 5.08-5.18 (2H), 7.28-7.41 (5H).
  • A solution of benzyl ester 9-B (0.76 g, 2.75 mmol) in CH[0265] 2Cl2 (10 mL) was treated at 0° C. and under N, with ethyl diazoacetate (5 equiv, 13.75 mmol, 1.4 mL) and a catalytic amount of HBF4 (0.1 equiv, 0.03 mL). The reaction solution was a stirred under N2 overnight while the bath temperature equilibrated to room temperature. Solvent was removed under reduced pressure to yield 1.8 g of product. Crude product was subjected to silica gel (35 mm×15.2 cm) flash column chromatography, eluting with 2%-10% EtOAc/hexanes, to provide 0.64 g (64%) of 9-C: 1H NMR (CDCl3) δ 0.81 (3H), 1.05 (3H), 1.12 (3H), 1.22-1.31 (3H), 1.32-1.43 (1H), 1.54-1.62 (2H), 1.65-1.92 (2H), 2.08-2.23 (1H), 2.85 (1H), 3.28 (1H), 3.47 (1H), 3.96-4.08 (2H), 4.14-4.25 (2H), 5.07-5.18 (2H), 7.28-7.40 (5H).
  • Benzyl ester 9-C (0.3 g, 0.83 mmol) was dissolved in absolute EtOH (10 mL). The solution was treated with a catalytic amount of 10% Pd/C (0.2 g) and hydrogenated at 20 psi for 1 h on a Parr hydrogenation apparatus. The suspension was filtered through a Celite cake and washed cake with EtOH. Solvent was removed from the filtrate under reduced pressure to yield 0.19 g (84%) of 9-D: [0266] 1H NMR (CDCl3) δ 0.90 (3H) 1.06 (3H), 1.16 (3H), 1.19-1.32 (3H), 1.33-1.45 (1H), 1.65-1.92 (2H), 2.03-2.18 (1H) 2.83 (1H), 3.29 (1H). 3.49 (1H), 3.95-4.11 (2H), 4.19 (2H).
  • Dissolved acid 9-D (0.18 g, 0.66 mmol), HOBT (1.2 equiv, 0.81 mmol, 0.11 g), DMAP (0.11 equiv, 0.074 mmol, 0.009 g), EDCl (1.2 equiv, 0.78 mmol, 0.15 g) and Et[0267] 3N (3.8 equiv, 2.5 mmol, 0.35 mL) in CH2Cl2 (10 mL) at 0° C. The reaction mixture was stirred for several minutes and-then 7-E-1 (1.0 equiv, 0.69 mmol, 0.28 g) was added. The mixture was stirred overnight while the bath temperature equilibrated to room temperature. The reaction mixture was concentrated to dryness under reduced pressure. The residue was quenched with acidic H2O (30 mL) and extracted with CHCl3 (3×15 mL). The organic layers were combined, washed with saturated NaHCO3 (30 mL), dried (MgSO4) and solvent was removed under reduced pressure. Crude product was purified by silica gel (20 mm×22.9 cm) flash chromatography using 90% CH2Cl2/EtOAc as eluent.
  • Fractions containing the compound were combined and solvent was removed under reduced pressure. The product was dissolved in CH[0268] 3CN:H2O, frozen and lyophilized to yield 0.18 g (44%) of 9-F (where R4=H, R5=CO2CH3 and R6=C6H4OCH2C6H3Cl2): 1H NMR (CDCl3) δ 0.82 (3H), 1.05 (3H), 1.12 (3H), 1.27 (2H), 1.31-1.43 (1H), 1.59-1.84 (2H), 2.05-2.21 (1H), 2.55 (1H) 3.03-3.23 (2H), 3.30 (1H), 3.47 (1H), 3.74 (3H), 3.93-4.09 (2H), 4.19 (2H), 4.90 (2H), 5.76 (1H), 7.11 (2H), 7.24-7.39 (3H), 7.47 (1H), 7.57 (2H); IR (mineral oil mull) 3292, 3193, 3123, 3064, 3033, 2951, 2922, 2870, 2855, 2854, 1748, 1657, 1656, 1606, 1579, 1562, 1537, 1515, 1461, 1431, 1414, 1376, 1348, 1324, 1271, 1207, 1196, 1153, 1129, 1023, 800, 781 cm−1; MS (FAB) for C31H38Cl2N2O7, m/z (relative intensity) 623 ([M+H]+, 49), 622 ([M+H]+, 29), 621 ([M+H]+, 71), 620 (M+, 71, 517 (12), 351 (16), 349 (22), 175 (16), 173 (26), 151 (27), 123 (100). Anal. Calcd for C31H38Cl2N2O7: C, 59.91; H, 6.16; N, 4.51; Cl, 11.41; Found: C, 59.67; H, 6.09; N, 4.63; Cl, 11.50. Corrected for 0.40% H2O found by Karl Fischer analysis.
  • To a solution of methyl ester 9-F (R[0269] 4=H, R5=CO2CH3, R6=C6H4OCH2C6H3Cl2) (0.11 g, 0.18 mmol) in MeOH (5 mL) was added LiOH2O (5 equiv, 0.88 mmol, 0.04 g) in H2O (1 mL). The reaction solution was allowed to stir for about 2 hrs. The solvent was removed in vacuo. The residue was dissolved in H2O (30 mL) and acidified with HCl. The resulting precipitate was filtered and washed with H2O. The precipitate was dissolved in 50S CH3CN:H2O (10 mL), was frozen and lyophilized to yield 0.083 g (80%) of Example 183 (9-G: where R4=H and R6=C6H4OCH2C6H3Cl2): 1H NMR (MeOD) δ 0.79 (3H), 1.00 (3H), 1.05 (3H), 1.23-1.41 (1H), 1.60-1.78 (2H), 1.91-2.05 (1H), 2.72 (1H), 2.98 (1H), 3.20 (1H), 3.46 (1H), 3.93-4.08 (2H), 4.66-4.78 (1H), 7.24 (2H), 7.38-7.51 (3H), 7.57 (2H), 7.73 (1H); IR (mineral oil mull) 3270, 3193, 3123, 3056, 3034, 2954, 2928, 2854, 1731, 1657, 1607, 1562, 1537, 1516, 1461, 1459, 1432, 1414, 1376, 1326, 1272, 1220, 1195, 1126, 800, 781 cm−1; MS (FAB) for C28H32Cl2N2O7, m/z (relative intensity) 581 (([M+H]+, 51), 580 ([M+H]+, 29), 579 ([M+H]+, 76), 578 (M+, 8), 337 (12), 335 (18), 175 (19), 173 (29), 161 (18), 151 (18), 123 (100). Anal. Calcd for C28H32Cl2N2O7: C, 58.04; H, 5.57; N, 4.84; Cl, 12.24; Found: C, 57.87; H, 5.44; N, 4.97; Cl, 12.29. Corrected for 2.17% H2O found by Karl Fischer analysis.
    Figure US20030130349A1-20030710-C00073
  • Preparation of Example 192:
  • Scheme 10, 10-G: wherein R[0270] 4=H, R5=CO2H, R6=4-[(2,6-dichlorophenyl)methoxy]phenyl Stereochemistry=(1S-cis)-L
  • (1S-cis)-O-[(2,6-Dichlorophenyl)methyl]-N-[(3-hydroxy-2,2,3-trimethylcyclopentyl)carbonyl]-L-tyrosine (Example 192). [0271]
  • Into a 100 mL oven dried round bottom flask was placed (1S-cis)-[3-carboxy-2,2,3-trimethyl]cyclopentane carboxylic acid methyl ester (10-A) (5.0 g, 23 mmol, 1 equiv), followed by dry THF (20 mL). The reaction flask was immersed in a −15° C. bath (ethylene glycol/dry ice) and then boron trifluoride-tetrahydrofuran (1 equiv, 1 M, 23 mmol, 23 mL) was slowly added. [Note: observed mild evolution of H[0272] 2 gas]. The reaction solution was allowed to stir for 24 hrs under N2. The solution was stirred in the −15° C. bath during the day, and the bath was permitted to reach room temperature overnight. Silica gel Thin-layer chromatography, using 1:1 hexanes/ethyl acetate as eluent, showed that the starting material was consumed. Consequently, the reaction solution was quenched with H2O (100 mL) and then treated with potassium carbonate (6 g). The THF phase was separated while the aqueous phase was extracted with EtOAc (3×80 mL). The organic phases were combined, washed with saturated NaCl (100 mL) and dried (MgSO4). The solvent was removed via rotary evaporator and then by overnight hi vacuum conditions to yield 4.7 g of crude product. Crude product was subjected to a silica gel (35 mm×25.4 cm) flash chromatography column. Elution with 50 EtOAc/hexanes yielded 4.0 g (87%) of 10-B as-a clear oil: 1H NMR (CDCl3) δ 0.79 (3H), 0.99 (3H), 1.10 (3H), 1.32-1.43 (1H), 1.48 (H), 1.63-1.88 (2H), 2.04-2.15 (1H), 2.8 (1H), 3.52 (2H), 3.66 (3H); IR (mineral oil mull) 3445, 2968, 2878, 1734, 1719, 1456, 1436, 1372, 1358, 1270, 1218, 1203, 1173, 1031, 1006 cm−1; MS (FAB) for C11H20O3, m/z (relative intensity) 202 ([M+2H]+, 11), 201 ([M+H]+, 100), 200 (M+, 0.9), 183 (11), 169 (8), 151 (8), 123 (29). Anal. for C11H20O3: C, 65.97; H, 10.07; Found: C, 65.94; H, 9.91. Corrected for 0.85% H2O found by Karl Fischer analysis.
  • (1S-cis)-[3-hydroxymethyl-2,2,3-trimethyl]-cyclopentane carboxylic acid methyl ester 10B (1.5 g, 7.5 mmol) was dissolved in CH[0273] 2Cl2 (35 mL). Cooled flask to 0° C., slowly added chloromethyl methyl ether (3.3 equiv, 25 mmol, 1.9 mL) and then added DIEA (5.87 mL, 4.5 equiv, 3.4 mmol). The bath temperature was allowed to equilibrate to room temperature while the reaction mixture was stirred for four days. Solvent was removed via a rotary evaporator. Crude product was dissolved in toluene, the insoluble precipitate was filtered and the concentrated filtrate was chromatographed on silica gel (35 mm×5.2 cm) flash chromatography using 2% EtOAc/hexanes as eluent to yield 1.37 g (75%) of 10-C (where R10-1=methoxymethyl): 1H NMR (CDCl3) δ 0.80 (3H), 1.02 3H), 1.12 (3H), 1.34-1.45 (1H), 1.57 (H), 1.67-1.87 (2H), 2.07-2.18 (1H), 2.82 (1H), 3.31 (1H), 3.36 (3H), 3.45 (1H), 3.67 (3H), 4.56-4.61 (2H).
  • Compound 10-C (1.57 g, 6.4 mmol) was dissolved in THF (20 mL) and treated with LiOH.H[0274] 2O (10 equiv, 64 mmol, 2.7 g) in H2O (30 mL), H2O2 (6 mL), H2O (16 mL) and MeOH (16 mL). The mixture was refluxed overnight. The solvent was removed in vacuo, crude residue was quenched with H2O (35 mL) and the pH was lowered to 5 with 10%, 6N or 12N hydrochloric acid. Extracted aqueous portion with EtOAc (3×20 mL) and then with CHCl3 (3×20 mL). Organic layers were combined, dried (MgSO4) and solvent removed on rotary evaporator.
  • Crude product was subjected to a silica gel (35 mm×15.2 cm) flash chromatography column using 80% hexanes/EtOAc as eluent to yield 1.1 g (75%) of 10-D (where R[0275] 10-1=methoxy methyl): 1H NMR (CDCl3) δ 0.89 (3H), 1.03 (3H), 1.16 (3H), 1.36-1.46, 1.68-1.89, 2.04-2.18 (1H), 2.85 (1H), 3.32 (1H), 3.36 (3H), 3.47 (1H), 4.57-4.62 (2H).
  • Compound 10-D (0.83 g, 3.6 mmol), HOBT (1.13 equiv, 4.1 mmol, 0.55 g), DMAP (0.11 equiv, 0.4 mmol, 0.048 g), EDCl (1.1 equiv, 4.0 mmol, 0.76 g) and Et[0276] 3N (3.6 equiv, 13 mmol, 1.8 mL) were mixed in CH2Cl2 (30 mL) at 0° C. The reaction mixture was stirred for several minutes, and then added O-[(2,6-dichlorophenyl)methyl]-L-tyrosine methyl ester hydrochloride 7-E-2.HCl (wherein R4=H, R5=CO2CH3, R6=4-[(2,6-dichlorophenyl)methoxy]phenyl) (1 equiv, 3.6 mmol, 1.4 g). The mixture was stirred overnight while bath temperature equilibrated to room temperature. Reaction mixture was concentrated to dryness under reduced pressure. The residue was quenched with acidic H2O (70 mL), and extracted with CHCl3 (3×35 mL). Organic layers were combined, washed with saturated NaHCO3 (40 mL), dried (MgSO4) and solvent was removed under reduced pressure. Crude product was subjected to a silica gel (35 mm×15.2 cm) flash chromatography column using 10% EtOAc/hexanes as eluent to yield 1.6 g (78%) of 10-F-1 (10-F (where R10-1=methoxymethyl, R4=H, R5=CO2CH3 and R6=4-[(2,6-dichlorophenyl)methoxy]phenyl-): 1H NMR (CDCl3, 400 MHz) δ 0.82 (3H), 1.03 (3H), 1.11 (3H), 1.33-1.42 (1H), 1.61 (2H), 1.63-1.82 (2H), 2.08-2.21 (1H), 2.56 (1H), 3.03-3.16 (2H), 3.31 (1H), 3.36 (3H), 3.46 (1H), 3.75 (3H), 4.57-4.61 (2H), 4.85-4.93 (1H), 5.26 (2H), 5.72 (1H), 6.96 (2H), 8.6 (2H), 7.22-7.28 (1H), 7.37 (2H); IR (mineral oil mull) 2951, 2879, 1746, 1668, 1657, 1565, 1511, 1468, 1439, 1382, 1371, 1241, 1216, 1197, 1179, 1148, 1108, 1096, 1047, 1019, 780, 768 cm−1; MS (FAB) for C29H37Cl2N1O6, m/z (relative intensity) 568 (M+H]+, 69), 566 ([M+H]I+, 100), 565 (M+, 12), 506 (41), 504 (61), 336 (41), 161 (40), 159 (62), 123 (83), 45 (53). Anal. Calcd for C29H37Cl2N1O6: C, 61.48; H, 6.58; N. 2.47; Cl, 12.52; Found: C, 61.30; H, 6.55; N. 2.80; Cl, 12.57. Corrected for 0.11% H2O found by Karl Fischer analysis.
  • Compound 10-F (0.74 g, 1.3 mmol) was dissolved in MeOH (30 mL) and treated with concentrated HCl (5 mL) and stirred at room temperature for 24 hrs. The solvent was removed in vacuo to yield a residue that was taken up in CHCl[0277] 3 and washed with saturated NaHCO3. [Note: upon treatment with saturated NaHCO3 a precipitate formed which was filtered and washed with CHCl3.] Filtrate volume was reduced in vacuo and subjected to a silica gel (35 mm×16.5 cm) flash chromatography column using 50% hexanes/EtOAc as eluent. Fractions containing pure compound were combined and solvent was removed under reduced pressure. The residue was dissolved in 50% CH3CN:H2O (10 mL), frozen and lyophilized to yield 0.23 g (34%) of 10-G (where R4=H, R5=CO2CH3 and R6=4-[(2,6-dichlorophenyl)methoxy]phenyl-: 1H NMR (CDCl3, 400 MHz) δ 0.84 (3H), 1.00 (3H), 1.10 (3H), 1.37-1.48 (1H), 1.69-1.82 (2H), 2.09-2.21 (1H), 2.55 (1H), 3.02-3.18 (2H), 3.51-3.59 (2H), 3.75 (3H), 4.84-4.91 (1H), 5.26 (2H), 5.75 (1H), 6.96 (2H), 7.05 (2H), 7.22-7.29 (1H), 7.37 (2H); IR (mineral oil mull) 3428, 3322, 2923, 2870, 2854, 1743, 1654, 1565, 1511, 1466, 1439, 1377, 1298, 1278, 1240, 1197, 1179, 1018, 1003, 780, 768 cm−1; MS (FAB) for C27H33Cl2N1O5, m/z (relative intensity) 524 ([M+H]+, 65), 523 ([M+H]+, 41), 522 ([M+H]-F, 100), 521 (M+, 19), 354 (21), 338 (21), 336 (29), 161 (21), 159 (34), 123 (38). Anal. Calcd for C27H33Cl2N1O5: C, 62.07; H, 6.37; N, 2.68; Cl, 13.57; Found: C, 61.85; H, 6.27; N, 2.85; Cl, 13.50. Corrected for 0.44% H2O found by Karl Fischer analysis.
  • Compound 10-G (0.19 g, 0.36 mmol) was dissolved in CH[0278] 3OH (4 mL) and treated with LiOH.H2O (10 equiv, 0.15 g, 3.6 mmol) in H2O (4 mL). Additional MeOH (2 mL) was added to ensure solubility. The solution was stirred for 2 h at room temperature. The solvent was removed under reduced pressure. The residue was dissolved in warm H2O (20 mL) (Note: room temperature H2O caused aqueous solution to gel). The pH of the solution was lowered to 2 with 1.0 N HCl and the resulting precipitate was filtered, washed with H2O and dried to yield 0.17 g (93.) of Example 192 (10-G: where R4=H. R5=CO2H and R6=4-[(2,6-dichlorophenyl)methoxy]phenyl-): unable to determine mp due to compound shrinkage at 80 C; 1H NMR (CDCl3) δ 0.81 (3H), 0.97 (3H), 1.06 (3H), 1.35-1.48 (1H), 1.65-1.88 (2H), 2.03-2.18 (1H), 2.55 (1H), 3.04-3.27 (2H), 3.48-3.58 (2H), 4.81-4.89 (1H), 5.24 (2H), 5.85 (1H), 6.96 (2H), 7.12 (2H), 7.19-7.29 (1H), 7.32-7.38 (2H); IR (mineral oil mull) 3421, 3332, 3058, 3030, 2954, 2920, 2871, 2855, 1729, 1653, 1612, 1585, 1565, 1511, 1466, 1439, 1377, 1299, 1241, 1196, 1179, 1018, 1003, 779, 769 cm4; MS (FAB) for C26H31Cl2N1O5 m/z (relative intensity) 510 ([M+H]+, 66), 509 ([M+H]+, 37), 508 ([M+H+, 100), 507 (M+, 15), 340 (12), 324 (15), 322 (22), 161 (23), 159 (36), 123 (45). Anal. Calcd for C26H31Cl2N1O5: C, 61.42; H, 6.15; N, 2.76; Cl, 13.95; Found: C, 61.33; H, 6.16; N, 2.93; Cl, 13.74. Corrected for 1.61% H2O found by Karl Fischer analysis.
  • Preparation of Example 198: Scheme 10, 10-F=wherein R10-1=methyl, R4=H, R5=CO2H, R6=4-[(2,6-dichlorophenyl)methoxy]phenyl Stereochemistry=(1S-cis)-L
  • (1S-cis)-O-[(2,6-Dichlorophenyl)methyl]-N-[(3-methoxymethyl-2,2,3-trimethylcyclopentyl)carbonyl]-L-tyrosine (Example 198). [0279]
  • Alcohol 10-B (1.0 g, 4.8 mmol), in Et[0280] 2O (20 mL), was treated with boron trifluoride dimethyl etherate (0.1 equiv, 0.48 mmol, 0.06 mL) and an excess amount of CH2N2Et2O. The reaction mixture was stirred overnight. The mixture was filtered and solvent was removed on rotary evaporator. Crude product was subjected to a silica gel (35 mm×16.5 cm) flash chromatography column using 1% EtOAc/pentane as eluent to yield 0.69 g (67%) of 10C (where R10-1=methyl): 1H NMR (CDCl3) δ 0.79 (3H), 1.00 (3H), 1.10 (3H), 1.32-1.43 (1H), 1.60-1.89 (2H) 2.04-2.20 (1H), 2.80 (1H), 3.16 (1H), 3.28 (1H), 3.31 (3H), 3.68 (3H).
  • Ester 10-C (0.37 g, 1.7 mmol) was dissolved in THF (40 mL) and treated with LiOH.H[0281] 2O (10 equiv, 17 mmol, 0.71 g) in H2O (20 mL), MeOH (10 mL) and H2O2 (10 mL). The mixture was heated at reflux for about 8 h. Solvent was removed via a rotary evaporator. Dissolved residue in H2O (50 mL), lowered pH to 5 with hydrochloric acid, extracted aqueous portion with EtOAc (3×25 mL) followed by CHCl3 (3×25 mL). Organic extracts were combined, dried (MgSO4), and solvent was removed under reduced pressure to yield 0.44 g of 10-D (wherein R10-1=Me): 1H NMR (CDCl3) δ 0.86 (3H), 1.00 (3H), 1,14 (3H), 1.34-1.44 (1H), 1.62-1.89 (2H), 2.01-2.15 (1H), 2.83 (1H), 3.16 (1H), 3.29 (1H), 3.30 (3H).
  • Compound 10-D (0.25 g, 1.2 mmol), HOBT (1.13 equiv, 1.41 mmol, 0.19 g), DMAP (0.11 equiv, 0.14 mmol, 0.017 g), EDC (1.1 equiv, 1.37 mmol, 0.25 g) and Et[0282] 3N (3.6 equiv, 4.49 mmol, 0.6 mL) were mixed in CH2Cl2 (10 mL) at 0° C. Stirred the reaction mixture for several minutes then added 7-E-2.HCl (0.8 equiv, 1.0 mmol, 0.4 g). The mixture was stirred overnight while the bath temperature equilibrated to room temperature. Concentrated reaction mixture to dryness under reduced pressure. Residue was treated with acidic H2O (30 mL) and extracted with CHCl3 (3×15 mL) Combined organic layers, washed with saturated NaHCO3 (20 mL), dried (MgSO4) and solvent was removed under reduced pressure. Crude product was purified by silica gel (20 mm×16.5 cm) flash chromatography using 10%-15% EtOAc/hexanes as eluent. Fractions containing the compound were combined and solvent was removed under reduced pressure. The residue was dissolved in 50% 6 CH2CN:H2O (50 mL), frozen and lyophilized to yield 0.24 g (45%) of 10-F (where R10-1=methyl, R4=H, R5=CO2CH3 and R6=4-[(2,6-dichlorophenyl)methoxy]phenyl-): 1H NMR (CDCl3) δ 0.79 (3H), 0.99 (3H), 1.09 (3H), 1.32-1.43 (1H), 1.61 (2H), 1.65-1.85 (2H), 2.08-2.23 (1H), 2.53 (1H), 3.01-3.18 (2H), 3.16 (1H), 3.28 (1H), 3.30 (3H), 3.74 (3H), 4.83-4.91 (1H), 5.25 (2H), 5.74 (1H), 6.96 (2H). 7.05 (2H), 7.23-7.29 (1H), 7.35-7.40 (2H): IR (mineral oil mull) 3317, 2956, 2924, 2871, 2857, 2855, 1745, 1652, 1612, 1565, 1511, 1466, 1439, 1378, 1298, 1278, 1240, 1197, 1178, 1106, 1096, 1017, 1000, 779, 768 cm−1; MS (FAB) for C28H35Cl2N1O5, m/z (relative intensity) 538 (([M+H], 65), 537 ([M+H]+, 40), 536 ([M+H]+, 100), 535 (M+, 16), 338 (16), 336 (24), 161 (18), 159 (28), 123 (69). Anal. Calcd for C28H35Cl2N2O5 nH2O: C, 62.69; H, 6.58; N, 2.61; Cl, 13.22; Found: C, 62.44; H, 6.35; N, 2.88; Cl, 13.11. Corrected for 0.37% H2O found by Karl Fischer analysis.
  • Ester 10-F (0.27 g, 0.5 mmol) was treated with LiOH.H[0283] 2O (10 equiv, 5.0 mmol, 0.21 g) in H2O (8 mL). The reaction mixture was stirred for 1 h. THF (3 mL) was added to complete dissolution of starting material and MeOH (3 mL) was added to convert reaction mixture to homogeneous solution. The volume was reduced under vacuum. H2O (40 mL) was added and the mixture was acidified with HCl. The resulting precipitate was filtered, washed with H2O, and dried under house vacuum to yield 0.26 g (99%) of Example 198 (10-F: where R10-1=methyl; R4=H; R5=CO2H and R6=4[(2,6 dichlorophenyl) methoxy]phenyl-stereochemistry=(1S-cis)-L): 1H NMR (CDCl3) δ 0.77 (3H), 0.98 (3H), 1.05 (3H), 1.31-1.42 (1H), 1.61-1.85 (2H), 2.01-2.16 (1H), 2.53 (1H), 3.07-3.28 (2H), 3.15 (1H), 3.27 (L H), 3.29 (3H), 4.79-4.87 (1H), 5.24 (2H), 5.77 (1H), 6.97 (2H), 7.12 (2H), 7.21-7.29 (1H), 7.32-7.39 (2H); IR (mineral oil mull) 3424, 3335, 3032, 2955, 2924, 2855, 1734, 1644, 1612, 1586, 1565, 1512, 1466, 1439, 1377, 1299, 1241, 1197, 1179, 1107, 1096, 1018, 873, 779, 769 cm−1; MS (FAB) for C27H33Cl2N1O5, m/z (relative intensity) 524 (([M+H]+, 62), 523 ([M+H]+, 36), 522 ([M+H]+, 96), 521 (M+, 13), 324 (14), 322 (22), 161 (31), 159 (42), 123 (100). Anal. Calcd for C27H33C12N1O5nH2O: C, 62.07; H, 6.37; N, 2.68; Cl, 13.57; Found: C, 62.04; H, 6.24; N, 2.90; Cl, 13.91. Corrected for 2.18% H2O found by Karl Fischer analysis.
  • Preparation of Example 203: Scheme 10, 10-F; where R10-1=methoxymethyl; R4=H; R5=CO2H and R6=4-[(2,6-Dichlorophenyl)methoxy]phenyl-, Stereochemistry=(1S-cis)-L
  • (1S-cis)-O [(2,6-Dichlorophenyl)methyl]-N-[(3-methoxymethoxymethyl-2,2,3-trimethylcyclopentyl) carbonyl]-L-tyrosine (Example 203) Compound 10-F-1 (10-F: (where R[0284] 10-1=methoxymethyl, R4=H, R5=CO2CH3 and R6=4 (2,6-dichlorophenyl)methoxy]phenyl-) (0.25 g, 0.44 mmol) was dissolved in MeOH (3 mL). Treated the reaction solution with LiOH.H2O (10 equiv, 4.4 mmol, 0.18 g) in H2O (5 mL). Additional MeOH (2 mL) was added to ensure solubility. [Note: reaction mixture turned clear within 1 hour of reaction time]. Solution was allowed to stir overnight at room temperature under N2. Solvent volume was reduced on a rotary evaporator, the remaining material was diluted with H2O (10 mL) and acidified with hydrochloric acid. The resulting precipitate was filtered, washed with H2O and dried under house vacuum to yield 0.23 g (95%) of Example 203. mp: unable to determine due to compound shrinkage at 60° C.; 1H NMR (CDCl3) δ 0.79 (3H), 1.01 (3H), 1.07 (3H), 1.32-1.46 (1H), 1.62-1.84 (2H), 2.01-2.18 (1H), 2.55 (1H), 3.02-3.24 (2H), 3.30 (1H), 3.35 (3H), 3.45 (1H), 4.55-4.61 (2H), 4.81-4.89 (1H), 5.24 (2H), 5.76 (1H), 6.97 (2H), 7.12 (2H), 7.18-7.27 (1H), 7.31-7.37 (2H); IR (mineral oil mull) 2930, 2872, 2855, 1736, 1638, 1612, 1511, 1466, 1439, 1377, 1241, 1196, 1179, 1147, 1108, 1045, 1020 cm−1; MS (FAB) for C28H35Cl2N1O6nH2O, m/z (relative intensity) 554 (([M+H]+, 67), 552 ([M+H]+, 100), 551 (M+, 16), 492 (38), 490 (56), 322 (33), 161 (40), 159 (64), 123 (73), 45 (61). Anal. Calcd for C28H35Cl2N1O6: C, 60.87; H, 6.39; N, 2.54; Cl, 12.83; Found: C, 60.73; H, 6.41; N, 2.69; Cl, 12.86. Corrected for 0.11% H2O found by Karl Fischer analysis.
    Figure US20030130349A1-20030710-C00074
  • Preparation 11-B Scheme 11, 11-B Stereochemistry=1S-cis
  • (1S-cis) 3-Formyl-2,2,3-trimethylcyclopentanecarboxylic acid methyl ester (11-B) (C[0285] 11H20O3)
  • To a solution of 10-B (2.57 g, 12.8 mmol) in methylene chloride (50 mL) is added a mixture of pyridinium chlorochromate (3.05 g, 14.15 mmol), magnesium sulfate (4 g, 33 mmol), and Celite. After overnight stirring, the mixture is eluted through a short column of silica gel (80 g) using methylene chloride (500 mL) as the eluant. Evaporation in vacuo gives 11-B as a colorless liquid (2.09 g, 82% yield). [0286]
  • [0287] 1H NMR (CDCl3) δ 9.65 (1H), 3.67 (3H), 2.81 (1H), 2.40 (1H), 2.23 (1H), 1.94 (1H), 1.42 (1H), 1.16 (3H), 1.05 (3H), 0.88 (3H).
  • Preparation 11-C Scheme 11, 11-C Stereochemistry=1S-cis, (E)
  • (1S-cis)-3-[2-[(1,1-dimethylethyoxy)carbonyl]ethenyl]-2,2,3-trimethylcyclopentanecarboxylic acid methyl ester (11-C) (C[0288] 17H28O4)
  • To a dry N[0289] 2 flushed 100 mL flask is added t-butyl diethyl phosphonoacetate (5 mL, 21.3 mmol) and THF (dry, 20 mL). The flask is immersed in an ice water bath and, five minutes later, NaH/oil (60% NaH, 0.5 g, 12.5 mmol) is added in portions. After thirty minutes, 11-B (2.08 g, 10.5 mmol) is mixed with THF (dry, 15 mL) and added via syringe over a five minute period. Four hours later, the still cold solution is diluted with toluene (200 mL), shaken with ice water (4×100 mL), and the organic layer evaporated to dryness in vacuo, giving a colorless oil which is chromatographed on silica gel (80 g) using a gradient from 0 to 46 ethylacetate/hexane. A colorless oil 11-C is obtained (1.99 g, 63% yield).
  • [0290] 1H NMR (CDCl3) δ 6.92 (1H), 5.68 (1H), 3.67 (3H), 2.84 (1H), 2.23 (1H), 1.99 (1H), 1.90 (1H), 1.49 (1H), 1.48 (9H), 1.06 (3H), 1.01 (3H), 0.71 (3H); IR (nujol) 1728, 1712, 1651, 1438, 1358, 1316, 1288, 1270, 1228, 1220, 1191, 1171, 1151, 1132, 1001 cm−1; MS (FAB) m/z (rel. intensity) 297 (M+H, 44), 297 (44), 242 (12), 241 (87), 224 (14), 223 (99), 195 (14), 191 (14), 135 (12), 57 (34), 41 (12); HRMS (FAB) calcd for C17H28O4 +H1 297.2065, found 297.2067; Anal. Calcd for C17H28O4: C, 68.89; H, 9.52; N; Found: C, 69.03; H, 9.18.
  • Preparation 11-D Scheme 11, 11-D Stereochemistry=1S-cis, (E)
  • (1S-cis)-3-[2-[(1,1-dimethylethoxy)carbonyl]ethenyl]-2,2,3-trimethylcyclopentanecarboxylic acid (11-D) (C[0291] 16H26O4)
  • A solution of LiOH.H[0292] 2O(0.65 g, 15.4 mmol) in H2O (15 mL) and aqueous H2O2 (5 mL, 30%) is added to a solution of 11-C (1.52 g, 5.13 mmol) in methanol (30 mL). After stirring for two days, the mixture is diluted with water (100 mL) and evaporated in vacuo until all of the methanol is gone. The aqueous remainder is then shaken with diethyl ether (3×40 mL) and then cooled in an ice water bath and brought to pH 5 using 1N aq. HCl. The resultant white precipitate 11-D is isolated by suction filtration (0.385 g, 26% yield)
  • [0293] 1H NMR (CDCl3): δ 6.94 (1H), 5.69 (1H), 2.88 (1H), 2.27-1.84 (3H), 1.53 (1H), 1.49 (9H), 1.11 (3H), 1.02 (3H), 0.81 (3H); IR (nujol) 2729, 2669, 1707, 1647, 1418, 1393, 1316, 1305, 1242, 1154, 999, 976, 960, 944, 856 cm−1; MS (FAB) m/z (rel. intensity) 283 (M+H, 34), 566 (13), 283 (34), 228 (13), 227 (99), 210 (13), 209 (96), 191 (14), 181 (14), 57 (51), 41 (16); Anal. Calcd for C16H26O4: C, 68.06; H, 9.28; Found: C, 67.84; H, 9.10.
  • Preparation 11-F-1 Scheme 11, 11-F: wherein R4=H, R5=CO2CH3 R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl Stereochemistry=[1S-[1α,3α(E)]]-L
  • [S-[1α,3α(E)]]-N-[[3-[2-[(1,1-dimethylethoxy)carbonyl]ethenyl]-2,2,3-trimethylcyclopentyl]carbonyl]l-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester (11-F-1) (C[0294] 33H38Cl2N2O6)
  • A solution of 11-D (0.45 g, 1.59 mmol) in methylene chloride (15 mL) is cooled in an ice water bath. To this stirred solution were added N,N-diisopropylethyl amine (2 mL, 11.48 mmol), EDC (0.335 g, 1.75 mmol), HOBt (0.25 g, 1.85 mmol), and 4-N,N-dimethylaminopyridine (0.02 g, 0.16 mmol), followed thirty minutes later by the 4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester hydrochloride 7-E-1.HCl (7-E: R[0295] 4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorobenzoyl)amino]-phenyl) (0.67 g, 1.66 mmol). After two days, the reaction mixture is evaporated to dryness, mixed with toluene (200 mL), and shaken with cold 0.5N aq. HCl (50 mL) followed by water (4×50 mL). The organic layer is then evaporated to dryness in vacuo to give an off-white solid which is recrystallized from ethyl acetate to give a white solid (11-F-1) (0.59 g, 58% yield).
  • [0296] 1H NMR (CDCl3) δ 7.84 (1H), 7.56 (2H), 7.35-7.25 (3H), 7.08 (2H), 6.90 (1H), 5.77 (1H), 5.65 (1H), 4.87 (1H), 3.73 (3H), 3.12 (2H), 2.59 (1H), 2.21 (1H), 2.05 (1H), 1.85 (1H), 1.49 (1H), 1.47 (9H), 1.03 (3H), 0.99 (3H), 0.69 (3H); MS (ES+) m/z 631.
  • Preparation 11-G-1 Scheme 11, 11-G: wherein R4=H, R5=CO2CH3 R6=4-[(2,6-Dichlorobenzoyl)amino]-phenyl Stereochemistry=[1S-[1α,3α(E)]]
  • [1S-[1α,3α(E)]]-N-[[3-(2-Carboxyethenyl)-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester (11-G-1) (C[0297] 29H30Cl2N2O6)
  • A solution of 11-F-1 (0.5 g, 0.8 mmol) in trifluoroacetic acid (3 mL) is stirred overnight under nitrogen. The solution is then evaporated to dryness in vacuo, diluted with toluene (50 mL) and evaporated to dryness again, giving 11-G-1 as an oil (0.45 g, 97% yield). [0298]
  • [0299] 1H NMR (300 MHz, DMSO-d6) 10.75 (1H), 10.60 (1H), 7.96 (1H), 7.64-7.44 (4H), 7.23-7.14 (3H), 6.88 (1H), 5.63 (1H), 4.9 (1H), 4.8 (1H), 4.50 (1H), 3.58 (3H), 3.03-2.70 (2H), 2.50 (1H), 2.1-1.2 (4H), 0.92 (3H), 0.83 (3H), 0.55 (3H).
  • Preparation of Example 182 Scheme 11, 11-G: wherein R4=H, R5=CO2H R6=4-[(2,6-Dichlorobenzoyl)amino]-phenyl Stereochemistry=[1S-[1α,3α( (E)]]
  • [1S-[1α,3α(E)]]-N-[[3-(2-Carboxyethenyl)-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine disodium salt (Example 182) (C[0300] 28H28Cl2Na2N2O6)
  • To a solution 11-G-1 (0.45 g, 0.78 mmol) in methanol (5 mL), in a flask cooled in an ice water bath, is added a solution of LiOH.H[0301] 2O (0.127 g, 3 mmol) in H2O (5 mL) After two days, the mixture is diluted with water (50 mL), evaporated in vacuo until the methanol is gone and then cooled to −10° C. and brought to pH 2 using 1N aq. HCl. The resultant white precipitate is isolated by suction filtration to give a white solid which is stirred with saturated aqueous NaHCO3 (2 mL) and then transferred to a C-18 reversed phase HPLC column and eluted with a gradient from 0.01% aq NaHCO3 to 10% acetonitrile/0.01% aq NaHCO3. Evaporation is accomplished in vacuo to give Example 182 as a white solid (0.25 g, 51% yield).
  • [0302] 1H NMR (300 MHz, DMSO-d6) δ 7.51-7.42 (5H), 7.06 (2H), 6.39 (1H), 5.49 (1H), 4.10 (1H), 2.99 (1H), 2.86 (1H), 2.56 (1H), 1.85 (2H), 1.61 (1H), 1.29 (1H), 0.90 (3H), 0.85 (3H), 0.51 (3H); IR (nujol) 3393, 3257, 3124, 3035, 1654, 1604, 1562, 1544, 1515, 1431, 1398, 1325, 799, 778, 722 cm−1; MS (FAB) m/z (rel. intensity) 605 (M+H, 44), 629 (9), 627 (14), 608 (8), 607 (30), 606 (14), 605 (44), 585 (14), 583 (21), 73 (45), 23 (99); KF Water: 7.09%.
    Figure US20030130349A1-20030710-C00075
  • Preparation of Example 184 Scheme 12, 12-E: wherein R4=H, R5=CO211, R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl Stereochemistry=(1S-cis)-L
  • (1S-cis)-N-[[3-Cyano-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]l-L-phenylalanine Ester 10-A (5.0 g, 23.3 mmol) was dissolved in dry pyridine (100 mL). At 0° C., the reaction solution was treated with methanesulfonyl chloride (MsCl) (1 equiv, 23.3 mmol, 1.8 mL). The mixture was stirred for two days, while the bath temperature equilibrated to room temperature. The reaction mixture was saturated with ammonia (NH[0303] 3) gas. Excess NH3 was removed via a rotary evaporator. The reaction mixture was treated with MsCl (10 equiv. 18 mL, 0.23 mol) and allowed to stir overnight. Solvent was removed under reduced pressure and resulting crude material was purified by a silica gel (35 mm×15.2 cm) flash column chromatography using 90% hexanes:EtOAc as eluent to yield 3.5 g of 12-B: 1H NMR (CDCl3) δ 1.08 (3H), 1.19 (3H), 1.32 (3H), 1.73-2.01 (2H), 2.25-2.41 (2H), 2.71 (1H), 3.70 (3H).
  • Ester 12-B (0.7 g, 3.6 mmol) was dissolved in THF (20 mL) and treated with LiOH.H[0304] 2O (10 equiv, 1.5 g, 35.8 mmol) in H2O (20 mL) and MeOH (10 mL) After 2 h, the solvent was removed in vacuo. The residue was dissolved in acidic H2O (50 mL), for example 1N HCl, 10% H2SO4, or 1N AcOH, and was extracted with CHCl3 (3×20 mL) to yield, upon usual work-up, 0.6 g (92%) of 12-C: 1H NMR (CDCl3) δ 1.17 (3H), 1.25 (3H), 1.34 (3H), 1.77-2.00 (2H), 2.19-2.41 (2H), 2.69-2.81 (1H).
  • Acid 12-C (0.6 g, 3.3 mmol), HOBT (1.13 equiv, 0.5 g), DMAP (0.11 equiv, 0.04 g), EDC (1.1 equiv, 0.7 g,), Et[0305] 3N (3.6 equiv, 1.6 mL) and CH2Cl2 (20 mL) were combined and the reaction was stirred for a couple of minutes. Then 7-E-1.HCl (0.75 equiv, 1.0 g) was added and the reaction mixture was stirred overnight at room temperature. The solvent was removed under reduced pressure and the residue was triturated with THF. The precipitate was filtered, dissolved in a mixture of CH3CN:H2O, frozen and lyophilized to yield 12-E (where R4=H, R5=CO2CH3 and R6=4-[(2,6-dichlorobenzoyl)amino]phenyl): mp 170-174° C.; 1H NMR (MeOD) δ 1.01 (3H), 1.17 (3H), 1.32 (3H), 1.72-1.87 (2H), 2.02-2.32 (2H), 2.67 (1H), 2.97 (1H), 3.18 (1H), 3.71 (3H), 4.73 (1H), 7.22 (2H), 7.38-7.50 (3H), 7.59 (2H); IR (mineral oil mull) 3282, 3252, 3314, 3192, 3125, 3075, 2954, 2925, 2870, 2855, 2235, 1750, 1738, 1653, 1610, 1562, 1541, 1516, 1458, 1431, 1415, 1379, 1332, 1274, 1260, 1243, 1231, 1214, 1195, 799 cm−1; MS (FAB) for C27H29Cl2N3O4, m/z (relative intensity) 532 ([M+H]I+, 68), 531 ([M+H]+, 35), 530 ([M+H]+, 100), 529 (M+, 7), 351 (26), 349 (40), 175 (21), 173 (33), 136 (35), 109 (20). Anal. Calcd for C27H29Cl2N3O4nH2O: C, 61.14; H, 5.51; N, 7.92; Cl, 13.37; Found: C, 61.32: H, 5.53; N, 7.94; Cl, 13.04. Corrected for 0.66% H2O found by Karl Fischer analysis.
  • To a solution of the obtained 12E (1.0 g, 1.9 mmol), in MeOH (25 mL) was added LiOH.H[0306] 2O (5 equiv, 9.4 mmol, 0.4 g) in H2O (10 mL). The reaction solution was allowed to stir for 4 h. The reaction solution was reduced in vacuo to dryness. The residue was treated with n acidic H2O (25 mL). The resulting precipitate was filtered and subjected to silica gel (35 mm×15.2 cm) flash chromatography column using CH3CN spiked with 0.1% acetic acid as an eluent. Fractions containing compound were combined and solvent was removed under reduced pressure. The residue was then dissolved in CH3CN:H2O, frozen and lyophilized to yield 0.62 g (63%) of Example 184 (12-E: where R4=H, R5=CO2H and R6=4-[(2,6-dichlorobenzoyl)amino]phenyl): mp: unable to determine due to compound shrinkage at 50° C.; 1H NMR (MeOD) δ 1.02 (3H), 1.18 (3H), 1.31 (3H), 1.72-1.87 (2H), 2.01-2.33 (2H), 2.69 (1H), 2.97 (1H), 3.23 (1H), 4.68-4.78 (1H), 7.24 (2H), 7.38-7.49 (3H), 7.58 (2H), 7.96 (1H); IR (mineral oil mull) 3338, 3291, 3260, 3200, 3132, 3079, 3039, 2954, 2914, 2854, 2253, 1746, 1672, 1657, 1611, 1579, 1560, 1544, 1516, 1466, 1457, 1431, 1416, 1397, 1379, 1328, 1282, 1271, 1222, 1210, 1196, 1125, 812, 782, 800 cm−1; MS (FAB) for C26H27CI2N3O4, m/z (relative intensity) 518 ([M+HI 68), 517 ([M+H]+, 35), 516 ([M+H]+, 100), 515 (M+, 7), 337 (16), 335 (24), 175 (18), 173 (27), 136 (23), 109 (12). Anal. Calcd for C26H27Cl2N3O4nH2O: C, 60.47; H, 5.27; N, 8.14; Cl, 13.73; Found: C, 60.33; H, 5.25; N, 8.03; Cl, 13.62. Corrected for 4.58% H2O found by Karl Fischer analysis
    Figure US20030130349A1-20030710-C00076
  • The Intermediate amine according to Example 56 (619 mg, 1.43 mmol) was dissolved in CH[0307] 2Cl2 (5 mL) containing pyridine (0.3 mL, 3.58 mmol). To this solution 2,4,6-trichlorobenzoyl chloride (246 mg, 1.58 mmol) was added and the solution stirred at RT for 6 h. The reaction was acidified with 1 N HCl (20 mL) and extracted with CH2Cl2 (3×20 mL). The combined organics were dried (Na2SO4), filtered, and the solvent removed in vacuo. The residue was chromatographed (SiO2, gradient elution: 100% hexanes→50% EtOAc/hexanes) to provide Example 58 (866 mg, 95%) as a colorless foam: 1H NMR (300 MHz, CDCl3), δ 7.56 (2H), 7.33 (2H), 7.08 (2H), 5.84 (1H), 4.8-4.9 (1H), 3.74 (3H), 3.0-3.2 (3H), 2.4-2.6 (2H), 2.0-2.2 (1H), 1.6-1.8 (1H), 1.43 (9H), 1.21 (3H), 1.14 (3H), 0.76 (3H) 13C NMR (75 MHz, CDCl3), δ 174.92, 172.55, 172.00, 161.81, 136.29, 135.91, 134.46, 133.00, 132.69, 129.75, 128.08, 120.51, 80.19, 56.60, 54.33, 53.04, 52.35, 46.32, 37.14, 32.26, 27.99, 22.90, 22.39, 21.90, 20.50; ESMS (m/z) 639 (MH+).
  • Example 58 (810 mg, 1.27 mmol) was dissolved in CH[0308] 2Cl2 (5 mL) containing TFA (5 mL). This reaction mixture was stirred for 6 h at RT. Evaporation of the solvent under reduced pressure provided a crude oil. The residue was chromatographed (SiO2, gradient elution: 100% hexanes→50% EtOAc/hexanes) to provide Example 92 (703 mg, 95 O) as a colorless foam: 1H NMR (300 MHz, Acetone-d6), δ 9.68 (1H), 7.66 (2H), 7.44 (2H), 7.17 (2H), 6.93 (1H), 4.88 (1H), 3.72 (3H), 3.17 (1H), 3.05 (1H), 2.79 (1H), 2.5-2.6 (1H), 2.1-2.2 (1H), 1.7-1.8 (1H), 1.4-1.5 (1H), 1.25 (3H), 1.21 (3H), 0.84 (3H); 13C NMR (75 MHz, Acetone-d6), δ 176.92, 172.14, 171.14, 160.87, 136.16, 134.58, 134.48, 132.16, 132.12, 128.93, 127.08, 119.23, 59.24, 55.25, 52.60, 51.06, 45.64, 36.03, 31.71, 21.92, 21.63, 20.83, 20.06; ESMS (m/z) 583 (MH+).
  • Example 92 (684 mg, 1.17 mmol) was dissolved in H[0309] 2O (8 mL) containing LiOH (127 mg, 5.27 mmol). After 6 h at RT the mixture was acidified with 3 N HCl (3 mL) and the precipitate filtered and washed with cold H2O (3 mL). Drying under high vacuum provided Example 93 (547 mg, 82%) as a colorless solid: 1H NMR (300 MHz, Acetone-d6), δ 9.96 (1H), 7.70 (2H), 7.58 (2H), 7.30 (2H), 4.8-4.9 (1H), 3.23 (1H), 3.07 (1H), 2.89 (1H), 2.5-2.6 (1H), 2.0-2.2 (1H), 1.6-1.7 (1H), 1.4-1.5 (1H), 1.26 (3H), 1.19 (3H), 0.81 (3H); 13C NMR (75 MHz, Acetone-d6), δ 178.52, 174.10, 173.62, 162.22, 138.12, 136.08, 134.47, 133.69, 130.77, 128.90, 120.54, 56.92, 54.56, 53.89, 47.24, 37.48, 33.37, 23.52, 23.04, 22.31, 21.73; ESMS (m/z) 569 ([M−H]).
    Figure US20030130349A1-20030710-C00077
  • The compound according to Example 56 (810 mg, 1.87 mmol) was dissolved in Pyr/CH[0310] 2Cl2 (5 mL of each) and O-nitrobenzoyl chloride (383 mg, 2.06 mmol) was added under dry N2. The reaction was stirred for 4 hours at RT. The solvent was evaporated under high vacuum and 1N HCl (20 mL) was added to the residue. This was extracted with CH2Cl2 (3×25 mL) and the combined organic phases were dried over Na2SO4. The solution was filtered, solvent evaporated and the residue chromatographed (SiO2, gradient elution: 100% hexanes→50% EtOAc/hexanes) to provide the intermediate (14-A) (808 mg, 74%): 1H NMR (300 MHz, CDCl3), δ 8.4-8.6 (1H), 8.03 (1H), 7.6-7.7 (3H), 7.49 (2H), 7.04 (2H), 5.86 (1H), 4.7-4.8 (1H), 3.71 (3H), 3.0-3.2 (2H), 2.4-2.6 (2H), 2.0-2.1 (1H), 1.6-1.7 (1H), 1.4-1.5 (1H), 1.42 (9H), 1.20 (3H), 1.12 (3H), 0.73 (3H); 13C NMR (75 MHz, CDCl3), & 174.94, 172.61, 171.97, 164.57, 146.15, 136.64, 133.77, 132.76, 132.38, 130,56, 129.62, 128.63, 124.47, 120.56, 80.16, 56.58, 54.23, 53.05, 52.28, 46.31, 37.07, 32.23, 27.96, -22.83, 22.31, 21.85, 20.50; FABMS (m/z) 582 (MH+).
  • The above compound, (14-A) (706 mg, 1.21 mmol), was dissolved in CH[0311] 2Cl2 (5 mL) and TFA (15 mL) was added with stirring at RT. After 4 hours the solvent was evaporated and the residue chromatographed (SiO2, gradient elution: 100% hexanes→100% EtOAc) to provide the mono methyl ester Example 101 (623 mg, 98%): 1H NMR (300 MHz, Acetone-d6), δ 9.79 (1H), 8.11 (1H), 7.7-7.9 (6H), 7.22 (2H), 4.81 (1H), 3.68 (3H), 3.15 (1H), 3.02 (1H), 2.86 (1H), 2.53 (1H), 2.0-2.2 (2H), 1.6-1.8 (1H), 1.43 (1H), 1.26 (3H), 1.20 (3H), 0.81 (3H); 13C NMR (75 MHz, Acetone-d6) δ 177.64, 173.39, 172.92, 165.03, 147.96, 138.68, 134.68, 134.11, 133.81, 131.70, 130.52, 129.97, 125.18, 120.62, 120.54, 56.90, 54.55, 53.88, 52.31, 47.16, 37.56, 33.40, 23.48, 23.00, 22.30, 21.66; ESMS (m/z) 526 (MH+).
  • Example 101 (605 mg, 1.15 mmol) was dissolved in H[0312] 2O (5 mL) containing LiOH (138 mg, 5.76 mmol). This was stirred for 4 hours. The solution thus obtained was acidified with 2 N HCl (10 mL) and the precipitate filtered. The filter cake was washed with cold H2O (3 mL) and then dried under high vacuum to provide Example 102 (539 mg, 92%) as a white amorphous powder:
  • [0313] 1H NMR (300 MHz, DMSO-d6), δ 10.66 (1H), 8.14 (1H), 7.85 (2H), 7.76 (2H), 7.57 (2H), 7.22 (2H), 4.4-4.5 (1H), 3.02 (1H), 2.88 (1H), 2.70 (1H), 2.3-2.4 (1H) 1.8-2.0 (1H), 1.5-1.6 (1H), 1.2-1.4 (1H), 1.17 (3H) 1.12 (3H), 0.67 (3H); 13C NMR (75 MHz, DMSO-d6), δ 177.00, 173.17, 172.03, 163.90, 146.50, 137.19, 134.04, 133.35, 132.69, 130.92, 129.38, 129.28, 124.23, 119.44, 55.50, 53.59, 51.90, 45.95, 36.10, 32.23, 22.30, 22.23, 21.72, 21.00; ESMS (m/z) 512 (MH+).
    Figure US20030130349A1-20030710-C00078
  • Preparation 10-A Scheme 15, 10-A Stereochemistry=(1R-cis)
  • (1R-cis)-1,2,2-Trimethylcyclopentane-1,3-dicarboxylic acid 3-methyl ester 10-A (C[0314] 11H18O4).
  • To methanol (0.5L), cooled in an ice-water bath under nitrogen, is added acetyl chloride (50 mL, 0.703 mol) over 30 minutes. After stirring for 30 minutes (1R, 3S)-camphoric acid 15-A (10 g, 0.5 mol) was added in one portion. The camphoric acid dissolves over ca. 10 minutes and the solution was allowed to warm slowly to room temperature and stir for 72 hours. Concentration in vacuo afforded a clear, pale yellow oil which was dissolved in ethyl acetate (0.6L). The solution was extracted with 0.5N aq. NaOH (4×0.35L). The combined aqueous phases were washed with pentane (0.35L) and the pH of the aq. layer was adjusted to ca. 4 with 1N aq. HCl. The aqueous layer was extracted with ether (4×0.35L) and the combined organic phases were concentrated in vacuo to give a colorless oil which slowly solidified to afford 96 g (90%) of 10-A as a white crystalline solid. An analytical sample can be obtained by recrystallizing 10-A from ether-pentane (1:1) to provide 10-A as clear hexagonal plates. [0315]
  • MP: 76-77° C.; 1H-NMR (CDCl[0316] 3) δ 3.69 (s, 3H), 2.79 (m, 1H), 2.54 (m, 1H), 2.20 (m, 1H), 1.84 (m, 1H), 1.54 (m, 1H), 1.45 (m, 1H), 1.27 (s, 3H), 1.26 (s, 3H), 0.86 (s, 3H); IR (nujol): 3201, 2925, 1730, 1700, 1237, 1210, 1150, and 1110 cm−1; Anal: Calcd. for C11H18O4: C, 61.66; H, 8.47; Found: C, 61.63; H, 8.75.
  • Preparation 15-C Scheme 15, 15-C Stereochemistry=(1R-cis)
  • (1R-cis)-1,2,2-Trimethylcyclopentane-1,3-dicarboxylic acid 1 (1,1-dimethylethyl)-3-methyl diester 15-C (C[0317] 15H26O4)
  • To 10-A (25 g, 0.117 mol) in a 500 mL Parr bottle, cooled in a dry ice-iPrOH bath under nitrogen, was condensed isobutylene until the bottle was approximately ½ full. In a separate Erlenmeyer flask, ether (6 mL) was cooled (dry ice-iPrOH bath) and conc. sulfuric acid (3 mL) was added. The mixture was allowed to cool for ca. 5 minutes, then was slowly added via disposable pipet to the isobutylene-15-B mixture. The Parr bottle was transferred to the shaker apparatus and shaken for 12 hours (pressure ca. 35 psi at the end of 12 hours). The bottle was surrounded by aluminum foil, dry ice was added to cool the bottle and contents, and the bottle was removed from the shaker when the pressure reading was ca. 0 psi. The isobutylene was condensed from the reaction vessel via a cold finger condenser over ca. 2 hours. The resulting thin oil was dissolved in pentane (0.5L), the organic phase was washed with water (2×0.25L), 0.5N aq. NaOH (2×50 mL), and water (2×0.25L). The organic phase was concentrated in vacuo to provide 15-C as a clear colorless oil which slowly solidified at room temperature. Recrystallization from petroleum ether gave 15-C as a fine white crystalline solid (26.86 g, 89%). [0318]
  • MP: 36-37.6° C.; [0319] 1H-NMR: (300 MHz, CDCl3): δ 3.68 (s, 31H) 2.78 (m, 1H), 2.52 (m, 1H), 2.16 (m, 1H), 1.78 (m, 1H), 1.45 (m, 1H), 1.45 (s, 9H), 1.24 (s, 3H), 1.17 (s, 3H), 0.81 (s, 3H).
  • Preparation 15-D Scheme 15, 15-D Stereochemistry=(1R-cis)
  • (1R-cis)-1,2,2-Trimethylcyclopentane-1,3-dicarboxylic acid 1-(1,1-dimethylethyl) ester 15-D (C[0320] 14H24O4)
  • To 15-C (10.25 g, 0.38 mol) in methanol (0.1L) was added in order water (0.1L, see considerable precipitation), LiOH di-hydrate (10 g, 2.38 mol), and 30% aq. hydrogen peroxide (0.1L,). The mixture was warmed in a 90° C. oil bath and allowed to stir for 20 hours. The mixture was cooled to room temperature and concentrated in vacuo to remove methanol and the residual white solid material was removed by filtration. The clear solution was washed with pentane (3×0.15L), the aqueous layer was cooled in an ice-water bath and carefully acidified to ca. pH 4 with 1N aq. HCl. The resulting white precipitate was isolated by filtration, washed with water (3×0.1L) and air dried to afford 15-D (9.6 g, 96%) as a fine white solid. [0321]
  • MP: 98-98.6° C.; [0322] 1H-NMR: (300 MHz, CDCl3): δ 2.82 (1H) 2.50 (1H), 2.13 (1H), 1.79 (1H), 1.46 (9H) 1.45 (1H), 1.28 (3H), 1.18 (3H), 0.89 (3H); IR (nujol): 3075, 3025, 3006, 1719, 1689, 1270, 1249, 1164, and 851 cm−1; EI/MS: 200 (3.8), 183 (7.3), 164 (7.1), 154 (20.3), 136 (14.6), 109 (32.8), 57 (base); Anal: Calcd. for C14H24O4-0.18H2O: C, 64.77; H, 9.46; Found: C, 64.79; H, 9.44; K.F.-Water: 0.86%.
    Figure US20030130349A1-20030710-C00079
  • Boc-piperazine (1.9 gm, 10.4 mmol) was dissolved in THF (10 mL) and CH[0323] 2Cl2 (1 mL) containing DIEA (5.6 mL, 32.2 mmol). To this solution 2,6-dichlorobenzoyl chloride (1.6 mL, 11.4 mmol) was added at 0° C. After stirring the reaction for 1 hour at 0° C., 1N HCl (30 mL) was added. The mixture was extracted with CH2Cl2 (2×20 mL) and the combined organics dried (Na2SO4), filtered and the solvent removed in vacuo. Chromatography of the residue (SiO2, gradient elution, hexanes→30% EtOAc/hexanes) provided Intermediate (16-A) as an off white solid (3.6 gm, 98%): mp=157-159° C.; ESMS (m/z) 359 (MH+).
    Figure US20030130349A1-20030710-C00080
  • Intermediate 16-A (553 mg, 1.54 mmol) was dissolved in CH[0324] 2Cl2 (5 mL) and treated with TFA (5 mL) After 2 hours the solvent was removed under reduced pressure. The residue was dissolved in CH2Cl2 (20 mL) containing DIEA (0.8 mL, 4.6 mmol). Boc-aspartic acid α t-butyl ester (534 mg, 1.85 mmol) was added with BOP-Cl (470 mg, 1.85 mmol). After stirring at RT for 24 hours the reaction was quenched with 1N HCl (25 mL). The mixture was extracted with CH2Cl2 (3×25 mL). The combined organics were dried (Na2SO4), filtered and the solvent removed in vacuo. The residue was purified by column chromatography (SiO2, gradient elution, hexanes→500 EtOAc/hexanes) to provide the intermediate aspartate (17-A) (405 mg, 50%): ESMS (m/z) 530 (MH+).
  • To a methanolic solution of the above mentioned intermediate 17-A (0.2 g, 0.378 mmol), HCl gas was bubbled for 5 minutes and the reaction mixture was left to stand overnight at room temperature. The methanol was evaporated and the residual gum was triturated with ether. The resultant solid was washed with ether and dried under high vacuum. The solid was suspended in THF (5 mL), and 17-B (1R,3S)-1-(tert-butoxycarbonyl)-1,2,2-trimethylcyclopentanecarboxylic acid (102 mg, 0.397 mmol), BOP (176 mg, 0.397 mmol) were added, followed by DIEA (0.207 mL, 1.19 mmol). The mixture was stirred overnight at room temperature. The solvent was evaporated and the residue was partitioned between 1N HCl (5 mL) and EtOAc (15 mL). The organic layer was separated and washed successively with 1N HCl (5 mL), brine (5 mL), Sat. NaHCO[0325] 3 (2×5 mL), Sat. LiCl (2×5 mL), and dried over MgSO4. Evaporation of EtOAc produced a colorless solid, which was purified on silica (Chromatotron, hexane:EtOAc (1:1) as eluant) to provide Example 176 (206 mg, 87%) as a colorless solid. ESMS: (m/z) 626 (MH+).
  • Example 176 (0.18 g, 0.287 mmol) was dissolved in CH[0326] 2Cl2 (1.0 mL) and TFA (1.0 mL). After stirring at room temperature for 1 hr the solvent was evaporated and the resultant gum triturated with ether. The solid was washed with ether and dried under vacuum to provide [1S-[1α(R*),3α]]-α-[[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]amino]-4-(2,6-dichlorobenzoyl)-γ-oxo-1-piperazinebutanoic acid methyl ester as a colorless solid (0.14-g, 87%). ESMS (m/z) 570 (MH+).
  • To a solution of the methyl ester obtained above (0.1 g, 0.18 mmol) in THF/CH[0327] 3OH (5 mL/1 mL) was added an aqueous (1 mL) solution of LiOH mono hydrate (19 mg, 0.378 mmol) and the mixture was stirred at room temperature for 2 h. The organics were evaporated and the residue was taken up in 2 ml of water and acidified with citric acid. The solid was filtered, washed with water and vacuum dried to provide Example 177 (90 mg, 92%). ESMS (m/z) 556 (MH+).
    Figure US20030130349A1-20030710-C00081
  • Intermediate 16-A (593 mg, 1.54 mmol) was dissolved in CH[0328] 2Cl2 (6 mL) and treated with TFA (6 mL). After 2 hours the solvent was removed under reduced pressure. The residue was dissolved in CH2CL. (20 mL) containing DIEA (0.9 mL, 4.95 mmol). Boc-aspartic acid β t-butyl ester (573 mg, 1.98 mmol) was added with BOP-Cl (504 mg, 1.98 mmol). After stirring at RT for 24 hours the reaction was quenched with 1N HCl (15 mL). The mixture was extracted with CH2Cl2 (3×20 mL). The combined organics were dried (Na2SO4), filtered and the solvent removed in vacuo. The residue was purified by column chromatography (SiO2, gradient elution, hexanes→50% EtOAc/hexanes) to provide the intermediate aspartate (18-A) (360 mg, 41 ESMS (m/z) 530 (MH+).
  • To a methanolic solution of the above mentioned intermediate (18-A) (0.25 g, 0.47 mmol), HCl gas was bubbled for 5 minutes and the reaction mixture was left to stand overnight at room temperature. The methanol was evaporated and the residual gum was triturated with ether. The resultant solid was washed with ether and dried under high vacuum. The solid was suspended in THF (5 mL), and 15-D (1R,3S)-1-(tert-butoxycarbonyl)-1,2,2-trimethylcyclopentanecarboxylic acid (127 mg, 0.495 mmol), BOP (219 mg, 0.495 mmol) were added, followed by DIEA (0.259 mL, 1.485 mmol). The mixture was stirred overnight at room temperature. The solvent was evaporated and the residue was partitioned between 1N HCl (5 ml) and EtOAc (15 ml). The organic layer was separated and washed successively with 1N HCl (5 ml), brine (5 ml), Sat. NaHCO[0329] 3 (2×5 ml), Sat. LiCl (2×5 ml), and dried over MgSO4. Evaporation of EtOAc produced a colorless solid, which was purified on silica (Chromatotron, hexane:EtOAc (1:1) as eluant) to provide Example 178 (249 mg, 77%) as a colorless solid. ESMS: (m/z) b26 (MH+).
  • Example 178 (0.19 g, 0.303 mmol) was dissolved in CH[0330] 2Cl2 (1.0 mL) and TFA (1.0 mL). After stirring at room temperature for 1 hr the solvent was evaporated and the resultant gum triturated with ether. The solid was washed with ether and dried under vacuum to provide [1S-[1α(R*),3α]]-β-[[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonylamino]-4-[2,6-dichlorobenzoyl)-γ-oxo-1-piperazinebutanoic acid methyl ester as a colorless solid (98 mg, 58%). ESMS (m/z) 570 (MH+).
  • To a solution of the methyl ester obtained above (60 mg, 0.105 mmol) in THF/CH[0331] 3OH (5 mL/1 mL) was added an aqueous (1 mL) solution of LiOH mono hydrate (9.5 mg, 0.221 mmol) and the mixture was stirred at room temperature for 2 h. The organics were evaporated and the residue was taken up in 2 ml of water and acidified with citric acid. The solid was filtered, washed with water and vacuum dried to provide Example 195 (48 mg, 836). ESMS (m/z) 556 (MH+).
    Figure US20030130349A1-20030710-C00082
  • Preparation 19-C-2 Scheme 19, 19-C: wherein R4=H, R5=CO2CH3 R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl, R19-2=H, R19-1=CH3, R19-3=OC(CH3)3, m=0 Stereochemistry=[1S-[1α,3α(S*)]]
  • [1S[1α,3α(S*)]] N-3-[[(1-(1,1-dimethylethyoxy)carbonylethyl) amino]carbonyl]-2,2,3-trimethylcyclopentyl)carbonyl-4-[(2,6-dichiorobenzoyl)amino]-L-phenylalanine methyl ester (19-C-1) (C[0332] 34H43Cl2N3O7)
  • A solution of (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichiorobenzoyl)amino]-L-phenylalanine methyl ester 19-A-1 (19-A: R[0333] 4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl) (0.468 g, 0.85 mmol), HATU (0.36 g, 0.95 mmol), and diisopropylethyl amine (dry, 2 mL, 11.5 mmol) are stirred together with DMF (dry, 5 mL) in a dry round bottom flask under a nitrogen atmosphere. After 30 minutes, D-alanine tbutyl ester hydrochloride (19-B, (R19-1=CH3, R19-2=H, R19-3=t-BuO, Stereochemistry D) (0.34 g, 1.87 mmol) is added. After three days, the mixture is evaporated to dryness in vacuo to give a yellow oil which is mixed with methylene chloride (100 mL) and shaken with water (5×50 mL). The organic layer is then evaporated to dryness, giving an off-white solid Recrystallization from ethyl acetate/diethyl ether gives 19-C-1, as a white solid (0.428 g, 74% yield). 1H NMR (300 MHz, DMSO-d6) δ 10.70 (1H), 7.90 (1H), 7.60-7.18 (8H), 4.48 (1H), 4.15 (1H), 3.57 (3H), 2.95 (2H), 2.75 (1H), 2.37 (1H), 1.87 (1H), 1.55 (1H), 1.36 (9H), 1.30 (1H), 1.22 (3H), 1.18 (3H), 1.09 (3H), 0.57 (3H).
  • Preparation 19-C-2 Scheme 19, 19-C: wherein R4=H, R5=CO2CH3 R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl R19-2=H, R19-1=CH3, R19-3=OH, m=0 Stereochemistry=[1S-[1α,3α(S*)]]
  • [1S-[1α,3α(S*)] J-N-[[3-[(1-Carboxyethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester (19-C-2) (C[0334] 30H35Cl2N3O7)
  • Di-ester 19-C-1 is stirred overnight with trifluoroacetic acid (5 mL) and then the mixture is diluted with toluene (100 mL) followed by evaporation to dryness in vacuo, giving acid 19-C-2 as a pale brown oil (0.34 g, 90% yield). [0335]
  • [0336] 1H NMR (300 MHz, DMSO-d6) δ 10.70 (1H), 7.96 (1H), 7.57-7.10 (8H), 4.47 (1H), 4.22 (1H), 3.57 (3H), 2.95 (2H), 2.65 (1H), 2.37 (1H), 1.89 (1H), 1.55 (1H), 1.30 (1H), 1.25 (3H), 1.18 (3H), 1.09 (3H), 0.57 (3H); MS (ES+) m/z 619.8.
  • Preparation of Example 211 Scheme 19, 19-D: wherein R4=H R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl R19-2=H, R19-1=CH3, R19-3=OH, m=0 Stereochemistry=[1S-[1α,3α(S*)]]
  • [1S-[1α,3α(S*)]]-N-[[3-[[(1-Carboxyethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine Example 211 (C[0337] 29H33Cl2N3O7)
  • A solution of LiOH[0338] 12O (0.15 g, 3.57 mm) in H2O (5 mL) is added to a solution of 19-C-2 in methanol (5 mL). After overnight stirring, the solution is brought to pH 7 with 1.2N aq. HCl (2 mL), evaporated in vacuo until the methanol is gone, and shaken with diethyl ether (3×30 mL). The aqueous layer is filtered, cooled in an ice bath and brought to pH 2 using 1.2N aq. HCl. The resultant white precipitate is filtered, washed with water (100 mL) and air dried to give Example 211 as a white solid (0.277 g, 78% yield).
  • [0339] 1H NMR (DMSO-d6) δ 10.67 (1H), 7.76 (1H), 7.57-7.44 (5H) 7.31 (1H), 7.20 (2H), 4.42 (1H), 4.22 (1H), 3.03-2.82 (2H), 2.64 (1H), 2.36 (1H), 1.89 (1H), 1.54 (1H) 1.32-1.26 (4H), 1.19 (3H), 1.09 (3H), 0.58 (3H); IR (nujol) 3293, 3261, 3078, 1740, 1672, 1612, 1562, 1551, 1527, 1518, 1429, 1415, 1334, 1276, 1197 cm−1; MS (ES−) m/z 606.3, 604.3; KF Water 7.76%.
  • Preparation 19-C-3 Scheme 19, 19-C: wherein R4=H, R5 CO2CH3 R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl, R19-2, R19-1=(CH2) 3, R19-3=NH2, m=0 Stereochemistry=[1S-[1α,3α(S*)]]
  • [1S-[1α,3α(S*)]]-N-[3-[[2-(Aminocarbonyl)-1-pyrrolidinyl]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester (19-C-3) (C[0340] 32H37Cl2N4O6)
  • In a similar manner to that reported for the synthesis of 19-C-1, 19-A-1 (19-A: R[0341] 4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl))(0.55 g, 1.00 mmol) is coupled to D-prolineamide (0.3 g, 2.63 mmol) using HATU (0.40 g, 1.05 mmol) and diisopropylethyl amine (3 mL, 17.2 mmol) in dry DMF. After overnight stirring, the reaction mixture is evaporated to dryness in vacuo, giving a pale yellow oil which is stirred with ethyl acetate (50 mL). A precipitate soon formed and it is isolated by suction filtration to give 19-C-3 as a white solid (0.632 g, 900 yield).
  • [0342] 1H NMR (DMSO-d6) δ 10.67 (1H), 7.95 (1H), 7.58-7.48 (5H), 7.19 (2H), 7.10 (1H), 6.65 (1H), 4.45 (1H), 4.18 (1H), 3.70-3.30 (1H), 3.57 (3H), 2.90 (2H), 2.58 (111), 2.28-1.40 (9H), 1.25 (3H), 1.12 (3H), 0.73 (3H).
  • Preparation of Example 212 Scheme 19, 19-D: wherein R4=H, R5=CO2Li R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl, R19-2, R19-1=(CH2) 3, R19-3=NH2, m=0 Stereochemistry=[1S-[1α,3α(S*)]]
  • [1S-[1α,3α(S*)]]-N-[3-[[2-(Aminocarbonyl)-1-pyrrolidinyl]carbonyl]-2,2,3-trimethylcyclopentyl]-carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine monolithium salt Example 212 (C[0343] 31H35Cl2N4O6Li)
  • A solution of LiOH.H[0344] 2O (0.2 g, 4.8 mmol) in H2O (4 mL) is added to a stirred solution of the methyl ester 19C-3 (0.58 g, 0.89 mmol) in methanol (10 mL). After overnight stirring, the mixture is evaporated in vacuo until the methanol is gone. The reaction mixture is then brought to pH 7 with 1N aq. HCl, filtered and the filtrate transferred to a C-18 reversed phase HPLC column and eluted with a 0-10% acetonitrile/water gradient. Evaporation is accomplished in vacuo to give the target compound as a white solid (0.445 g, 78% yield).
  • [0345] 1H NMR (300 MHz, DMSO-d,) δ 10.55 (1H), 7.56-7.43 (5H), 7.04 (3H), 6.79 (1H), 6.62 (1H), 4.18 (1H), 3.92 (1H) 3.62 (1H) 3.43 (1H), 2.96 (2H), 2.44 (1H), 2.25-1.5 (8H) 1.23 (3H), 1.1 (3H), 0.75 (3H); IR (nujol) 3392, 3288, 3194, 3124, 3068, 1660, 1604, 1562, 1539, 1515, 1431, 1403, 1325, 799, 686 cm−1; MS (FAB) m/z (rel. intensity) 631 (M+H, 1), 659 (29), 653 (24), 643 (26), 639 (34), 637 (50), 279 (33), 133 (26), 109 (61), 70 (35), 23 (99); HRMS (FAB) m/z calcd for C31H36Cl2N4O6+H+ 631.2090, found 631.2086; KF Water: 9.90%.
  • Preparation 19-C-4 Scheme 19, 19-C: wherein R4=H, R5=CO2CH3 R6=4-[(2,6-Dichlorobenzoyl)amino]-phenyl, R19-2, R19-1=(CH2) 3, R19-3=NH2, m=0 Stereochemistry=[1S-[1α,3α(R*)]]
  • [1S-[1α,3α(R*)]]-N-[3-[[2-(Aminocarbonyl)-1-pyrrolidinyl]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenyl alanine methyl ester (19-C-4) (C[0346] 32H37Cl2N4O6)
  • In a similar manner to that reported for the synthesis of 19-C-3,19-A-1 (19-A: R[0347] 4=H, R5 CO2CH3, R6=4-[(2,6-Dichlorobenzoyl)amino]-phenyl))(0.50 g, 0.91 mmol) is coupled to L-prolineamide (0.2 g, 1.75 mmol) using HATU (0.41 g, 1.08 mmol) and diisopropylethyl amine (1 mL, 5.74 mmol) in dry DMF (5 mL). After overnight stirring, the mixture is evaporated to dryness in vacuo to give a yellow oil which is shaken with methylene chloride (100 mL) and water (50 mL); this gives a white precipitate which is filtered and the isolated solid is washed with water (3×50 mL). Recrystallization from ethyl acetate gives 19-C-4 as a white solid (0.31 g, 53% yield).
  • [0348] 1H NMR (DMSO-d): δ 10.70 (1H), 7.92 (1H), 7.50 (5H) 7.19 (2H), 7.04 (1H), 6.74 (1H), 4.48 (1H), 4.11 (1H), 3.58 (3H), 3.47 (1H), 2.92 (2H), 2.56 (1H), 2.35 (1H), 2.10-1.45 (8H), 1.24 (3H), 1.09 (3H), 0.71 (3H); MS (ES+, m/z) 644.9.
  • Preparation of Example 213 Scheme 19, 19-D: wherein R4=Hi R5=CO2Na R6=4-[(2,6-Dichlorobenzoyl)amino]-phenyl, R19-2, R19-1=(CH2)3, R19-3=NH2, In=0 Stereochemistry=[1S-[1α(S*),3α(S*)]]
  • [1S-[1α,3α(R*)]]-N-[3-[[2-(Aminocarbonyl)-1-pyrrolidinyl]carbonyl]-2,2,3-trimethylcyclopentyl]-carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine monosodium salt Example 213 (C[0349] 31H35Cl2N4O6Na)
  • The ester 19-C-4 (0.258 g., 0.4 mmol) is hydrolyzed in a manner similar to that described above for Example 212. The reaction mixture is then brought to pH 7, filtered and the filtrate transferred to a C-18 reversed phase HPLC column and eluted with a 0-10% acetonitrile/0.01 aqueous Na[0350] 2CO3 gradient. Evaporation is accomplished in vacuo to give the target compound as a white solid (0.2 g, 76% yield).
  • [0351] 1H NMR (300 MHz, DMSO-d,): δ 10.57 (1H), 7.56-7.44 (4H), 7.02 (3H), 6.77 (2H), 4.10 (1H), 3.91 (1H), 3.6 (1H), 3.50 (1H), 3.95 (1H), 2.44 (1H), 2.10-1.50 (7H), 1.23 (3H), 1.07 (3H), 0.73 (3H); IR (nujol) 3392, 3288, 3194, 3124, 3068, 1660, 1604, 1562, 1539, 1515, 1431, 1403, 1325, 799, 686 cm−1; MS (FAB) m/z (rel. intensity) 631 (M+H, 1), 659 (29), 653 (24), 643 (26), 639 (34), 637 (50), 279 (33), 133 (26), 109 (61), 70 (35), 23 (99); HRMS (FAB) calcd for C31H36Cl2N4O6+H+ 631.2090, found 631.2086; KF Water: 9.90%.
  • Preparation 19-C-5 Scheme 19, 19-C: wherein R4=H, R5=CO2CH3 R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl, R19-2=H, R19-1=H, R19=OCH3, m=1 Stereochemistry=(1S-cis)-L
  • (1S-cis)-N-[[3-[[[2-(Methyloxycarbonyl)ethyl]amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester (19-C-5) (C[0352] 31H37Cl2N3O7)
  • A solution of 19-A-1 (19-A: R[0353] 4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl) (0.6 g, 1.09 mmol), HATU (0.5 g, 1.32 mmol), and diisopropylethyl amine (dry, 2 mL, 11.5 mmol) are stirred together with DMF (dry, 5 mL) in a dry round bottom flask under a nitrogen atmosphere. After 30-minutes, O-alanine methyl ester hydrochloride (19-B, R19-1=H, R19-2=H, R19-3=CH3O) (0.3 g, 2.15 mmol) is added. After 18 hours, the mixture is evaporated to dryness in vacuo to give a yellow oil which is mixed with ehtyl acetate (150 mL) and shaken with water (50 mL). The organic layer is washed with water and brine (2×60 mL, 5:1), satd aq. NaHCO3 (50 mL), water and brine (60 mL, 5:1), the organic layer is mixed with pentane (30 mL) and is cooled to −20° C. (3 days). The resulting solid is isolated by filtration, washed with diethyl ether (2×50 mL), and is air dried to give 19-C-5 as a white solid (0.63 g, 91%).
  • [0354] 1H NMR (CDCl3) δ 7.62 (1H), 7.57 (2H), 7.32 (3H), 7.10 (2H), 6.26 (1H), 5.81 (1H), 4.88 (1H), 3.75 (3H), 3.68 (3H), 3.48 (2H), 3.12 (2H), 2.51 (3H), 2.28 (2H), 1.80 (1H), 1.49 (1H), 1.26 (3H), 1.15 (3H), 0.74 (3H); MS (ES+) m/z 633.8.
  • Preparation of Example 214 Scheme 19, 19-D: wherein R4=H, R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl, R19-2=H, R19-1=H, R19-3=OH, m=1 Stereochemistry=(1S-cis)-L
  • (1S-cis)-N-[[3-[[(2-Carboxyethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl ]carbonyl]-4-[[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (Example 214) (C[0355] 29H33Cl2N3O7)
  • To a solution of the dimethyl ester 19-C-5 (19-C: R[0356] 4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorobenzoyl)amino]-phenyl, R19-2=H, R19-1=H, R19-3=OCH3, n=1, Stereochemistry=(1S-cis)-L) (0.52 g, 0.82 mmol) in methanol (10 mL) is added a solution of LiOH.H2O (0.18 g, 4.29 mmol) in water (4 mL). After overnight stirring, the reaction mixture is brought to pH 8 using 1N aq. HCl and then evaporated to dryness in vacuo. The mixture is chromatographed on a reversed phase (C-18) HPLC column using a gradient (0 to 10% acetonitrile/(36 methanol in H2O)). The selected eluant is evaporated to dryness and dissolved in water (50 mL), cooled in an ice water bath, and brought to pH 3 using 1N aq. HCl. The resultant white precipitate is isolated by suction filtration to give Example 214 as a white solid (0.43 g, 85% yield).
  • [0357] 1H NMR (DMSO-d6) δ 10.6 (1H), 7.91 (1H), 7.55-7.42 (5H) 7.08 (2H), 6.78 (1H), 4.02 (1H), 3.10 (2H), 2.90 (2H), 2.48 (1H), 2.25 (1H), 1.93 (3H), 1.56 (1H), 1.24 (1H), 0.99 (3H), 0.93 (3H), 0.46 (3H); IR (nujol) 3327, 3080, 1726, 1672, 1622, 1614, 1595, 1558, 1515, 1429, 1338, 1279, 1262, 1197, 783 cm−1; MS (FAB) m/z (rel. intensity) 606 (M+H, 99), 609 (24), 608 (69), 607 (42), 606 (99), 605 (15), 517 (16), 254 (691, 175 (16), 173 (25), 109 (51); HRMS (FAB) m/z calcd for C29H33Cl2N3O7+H1 606.1774, found 606.1758.
  • Preparation 19-C-6 Scheme 19, 19-C: wherein R4=H, R5=CO2CH3 R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl, R19-2, R19-1=(CH2)3, R19-3=NHCH3, m=0 Stereochemistry=[1S-[1α,3α(R*)]]
  • [1S-[1α,3α(R*)]]-4-[(2,6-Dichlorobenzoyl)amino]-N-[3-[[2-[(methylamino)carbonyl]-1-pyrrolidinyl]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-L-phenylalanine methyl ester (19-C-6) (C[0358] 33H40Cl2N4O6)
  • In a similar manner to that reported for the synthesis of 19-C-1,19-A-1 (19-A: R[0359] 4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorobenzoyl)amino]-phenyl) (0.53 g, 0.96 mmol), HATU (0.4 g, 1.05 mmol), and diisopropylethyl amine (dry, 1.5 mL, 8.6 mmol) are stirred together with DMF (dry, 5 mL) in a dry round bottom flask under a nitrogen atmosphere. After 30 minutes, N-methylprolineamide hydrochloride (19-B: R19-1, R19-2=CH2 CH2CH2, R19-3=NHCH3, n=0, stereochemistry L) (0.5 g, 3.04 mmol) is added. After two days, the mixture is evaporated to dryness in vacuo to give a pale brown oil which is transferred to a silica gel column (20 g) and eluted with a gradient from 0 to 10% methanol/chloroform to give, after solvent evaporation, 19-C-6 as a pale oil (0.55 g, 86%)
  • [0360] 1H NMR (300 MHz, DMSO-d6) δ 10.68 (1H), 7.95 (1H), 7.58-7.44 (7H), 7.19 (2H), 4.48 (1H), 4.11 (2H), 3.57 (3H), 3.06 (4H), 2.63 (3H), 2.46-1.69 (11H), 1.31 (3H), 1.12 (3H), 0.79 (3H).
  • Preparation of Example 215 Scheme 19, 19-D: wherein R4=H, R5=CO2Na R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl, R19-1, R19-2 CH;CH2CH2, R19-3=NHCH3, m=0 Stereochemistry [1S-[1α,3α(R*)]]
  • [1S-[1α,3α(R*)]]-N-[3-[[2-[(Methylamino)carbonyl]-1-pyrrolidinyl]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine monosodium salt Example 215 (C[0361] 32H37Cl2NaN4O6)
  • A solution of LiOH.H[0362] 2O (0.2 g, 4.8 mmol) in H2O (6 mL) is added to a stirred solution of the methyl ester 19-C-6 (19-C: R4=H, R5=CO2CH3, R6=4-[2,6-Dichlorobenzoyl)amino]phenyl) (0.53 g, 0.8 mmol) in methanol (10 mL). After overnight stirring, the mixture is diluted with H2O (50 mL), evaporated in vacuo until the methanol is gone. The reaction mixture is then brought to pH 8 using 1N aq. HCl, filtered, and the filtrate transferred to a C-18 reversed phase HPLC column and eluted with a 10′-18 acetonitrile/0.02% aq sodium bIcarbonate gradient. The eluant is evaporated to dryness, stirred with isopropanol (3×10 mL) and filtered through a sintered glass funnel. The combined isopropanol filtrates are evaporated to dryness, mixed with water and evaporated to dryness again (2×10 mL H2O) to give Example 215 as a white solid (0.4 g, 74% yield).
  • [0363] 1H NMR (300 MHz, DMSO-d6) δ 10.57 (1H), 7.51 (6H), 7.04 (2H), 6.80 (1H), 4.18 (1H), 3.90 (1H), 3.55 (2H), 3.00 (2H), 2.53 (3H), 2.47 (1H), 2.12-1.50 (8H), 1.24 (3H), 1.09 (3H), 0.72 (3H).
  • HRMS (FAB) m/z calcd for C[0364] 32H37CL2N4O6NaI +HI 667.2066; found 667.2056.
    Figure US20030130349A1-20030710-C00083
  • (1R-cis)-1,2,2-Trimethylcyclopentane-1,3-dicarboxylic acid 1-methyl ester (20-B) [0365]
  • A solution of (1R)-camphoric anhydride, prepared by the method of Bell, K. H. [0366] Aust. J. Chem. 1981, 34 665-670, (5 g, 27.44 mmol) in toluene (400 mL) in a round bottom flask is flushed with N, and then cooled in a dry ice/ethanol bath. To this cooled solution is added (via dropwise addition) a one molar solution of potassium t-butoxide (30 mL, 30 mmol) over a twenty minute period. After stirring overnight, the room temperature reaction mixture is again cooled on a dry ice/ethanol bath. To the cooled solution is added methyl trifluoromethanesulfonate (3.5 mL, 30.9 mmol). After an additional 12 hours of stirring, the reaction mixture is acidified with trifluoroacetic acid (50 mL, 649 mmol), and is allowed to stir for another 16 hours. The mixture is then diluted with toluene (200 mL), shaken with water (4×120 mL), and evaporated to dryness, giving 20-B as a pale brown solid (2.35 g, 10.97 mmol, 40% yield).
  • [0367] 1H NMR (300 MHz, DMSO-d6) δ 3.60 (s, 3H), 2.75 (m, 1H) 2.40 (m, 1H), 2.00 (m, 1H), 1.74 (m, 1H), 1.41 (m, 1H), 1.19 (s, 3H), 1.15 (s, 3H), 0.70 (s, 3H).
  • Preparation 20-D Scheme 20, 20-D: wherein R4=H, R5=CO2CH3 R6=4-[(2,6-Dichlorophenyl) methoxy]-phenyl Stereochemistry=[1S-cis]-L
  • (1S-cis)-O-[(2,6-Dichlorophenyl)methyl]-N-[[3-(methoxycarbonyl)-2,2,3-trimethylcyclopentyl]-carbonyl]-L-tyrosine methyl ester (20-D) [0368]
  • A solution of camphoric acid methyl ester 20-B (0.35 g, 1.63 mmol), EDC (0.35 g, 1.83 mmol), HOBT (0.25 g, 1.85 mmol), 4-dimethylaminopyridine (0.05 g, 0.41 mmol), in methylene chloride (10 mL) is stirred together in a 25 mL, 2-neck flask cooled in an ice water bath. To this mixture is added (2,6-dichlorophenyl)methyl)-L-tyrosine methyl ester hydrochloride (0.65 g, 1.83 mmol), creating a thick, heterogeneous mixture which becomes homogeneous after addition of triethylamine (0.3 mL, 2.15 mmol). After three days, the reaction mixture is diluted with methylene chloride (150 mL) and shaken with water (2×100 mL), aqueous HCl (0.5N, 2×100 mL), water (3×100 mL), aqueous NaHCO[0369] 3 (2×100 mL) and water (1×100 mL). The organic layer is then evaporated to dryness, giving 20-D as an off-white foam (0.7 g, 78% yield).
  • [0370] 1H NMR (300 MHz, CDCl3) δ 7.38 (m, 2H), 7.26 (m, 1H), 7.05 (m, 2H), 6.96 (m, 2H), 5.76 (m, 1H), 5.25 (s, 2H), 4.88 (m, 1H), 3.75 (s, 3H), 3.67 (s, 3H), 3.17-3.03 (m, 2H), 2.61-2.50 (m, 2H), 2.26-2.13 (m, 1H), 1.84-1.70 (m, 1H), 1.54-1.45 (m, 1H), 1.22 (s, 3H), 1.19 (s, 31H), 0.76 (s, 3H), MS (ES+) m/z 549.8
    Figure US20030130349A1-20030710-C00084
  • (1R-cis)-N-[[3-Methoxycarbonyl-1,2,2-trimethylcyclopentyl]carbonyl-L-proline 1,1-dimethylethyl ester 21-A [0371]
  • The camphoric acid mono ester 10-A (1.41 g, 6.58 mmol) in DMF (dry, 5 mL) in a dry flask under N2 is cooled in an ice water bath. Diisopropylethyl amine (4.6 mL, 26.41 mmol) is added followed by HATU (2.6 g, 6.83 mmol). After thirty minutes, L-proline t-butyl ester (1.3 mL, 7.44 mmol) is added. After overnight stirring, the mixture is evaporated to dryness, to give a pale yellow oil. Recrystallization from CHCl[0372] 3 to give 21-A as a white solid (1.6 g, 66% yield). 1H NMR (300 MHz, CDCl3) δ 4.32 (1H), 3.67 (3H), 3.59 (2H), 2.71 (1H), 2.39 (1H), 2.20 (2H), 2.10-1.70 (5H), 1.43 (9H), 1.38 (3H)l 1.20 (3H), 0.93 (3H); IR (nujol) 1741, 1733, 1617, 1430, 1396, 1359, 1343, 1218, 1202, 1185, 1174, 1157, 1127, 1014, 771 cm−1; MS (FAB) m/z (rel. intensity) 368 (M+H, 99), 369 (22), 368 (99), 312 (15), 280 (12), 197 (72), 169 (23), 137 (29), 109 (38), 70 (11), 57 (14). Anal. Calcd for C20H33NO5: C, 65.37; H, 9.05; N, 3.81. Found: C, 65.53; H, 8.88; N, 3.83.
  • (1R-cis)-[3-Carboxy-1,2,2-trimethylcyclopentyl]carbonyl-L-proline 1,1-dimethylethyl ester 21-B [0373]
  • To a solution of the diester 21-A (0.74 g, 2.01 mmol) in methanol (5 mL) is added a solution of LiOH.H[0374] 2O (0.15 g, 3.62 mmol) in aq H2O2 (306, 2 mL) and water (5 mL). After overnight stirring, the mixture is evaporated until all of the methanol is gone, and then cooled in an ice water bath and brought to pH 5 using 1N aq. HCl. The resultant white precipitate is isolated by suction filtration (with water washes, 3×30 mL) to give 21-B as a white solid (0.45 g, 63% yield).
  • [0375] 1H NMR (300 MHz, CDCl3) δ 4.34 (1H), 3.60 (2H), 2.75 (1H) 2.41 (1H), 2.18 (1H), 2.00-1.70 (5H), 1.44 (9H), 1.43 (3H), 1.22 (3H), 1.02 (3H).
  • [1R-[1α,3α(S*)]]-N-[[3-[[[1-Carbomethoxy-2-[4-[(2,6-dichlorophenyl)methoxy]phenyl]ethyl]amino]carbonyl]-1,2,2-trimethylcyclopentyl]carbonyl]-L-proline 1,1-dimethylethyl ester 21-C [0376]
  • A solution of the acid 21-B (0.41 g, 1.16 mmol) in methylene chloride (20 mL) under N[0377] 2 is cooled in an ice water bath. To this is added diisopropylethyl amine (2 mL), EDC (0.26 g, 1.36 mmol), HOBT (0.19 g, 1.41 mmol) and dimethylaminopyridine (0.02 g, 0.16 mmol). Forty minutes later, 7-E-2 (7-E. R4=H, R5=CO2CH3, R6=0.4-[(2,6-Dichlorophenyl)methoxy)phenyl) (0.45 g, 1.27 mmol) is added in one portion. After three days, the mixture is evaporated to dryness, giving a colorless oil which is mixed with THF (100 mL) and water (50 mL) and then shaken sequentially with water (2×50 mL), aq. HCl (0.5N, 4×30 mL), satd. aq. NaHCO3 (2×50 mL), and water (2×50 mL). The organic layer is then evaporated to dryness, giving a colorless oil (0.91 g) which is chromatographed on silica gel with 10% methanol/chloroform to give 21-C as a white, foamy solid (0.57 g, 70% yield).
  • [0378] 1H NMR (300 MHz, CDCl3) δ 7.31-6.87 (7H), 5.80 (1H) 5.18 (2H), 4.81 (1H), 4.25 (1H), 3.67 (3H), 3.53 (2H), 3.04 (2H), 2.44-1.58 (9H), 1.37 (9H), 1.33 (3H), 1.13 (3H), 0.90 (3H). IR (nujol) 1739, 1658, 1622, 1612, 1585, 1511, 1439, 1298, 1241, 1204, 1177, 1153, 1122, 1017, 768 cm−1; MS (FAB) m/z (rel. intensity) 689 (M+H, 24), 689 (24), 520 (69), 519 (32), 518 (99), 336 (44), 280 (21), 161 (24), 159 (40), 109 (75), 57 (22).
  • Preparation of Example 200
  • [1R-[1α,3α(S*)]]-N-[[3-[[[1-Carboxy-2-[4-[(2,6-dichlorophenyl)methoxy]phenyl]ethyl]amino]carbonyl]-1,2,2-trimethylcyclopentyl]carbonyl]-L-proline 1,1-dimethylethylester, monosodium salt (Example 200) [0379]
  • To a solution of 21-C (0.43 g, 0.62 mmol) in methanol is added a solution of LiOH.H[0380] 2O (0.083 g, 1.98 mmol) in aqueous H2O2 (30%, 3 mL) plus H2O (3 mL) After overnight stirring, the mixture is diluted with water (50 mL) and evaporated in vacuo until the methanol is gone. The aqueous layer is then washed with diethyl ether (3×30 mL) and brought to pH 6 using 1 N aq. HCl. The resultant white precipitate is isolated by suction filtration to give a white solid (0.4 g). This is stirred overnight with NaHCO3 (0.1 g, 1.2 mmol) in H2O (5 mL). The aqueous solution is brought to pH 7-8 using 1 N HCl and then transferred to a C-18 HPLC column and eluted with a gradient of 0-12% acetonitrile/aq. Na2CO3 (0.02%). Evaporation is accomplished in vacuo to give [1R-[1α,3α(S*)]]-N-r[3-[[[1-Carboxy-2-[4-[(2,6-dichlorophenyl)methoxy] phenyl]ethyl]amino]carbonyl]-1,2,2-trimethylcyclopentyl]carbonyl]-L-proline 1,1-dimethylethylester, monosodium salt as a white solid (0.2 g, 460%)
  • [0381] 1H NMR (DMSO-d6) δ 7.55-7.41 (3H), 7.01 (2H), 6.82 (2H), 6.80 (1H), 5.13 (2H), 4.08 (1H), 3.90 (1H), 3.65 (1H), 3.40 (1H), 3.31 (3H), 2.95 (2H), 2.44 (1H), 2.25-1.5 (8H), 1.34 (9H), 1.23 (3H), 1.08 (3H), 0.76 (3H), IR (nujol) 3405, 1735, 1610, 1565, 1511, 1439, 1299, 1240, 1195, 1175, 1153, 1093, 1018, 779, 769 cm−1; MS (FAB) m/z (rel. intensity) 675 (M+H, O), 720 (27), 701 (25), 700 (67), 699 (39), 698 (99), 336 (32), 280 (23), 159 (20), 109 (53), 23 (28). HRMS (FAB) m/z calcd for C35H4Cl2N2O7 +Na 697.2424, found 697.2418.
    Figure US20030130349A1-20030710-C00085
  • Preparation 22-C-1 Scheme 22, 22-C: wherein R4=H R5=CO2CH3 R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl, Stereochemistry=(1S-cis)-L
  • (1S-cis)-N-[[3-[[(2-Cyanoethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester (22-C-1) (C[0382] 30H34Cl2N4O5)
  • A solution of 19-A-1 (19-A: R[0383] 4=Hi R5=CO2CH3, R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl) (0.5 g, 0.91 mmol), HOBT (0.12 g, 0.91 mmol), and DCC (0.18 g, 0.91 mmol are stirred together in CH2Cl2 (dry, 10 mL) in a dry round bottom flask under a nitrogen atmosphere. After 30 minutes, 2-cyanoethyl amine 22-B (0.064 g, 0.91 mmol) is added. After 24 hours a precipitate is observed and methanol (5 mL) is added to achieve a homogenous solution and an additional portion of 2-cyanoethyl amine 22-B (0.064 g, 0.91 mmol) is added. After an additional 8 days of stirring the solvent is removed in vacuo and the residue is dissolved in THF and purified by chromatography on a column of silica gel to give 22-C-1 (0.42 g, 78%) as a white solid.
  • [0384] 1H-NMR (300 MHz, MeOH-d4): δ 7.58 (2H), 7.43 (3H) 7.22 (2H), 5.48 (1H), 4.72 (1H), 3.69 (3H), 3.58 (2H), 3.29 (1H), 3.02 (1H), 2.65-2.77 (3H), 2.40 (1H), 2.04 (1H), 1.76 (1H), 1.47 (1H), 1.27 (3H), 1.20 (3H), 0.74 (3H); Anal. Calcd for C30H34Cl2N4O7-0.54H2O: C, 58.95; H, 5.78; N, 9.17; Found: C, 59.04; H, 5.75; N, 9.22; KF Water 1.59%.
  • Preparation of Example 216 Scheme 22, 22-D: wherein R4=H R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl, Stereochemistry=(1S-cis)-L
  • (1S-cis)-N-[[3-[[(2-Cyanoethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (Example 216) (C[0385] 29H32Cl2N4O5)
  • To the L-phenylalanine methyl ester 22-C-1 (22-C: R[0386] 4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl) (0.16 g, 0.266 mmol), in methanol (10 mL) is added a mixture of LiOH.H2O (0.056 g, 1.33 mmol), in H2O (3 mL). The mixture is allowed to stir at room temperature for 2 hours, then the solvent is removed in vacuo. The resulting solid is dissolved in water (10 mL) and the pH is adjusted to ca. 2 with 1N aq. HCl to give a white precipitate the precipitate is isolated by filtration, washed with water (10 mL) and then dissolved in acetonitrile (25 mL). The organic phase is dried (Na2SO4) and the solvent is removed in vacuo to give a sticky solid which is dissolved in acetonitrile/water (25 mL, 1:3) and lyophilized to give 0.104 g (67%) of the target compound as a white solid.
  • [0387] 1H-NMR (300 MHz, MeOH-d4): δ 7.82 (1H), 7.57 (2H), 7.46 (3H), 7.24 (2H), 4.72 (1H), 3.40 (2H), 3.30 (1H), 2.99 (1H), 2.63-2.77 (3H), 2.40 (1H), 2.03 (1H), 1.74 (1H), 1.48 (1H), 1.27 (3H), 1.20 (3H), 0.75 (3H); IR (nujol): 3317, 3262, 1762, 1673, 1638, 1608, 1546, 1515, 1432, 1325, 1203, 811, 801, 780 cm−1; MS (FAB) m/z (rel. intensity) 587 (M+, base), 517 (12.4), 335 (9.8), 252 (8.5), 235 (80); Arial. Calcd for C29H32Cl2N4O7.2.14H20 : C, 55.64; H, 5.84; N, 8.95; Found: C, 55.74; H. 5.72; N, 8.99; KF Water 6.16%.
    Figure US20030130349A1-20030710-C00086
  • Preparation 23-C-1 Scheme 23, 23-C: wherein R4=H, R5=CO2CH3 R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl, Stereochemistry=(1S-cis)-L
  • (1S-cis)-4-[(2,6-Dichlorobenzoyl)amino]-N-[[2,2,3-trimethyl-3-[[(phenylmethoxy)amino]carbonyl]cyclopentyl]carbonyl]-L-phenylalanine methyl ester (23-C-1) (C[0388] 34H37Cl2N3O6).
  • A solution of 19-A-1 (19-A: R[0389] 4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl) (0.5 g, 0.91 mmol), HOBT (0.14 g, 1.04 mmol), EDC (0.19 g, 1.0 mmol), triethylamine (0.46 mL, 3.28 mmol), and DMAP (0.012 g, 0.1 mmol) are stirred together in CH2Cl2 (dry, 15 mL) in a dry round bottom flask under a nitrogen atmosphere. Stir for 30 minutes at room temperature and benzyloxyamine-HCl 23-B (0.26 g, 1.64 mmol) is added in one portion. The resulting mixture is allowed to stir for 72 hours at room temperature and the solvent is removed in vacuo. The residue is dissolved in CHCl3 (50 mL) and the solution is washed with 1N aq. HCl (50 mL), saturated aq. NaHCO3 (50 mL), and the organic layer is dried (MgSO4). Concentration in vacuo gives the crude product as a sticky oil which is purified by flash chromatography on a column of silica gel (5% MeOH, 956 CH2Cl2) to give 0.27 g (45%) of 23-C-1 (23-C: R4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl) as a white solid.
  • [0390] 1H-NMR (300 MHz, CDCl3): δ 8.13 (1H), 7.57 (2H), 7.49 (1H), 7.35 (6H), 7.10 (2H), 5.78 (1H), 4.89 (2H), 3.76 (3H), 3.14 (2H), 2.47 (1H), 2.18 (2H), 1.30-1.90 (4H), 1.27 (3H), 1.15 (3H), 0.80 (3H); Anal. Calcd for C34H37Cl2N3O6.0.24H2O: C, 61.97; H, 5.73; N, 6.38; Found: C, 62.02; H, 5.75; N, 6.39; KF Water 0.66%.
  • Preparation of Example 217 Scheme 23, 23-D: wherein R4=H, R5=CO2H R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl, R23=Phenylmethyl Stereochemistry=(1S-cis)-L
  • (1S-cis)-4-[(2,6-Dichlorobenzoyl)amino]-N-[[2,2,3-trimethyl-3-[[(phenylmethoxy)amino]carbonyl]cyclopentyl]carbonyl-L-phenylalanine (Example 217) (C[0391] 33H35Cl2N3O6).
  • A solution of 23-C-1 (23-C: R[0392] 4=H, R5=CO2CH3, R6 4-[(2,6-Dichlorobenzoyl)amino]phenyl) (0.075 g, 0.11 mmol) in methanol (10 mL) is treated with a solution of LiOH—H2O (0.024 g, 0.57 mmol) in water (5 mL) over 5 minutes. The mixture is allowed to stir for 4 hours at room temperature, then the solvent is removed in vacuo. The crude residue is dissolved in water (10 mL), is filtered through a sintered glass funnel and then the solution is brought to ca. pH 4 by the addition of 1N aq. HCl. The resulting solid is isolated by suction filtration, washed with water (2×10 mL), and is then dissolved in acetonitrile-water (25 mL, 1:3). The solution is frozen and lyophylized to give Example 217 (23-D: R4=H, R5=CO2H, R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl, R23=phenylmethyl) (0.048 g, 68%) as a white solid.
  • [0393] 1H-NMR (300 MHz, MeOH-d4): δ 7.85 (1H), 7.57 (2H), 7.32-7.49 (8H), 7.24 (2H), 4.81 (2H), 4.70 (1H), 3.21 (1H), 2.98 (1H), 2.69 (1H), 2.24 (1H), 1.98 (1H), 1.72 (1H), 1.40 (1H), 1.25 (3H), 1.15 (3H), 0.71 (3H); IR (nujol): 3264, 3195, 3063, 3032, 1731, 1658, 1607, 1502, 1538, 1516, 1432, 1326, 1195, 800 cm−1; MS (ES+) m/z 640 (M+H+); Anal. Calcd for C33H35Cl2N3O6-1.19H2O: C, 59.87; H, 5.69; N, 6.35; Found: C, 59.70; H, 5.78; N, 6.37; KF Water 3.24%.
  • Preparation of Example 218 Scheme 23, 23-D: wherein R4=H, R5=CO2CH3 R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl, R23=H Stereochemistry (1S-cis)-L
  • (1S-cis)-4-[(2,6-Dichlorobenzoyl)amino]-N-[[3-[(hydroxyamino)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-L-phenylalanine methyl ester (Example 218) (C[0394] 27H31Cl2N3O6)
  • A solution of 23-C-1 (23-C: R[0395] 4=H R5=CO2CH3, R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl) (0.7 g, 1.07 mmol) in THF (120 mL) is hydrogenated over Pd(OH)2 (0.42 g) under 46 psi of hydrogen for 2.75 hours. The catalyst is removed by filtration through a pad of Celite®, the filter cake is rinsed with THF (75 mL) and the solvent is removed in vacuo to afford the crude product as a sticky solid. The crude material is purified by flash chromatography on a column of silica gel (EtOAc/HOAc, 99.9:0.1) to give Example 218 (23-D: R4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl, R23=H) (0.34 g, 56%) as a white solid.
  • [0396] 1H-NMR (300 MHz, MeOH-d4): δ 7.58 (2H), 7.31-7.49 (3H), 7.22 (2H), 4.72 (1H), 3.69 (3H), 3.18 (1H), 2.99 (1H), 2.73 (1H), 2.29 (1H), 2.02 (1H), 1.75 (1H), 1.42 (1H), 1.27 (3H), 1.18 (3H), 0.75 (3H); IR (nujol): 3313, 3292, 3245, 3194, 3129, 3073, 1749, 1668, 1653, 1606, 1547, 1517, 1459, 1434, 1336, 1211, 1021, 801, 779 cm−1; MS (FAB) m/z (rel. intensity): 564 (M++H, 71), 548 (3), 531 (base), 109 (95).
  • Preparation of Example 219 Scheme 23, 23-D: wherein R4=H, R5=CO2H R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl, R23=H Stereochemistry=(1S-cis)-L
  • (1S-cis)-4-[(2,6-Dichlorobenzoyl)amino]-N-[[3-[(hydroxyamino)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-L-phenylalanine (Example 219) (C[0397] 26H29Cl2N3O6)
  • A solution of Example 218 (0.14 g, 0.25 mmol) in methanol (8 mL) is treated with a solution of LiOH—H[0398] 2O (0.053 g, 1.27 mmol) in water (4 mL) over 15 minutes. The mixture is allowed to stir for 1.5 hours at room temperature, then the solvent is removed in vacuo. The residue is dissolved in water (25 mL), the pH is, adjusted to ca. 4 with 1N aq. HCl, and the mixture is extracted with ethyl acetate (3×25 mL). The combined organic extracts are dried (MgSO4), and concentrated in vacuo to give the crude material as a sticky solid. The crude product is dissolved in acetonitrile/water (25 mL, 1:3), and the solution is frozen and lyophylized to provide Example 219 (23-D: R4=H, R5=CO2H, R6=4-[(2,6-Dichlorobenzoyl)amino]-phenyl, R23=H) (0.098 g, 71%) as a beige solid.
  • [0399] 1H-NMR (300 MHz, MeOH-d4): δ 7.57 (2H), 7.40-7.47 (3H) 7.24 (2H), 4.71 (1H), 3.20 (1H), 2.99 (1H), 2.69 (1H), 2.29 (1H), 2.03 (1H), 1.75 (1H), 1.43 (1H), 1.28 (3H), 1.18 (3H), 0.76 (3H); IR (nujol): 3262, 3197, 3127, 3070, 1725, 1657, 1607, 1584, 1562, 1535, 1516, 1432, 1326, 1234, 1194, 800, 781 cm−1; MS (FAB) m/z (rel. intensity): 550 (M++H, 70), 517 (75), 198 (base); Anal. Calcd for C26H29Cl2N3O6-0.95H2O: C, 55.02; H, 5.49; N, 7.40; Found: C, 55.29; H, 5.93; N, 7.26; KF Water 3.020.
    Figure US20030130349A1-20030710-C00087
  • Boc-Tryptophan-O-methyl ester 24-A (636 mg, 2.00 mmol, 1 eq) was dissolved in dry DMF. To this solution NaH (88 mg, 2.20 mmol, 1.1 eq) was added with evolution of H[0400] 2. To this mixture benzyl bromide (285 μL, 2.40 mmol, 1.2 eq) was added and the reaction stirred for 3 hours at room temperature. The reaction was quenched with brine (15 mL) and extracted with Et2O (3×15 mL). The combined organics were dried (Na2SO4), filtered and the solvent removed in vacuo. The residue was purified by column chromatography (SiO2, Hexanes to 30% EtOAc/Hexanes gradient elution) to provide 426 mg (52 W) of the benzyl indole 24-B.
    Figure US20030130349A1-20030710-C00088
  • The final compound Example 196 was produced as described in Example 2. ESMS (m/z): 475 (M−H)[0401] .
    Figure US20030130349A1-20030710-C00089
  • The preparation of Example 190 is as follows. Boc Tyr(2,6-dichlorobenzyl)-OH (25-A) (1.31 gm, 2.97 mmol) was dissolved in THF (5 mL) and cooled to −78° C. under dry N[0402] 2. BH3 THF (5.9 mL, 5.9 mmol, 1N) was added and the reaction warmed slowly to room temperature with stirring for 3 hours. The reaction was cooled to 0° C. and quenched with H2O (1 mL) and warmed to room temperature. After the addition of 1 N HCl (25 mL), the mixture was extracted with EtOAc (3×25 ml) and the combined organic phases were dried over Na2SO4. The solution was filtered, solvent evaporated and the residue chromatographed (SiO2, gradient elution: 100% hexanes→50% EtO Ac/hexanes) to provide the intermediate 25-B (665 mg, 53%): ESMS (m/z) 448 (M+Na)+.
  • The above compound, (25-B) (270 mg, 0.634 mmol), was dissolved dry DMF (5 mL) containing methyl iodide (51 μl, 0.824 mmol). To this solution was added NaH (28 mg, 0.697 mmol: in 60% oil) and the mixture was stirred for 5 minutes. The reaction was quenched with the addition of H[0403] 2O (1 mL) followed by 1N HCl (10 ml). The mixture was extracted with EtOAc (3×15 mL) and the combined organics were dried over Na2SO4. The solution was filtered, solvent evaporated and the residue chromatographed (SiO2, gradient elution: 100% hexanes→25% EtOAc/hexanes) to provide intermediate 25-C (130 mg, 47%): ESMS (m/z)462 (M+Na)+.
  • 25-C (115 mg, 0261 mmol) was dissolved in 3 N HCl/EtOAc (3 mL) and stirred for 1 h. The solvent was removed under reduced pressure and dried thoroughly under high vacuum. The residue was dissolved in THF (5 mL) and DIEA (228 μL, 1.31 mmol) and (1R)-camphoric anhydride (57 mg, 0.314 mmol) was added. The reaction was warmed to 60° C. with stirring for 48 h. After cooling to room temperature, 1N HCl (15 mL) was added and the mixture extracted with EtOAc (3×15 mL). The combined organics were dried over Na[0404] 2SO4, filtered and the solvent removed under reduced pressure. The residue was then chromatographed (SiO2, gradient elution: 100% hexanes→100% EtOAc) to provide Example 190 (113 mg. 83%): ESMS (m/z) 422 (MH+).
    Figure US20030130349A1-20030710-C00090
  • Example 56 (0.27 gm, 0.62 mmol) was dissolved in CH[0405] 2Cl2 (10 mL) and 2,6-dichlorophenylisocyanate (0.18 gm, 0.94 mmol) and DIEA (327 μL, 1.86 mmol) was added and the reaction stirred overnight. After the addition of 1 N HCl (20 mL), the mixture was extracted with EtOAc (3×25 mL) and the combined organic phases were dried over Na2SO4. The solution was filtered, solvent evaporated and the residue chromatographed (SiO2, gradient elution: 100% hexanes→33% EtOAc/hexanes) to provide Example 227 (310 mg, 82%): ESMS (m/z) 620 (MH+).
  • Example 227 (250 mg, 0.40 mmol) was dissolved in CH[0406] 2Cl2 (1.5 mL) and TFA (1.5 mL). After 1 h, the solvent was removed and the residue triturated with Et2O (3×5 mL) to form a gum. The residue was purified by column chromatography (SiO2, gradient elution: 100% hexanes 25% acetone/hexanes) to provide Example 228 (170 mg, 73%): ESMS (m/z) 564 (MH+).
  • Example 228 (130 mg, 0.23 mmol) was dissolved in THF/CH[0407] 3OH (5 mL/mL, respectively) and LiOH (22 mg, 0.53 mmol) was added in H2O (1 mL). After 2 h the solvent was evaporated and the residue dissolved in H2O (3 mL). The solution was precipitated with the addition of 1 N HCl (2 mL). The solvent was collected by vacuum filtration and washed with cold H2O (2×2 mL). The solid material was then thoroughly dried under high vacuum to afford Example 229 (80 mg, 64%) as a white solid: ESMS (m/z) 550 (MH+).
    Figure US20030130349A1-20030710-C00091
  • Scheme 27 is as follows: [0408]
  • Example 54 (387 mg, 0.704 mmol) and morpholine (0.14 mL, 1.55 mmol) were dissolved in CH[0409] 2Cl2 (15 mL). This solution was treated with BOP-Cl (215 mg, 0.845 mmol) and stirred under dry N2 at room temperature. After 18 h the reaction was treated with 1 N HCl (10 mL) and extracted with CH2Cl2 (3×15 mL). The combined organics were dried over Na2SO4, filtered and the solvent evaporated. The residue was then purified by column chromatography (SiO2, gradient elution: 100% hexanes→100% EtOAc) to provide Example 226 (160 mg, 37%) as a colorless oil: ESMS (m/z) 618 (M+Na)+.
  • The methyl ester (Example 226) (160 mg, 0.258 mmol), was dissolved in THF (5 mL) and LiOH (12 mg, 0.52 mmol) was added in H[0410] 2O (5 mL). After 4 h 1 N HCl (3 mL) was added and the precipitate collected by vacuum filtration washing with cooled H2O (3×3 mL) The product was thoroughly dried under high vacuum to provide Example 225 (148 mg, 95%) as an amorphous powder: ESMS (m/z) 602 (M−H).
    Figure US20030130349A1-20030710-C00092
  • Intermediate used in solid phase synthesis [0411]
  • Intermediate Urea. [0412]
  • Camphoric acid methyl ester (10-A) (2.15 g, 11.65 mmol) was dissolved in THF (25 mL). To this solution was added DPPA (diphenylphosphoryl azide) (3.33 g, 12.1 mmol) and DIEA (1.73 g, 13.4 mmol). The reaction was warmed at 45° C. with stirring. After 2.5 h tert-butyl alcohol was added and heated at 85° C. for an additional 2.5 h. The reaction was worked-up by removing the volatile components under reduced pressure. The residue was then purified by flash chromatography (SiO[0413] 2, gradient elution: 2% EtOAc/hexanes→20% EtOAc/hexanes) to provide the symmetrical urea diester (28-A) (2.0 g, 43%): ESMS (m/z) 397 (MH+).
  • The intermediate ester (28-A) (2 g, 5.0 mmol) was dissolved in THF/CH[0414] 3OH (5 mL/2 mL, respectively) and LiOH (490 mg, 11.6 mmol) was added in H2O (1 mL). After 2 h the solvent was evaporated, and the residue dissolved in H2O (5 mL). The solution was precipitated with the addition of 1 N HCl (1.5 mL). The solvent was collected by vacuum filtration and washed with cold H2O (2×2 mL). The solid material was then thoroughly dried under high vacuum to afford the intermediate symmetrical urea diacid (28-B) (1.6 g, 89%) as a white solid: ESMS (m/z) 369 (MH+).
    Figure US20030130349A1-20030710-C00093
  • To a solution of N-(tert-butoxycarbonyl)-4-(2,6-dichlorobenzamido)-L-phenylalanine (29-A) (9.25 g, 20.3 mmol) in DMF (100 mL) was added Merrifield resin (10.0 g, 10.0 meq/g) and anhydrous potassium fluoride (1.57 g, 20.0 mmol). The reaction mixture was stirred for 1 day at 80° C. and the resulting resin bound amino acid was collected by filtration, washed sequentially with DMF (2×200 mL), 50% aqueous DMF (3×200 mL), CH[0415] 3OH (3×300 mL), CH2Cl2 (3×300 mL) and CH3OH (3×100 mL) then dried in vacuo to provide the resin bound N-(tert-butoxycarbonyl)-4-(2,6-dichlorobenzamido)-L-phenylalanine (29-B) (0.53 meq/g). Substitution of the Boc(L)Phe[4-(2,6-dichlorobenzamido)]-OH onto the resin was estimated using the picric acid method.
  • To the obtained resin (29-B) (150 mg, 0.107 mmol) was added 50% TFA/CH[0416] 2Cl2 (3 mL) and the mixture was shaken for 30 min. The resin was collected by filtration, washed sequentially with CH2Cl2 (2×10 mL), CH3OH (2×10 mL), and CH2Cl2 (2×10 mL). To the washed resin was added the symmetrical urea diacid (28-B) (118 mg, 0.320 mmol), 0.5 M DMF solution of HBTU-HOBT (0.70 mL, 0.320 mmol), DIEA (0.139 mL, 0.799 mmol) and DMF (3.0 mL) and the mixture was vortexed for 2 hrs. at room temperature. The resin was collected by filtration, washed sequentially with DMF (2×10 mL), CH2Cl2 (2 10 mL), CH3OH (2×10 mL), CH2Cl2 (2×10 mL). To the resin bound substrate was added THF (1.6 mL), CH3OH (0.5 mL) and 2N LiOH (0.310 mL) and the mixture was shaken for 15 mins. The supernatant was collected by filtration and the resin washed with THF/5% CH3OH (2×2 mL) and the combined filtrate was evaporated on a Pierce block evaporator. The concentrate was diluted with H2O (1 mL) and the aqueous solution acidified with 1N HCl (1.5 mL). The precipitate was collected by centrifugation and the solid washed with H2O (3×3 mL). The solid material was dried under high vacuum to furnish Example 236 (25 mg, 33%): ESMS (m/z) 701 (M−H).
    Figure US20030130349A1-20030710-C00094
  • Scheme 2, III-a: wherein R4=H, R5a=—CH2CO2Et, R6=4-[(2,6-Dichlorophenyl)methoxy]-phenyl-Stereochemistry=(S)
  • (Intermediate for Examples 201 and 204) [0417]
  • (S)-4-[(2,6-Dichlorophenyl)methoxy]-β-[(1,1-Dimethylethoxy)carbonyl]amino]benzenebutanoic acid ethyl ester (30-A) (C[0418] 24H29Cl2NO5).
  • To a mixture of Boc-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine (25-A) (5.0 g, 11.36 mmol) and N-methylmorpholine in dry Et[0419] 2O at −10° C. under Ar is added isobutyl chloroformate (1.49 mL, 11.36 mmol). The reaction is warmed to room temperature, stirred for 1 h, and filtered. The filtrate is reacted at 0° C. with an excess of ethereal CH2N2. The solution is stirred for 1 h at 0° C., and then is concentrated. The residue is dissolved in absolute EtOH, and a solution of C6H5CO2Ag (2.86 g, 12.38 mmol) in Et3N (14 mL) is added slowly. The resulting mixture is stirred for 1 h at room temperature under Ar and filtered. The filtrate is concentrated to a dark brown paste. The product is purified by silica flash chromatography (9:1 and 8:2 hexanes/EtOAc), from which is isolated 2.95 g (6.09 mmol, 54%) of 30-A: TLC Rf=0.32 (7:3 hexanes/EtOAc); [α]D (C=0.9, CHCl3)=−2°; IR (mull) 3360, 2984, 2954, 2925, 2869, 2855, 1721, 1678, 1585, 1524, 1510, 1467, 1447, 1441, 1378, 1373, 1299, 1263, 1251, 1236, 1197, 1177, 1163, 1020, 1016, 783 cm−1; 1H NMR δ 1.27 (3H), 1.41 (9H), 2.38-2.57 (2H), 2.73-2.96 (2H), 4.10-4.20 (3H), 7.22-7.26 (1H), 7.37 (2H); MS (FAB) m/z 482, 426, 382, 364, 348, 338, 319, 294, 268, 216, 159, 133, 116, 107, 57; Anal. C, 59.67; H, 6.09; Cl, 14.59; N, 3.03 (calcd C, 59.75; H, 6.06; Cl, 14.70; N, 2.90).
  • (S)-4-[(2,6-Dichlorophenyl)methoxy]-β-aminobenzenebutanoic acid ethyl ester (C[0420] 19H21C2NO3). A solution of the Boc-aminoester (30-A) (0.74 g, 1.53 mmol) in 1:1 CH2Cl2/TFA at 0° C. under Ar is stirred for 30 min at 0° C. and for 1.5 h at room temperature. It is concentrated, azeotroped thrice wish toluene, and dried to give the aminoester (30-B) as a solid: TLC Rf=0.15 (EtOAc); 1H NMR (CHCl3) δ 1.24 (3H), 2.63-2.73 (2H), 2.77-2.92 (1H), 3.07-3.23 (1H), 3.64-3.82 (1H), 4.15 (2H), 5.23 (2H), 6.97 (2H), 7.13 (2H), 7.16-7.25 (1H), 7.36 (2H), 8.16 (2H); MS (FAB) m/z 382, 365, 348, 294, 268, 224, 159, 133, 116, 70.
    Figure US20030130349A1-20030710-C00095
  • Scheme 2, III-a: wherein R[0421] 4=(CH3)3CO]C(O)—, R5a=-CO2Me, R6=4-aminophenyl-, Stereochemistry=L
  • (Intermediate for Examples 208, 209, 210) [0422]
  • N-[(1,1-Dimethylethoxy)carbonyl]-4-nitro-L-phenylalanine methyl ester (31-B) (C[0423] 15H20N2O6) A solution of N-Boc-4-nitrophenylalanine (31-A) (25.2 g, 81.28 mmol) and DMAP (0.82 g, 6.7 mmol) in dry DMF is cooled to 0° C. under Ar, and treated with MeOH (7.55 mL, 186 mmol) and DCC (18.975 g, 91.04 mmol). The reaction mixture is stirred overnight at room temperature and filtered. The filtrate is washed with satd NaHCO3 and brine. The aqueous washes are back-extracted with CH2Cl2. The organics are dried, filtered and concentrated to a yellow solid. This product is purified by silica flash chromatography (3:1 hexanes/EtOAc) to give 24.6 g (75.85 mmol, 93%) of 31-B: TLC Rf=0.36 (7:3 hexanes/EtOAc); 1H NMR (CHCl3) δ 1.41 (9H), 3.12 (1H), 3.28 (1H), 3.73 (3H), 4.63 (1H), 5.05 (1H), 7.31 (2H), 8.16 (2H); 13C NMR δ 28.25, 38.38, 52.56, 54.08, 80.35, 123.67, 130.25, 144.03, 147.12, 154.90, 171.64.
  • 4-Amino-N-[(1,1-dimethylethoxy)carbonyl]-L-phenylalanine methyl ester (31-C) (C[0424] 15H22N2O4). A solution of the above product (2.87 g, 8.85 mmol) in MeOH is treated at room temperature under N2 with 100 Pd/C (0.190 g), and hydrogenated at 40 psi for 3.5 h. The reaction mixture is filtered, and the filtrate is concentrated to give (31-C) as a dark foam: TLC Rf=0.34 (1:1 EtOAc/hexane); 1H NMR (CHCl3) δ 1.42 (9H), 2.97 (2H), 3.48 (2H), 3.70 (3H), 4.51 (1H), 4.93 (1H), 5.05 (1H), 6.61 (2H), 6.90 (2H); MS (EI) m/z 294, 238, 221, 207, 193, 177, 161, 135, 118, 106, 91, 77, 57.
    Figure US20030130349A1-20030710-C00096
  • Scheme 2, III-a: wherein R4=H.HCl, R5a=—CO2CH3, R6=4-benzoylphenyl-, Stereochemistry=L
  • (Intermediate to Examples 40, 191 and 197) [0425]
  • 4-Benzoyl-L-phenylalanine methyl ester, HCl salt (32-B) (C[0426] 17H17NO3.HCl). To cold MeOH (100 mL) under N2 is added AcCl (10 mL). The solution is stirred at room temperature for 30 min. 4-Benzoyl-L-phenylalanine (32-A) (0.99 g, 3.7 mmol) is treated with the methanolic HCl solution (60 mL) at room temperature for 26 h. The reaction mixture is concentrated to give 1.05 g of the aminoester.HCl salt (32-B) as a solid: TLC (UV) Rf=0.40 (95:5 CHCl3MeOH); HPLC tR=3.0 min (isocratic 650:350:1 CH3CN/H2O/TFA); 1H NMR (CD3OD) δ 7.80-7.70 (4H), 7.68-7.60 (1H), 7.55-7.41 (4H); 4.84 (2H), 4.44 (1H), 3.82 (3H), 3.44-3.27 (2H); 13C NMR (CD3OD) δ 196.66, 168.86, 139.22, 137.29, 136.91, 132.59, 130.34, 129.62, 129.35, 128.23, 53.56, 52.39, 35.83.
    Figure US20030130349A1-20030710-C00097
  • (1S)-1,8,8-Trimethyl-3-oxabicyclo[3.2.1]octane-2,4-dione [(1S)-camphoric anhydride] (33-A) (1S-cis)-1,2,2-Trimethylcyclopentane-1,3-dicarboxylic acid) [(1S,3R)-camphoric acid] (1.0 g, 5 mmol) and acetic anhydride (10 mL) were heated at reflux for 3 h. The reaction was cooled and the solvent was removed on a rotovap (bath temp 60° C.). To the remainder of the material, saturated NaHCO[0427] 3 (2 mL) was added. The aqueous portion was extracted with CH2Cl2 (3×5 mL), dried and concentrated in vacuo to 1.08 g. This was triturated with methyl-t-butyl ether to afford after filtering 0.94 g (103%) of (1S)-Camphoric anhydride (33-A): mp 222-223° C.; [α]D+3.8° (c=0.8, toluene); 1H NMR (CDCl3) δ 1.01, 1.10, 1.27, 1.89-2.35, 2.84; 13C NMR (CDCl3) ppm 172.7, 170.0, 54.33, 53.8, 43.7, 33.5, 24.5, 20.8, 20.2, 14.1; IR (mineral oil mull) 2925, 1804, 1763, 1180, 1128, 1043, 983, 943 cm−1; MS for C10H14O3, m/z (relative intensity) 169 (1), 138 (37), 123 (17), 110 (16), 95 (100); Anal. Calcd for C10H14O3: C, 65.92; H, 7.75. Found: C, 65.86; H, 7.74.
    Figure US20030130349A1-20030710-C00098
    Figure US20030130349A1-20030710-C00099
  • Preparation of Intermediate (34B) (C5H3ClINO).
  • To a solution of 2-chloro-3-hydroxypyridine (34-A) (10.2 g, 78.7 mmol) and K[0428] 2CO3 (38.87 g, 0.27 mmol) in H2O (120 mL) at rt is added 12 (24.33 g, 95.8 mmol). The solution is stirred at rt for 4 h, and the reaction then is quenched with satd aqueous Na2S2O5.5H2O. The pH of the reaction mixture is adjusted to pH 2 with 12 M aqueous HCl. The mixture is extracted with EtOAc (3×100 mL), and the combined EtOAc portions dried (MgSO4), filtered and concentrated to a yellow solid. Recrystallization of this solid from 120:25 heptane/EtOAc (145 mL) gives 11.2 g (43.8 mmol, 56%) of intermediate (34-B): IR 3113, 3068, 3056, 3021, 2991, 2955, 2925, 2871, 2855, 2832, 2808, 2749, 2668, 2601, 2530, 1554, 1457, 1398, 1304, 1289, 1226, 1086, 828, 711, 617 cm−1; 1H NMR (DMSO-d) δ 7.06 (1H), 7.59 (1H), 11.06 (1H); 13C NMR (DMSO-d6) δ 101.18, 127.02, 134.98, 138.07, 150.68; Anal. C, 23.32; H, 1.23; Cl, 13.73; N, 5.42 (calcd C, 23.51; H 1.18, Cl, 13.88; N, 5.48).
  • Preparation of Intermediate 34-C (C[0429] 12H7Cl3INO).
  • To a mixture of (34-B) (5.11 g, 20.0 mmol), Ph[0430] 3P (5.30 g, 20.0 mmol) and 2,6-dichlorobenzyl alcohol (3.54 g, 20.0 mmol) in dry THF (100 mL) at 0° C. is added dropwise DEAD (3.15 mmol, 20.0 mmol). The reaction mixture is stirred an additional 1.5 h at 0° C. and 1.5 h at rt, and then is concentrated. The reaction product is purified by silica flash chromatography (85:15 hexanes/EtOAc) co give 7.61 g (18.36 mmol, 92%) of (34-C) as a white solid: TLC Rf=0.57 (7:3 hexanes/EtOAc); 1H NMR (DMSO-d) δ 5.34 (2H), 7.48 (1H), 7.55-7.63 (3H), 7.85 (1H); MS (ES) m/z 413.8, 327.9, 288.0, 255.9, 183.0, 150.9, 136.9.
  • Preparation of Intermediate (34-E) (C[0431] 16H23NO7S).
  • To a soluition of N-Boc-L-serine methyl ester (34-D) (10.0 g, 45.6 mmol) in anhydrous pyridine (78 mL) at −10° C. under Ar is added TsCl (10.0 g, 52.4 mmol). The reaction mixture is stirred for 3 h at −10° C., and then kept at −15° C. for 66 h. The reaction is quenched with ice, stirred for 2 h, and then extracted with EtOAc (4×300 mL). The combined EtOAc portions are washed with 0.2 M aqueous KHSO[0432] 4 (3×300 mL), H2O (300 mL), satd aqueous NaHCO3 (300 mL), and H2O (300 mL); and then dried (Na2SO4) filtered and concentrated to a pale yellow-colored oil. The reaction product is purified by silica flash chromatography (3:1 hexanes/EtOAc) to give 13.0 g (34.8 mmol, 76%) of (34-E) as an off-white colored solid: TLC Pf=0.25 (7:3 hexanes/EtOAc); IR 3400, 2407, 2313, 2291, 1928, 1741, 1708, 1513, 1350, 1245, 1174, 1159, 1060, 995, 941 cm−1; 1H NMR (CDCl3) δ 1.41 (9H), 2.44 (3H), 3.69 (3H), 4.28 (1H), 4.39 (1H), 4.49 (1H), 5.29 (1H), 7.34 (2H), 7.75 (2H); MS (FAB) m/z 747, 527, 374, 319, 318, 274, 146, 102, 57, 41, 29; Anal. C, 51.41; H, 6.32; N, 3.87; S, 8.27 (calcd C, 51.46; H, 6.21; N, 3.75; S, 8.59).
  • Preparation of Intermediate (34-F) (C[0433] 9H16INO4).
  • To a solution of (34-E) (12.82 g, 34.3 mmol) in dry acetone (40 mL), in an amber reaction flask at rt under Ar, is added dropwise a solution of NaI (7.73 g, 51.5 mmol) in dry acetone (40 mL). The reaction mixture is stirred at rt for 42 h, and then is concentrated. The residue is dissolved in CHCl[0434] 3 (300 mL). This CHCl3 solution is extracted with H2O (2×300 mL), aqueous 1 M Na2S2O3.5H2O (300 mL), and H2O (3×300 mL); and then is dried (Na2SO4), filtered, and concentrated to give a yellow oil. The product is purified by silica flash chromatography (4:1 hexanes/EtOAc) to give 9.49 g (28.8 mmol, 84%) of (34-F) as a white solid: TLC Rf=0.52 (7:3 hexanes/EtOAc); 1H NMR (CDCl3) δ 1.45 (9H), 3.56 (2H), 3.79 (3H), 4.51 (1H); MS (FAB) m/z 330, 274, 230, 211, 170, 146, 102, 57, 41.
  • Preparation of Intermediate (34-G) (C[0435] 21H23Cl3N2O5).
  • To a dry amberized reaction flask under Ar, containing activated Zn dust (0.777 g, 11.89 mmol) and (34-F) (3.91 g, 11.9 mmol), is added dry THF (11.8 mL) and CH[0436] 3C(O)N(Me)2 (11.8 mL). Residual O2 is removed by bubbling Ar through the suspension for 5 min. The reaction mixture is stirred at 65±5° C. for 2 h, and then is cooled to 0° C. The PdCl2[P(Ph)3]2 catalyst (0.41 g) is added, folowed immediately by an O2-free solution of (34-C) (2.46 g, 5.94 mmol) in dry 1:1 THF/CH3C(O)N(Me)2 (17.8 mL). The resulting reaction mixture is stirred at 65±5° C. under Ar for 5 h. It is quenched with satd aqueous NH4Cl (150 mL). The resulting mixture is extracted with EtOAc (3×300 mL). The combined EtOAc portions are washed with brine (300 mL), dried (Na2SO4), filtered and concentrated to a yellow-green colored oil. The product is purified by silica flash chromatography (3:1 hexanes/EtOAc) to give 1.90 g (3.88 mmol, 65%) of (34-G): TLC Rf=0.32 (7:3 hexanes/EtOAc); IR 3391, 1734, 1702, 1567, 1561; 1508, 1439, 1287, 1256, 1225, 1214, 1198, 1179, 1167, 1152, 1099, 1087, 1070, 1022, 992, 989, 846, 784, 772, 718 cm−1; 1H NMR (DMSO-d6) δ 1.31 (9 U), 2.94-3.03 (2H), 3.60 (3H), 4.32 (1H), 5.30 (2H), 7.28 (2H); 7.45-7.58 (3H), 7.76 (1H); MS (ES) m/z 490.8, 434.8, 388.9.
  • Preparation of Intermediate (34-H) (C[0437] 16H15Cl3N2O3.HCl).
  • A solution of (34-G) (1.90 g, 3.88 mmol) in 4 M HCl in 1,4-dioxane (35 mL) is stirred at rt under Ar for 20 h. The reaction mixture is concentrated, diluted with H[0438] 2O (40 mL), and extracted with Et2O (3×40 mL). The Et2O portions are discarded. The aqueous solution is lyophilized to give 1.39 g (3.26 mmol, 84%) of (34-H) as a beige-colored solid: 1H NMR (DMSO-d6) δ 3.27 (2H), 3.72 (3H), 4.37 (1H), 5.32 (2H), 7.37 (1H), 7.48 (1H), 7.58 (1H), 7.81 (1H), 8.62 (3H); 13C NMR (DMSO-d6) δ 36.29, 51.98, 53.14, 66.74, 123.33, 125.04, 129.38, 131.21, 132.50, 136.63, 138.99, 147.02, 149.88, 169.75.
  • Preparation of Intermediate (34-I) (C[0439] 30H37Cl3N2O6).
  • To a reaction mixture containing acid (15D) (0.513 g, 2.0 mmol), EDC (0.403 g, 2.06 mmol), HOBt (0.284 g, 2.10 mmol), DMAP (0.076 g, 0.62 mmol) and (34-H) (0.878 g, 2.06 mmol) in CH[0440] 2Cl2 (20.4 mL) at 0° C. is added Et3N (1.02 mL, 7.24 mmol). The mixture is stirred for 2 h at 0° C. and 44 h at rt. It is diluted with CH2Cl2 (200 mL). The CH2Cl2 mixture is washed with H2O (3×200 mL), 0.5 M aqueous HCl (2×200 mL), satd aqueous NaHCO3 (2×200 mL), and H2O (2×200 mL). The combined aqueous washes are extracted with one portion of CH2Cl2 (200 mL). The combined CH2Cl2 portions are dried (Na2SO4), filtered, and concnetrated to give a yellow-colored oil. The product is purified by silica flash chromatography (3:2 hexanes/EtOAc) to give 0.919 g (1.46 mmol, 73%) of (34-I): TLC Rf=0.15 (7:3 hexanes/EtOAc); 1H NMR (CDCl3) δ 0.82 (3H), 1.18 (3H), 1.31 (3H), 1.38-1.48 (1H), 1.44 (9H), 1.58-1.81 (1H), 2.14-2.27 (1H), 2.49-2.70 (2H), 3.17 (1H), 3.29 (1H), 3.69 (3H), 4.93 (1H), 5.33 (2H), 7.06 (2H), 7.26-7.39 (4H); 13C NMR (CDCl3) δ 14.19, 20.40, 21.93, 22.19, 22.91, 28.07, 32.36, 37.04, 46.41, 51.76, 52.28, 54.50, 56.78, 60.38, 61.17, 66.73, 80.07, 123.08, 123.23, 128.60, 130.92, 130.96, 137.08, 140.97, 149.43, 149.77, 171.71, 172.70, 175.11.
  • Preparation of Intermediate (34-J) (C[0441] 26H29Cl3N2O6).
  • To the solid (34-I) (0.910 g, 1.45 mmol) at 0° C. under Ar is added slowly TFA (9 mL). The resulting solution is stirred for 30 min at 0° C. and 2 h at rt. The reaction mixture is concentrated in vacuo, thrice azeotroped with PhCH[0442] 3, and dried under vacuum to give (34-J) as an amber-colored foam: TLC Rf=0.37 (750;250:5 hexanes/EtOAc/HCO2H); IR 3321, 3061, 1746, 1728, 1696, 1655, 1584, 1565, 1523, 1497, 1440, 1355, 1286, 1209, 1200, 1171, 1119, 1094, 1088, 995, 781, 769, 731, 717, 695 cm−1; 1H NMR (CDCl3) δ 0.87 (3H) 1.26 (3H), 1.33 (3H), 1.46-1.58 (1H), 1.73-1.87 (1H), 2.14-2.30 (1H), 2.53-2.65 (2H), 2.69 (1H), 3.19 (1H), 3.29 (1H), 3.69 (3H), 4.93 (1H), 5.33 (2H), 7.07 (1H), 7.23-7.40 (4H), 10.18 (1H).
  • Preparation of Example 194 (C[0443] 26H29Cl3N2O6).
  • To a solution of (34-J) (0.908 g, 1.41 mmol) in THF (28.9 mL) is added a solution of LiOH.H[0444] 2O (0.291 g, 6.94 mmol) in H2O (14.4 mL). The reaction mixture is stirred for 5 h, and then is diluted with H2O (70 mL). It is cooled to 0° C., acidified with 1 M aqueous HCl, and extracted with EtOAc (3×200 mL). The combined EtOAc portions are washed with brine (200 mL), dried (Na2SO4), filtered and concentrated to a viscous colorless oil. The product is purified by silica flash chromatography (200:50:1 hexanes/EtOAc/HCO2H), azeotroped thrice from PhCH3, dissolved in 1:1 CH3CN/H2O (40 mL), and lyophilized to give 0.752 g (1.35 mmol, 96%) of Example 194 as a white solid: mp 120-122° C.; TLC Rf=0.28 (200:1 EtOAc/HCO2H); IR 3323, 3064, 2730, 2668, 1714, 1700, 1648, 1584, 1564, 1522, 1440, 1354, 1284, 1235, 1198, 1162, 1118, 1096, 1089, 995, 862, 828, 780, 769, 716 cm−1; 1H NMR (DMSO-d6) δ 0.63 (3H) 1.09 (3H), 1.12 (3H), 1.24-1.37 (1H), 1.46-1.58 (1H), 1.78-1.94 (1H), 2.27-2.43 (2H), 2.62 (1H), 2.96-3.14 (2H), 3.31 (1H), 4.57 (1H), 5.30 (2H), 7.29 (1H), 7.45-7.58 (3H), 7.75 (1H), 7.85 (1H) MS (EI) m/z 556, 538, 379, 353, 335, 301, 159, 142, 123, 109, 95.
    Figure US20030130349A1-20030710-C00100
    Figure US20030130349A1-20030710-C00101
  • Preparation 35-B: Scheme 35
  • To a cooled (0-5° C.) mixture of Wang polystyrene resin 35-A (Advanced Chemtech, 2.0 g, ca. 1.5 mmol), N-Boc-4-iodo-L-phenylalanine (4.00 g, 10 mmol), and PPh[0445] 3 (1.30 g, 5.0 mmol) in THF (20 mL) was added diethyl azodicarboxylate (0.80 mL, 5.0 mmol) in 4 approximately equal portions at 5 min intervals. When the orange color had discharged the mixture was warmed to ambient temperature and stirred for 5 h. The mixture was diluted with THF (30 mL) and filtered. The resin was washed with DMF (5×50 mL), THF (5×50 mL), and MeOH (3×50 mL) and then dried in vacuo to afford the esterified resin 35-B (2.68 g) as a colorless powder: 13C NMR (100 MHz, CD2Cl2, 4 mm MAS probe) δ 171.86, 155.33, 137.85, 136.40, 131.87, 128.00, 92.74, 80.09, 54.05, 38.05, 28.51.
  • Preparation 35-C-1: Scheme 35 where R[0446] 35 is 2,4,6-trichlorophenyl.
  • N[0447] 2 was bubbled through a mixture of N-Boc-4-iodo-L-phenylalanine functionalized Wang resin (35-B) (500 mg, ca. 0.3 mmol), PPh3 (105 mg, 0.4 mmol), 2,4,6-trichloroaniline (490 mg, 2.5 mmol) and DIEA (1.74 mL, 10 mmol) in NMP (10 mL) for 10 min. Pd2 dba3 (92 mg, 0.1 mmol) was added and the reaction mixture was placed under a CO atmosphere and heated (bath temp. 70° C.) for 18 h. Upon cooling to ambient temperature the mixture was diluted with 3% (w/v) sodium diethyldithiocarbamate in 95:5 NMP:DIEA (10 mL). After an additional 10 min the mixture was filtered and the resin washed with NMP (5×10 mL), THF (3×10 mL), and MeOH (3×10 mL) and dried in vacuo to afford 35-C-1 as a colorless powder.
  • Preparation 35-D-1: Scheme 35 where R[0448] 35 is 2,4,6-trichlorophenyl.
  • Resin 35-C-1 was swollen with methylene chloride (0.5 mL) and diluted with 95:5 TFA:H[0449] 2O (10 mL) After 90 min the mixture was filtered and the resin washed with TFA (3×5 mL), CH2Cl2 (3×5 mL) and MeOH (3×5 mL). The combined filtrates were concentrated in vacuo and the residue lyophilized from glacial acetic acid to provide the amino acid 35-D-1 (152 mg, 91%) as a powder which was used without purification: MS (FAB) m/z (rel. intensity) 387 (M+H, 42), 427 (26), 426 (80), 389 (46), 387 (42), 366 (33), 279 (99), 177 (54), 146 (18), 119 (26), 23 (26); HRMS (FAB) calcd for C16H13Cl3N2O3+H1 387.0070, found 387.0084.
  • Preparation 35-E-1: Scheme 35 where R[0450] 35 is 2,4,6-trichlorophenyl.
  • The amino acid 35-D-1 was dissolved in methanolic HCl (20 mL) and heated at 55° C. for 18 h. Concentration in vacuo afforded the methyl ester 35-E-1 which was used without purification: MS (ES-+) for C[0451] 17H15Cl3N2O3 m/z 400.9 (M+H)+.
  • Preparation 35-F-1: Scheme 35 where R[0452] 35 is 2,4,6-trichlorophenyl.
  • To a cooled (0-5° C.) solution of the methyl ester 35-E-1,15-D (97 mg, 0.38 mmol), and 1-hydroxy-7-azabenzotriazole (52 mg, 0.38 mmol) in CH[0453] 2Cl2/DMF (1:2, 6 mL) was added EDC (73 mg, 0.38 mmol) followed by DIEA (0.23 ml, 1.14 mmol). The solution was gradually allowed to warm to ambient temperature and stirred an additional 16 h. Volatiles were removed in vacuo and the residue partioned between ethyl acetate and 0.25N aq. HCL. The organic layer was washed with saturated aq. NaHCO3 and brine, dried (MgSO4), filtered and concentrated in vacuo. Purification of the residue by flash chromatography using ethyl acetate/CH2Cl2/hexane (1:1:6) as eluant afforded 35-F-1 (115 mg) as an amorphous powder: 1H NMR (300 MHz, CDCl3) δ 7.96 (1H), 7.86 (2H), 7.20 (2H), 5.86 (1H), 4.92 (1H), 3.72 (3H), 3.17 (2H), 2.49 (2H), 2.10 (1H), 1.69 (1H), 1.41 (10H), 1.25 (3H), 1.19 (3H), 0.76 (3H); 13C NMR (75 MHz, CDCl3) δ 174.90, 172.66, 171.88, 165.41, 140.88, 134.42, 133.50, 132.08, 131.27, 129.58, 128.44, 127.97, 80.25, 60.39, 56.64, 54.43, 52.91, 52.47, 46.38, 37.71, 32.33, 28.03, 22.99, 22.51, 21.93, 20.59, 14.17; MS (FAB) m/z (rel. intensity) 639 (M+H, 17), 641 (17), 639 (17), 583 (16), 403 (27), 401 (28), 189 (23), 137 (18), 109 (99), 57 (59), 41 (20); HRMS (FAB) calcd for C31H37Cl3N2O6+H, 639.1795, found 639.1779.
  • Preparation of Example 220
  • (1S-cis)-N-[[(3-Carboxy-2,2,3-trimethylcyclopentyl]-carbonyl]-4-[[[(1,1′-biphenyl)-4-yl)amino]-carbonyl]-L-phenylalanine [0454]
  • Example 220 was prepared as described in Schemes 35 and 2 starting from 4-aminobiphenyl and 35-B: physical properties as follows: [0455] 1H NMR (300 MHz, CD3OD) δ 7.87 (1H), 7.76 (2H), 7.62 (4H), 7.44 (4H), 7.30 (1H), 4.79 (1H), 3.30 (1H), 3.01 (1H), 2.72 (1H), 2.54 (1H), 2.02 (1H), 1.62 (1H), 1.58 (1H), 1.28 (3H), 1.20 (3H), 0.78 (3H); 13C NMR (75 MHz, CD3OD) δ 179.75, 175.49, 174.98, 168.82, 143.38, 142.03, 139.42, 138.75, 134.74, 130.70, 130.01, 128.86, 128.38, 128.31, 127.87, 122.68, 57.56, 54.69, 47.81, 38.43, 33.87, 23.94, 23.25, 22.57, 21.99; MS (FAB) m/z (rel. intensity) 543 (M+, 17), 109 (41), 83 (43), 81 (37), 71 (45), 69 (82), 67 (35), 57 (81), 55 (99), 43 (80); HRMS (EI) calcd for C32H34N2O6 542.2416, found 542.2429. Anal. Calcd for C32H34N2O6.0.5H2O: C, 69.67; H, 6.39; N, 5.08. Found: C, 69.72; H, 6.65; N, 4.75.
  • Preparation of Example 221
  • (1S-cis)-[[(3-Carboxy-2,2,3-trimethylcyclopentyl]carbonyl]-4-[[(4-chlorophenyl)amino]-carbonyl]-L-phenylalanine [0456]
  • Example 221 was prepared as described in Schemes 35 and 2 starting from 4-chloroaniline and 35-B: physical properties as follows: [0457] 1H NMR (300 MHz, CD3OD) δ 7.85 (2H), 7.68 (2H), 7.40 (2H), 7.36 (2H), 4.82 (1H), 3.29 (1H), 3.05 (1H), 2.81 (1H), 2.54 (1H), 1.96 (1H), 1.70 (1H), 1,24 (1H), 0.91 (3H), 0.86 (3H), 0.77 (3H); 13C NMR (75 MHz, CD3OD) δ 179.74, 175.55, 174.65, 168.73, 143.39, 138.93, 134.-5, 130.66, 130.55, 128.86, 123.68, 57.54, 54.62, 47.81, 38.33, 35.92, 33.86, 30.30, 29.00, 26.34, 23.21, 22.51, 21.98, 21.16; HRMS (FAB) calcd for C26H29ClN2O6+H1 501.1792, found 501.1790.
  • Preparation of Example 222
  • (1S-cis)-[[(3-Carboxy-2,2,3-trimethylcyclopentyl]-carbonyl]-4-[[(2-trifluoromethylphenyl)amino]-carbonyl]-L-phenylalanine [0458]
  • Example 222 was prepared as described in Schemes 35 and 2 starting from 2-(triflouromethyl)aniline and 35-B. Physical properties as follows: [0459] 1H NMR (300 MHz, CD3OD) δ 7.86 (2H), 7.66 (4H), 7.40 (2H), 4.78 (1H), 3.30 (1H), 3.10 (1H), 2.72 (1H), 2.49 (1H), 1.98 (1H), 1.62 (1H), 1.48 (1H), 1.28 (3H), 1.20 (3H), 0.78 (3H); 13C NMR (75 MHz, CD3OD) δ 179.70, 175.77, 173.54, 169.59, 143.34, 136.74, 124.18, 133.85, 131.74, 130.73, 128.89, 128.30, 127.13, 123.51, 57.56, 55.05, 62.54, 57.56, 55.05, 54.58, 50.06, 47.80, 38.21, 33.84, 23.89, 23.19, 22.52, 22.30, 21.85; IR (mull) 3302, 1708, 1656, 1613, 1592, 1530, 1508, 1320, 1294, 1260, 1206, 1173, 1123, 1059, 767 cm−1; MS (FAB) m/z (rel. intensity) 535 (M+, 99), 536 (32), 535 (99), 517 (25), 353 (46), 109 (57), 69 (14), 57 (13), 55 (14), 43 (13); HRMS (FAB) m/z calcd for C27H29F3N2O6+H1 535.2056, found 535.2049. Anal. Calcd for C27H29F3N2O.0.5H2O: C, 59.66; H, 5.56; N, 5.15. Found: C, 59.75; H, 5.73; N, 4.72.
  • Preparation of Example 223
  • (1S-cis)-[[(3-Carboxy-2,2,3-trimethylcyclopentyl]carbonyl]-4-[[(2,4,6-trichlorophenyl)amino]carbonyl]-L-phenylalanine [0460]
  • 35-F-1 (110 mg, 0.16 mmol) was deprotected as described in Scheme 2 to afford example 223 (90 mg) as an amorphous powder: [0461] 1H NMR (300 MHz, CD3OD) δ 7.90 (2H), 7.60 (1H), 7.41 (2H), 4.82 (1H), 3.29 (1H), 3.10 (1H), 2.72 (1H), 2.63 (1H), 1.92 (1H), 1.68 (1H), 1.44 (1H), 0.88 (6H), 0.77 (3H); 13C NMR (75 MHz, CD3OD) δ 179.72, 175.52, 174.72, 168.90, 143.97, 136.86, 135.15, 133.59, 133.02, 130,82, 129.66, 129.09, 62.70, 57.53, 54.65, 38.41, 33.85, 23.91, 23.20, 22.53, 21.97; IR (mull) 3263, 3079, 1709, 1657, 1614, 1573, 1556, 1524, 1495, 1287, 1246, 1205, 1190, 869, 857 cm−1; MS (ES+) for C26H27Cl3N2O6 m/z 568.9 (M+H)+; Anal. Calcd for O26H27Cl3N2O6: C, 54.80; H, 4.78; N, 4.92. Found: C, 55.00; H, 5.08; N, 4.64.
  • Preparation of Example 224
  • [1S-[1α(R*),3α]]-4-[[[(1-Carboxy-3-methylbutyl]-amino]carbonyl]-N-[[(3-carboxy-2,2,3-trimethylcyclopentyl]carbonyl]-L-phenylalanine [0462]
  • Example 224 was prepared as described in Schemes 35 and 2 starting from methyl L-leucinate and 35-B: Physical properties as follows: [0463] 1H NMR (300 MHz, CD3OD) δ 7.78 (2H), 7.34 (2H), 4.87 (1H), 4.72 (1H), 3.30 (1H), 3.02 (1H), 2.82 (1H), 2.68 (1H), 1.98 (1H), 1.74 (5H), 1.23 (3H), 0.97 (9H), 0.77 (3H); 13C NMR (75 MHz, CD3OD) δ 179.73, 176.33, 175.23, 174.66, 170.35, 143.05, 133.87, 130.52, 128.72, 62.41, 57.53, 54.63, 52.77, 47.79, 41.51, 38.30, 26.42, 23.90, 23.56, 23.18, 22.51, 21.93; MS (FAB) m/z (rel. intensity) 505 (M+, 99), 506 (27), 505 (99), 487 (20), 109 (29), 71 (20), 69 (34), 57 (34), 55 (33), 43 (36); HRMS (FAB) calcd for C26H36N2O8H1 505.2549, found 505.2570.
    Figure US20030130349A1-20030710-C00102
  • Where R[0464] 36-1 and R36-2 are independently defined as —CH3, t-Bu, or —CH2C6H5 and relative configuration is depicted by bold and dotted lines.
  • Scheme 36 teaches a general method (etherification, epimerization and ester deprotection) for the preparation of selectively protected camphoric acid isomers 36-B, 36-D, 36-G and 36-F. [0465]
    Figure US20030130349A1-20030710-C00103
  • Where R[0466] 37-1 and R37-2 are independently defined as —H or —CH3 and R37-3 and R37-4 are independently defined as —H, —CH3, t-Bu, or —CH2C6H5.
  • Scheme 37 teaches methods for the coupling of camphoric acid monoester isomers to isomers of 37-B and ester deprotection for the preparation of isomers of Example 54. 37-C-1 through 37-C-15. [0467]
  • Preparation of Example 237 Scheme 37: 37-C-1 where R3 is H, R1 is CH3, and the stereochemistry is (1S-cis) and D-Phenylalanine
  • (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine (C[0468] 26H28Cl2N2O6), 37-C-1 was prepared as follows:
  • To a solution of (1S-cis)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic acid 1-(1,1-dimethylethyl)-3-methyl diester (502.7 mg), 15-C, in methylene chloride (8 mL) at 0° C. was added DIEA (1 mL), EDC (413.1 mg), HOBT (291.1 mg), and dimethylaminopyridine (26.4 mg). The reaction was stirred at 0° C. for 15 minutes and then 4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine methyl ester, hydrochloride salt (803 mg), 37-B-2, was added and stirred at ambient temperature for 50 hours. The reaction was diluted with water and extracted with methylene chloride. The extracts concentrated in vacuo and the crude material purified by flash chromatography over silica gel. The crude material was applied to the column by concentrating it on a plug of silica gel and adding this plug to the top of the column. The column was eluted with methanol in methylene chloride to obtain (1S-cis)-N-[[3-[(1,1-Dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine methyl ester (890 mg). Physical properties as follows: m.p. 265-270° C.; [0469] 1H NMR (CDCl3) δ 7.63 (2H), 7.35 (3H), 7.19 (2H), 4.84 (1H), 3.76 (3H), 3.18 (1H), 3.01 (1H), 2.61 (1H), 2.47 (1H), 2.06 (1H), 1.74 (1H), 1.44 (10H), 1.14 (3H), 1.03 (3H), 0.66 (3H); MS-ESI (m/z): 603 ([M−H]); MS-ESI (m/z): 605 ([M+H+]). The deprotection of the carboxylic acids follows that of Examples 53, and 54 to obtain (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2, 6-dichlorobenzoyl)amino]-D-phenylalanine. Physical properties as follows: m.p. 263-267° C.; 1H NMR (300 MHz, DMSO-d6). δ 10.6 (1H), 7.83 (1H), 7.52 (5H), 7.2 (2H), 4.45 (1H), 3.1 (1H), 2.82 (1H), 2.62 (1H), 2.3 (1H), 1.94 (1H), 1.55 (1H), 1.3 (1H), 1.05 (3H), 0.83 (3H), 0.45 (3H); MS-ESI (m/z): 533 ([M−H); MS-ESI (m/z): 535 ([M+H+]).
  • Preparation of Example 238
  • Scheme 37: 37-C-2 where R[0470] 3 is H, R1 is CH3, and the stereochemistry is (1S-trans) and D-Phenylalanine (1S-trans)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine (C26H28Cl2N2O6), 37-C-2 was prepared as follows:
  • The preparation follows that of Preparation 37-C-1. The starting materials are (1S-trans)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic acid 1-(1,1-dimethylethyl)-ester, 36-G-1, and 4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine methyl ester, hydrochloride salt, 37-B-2. Physical properties as follows: m.p. 158-168° C.; [0471] 1H NMR (300 MHz, DMSO-d6). δ 12.3 (1H), 10.5 (1H), 7.95 (1H), 7.5 (5H), 7.19 (2H), 4.43 (1H), 3.04 (1H), 2.80 (1H), 2.66 (1H), 1.97 (2H), 1.66 (1H), 1.42 (1H), 0.96 (3H), 0.70 (3H), 0.44 (3H); MS-ESI— (m/z): 533 ([M−H]); MS-ESI (m/z): 535 ([M+H+]).
  • Preparation of Example 239
  • Scheme 37: 37-C-3 where R[0472] 3 is H, R1 is CH3, and the stereochemistry is (1S-trans) and L-Phenylalanine (1S-trans)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C26H28Cl2N2O6), 37-C-3 was prepared as follows:
  • The preparation follows that of Preparation 37-C-1. The starting materials are (1S-trans)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic acid 1-(1,1-dimethylethyl)-ester, 36-G-1, and 4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester, hydrochloride salt, 37-B-1. Physical properties as follows: m.p. 172-178° C.; [0473] 1H NMR (300 MHz, DMSO-d6) δ 10.6 (1H), 7.93 (1H), 7.5 (5H), 7.19 (2H), 4.43 (1H), 2.98 (1H), 2.86 (1H), 2.68 (1H), 2.01 (2H), 1.89 (1H), 1.63 (1H), 1.43 (1H), 1.02 (3H), 0.99 (3H), 0.72 (3H); MS-ESI (m/z): 533 ([M−H]
  • Preparation of Example 240
  • Scheme 37: 37-C-4 where R[0474] 3 is H, R1 is CH3, and the stereochemistry is (1R-trans) and L-Phenylalanine (1R-trans)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C26H28Cl2N2O6), 37-C-4 was prepared as follows:
  • The preparation follows that of Preparation 37-C-1. The starting materials are (1R-trans)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic acid 1-(1,1-dimethylethyl)-ester, 36-G-2, and 4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester, hydrochloride salt, 37-B-1. Physical properties as follows: m.p. 168-170° C.; [0475] 1H NMR (300 MHz, DMSO-d6) δ 12.3 (2H), 10.6 (1H), 7.96 (1H), 7.51 (5H), 7.20 (2H), 4.43 (1H), 3.05 (1H), 2.80 (1H), 1.97 (2H), 1.66 (1H), 1.42 (1H), 0.96 (3H), 0.70 (3H), 0.44 (3H); MS-ESI (m/z): 533 ([M−H]); MS-ESI (m/z) 535 ([M+H+]).
  • Preparation of Example 241
  • Scheme 37: 37-C-5 where R[0476] 3 is H, R1 is CH3, and the stereochemistry is (1R-trans) and D-Phenylalanine (1R-trans)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine (C26H28Cl2N2O6) 37-C-5 was prepared as follows:
  • The preparation follows that of Preparation 37-C-1. The starting materials are (1R-trans)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic acid 1-(1,1-dimethylethyl)-ester, 36-G-2, and 4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine methyl ester, hydrochloride salt, 37-B-2. Physical properties as follows: m.p. 158-165° C.; [0477] 1H NMR (300 MHz, DMSO-d6). 10.6 (1H), 7.93 (1H), 7.51 (5H), 7.19 (2H), 4.43 (1H) 2.98 (1H), 2.86 (1H) 2.68 (1H), 2.02 (1H), 1.88 (1H), 1.62 (1H)J, 1.43 (1H), 1.02 (3H), 0.991 (3H), 0.73 (3H); MS-ESI (m/z): 533 ([M−H]); MS-ESI (m/z): 5351 (M+H+])
  • Preparation of Example 242
  • Scheme 37: 37-C-6 where R[0478] 3 is H, R1 is CH3, and the stereochemistry is (1R-cis) and D-Phenyalaanine (1R-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine (C26H28Cl2N2O6), 37-C-6 was prepared as follows:
  • The preparation follows that of Preparation 37-C-1. The starting materials are (1S-cis)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic acid 1-(1,1-dimethylethyl)-ester, 36-D, and 4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine methyl ester, hydrochloride salt, 37-B-2. Physical properties as follows: m.p. 166-170° C.; [0479] 1H NMR (300 MHz, DMSO-d,) δ 10.65 (1H), 7.81 (1H), 7.51 (5H), 7.20 (2H), 4.44 (1H), 2.99 (1H), 2.85 (1H), 2.68 (11H), 2.34 (1H), 1.88 (1H), 1.52 (1H), 1.30 (1H), 1.15 (3H), 1.10 (3H), 0.65 (3H); MS-ESI (m/z): 533 ([M−H]); MS-ESI (m/z): 535 ([M+H+]).
  • Preparation of Example 243
  • Scheme 37: 37-C-7 where R[0480] 3 is H, R1 is CH3, and the stereochemistry is (1R-cis) and L-Phenylalanine (1R-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C26H28Cl2N2O6), 37-C-7 was prepared as follows:
  • The preparation follows that of Preparation 37-C-1. The starting materials are (1S-cis)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic acid 1-(1,1-dimethylethyl)-ester, 36-D, and 4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester, hydrochloride salt, 37-B-1. Physical properties as follows: m.p. 171-172° C.; [0481] 1H NMR (300 MHz, DMSO-d6) δ 10.6 (1H), 7.82 (1H), 7.51 (5H), 7.18 (2H), 4.45 (1H), 3.1 (1H), 2.82 (1H), 2.62 (1H), 2.32 (1H), 1.93 (1H), 1.58 (1H), 1.34 (1H), 1.06 (3H), 0.83 (3H), 0.45 (3H); MS-ESI (m/z): 533 ([M−H]); MS-ESI (m/z) 535 ([M+H+]).
  • Preparation of Example 244
  • Scheme 37: 37-C-8 where R[0482] 3 is CH3, R3 is H, and the stereochemistry is (1S-cis) and L-Phenylalanine (1S-cis)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C26H28Cl2N2O6), 37-C-8 was prepared as follows:
  • To a solution of (1S-cis)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic acid 3-methyl ester, 36-B (501.3 mg) in DMF (5 mL) and DIEA (3 mL) was added O-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (1.0445 g) and the reaction stirred at ambient temperature for 1 hour. To the reaction was added 4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester, hydrochloride salt (1.0093 g), 37-B-1, and the reaction stirred for 3 days. The reaction was then diluted with water and extracted with AcOEt. The concentrated extract was purified by flash chromatography on silica gel eluting with methanol in methylene chloride to obtain (1S-cls)-N-[(3-Methoxycarbonyl)-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester (2.272 g). A solution of LiOH (552 mg) in H[0483] 2O (20 mL) and 4 mL of 30% hydrogen peroxide was added to a solution of the above (1S-cis)-N-[((3-Methoxycarbonyl)-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester (C28H32Cl2N2O6) (2.272 g) in 15 mL of methanol. The solution was stirred for 6 days. The methanol is then removed in vacuo. The aqueous layer is further diluted with water and extracted with diethyl ether and the extract discarded. The aqueous layer is acidified to pH=3-4 with 0.6N HCl resulting in a precipitate. The precipitate is filtered washing with water to obtain (1S-cis)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (1.1265 g. Physical properties as follows: m.p. 152-157° C. 1H NMR (300 MHz, DMSO-d6). δ 12.3 (1H), 10.63 (1H), 7.50 (5H) 7.27 (1H), 7.18 (2H), 4.45 (1H), 2.99 (2H), 2.63 (1H), 2.25 (1H), 1.93 (1H), 1.69 (1H), 1.28 (1H), 1.16 (3H), 1.05 (3H), 0.50 (3H); MS-ESI (m/z): 533 ([M−H]); MS-ESI (m/z): 535 ([M+H+]).
  • Preparation of Example 245
  • Scheme 37: 37-C-9 where R[0484] 3 is CH3, R1 is H, and the stereochemistry is (1S-cis) and D-Phenylalanine (1S-cis)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine (C26H28Cl2N2O6) 37-C-9 was prepared as follows:
  • The preparation follows that of Preparation 37-C-8. The starting materials are (1S-cis)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic acid 3-methyl ester, 36-B, and 4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine methyl ester, hydrochloride salt, 37-B-2. Physical properties as follows: m.p. 155-163° C.; [0485] 1H NMR (300 MHz, DMSO-d.) δ 12.3 (1H), 10.6 (1H), 7.51 (5H), 7.29 (1H), 7.18 (2H), 4.44 (1H), 3.07 (1H), 2.94 (1H), 2.25 (1H), 2.62 (1H), 1.94 (1H), 1.67 (1H), 1.28 (1H), 1.04 (3H), 0.99 (3H), 0.48 (3H); MS-ESI (m/z): 533 ([M−H]); MS-ESI (m/z): 535 ([M+H+]).
  • Preparation of Example 246
  • Scheme 37: 37-C-10 where R[0486] 3 is CH3, R1 is H, and the stereochemistry is (1S-trans) and D-Phenylalanine (1S-trans)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[[(2,6-dichlorobenzoyl)amino]-D-phenylalanine (C26H28Cl2N2O6) 37-C-10 was prepared as follows:
  • To a solution of (1S-trans)-[3-(Phenylmethoxy)carbonyl]-1,2,2-trimethylcyclopentane-1-carboxylic acid, 36-F-1, (503.7 mg) in DMF (5 mL) and DIEA (3 mL) was added O-(7-Azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (731.7 mg) and the reaction stirred at ambient temperature for 1 hour. To the reaction was added 4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine methyl ester, hydrochloride salt (698.6 mg), 37-B-2, and the reaction stirred for 5 days. The reaction was then diluted with water and extracted with AcOEt. The concentrated extract was purified by flash chromatography on silica gel eluting with methanol in methylene chloride to obtain (1S-trans)-N-[(3-(Phenylmethoxy)carbonyl)-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine methyl ester (1.1024 g). Physical properties as follows: [0487] 1H NMR (CDCl3) δ 7.82 (1H), 7.58 (2H), 7.28 (8H), 7.05 (2H), 6.07 (1H), 5.06 (2H), 4.74 (1H), 3.66 (3H), 3.07 (3H), 2.05 (3H), 1.53 (1H), 1.21 (3H), 0.90 (3H), 0.75 (3H); MS-ESI (m/z): 637 ([M−H]); MS-ESI (m/z): 661 ([M+Na]+).
  • The product from above was dissolved in THF (10 mL) and 10% palladium on carbon (75 mg) was added and the mixture hydrogenated at atmospheric pressure for 26 hours. The reaction was then filtered and the filtrate concentrated to obtain (1S-trans)-N-1(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2, 6-dichlorobenzoyl)amino]-D-phenylalanine methyl ester (896.3 mg). Physical properties as follows; MS-ESI (m/z): 547 ([M−H[0488] ]); MS-ESI (m/z): 571 ([M+Na]+).
  • A solution of LiOH (213.2 mg) in H[0489] 2O (10 mL) and 2 mL of 30% hydrogen peroxide was added to a solution of (1S-trans)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine methyl ester (896.3 mg) in 10 mL of methanol. The solution was stirred for 26 hours. The methanol is then removed in vacuo. The aqueous layer is further diluted with water and extracted with diethyl ether and the extract discarded. The aqueous layer is acidified to pH=3-4 with 0.6N HCl resulting in a precipitate. The precipitate is filtered washing with water to obtain (1S-trans)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine (692.2 mg). Physical properties as follows: m.p. 145-150° C.; 1H NMR (300 MHz, DMSO-d6). δ 12.3 (1H), 10.6 (1H), 7.51 (5H), 7.29 (1H), 7.18 (2H), 4.44 (1H), 3.07 (1H), 2.94 (1H), 2.25 (1H), 2.62 (1H), 194 (1H), 1.67 (1H), 1.28 (1H), 1.04 (3H), 0.99 (3H), 0.48 (3H); MS-ESI (m/z): 533 ([M−H]); MS-ESI (m/z): 535 ([M+H+]).
  • Preparation of Example 247
  • Scheme 37: 37-C-11 where R[0490] 3 is CH3, R1 is H, and the stereochemistry is (1S-trans) and L-Phenylalanine (1S-trans)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C26H28Cl2N2O6), 37-C-11 was prepared as follows:
  • The preparation follows that of Preparation 37-C-10. The starting materials are (1S-trans)-[3-(Phenylmethoxy)carbonyl]-1,2,2-trimethylcyclopentane-1-carboxylic acid, 36-F-1, and 4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester, hydrochloride salt, 37-B-1 Physical properties as follows: m.p. 145-153° C.; [0491] 1H NMR (300 MHz, DMSO-d6). δ 12.3 (1H)l, 10.6 (1H), 7.51 (5H), 7.29 (1H), 7.18 (2H), 4.42 (1H), 3.01 (2H), 2.70 (1H), 1.88 (2H), 1.73 (1H), 1.36 (1H), 1.04 (3H), 0.94 (3H), 0.78 (3H); MS-ESI (m/z): 533 ([M−H]); MS-ESI (m/z): 535 ([M+H+]).
  • Preparation of Example 248
  • Scheme 37: 37-C-12 where R[0492] 3 is CH3, R1 is H, and the stereochemistry is (1R-cis) and L-Phenylalanine (1R-cis)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C26H28Cl2N2O6), 37-C-12 was prepared as follows:
  • The preparation follows that of Preparation 37-C-8. The starting materials are: (1R-cis)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic acid 3-methyl ester, 10-A, and 4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester, hydrochloride salt, 37-B-1. Physical properties as follows: m.p. 154-160° C. [0493] 1H NMR (300 MHz, MeOH-d4). δ 7.58 (2H), 7.42 (3H), 7.24 (3H), 4.74 (1H), 3.3 (1H), 3.03 (1H), 2.75 (1H), 2.41 (1H), 2.12 (1H), 1.80 (1H), 1.43 (1H), 1.14 (3H), 1.11 (3H), 0.63 (3H); MS-ESI (m/z): 533 ([M−H]); MS-ESI (m/z) 535 ([M+H+]).
  • Preparation of Example 249 Scheme 37: 37-C-13 where R3 is CH3, R1 is H, and the stereochemistry is (1R-cis) and D-Phenylalanine
  • (1R-cis)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine (q2[0494] 6H28Cl2N2O6), 37-C-13 was prepared as follows:
  • The preparation follows that of Preparation 37-C-8. The starting materials are (1R-cis)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic acid 3-methyl ester, 10-A, and 4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine methyl ester, hydrochloride salt, 37-B-2. Physical properties as follows: m.p. 155-159° C.; [0495] 1H NMR (300 MHz, DMSO-d6). δ 12.4 (1H), 10.6 (1H), 7.51 (5H), 7.19 (3H), 4.42 (1H), 2.99 (2H), 2.64 (1H), 2.26 (1H), 1.94 (1H), 1.68 (1H), 1.29 (1H), 1.17 (3H), 1.05 (3H), 0.51 (3H); MS-ESI (m/z): 533 ([M−H].
  • Preparation of Example 250
  • Scheme 37: 37-C-14 where R[0496] 3 is CH3, R1 is H, and the stereochemistry is (1R-trans) and L-Phenylalanine (1R-trans)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C26H28Cl2N2O6), 37-C-14 was prepared as follows:
  • The preparation follows that of Preparation 37-C-10. The starting materials are (1R-trans)-[3-(Phenylmethoxy)carbonyl]-1,2,2-trimethylcyclopentane-1-carboxylic acid, 36-F-2, and 4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester, hydrochloride salt, 37-B-1 . . Physical properties as follows: m.p. 148-155° C.; [0497] 1H NMR (300 MHz, DMSO-d6). δ 12.3 (2H), 10.6 (1H), 7.53 (5H), 7.27 (1H)f 7.21 (2H), 4.38 2 (1H), 3.03 (2H), 2.73 (1H), 1.92 (2H), 1.79 (1H), 1.43 (1H), 1.04 (3H), 0.75 (3H), 0.71 (3H); MS-ESI (m/z): 533 ([M−H]); MS-ESI (m/z): 535 ([M+H]).
  • Preparation of Example 251
  • Scheme 37: 37-C-15 where R[0498] 3 is CH3, R1 is H, and the stereochemistry is (1R-trans) and D-Phenylalanine (1R-trans)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine (C26H28Cl2N2O6), 37-C-15 was prepared as follows:
  • The preparation follows that of Preparation 37-C-10. The starting materials are (1R-trans)-[3-(Phenylmethoxy)carbonyl]-1,2,2-trimethylcyclopentane-1-carboxylic acid, 36-F-2, and 4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine methyl ester, hydrochloride salt, 37-B-2.. Physical properties as follows: m.p. 134-140° C.; [0499] 1H NMR (300 MHz, DMSO-d6). δ 12.3 (1H), 10.6 (1H), 7.50 (5H), 7.43 (1H), 7.21 (2H), 4.43 (1H), 3.03 (2H), 2.73 (1H), 1.92 (2H), 1.78 (1H), 1.41 (1H), 1.06 (3H), 0.96 (3H), 0.80 (3H); MS-ESI (m/z): 533 ([M−H]); MS-ESI (m/z): 535 ([M+H+]),
  • Preparation of Example 252
  • The synthesis for Example 252, (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-3-bromo-4-[(2, 6-dichlorobenzoyl)amino]-L-phenylalanine (C[0500] 26H28Cl2N2O6) is taught by Scheme 2 (Method B) as follows:
  • To a solution of (1R-cis)-1,2,2-Trimethylcyclopentane-1,3-dicarboxylic acid 1-(1,1-dimethylethyl) ester (153.9 mg), 15-D, in methylene chloride (6 mL) at 0° C. was added DIEA (1 mL), EDC (113.2 mg), HOET (80.3 mg), and dimethylaminopyridine (20.1 mg). The reaction was stirred at 0° C. for 20 minutes and then 3-Bromo-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester hydrochloride salt (259.4 mg) was added as a solution in methylene chloride (4 mL) and stirred at ambient temperature for 50 hours. The reaction was diluted with water and extracted with methylene chloride. The extracts were washed with 0.5 N HCl, dried over sodium sulfate and concentrated in vacuo. The crude material was purified by flash chromatography over silica gel eluting with AcOEt in hexane to obtain (1S-cis)-N-[[3-[(1,1-Dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-3-bromo-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester (153.8 mg). Physical properties as follows: [0501] 1H NMR (CDCl3) δ 8.33 (1H), 7.82 (1H), 7.31 (4H), 7.08 (1H), 5.87 (1H), 4.85 (1H), 3.74 (3H), 3.10 (2H), 2.52 (2H), 2.14 (1H), 1.73 (1H), 4.42 (10H), 1.23 (3H), 1.14 (3H), 0.79 (3H). MS-ESI (m/z): 681 ([M−H—I); MS-ESI (m/z): 683 ([M+H+]).
  • The deprotection of the carboxylic acids follows that of Examples 53, and 54 to obtain (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-3-bromo-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine. Physical properties as follows: m.p. 150-152° C.; [0502] 1H NMR (CD3OD) δ 7.95 (1H), 7.67 (1H), 7.57 (1H), 7.45 (3H), 7.35 (1H), 4.73 (1H), 3.25 (1H), 2.99 (1H), 2.75 (1H), 2.52 (1H), 2.00 (1H), 1.70 (1H), 1.44 (1H), 1.24 (3H), 1.21 (3H), 0.79 (3H); MS-ESI (m/z): 611 ([M−H]); MS-ESI (m/z): 613 ([M+H+]).
    Figure US20030130349A1-20030710-C00104
    Figure US20030130349A1-20030710-C00105
  • Preparation of Example 253
  • (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichloro-4-[(1,1-dimethyl)ethyl]benzoyl)amino]-L-phenylalanine (C[0503] 30H37Cl1N2O6).
  • 4-[(1,1-Dimethyl)ethyl]acetanilide (C[0504] 12H17NO, 38-B)
  • To a solution of 4-tert-butylaniline 38-A (14.9 g, 99.8 mmol) and pyridine (11 mL, 0.14 mol) in CH[0505] 2Cl2 (50 mL), under N2 and at 0° C., is added dropwise acetic anhydride (12 mL, 0.13 mmol). The reaction mixture is stirred at rt for 20 h, and is then quenched with 0.5 M aqueous HCl (100 mL). The reaction mixture is extracted with CH2Cl2. The combined CH2Cl2 extracts are washed with 0.5 M NaOH and brine, and then are dried, filtered and concentrated to give 38-B as an orange-colored solid (18.5 g). The solid is recrystallized from MeOH/Heptane to give a white solid (10.3 g, 54%): mp 172-173 0C; TLC (85:15 hexane/acetone) Rf=0.19; 13C NMR (CD3OD)δ 171.48, 148.15, 137.19, 126.52, 121.03, 35.14, 31.81, 23.74; MS (+ESI) m/z 190.
  • N,2-Dichloro-4-[(1,1-dimethyl)ethyl]acetanilide (C[0506] 12H15Cl2NO, 38-C)
  • To a suspension of 38-B (17.5 g, 91.5 mmol) and anhydrous NaOAc (19 g, 0.23 mmol) in HOAc (100 mL) under N[0507] 2 at 10° C. is added portionwise a solution of Cl2 (7 g) in HOAc (100 ml). Upon complete addition (approximately 15 min) the reaction mixture is allowed to warm to rt and is stirred for 1 h. A second portion of Cl2 (approximately 8 g) in HOAc (100 mL) is added and the resulting mixture is stirred at rt for 4.5 h. Finally, Cl2 gas is bubbled directly into the stirred mixture for 30 min. This mixture is stirred at rt for 17 h. It is concentrated under reduced pressure, chasing the residual HOAc with two portions of toluene. The solid is dissolved in EtOAc. The solution is filtered to give, after evaporation, an orange-colored oil (24.7 g). The oil is purified by silica chromatography (90:10 heptane/EtOAc) to give an orange-colored oil (18.4 g) that is recrystallized from pentane to give 38-C as a white solid (12.8 g, 54%): mp 64-65° C.; TLC (90:10 heptane/EtOAc) Rf=0.21; 13C NMR (CDCl3)δ 167.84, 155.65, 137.57, 133,57, 130.23, 127.91, 125.61, 35.11, 31.03, 21.52; MS (FAB) m/z 228, 226.
  • 2-Chloro-4-[(1,1-dimethyl) ethyl]acetanilide (C[0508] 12H16Cl1NO, 38-D).
  • To a solution of 18.6 g (71.5 mmol) of 38-C in absolute EtOH (100 mL) is added 10 M NaOH (7.1 mL) An exothermic reaction ensues. After the temperature had moderated the mixture is heated at reflux for 1 h. The pH of the cooled mixture is adjusted to pH 7-8 with concentrated HCl. The resulting mixture is partially concentrated (to remove EtOH), and then is diluted with CH[0509] 2Cl2 and and brine. The CH2Cl2 layer is separated. The aqueous solution is extracted twice additionally with CH2Cl2. The combined CH2Cl2 extracts are dried, filtered and concentrated to brown-colored oil. The oil is purified by silica chromatography (steps of 90:10 and 85:15 heptane/acetone) to give a solid (12.6 g), that is recrystallized from MeOH/pentane to give 38-D (7.49 g, 47%) as a white solid: mp 152-153° C.; TLC (85:15 heptane/acetone) Rf=0.33; 13C NMR (CDCl3)δ 168.23, 148.25, 131.92, 125.89, 124.70, 122.59, 121.64, 34.49,31.17, 24.70; MS (FAB) m/z 226.0995.
  • 2-Chloro-4-[(1,1-dimethyl)ethyl]aniline(C[0510] 10H14ClN, 38-E).
  • To a suspension of 38-D (6.0 g, 26 mmol) in EtOH (90 mL) is added 10 N NaOH (10 mL). The resulting mixture is heated at reflux. The suspended solid dissolves gradually. After 17 h at reflux the solution is cooled to 0° C. and is neutralized to pH 7 with concentrated HCl. The mixture is concentrated partially (to remove EtOH). the resulting aqueous mixture is diluted with brine, and is extracted with five portions of CH[0511] 2Cl2. The combined CH2Cl2 extracts are dried, filtered and concentrated to give 38-E as an orange-colored oil (5.5 g): TLC (85:15 heptane/acetone)=0.53; 1H NMR (CDCl3)δ 7.25 (1H), 7.09 (1H), 6.72 (1H), 3.82 (2H), 1.27 (9H); MS (+ESI; MeOH) m/z 186, 184.
  • 2-Chloro-4-[(1,1-dimethyl)ethyl]benzonitrile (C[0512] 11H12ClN, 38-F).
  • To a solution of aniline 38-E (5.5 g, 30 mmol) in 10:6 HOAc/H[0513] 2O (32 mL) is added concentrated H2SO4 (4.7 mL, 85 mmol). The brown-colored solution is cooled to 10° C. and is treated dropwise with a solution of NaNO2 (2.3 g, 33 mmol) in H2O (5 mL). After this addition is complete the reaction mixture is stirred at 10° C. for 1 h, yielding a yellow-colored solution. During this time a solution of KCN (9.8 g, 150 mmole) in H2O (25 mL) is added to a cold (ice bath), mechanically stirred solution of CuSO4.5H2O (9.0 g, 36 mmol) in H2O (25 mL). To this mixture is added NaHCO3 (20 g, 0.24 mmol) and benzene (30 mL), and the entire mixture is heated to 50-55° C. to dissolve all of the solids. The this solution is added dropwise the solution of the diazonium salt over 20 min under N2 and at 50-55° C. The reaction mixture is kept for 30 min at 50-55° C. for 0.5 h after the addition. The mixture is cooled, and extracted thrice with benzene. The combined benzene extracts are washed with 1N NaOH and brine, and then dried, filtered and concentrated to give a reddish-brown oil (6.8 g). The oil is purified by silica flash chromatography (steps of 95:5 and 90:10 heptane/CH2Cl2) to give 38-F (2.4 g, 41's): TLC (75:25 heptane/CH2Cl2) f=0.31; 1H NMR (CDCl3)δ 7.58 (1H), 7.49 (1H), 7.37 (1H), 1.31 (9H); 13C NMR (CDCl3)δ 158.53, 136.59, 133.64, 127.22, 124.49, 116.29, 110.21, 35.49, 30.80.
  • 2-Chloro-4-[(1,1-dimethyl)ethyl]benzoic acid (C[0514] 11H13ClO2, 38-G).
  • A solution of 38-F (2.28 g, 11.8 mmol), H[0515] 2O (7.4 mL), 10 N NaOH (5.9 mL), and 30. H2O2 (6.7 mL) in EtOH (80 mL) is refluxed for 28 h. The solution is cooled to 0° C. and neutralized to pH 7 with concentrated HCl.
  • A solution of NaHSO[0516] 3 (7 g), dissolved in the minimal amount of H2O, is added. The reaction mixture is concentrated partially (to remove most of the EtOH), basified to pH 12 with 1 N NaOH, and extracted twice with CH2Cl2. The combined CH2Cl2 extracts are discarded. The aqueous solution is acidified with concentrated HCl to pH 3, and then is extracted with CH2Cl2. The combined CH2Cl2 extracts are dried, filtered and concentrated to give 38-G (2.07 g, 83%) as a white crystalline solid: 1H NMR 11.62 (1H), 8.02 (1H), 7.51 (1H), 7.39 (1H), 1.36 (9H); 13C NMR (CDCl3)δ 171.17, 158.05, 134.84, 132.58, 128.75, 125.25, 123.96, 35.18, 30.91; MS (+ESI) m/z 237, 235 [M+Na]+; MS (−ESI; MeOH) m/z 213, 211.
  • (1S-cis)-N-[[3-(tert-Butoxycarbonyl)-2,2,3-trimethylcyclopentyl]carbonyl]-4-((2,6-dichloro-4-tert-butylbenzoyl)amino]-L-phenylalanine Methyl Ester (C[0517] 35H47ClN2O6, 38-H).
  • A mixture of (1S-cis)-N-[[3-(tert-Butoxycarbonyl)-2,2,3-trimethylcyclopentyl]-carbonyl]-4-nitro-L-phenylalanine methyl ester (406 mg, 0.88 mmol) and 10% Pd/C (39 mg) in 1:1 MeOH:THF (10 mL) is hydrogenated (30 psi H[0518] 2) for 1 h. The reaction mixture is filtered and concentrated to give the aniline as a colorless oil. This aniline is coupled directly with acid 15-D (190 mg, 0.89 mmol), as described by the general procedure for the synthesis of intermediates 7-F, to give after silica flash chromatography (steps of 99:1, 98:2, and 98:3 CHCl3/Acetone) to give 38-H (224 mg, 41%): TLC (95:5 CHCl3/Acetone) Rf=0.52; 13C NMR (CDCl3) δ 175.02, 172.53, 172.14, 164.53, 155.92, 136.86, 132.21, 131.96, 130.38, 130.31, 129.84, 127.48, 124.52,120.26, 80.19, 56.69, 54.44, 53.11, 52.36, 46.39, 37.20, 35.03, 32.36, 31.00, 28.07, 22.98, 22.48, 21.98, 20.61; MS (FAB) m/z 627.3201.
  • (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichloro-4-[(1,1-dimethyl)ethyl]benzoyl)amino]-L-phenylalanine (C[0519] 30H37C1N2O6, Example 253). A solution of 38-H (101 mg, 0.16 mmol) in TFA (2 mL) is stirred at rt for 2 h. The solution is diluted with CH2Cl2 and concentrated thrice under reduced pressure. The residue is diluted with toluene and again concentrated under reduced pressure to an oil. The oil is dissolved in MeOH (1.0 mL) and then treated with H2O (0.65 mL) and 1.00 M LiOH 0.35 mL). After 16 h a second portion of 1.00 M LiOH is added and the hydrolysis is allowed to proceed for an additional 4 h. The solution was diluted with H2O and the pH adjusted to ca. 8-9. The neutralized solution is diluted with MeOH and then concentrated. The aqueous concentrate is diluted with additional H2O, basified to pH 13 (1N NaOH), and extracted with Et2O. The Et2O extract is discarded. The aqueous phase is acidified to pH 2 (conc. HCl) and is extracted with EtOAc. The combined EtOAc extracts are dried, filtered and concentrated to give Example 253 (93 mg) as a colorless oil: 1H NMR (CD3OD) δ 7.61 (2H), 7.55-7.44 (3H), 7.25 (2H), 4.69-4.78 (1H), 3.23 (1H), 3.00 (1H), 2.80-2.70 (1H), 2.60-2.46 (1H), 2.10-1.94 (1H), 1.78-1.61 (1H), 1.52-1.39 (1H), 1.36 (9H); 1.27 (3H), 1.23 (3H), 0.81 (3H); MS (+ESI) m/z 557, 555.
    Figure US20030130349A1-20030710-C00106
    Figure US20030130349A1-20030710-C00107
  • Preparation of Example 254
  • [1S-[1α(R*),3α]]-α-[[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]amino]-3-[(2,6-dichlorophenyl)methoxy]-6-pyridinepropanoic acid (C[0520] 25H28Cl2N2O6).
  • (±)-2-Chloro-3-[(2-tetrahydropyranyl)oxyl-6-iodopyridine (C[0521] 10H11ClINO2, 39-A):
  • To a solution of chloroiodopyridinol 34-B (1.00 g, 3.91 mmol) and dihydropyran (1.0 mL, 10.6 mmol) in CH[0522] 2Cl2 (10 mL) under Ar at rt is added pyridinium chloride (0.050 g). The reaction mixture is stirred for 72 h. It is diluted with CH2Cl2, and is washed with satd aq NaHCO3 and brine. The CH2Cl2 solution is dried, filtered and concentrated to an oil, that is purified by silica flash chromatography (19:1 hexanes/EtOAc) to give 1.06 g (3.12 mmol, 80%) of 39-A: TLC (19:1 hexanes/EtOAc) Rf=0.24; 1H NMR (CDCl3, 300 MHz) δ7.55 (1H), 7.17 (1H), 5.50 (1H), 3.77 (1H), 3.61 (1H), 2.07-1.57 (6H); MS (+ESI) m/z 361.9, 339.9.
  • (2S)-2-Chloro-α-[[(1,1-dimethylethoxy)carbonyl]amino]-3-[(2-tetrahydropyranyl)-oxy]-6-pyridinepropanoic Acid Methyl Ester (C[0523] 19H27ClN2O6, 39-B):
  • To an amberized flask containing Reference Example 57 (1.81 g, 5.52 mmol) and activated Zn dust (0.349 g, 5.51 mmol) under Ar is added THF (2 mL) and 1,2-dibromoethane (0.018 mL, 0.21 mmol). The suspension is brought to reflux for several minutes, cooled to approximately 30° C., and TMSCl (0.17 mL of a 1 M solution in THF) is added. The reaction mixture is stirrred at 40±5° C. for 30 min, cooled in an ice bath, and solid PdCl[0524] 2 (PPh3)2 (0.192 g) is added. A degassed solution of the iodide 39-A (0.936 g, 2.76 mmol) in 1:1 THF/dimethylacetamide (5.6 mL) is added. This reaction mixture is stirred for 4 h at 45±5° C. It is then cooled to 0° C., quenched with satd aq NH4Cl, and extracted with EtOAc. The combined EtOAc portions are washed with satd aq NH4Cl and brine, and are dried, filtered and concentrated to a green-yellow colored foam. This foam is purified by silica flash chromatography (7:3 hexanes/EtOAc) to give 0.879 g (1.85 mmol, 60%) of 39-B: TLC (7:3 hexanes/EtOAc) Rf 0.21; 1H NMR (CDCl3, 300 MHz) δ7.39 (1H), 7.00 (1H), 5.46 (1H), 4.61 (1H), 4.13 (11H), 3.80 (3H), 3.62 (1H), 3.20 (1H), 2.13-1.53 (6H), 1.42 (9H); MS (+ESI) m/z 474.0
  • (S)-α-[[(1,1-Dimethylethoxy)carbonyl]amino]-3-[(2-tetrahydropyranyl)oxy]-6-pyridinepropanoic Acid Methyl Ester (C[0525] 19H28N2O6, 39-C):
  • A suspension of pre-reduced Pd/CaCO[0526] 3 (3.5 g) and 39-B (1.15 g, 2.77 mmol) in EtOH (40 mL) is hydrogenated (30 psi H2) for 19 h at rt. The mixture is filtered, and the filtrate is evaporated to give a yellow-colored foam that is purified by silica flash chromatography (600:400:1 hexanes/EtOAc/iPrOH) to give 0.367 g (0.96 mmol, 35%) of 39-C: TLC (1:1 hexanes/EtOAc) Rf 0.27; 1H NMR (CDCl3, 300 MHz) δ8.30 (1H), 7.29 (1H), 7.03 (1H), 5.81 (1H), 5.39 (1H), 4.65 (1H), 3.86 (1H), 3.73 (3H), 3.62 (1H), 3.21 (2H), 1.96-1.53 (6H), 1.42 (9H); MS (+ESI) m/z 381.1.
  • (S)-α-[[(1,1-Dimethylethoxy)carbonyl]amino]-5-hydroxy-2-pyridinepropanoic Acid Methyl Ester (C[0527] 14H20N2O5, 39-D):
  • A solution of 39-C (0.346 g, 0.91 mmol) and pyridinium p-toluenesulfonate (0.031 g, 0.12 mmol) in EtOH (8 mL) is stirred at 55±5° C. for 20 h. The reaction mixture is cooled to rt, and concentrated in vacuo. The residue is taken up in EtOAc (150 mL). This solution is washed with brine, dried, filtered and concentrated to a pale yellow-colored oil that is purified by silica flash chromatography (500:500:1 hexanes/EtOAc/iPrOH). Evaporation of the column fractions gives recovered 39-C (0.27 mmol) and 0.132 g (0.45 mmol, 49%) of 39-D: TLC (1:1 hexanes/EtOAc) R[0528] f=0.18; 1H NMR (CDCl3, 300 MHz) δ8.13 (1H), 7.13 (1H), 7.03 (1H), 5.71 (1H), 4.65 (1H), 3.70 (3H), 3.20 (2H), 1.39 (9H); MS (+ESI) m/z 297.1.
  • (S)-5-[(2,6-Dichlorophenyl)methoxy]-α-[((1,1-dimethylethoxy)carbonyl]amino]-2-pyridinepropanoic Acid Methyl Ester (C[0529] 21H4Cl2N2O5, 39-E):
  • To a solution of 39-D (0.126 g, 0.43 mmol), 2,6-dichlorobenzylalcohol (0.075 g, 0.43 mmol) and PPh[0530] 3 (0.113 g, 0.43 mmol) in dry THF (4 mL) at 0° C. under Ar is added DEAD (0.068 mL). The reaction mixture is permitted to warm to rt, and is stirred for 18 h. It is concentrated. The residue is purified by silica flash chromatography (700:300:1 hexanes/EtOAc/iPrOH) to give 0.149 g (0.33 mmol, 76%) of 39-E: TLC (7:3 hexanes/EtOAc) Rf=0.34; 1H NMR (CDCl31 300 MHz) δ8.31 (1H), 7.37 (2H), 7.25 (2H), 7.08 (1H), 5.81 (1H), 5.29 (2H), 4.65 (1H), 3.70 (3H) 3.24 (2H), 1.63 (1H) 1.43 (9H); 13C NMR (CDCl3, 75 MHz) δ172.47, 155.50, 153.82, 149.71, 137.33, 137.00, 131.51, 130.72, 128.56, 123.99, 122.78, 79.74, 65.64, 53.25, 52.27, 38.43, 28.33; MS (+ESI) m/z 454.9.
  • (S)-α-Amino-5-[(2,6-dichlorophenyl)methoxy]-2-pyridinepropanoic Acid Methyl Ester Dihydrogen Chloride Salt (C[0531] 16H16Cl2N2O3.2HCl, 39-F):
  • A solution of carbamate 39-E (0.546 g, 1.20 mmol) in 4 M HCl in dioxane (12 mL) is stirred at rt under Ar for 16 h. The reaction mixture is concentrated in vacuo. The residue is dissolved in H[0532] 2O (40 mL), and this solution is extracted with Et2O. The aqueous solution is frozen and lyophilized to give 0.485 g (1.13 mmol, 94%) of 39-F as a light yellow-colored solid: 1H NMR (CD3SOCD3, 300 MHz) δ8.75 (3H), 8.47 (1H), 7.81 (1H), 7.57 (3H), 7.48 (1H), 5.35 (2H), 4.49 (1H), 3.67 (3H), 3.42 (2H); 13C NMR (CD3SOCD3, 75 MHz) δ169.42, 154.95, 146.54, 136.57, 134.35, 132.50, 131.30, 129.36, 126.72, 126.52, 66.40, 53.32, 51.79, 34.81.
  • [1S-[1α(R*),3α]]-3-[(2,6-Dichlorophenyl)methoxy]-α-[[[3-[(1,1-dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]amino]-6-pyridinepropanoic Acid Methyl Ester (C[0533] 3H38Cl2N2O6, 39-G):
  • To a mixture of 15-D (0.141 g, 0.55 mmol), EDC (0.108 g, 0.57 mmol), HOBt (0,079 g, 0.58 mmol), DMAP (0.020 g, 0.16 mmol) and amine 39-F (0.246 g, 0.57 mmol) in CH[0534] 2Cl2 (6 mL) at 0° C. under Ar is added Et3N (0.18 mL, 1.26 mmol). The yellow-colored reaction mixture is stirred at rt for 90 h. It is diluted with CH2Cl2, and washed with H2O, 0.5 M aq HCl, H2O, satd aq NaHCO3, and H2O. The CH2Cl2 solution is dried, filtered and concentrated to a pale yellow-colored foam, that is purified by silica flash chromatography (600:400:1 hexanes/EtOAc.iPrOH) to give 0.195 g (0.33 mmol, 60%) of 39-G: TLC (3:2 hexanes/EtOAc) Rf=0.49; 1H NMR (CDCl3, 300 MHz) δ 8.28 (1H), 7.40-7.28 (4H), 7.13 (1H), 5.31 (2H), 4.92 (1H), 3.68 (3H), 3.33 (1H), 3.25 (1H), 2.65 (1H), 2.53 (1H), 2.21 (1H), 1.67 (1H), 1.44 (9H, 1.31 (3H), 1.18 (3H), 0.83 (3H).
  • [1S-[1α(R*),3α]]-α-[[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]amino]-3-[(2,6-dichlorophenyl)methoxy3-6-pyridinepropanoic Acid Methyl Ester (C[0535] 2H30Cl2N2O6, 39-H):
  • A solution of diester 39-G (0.195 g, 0.33 mmol) in TFA (4 mL) under Ar is stirred at 0° C. for 1 h and at rt for 2 h. The solution is concentrated, azeotroped thrice from toluene, and dried under vacuum to give 39-H as a yellow-colored glass: TLC (400:600:5 hexanes/EtOAc/HCO[0536] 2H) 4-0.29; 1H NMR (CDCl3, 300 MHz) δ8.46 (1H), 7.87 (1H), 7.74 (1H), 7.51 (1H), 7.42-7.26 (2H), 5.42 (2H), 5.01 (1H), 3.80 (3H), 3.62 (1H), 3.45 (1H), 2.70 (1H), 2.48 (1H), 1.97 (1H), 1.65 (1H), 1.49 (1H), 1.28 (3H), 1.24 (3H), 0.76 (3H).
  • [1S-[1α(R*)-α-[[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]amino)-3-[(2,6-dichlorophenyl)methoxy]-6-pyridinepropanoic acid (C[0537] 5H2Cl2N2O6, Example 254):
  • A solution of 39-H (0.33 mmol) and LiOH.H[0538] 2O (0.068 g, 1.62 mmol) in 2:1 THF/H2O (10.5 mL) is stirred under Ar for 5 h at rt. The reaction mixture is diluted with cold H2O, acidified with aq 1 M HCl, and extracted with EtOAc. The combined EtOAc extracts are washed with brine, and are dried, filtered and concentrated to a pale yellow-colored foam that is purified by silica flash chromatography (600:400:2 hexanes/EtOAc/HCO2H). The purified product is azeotroped thrice from toluene to remove HCO2H. It is dissolved in MeCN (10 mL) and the solution is diluted with H2O (10 mL). The solution is frozen and lyophilized to give, as an beige-colored solid, 0.170 g (0.32 mmol, 98%) of Example 254: mp 118-122° C.; TLC (150:50:1 EtOAc/hexanes/HCO2H) Rf=0.41;
  • [0539] 1H NMR (CD3SOCD3, 300 MHz) δ 8.43 (1H), 7.86 (2H) 7.58-7.47 (3H), 7.32 (1H), 5.28 (2H), 4.64 (1H), 3.24-2.99 (2H), 2.61 (1H), 2.32 (1H), 1.83 (1H), 1.51 (1H), 1.30 (1H), 1.12 (3H), 1.09 (3H), 0.63 (3H).
  • EXAMPLES Example 1 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-(phenylmethyl)-L-tyrosine methyl ester (C27H33NO6)
  • [0540]
    Figure US20030130349A1-20030710-C00108
  • DIEA (0.65 g) was added dropwise to a mixture of (1R)camphoric anhydride (0.18 g) and O-benzyl-L-tyrosine methyl ester hydrochloride (0.33 g) in DMF (2 ml) at 0° C. The mixture was stirred at 40° C. for 15 hr, cooled, diluted with AcOEt, and acidified with 1N HCl to pH 5. The organic layer was washed with H[0541] 2O, brine, dried over Na2SO4, and the solvent was removed in vacuo. The residue was purified by column chromatography on silica gel (eluent; 98:2, CHCl3/MeOH) to give (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-(phenylmethyl)-L-tyrosine methyl ester (C27H33NO6) (0.45 g) as a gum. MS (m/z): 468 (MH+)
  • Example 2 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-(phenylmethyl)-L-tyrosine (C26H31NO6)
  • [0542]
    Figure US20030130349A1-20030710-C00109
  • LiOH (72 mg) was added to a mixture of (1S-cis) —N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-(phenylmethyl)-L-tyrosine methyl ester (C[0543] 27H33NO6) (403 mg), THF (3 ml), and H2O (3 ml). The mixture was stirred at room temperature for 4 hr, acidified with 1N HCl, and extracted with AcOEt. The extract was dried over Na2SO4 and the solvent was removed in vacuo to give (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-(phenylmethyl)-L-tyrosine (391 mg), mp 146-149° C., MS (m/z): 452 ([M−H]-).
  • Example 3 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-1-[(3,4-dichlorophenyl)methyl]-L-histidine Methyl Ester (C24H29Cl2N3O5)
  • [0544]
    Figure US20030130349A1-20030710-C00110
  • DIEA (4.48 g) was added to a mixture of 1-(3,4-dichlorobenzyl)-L-histidine methyl ester (5.56 g) and (1R)-camphoric anhydride (2.53 g) in DMF (50 ml). The mixture was stirred at 40° C. for 17 hr, cooled, diluted with H[0545] 2O, acidified with 5% HCl to pH 5, and extracted with CHCl3. The extract was washed with H2O, brine, and dried over NaSO4. The solvent was removed in vacuo and the residue was purified by flash chromatography on silica gel (eluent: 100:1, CHCl3/MeOH), followed by recrystallization from AcOEt/MeOH to give (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-1-[(3,4-dichlorophenyl)methyl]-L-histidine methyl ester (5.90 g), mp 174-175° C. (dec), MS (m/z): 510 (MH+).
  • Example 4 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-1-[(3,4-dichlorophenyl)methyl]-L-histidine (C23H27Cl2N3O5)
  • [0546]
    Figure US20030130349A1-20030710-C00111
  • 1N NaOH (7.3 ml) was added dropwise to a mixture of (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-1-((3,4-dichlorophenyl)methyl]-L-histidine methyl ester (C[0547] 24H19Cl2N3O5) (1.50 g) in MeOH (20 ml) at 0° C. The mixture was stirred at room temperature for 15 hr, concentrated in vacuo, diluted with H2O, and acidified with 1N HCl to pH 5-6. The resulting precipitate was collected by filtration, washed with H2O, dried, and recrystallized from DMF/H2O to give (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-1-[(3,4-dichlorophenyl)methyl]-L-histidine (0.87 g), mp 148-149° C. (dec), MS (m/z): 496 (MH+)
  • Example 5 [1S-[1α(R*),3α]]-α-[[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]amino]-2-naphthalenepropanoic Acid Phenylmethyl Ester (C30H33NO5)
  • [0548]
    Figure US20030130349A1-20030710-C00112
  • Benzyl (S)-2-amino-3-(2-naphthyl) propionate tosylate (0.20 g) was partitioned between AcOH and sat. NaHCO[0549] 3. The organic layer was washed with H2O, brine, dried over Na2SO4, and the solvent was removed in vacuo. DMF (5 ml) and (1R)-camphoric anhydride (0.20 g) were added to the residue. The mixture was stirred at 30-40° C. for 17 hr, cooled, and poured into H2O. The resulting mixture was extracted with AcOH. The extract was washed with H2O, brine, dried over Na2SO4, and the solvent was removed in vacuo. The residue was purified by column chromatography on silica gel (eluent; 9:1, CHCl3/AcOEt) to give [1S-[1α(R*),3α]-α-[[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]amino]-2-naphthalenepropanoic acid phenylmethyl ester (0.51 g) as an oil. MS (m/z) 488 (MH+).
  • Example 6 (1S-[1α(R*),3α]]-α-[[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]amino]-2-naphthalenepropanoic Acid (C23H27NO5)
  • [0550]
    Figure US20030130349A1-20030710-C00113
  • 10% Pd-C (0.05 g) was added to a solution of [1S-[1α(R*),3α]]-α-[[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]amino]-2-naphthalenepropanoic acid phenylmethyl ester (C[0551] 30H33NO5) (0.20 g) in MeOH (10 ml) and the mixture was subjected to hydrogenolysis at a hydrogen pressure of 50 psi. The catalyst was filtered off and the filtrate was evaporated in vacuo to give [1S-[1α(R*),3α]]-α-[[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]amino]-2-naphthalenepropanoic acid (0.18 g), MS (m/z): 398 (MH+).
  • Examples 7 through 51 were prepared in a similar manner as described in Examples 1-6, and are shown in Tables 1, 2, and 3. [0552]
    TABLE 1
    Examples 7 through 20:
    Figure US20030130349A1-20030710-C00114
    Ex. physicochemical
    No. * R0 R15 Z1 R13 R18 property
    7 S H H single t-BuO— H M.P.: 98—100° C.
    bond MS(m/z)
    420 (MH+)
    8 S CH3 H —OCH2
    Figure US20030130349A1-20030710-C00115
    H gum MS(m/z): 518 (MH+)
    9 S H H —OCH2
    Figure US20030130349A1-20030710-C00116
    H M.P.: 92-93° C. MS (m/z): 502 ([M − H])
    10 S H H —OCH2
    Figure US20030130349A1-20030710-C00117
    H MS (m/z): 504 (MH+)
    11 S CH3 H —OCH2
    Figure US20030130349A1-20030710-C00118
    H gum MS (m/z): 536 (MH+)
    12 S H H —OCH2
    Figure US20030130349A1-20030710-C00119
    H M.P.: 95-97° C. MS (m/z): 520 ([M − H])
    13 S H H —OCH2
    Figure US20030130349A1-20030710-C00120
    H M.P.: 102-105° C. MS (m/z): 460 (MH+)
    14 S H H —OCH2
    Figure US20030130349A1-20030710-C00121
    H M.P.: 94-97° C. MS (m/z): 476 (MH+)
    15 S H I —OCH2
    Figure US20030130349A1-20030710-C00122
    I M.P. 210-212° C. (dec.) MS (m/z): 784, 786 (MH+)
    16 S H H —OCH2
    Figure US20030130349A1-20030710-C00123
    H MS (m/z): 520 ([M − H])
    17 S H PhCH2O —OCH2 Ph H MS (m/z):
    558 ([M − H])
    18 S H H —OCH2
    Figure US20030130349A1-20030710-C00124
    H MS (m/z): 530 (MH+)
    19 R CH3 H —OCH2 Ph H gum
    MS (m/z):
    468 (MH+)
    20 R H H —OCH2 Ph H MS (m/z):
    454 (MH+)
  • [0553]
    TABLE 2
    Examples 21 through 38:
    Figure US20030130349A1-20030710-C00125
    Ex. physicochemical
    No. R0 Z2 R16 property
    21 CH3 single —C(Ph)3 gum
    bond MS (m/z):
    594 (MH+)
    22 H single —C(Ph)3 MS (m/z): 58O (MH+)
    bond
    23 CH3 CH2
    Figure US20030130349A1-20030710-C00126
    gum MS (m/z): 510 (MH+)
    24 H CH2
    Figure US20030130349A1-20030710-C00127
    MS (m/z): 496 (MH+)
    25 CH3 CH2
    Figure US20030130349A1-20030710-C00128
    gum MS (m/z): 510 (MH+)
    26 H CH2
    Figure US20030130349A1-20030710-C00129
    MS (m/z) 496 (MH+)
    27 CH3 CH2
    Figure US20030130349A1-20030710-C00130
    gum MS (m/z): 476(MH+)
    28 H CH2
    Figure US20030130349A1-20030710-C00131
    MS (m/z): 462 (MH+)
    29 H CH2 Ph M.P. 258-259° C.
    (dec.)
    MS (m/z): 428 (MH+)
    30 CH3 CH2
    Figure US20030130349A1-20030710-C00132
    gum MS (m/z): 472 (MH+)
    31 H CH2
    Figure US20030130349A1-20030710-C00133
    MS (m/z): 458 (MH+)
    32 H CH2 PhCH2O MS (m/z): 458 (MH+)
    33 CH3 CH2
    Figure US20030130349A1-20030710-C00134
    gum MS (m/z): 460 (MH+)
    34 H CH2
    Figure US20030130349A1-20030710-C00135
    MS (m/z): 446 (MH+)
    35 CH3 CH2
    Figure US20030130349A1-20030710-C00136
    gum MS (m/z): 510 (MH+)
    36 H CH2
    Figure US20030130349A1-20030710-C00137
    MS (m/z): 496 (MH+)
    37 CH3 CH2
    Figure US20030130349A1-20030710-C00138
    gum MS (m/z): 492 (MH+)
    38 H CH2
    Figure US20030130349A1-20030710-C00139
    MS (m/z): 478 (MH+)
  • Example 39 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-[(2,6-dichlorophenyl)methyl]-D-tyrosine (C26H29Cl2NO6)
  • [0554]
    Figure US20030130349A1-20030710-C00140
  • Example 39 is an isomer of Example 12 and is therefore synthesized in a manner similar to that of Example 12. [0555]
    TABLE 3
    Examples 40 through 51:
    Figure US20030130349A1-20030710-C00141
    physicochemicali
    Ex. No. * R0 R6 property
    40 S H
    Figure US20030130349A1-20030710-C00142
    MS(m/z): 452 (MH+)
    41 S H
    Figure US20030130349A1-20030710-C00143
    M.P.: 163-165° C. MS (m/z): 354 (MH+)
    42 S H
    Figure US20030130349A1-20030710-C00144
    MS (m/z): 398 (MH+)
    43 S H
    Figure US20030130349A1-20030710-C00145
    MS (m/z): 509 ([M − H])
    44 S H Ph MS (m/z):
    348 (MH+(
    45 S CH3
    Figure US20030130349A1-20030710-C00146
    gum MS (m/z): 468 (MH+)
    46 S H
    Figure US20030130349A1-20030710-C00147
    M.P.: 88-90° C. MS (m/z): 452 ([M − H])
    47 S H
    Figure US20030130349A1-20030710-C00148
    MS (m/z): 502 ([M − H])
    48 S H
    Figure US20030130349A1-20030710-C00149
    MS (m/z): 422 ([M − H])
    49 S H
    Figure US20030130349A1-20030710-C00150
    MS (m/z): 475 ([M − H])
    50 S H
    Figure US20030130349A1-20030710-C00151
    MS (m/z): 537 (MH+)
    51 R H
    Figure US20030130349A1-20030710-C00152
    MS (m/z): 364 (MH+)
  • Example 52 (1S-cis)-N-[[3-[(1,1-Dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine Methyl Ester (C27H30Cl2N2O6)
  • [0556]
    Figure US20030130349A1-20030710-C00153
  • HCl gas was bubbled through a solution of N-(tert-butoxycarbonyl)-4-(2,6-dichlorobenzamido)-L-phenylalanine (800 mg) in MeOH (15 ml) for 5 minutes and the mixture was stirred for 3 hr. at room temperature. Excess HCl was removed by bubbling N[0557] 2 through the mixture and the solvent was removed in vacuo. The residue was washed with ether and dried. To the resulting solid was added THF (10 ml) containing DIEA (1.3 ml), BOP Reagent (938 mg) and (1S,3R)-3-(tert-butoxycarbonyl)-2,2,3-trimethylcyclopentanecarboxylic acid (480 mg), which was prepared by the saponification of methyl (1S,3R)-3-(tert-butoxycarbonyl)-2,2,3-trimethylcyclopentanecarboxylate derived from methyl (1S,3R)-3-carboxy (or chlorocarbonyl)-2,2,3-trimethylcyclopentanecarboxylate and t-BuOH. The mixture was stirred overnight under N, and the solvent was removed in vacuo.
  • 1N HCl (10 ml) was added to the residue and the mixture was extracted with AcOEt. The extract was washed with 1N HCl, brine; sat. NaHCO[0558] 3, brine, sat. LiCl, and brine, dried over MgSO4, and evaporated in vacuo.
  • The residue was purified by column chromatography on silica gel (eluent; 3:2, Hexane/AcOEt) to give (1S-cis)-N-[[3-[(1,1-Dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester (C[0559] 27H30Cl2N2O6) (1.05 g) as a colorless solid.
  • [0560] 1H NMR (300 MHz, CDCl3), δ 7,58 (2H), 7.3-7,4 (3H), 7.11 (2H), 5.78 (1H), 4.8-5.0 (1H), 3.75 (3H), 3.10 (2H), 2.50 (1H), 2.0-2.2 (1H), 1.6-1.8 (1H), 1.44 (9H), 1.23 (3H), 1.16 (3H), 0.80 (3H); 13C NMR (75 MHz, CDCl3), δ 175.07, 172.59, 172.17, 162.54, 136.32, 132.83, 132.48, 131.11, 130.01, 128.28, 120.56, 80.28, 56.76, 54.53, 53.12, 52.48, 46.46, 37.26, 32.41, 28.14, 23.06, 22.55, 22.06, 20.68; ESMS (m/z) 605 (MH+); Anal. Calcd for C31H38Cl2N2O6.½H2O: C, 60.53; H. 6.35; N, 4.56; Found: C, 60.71; H, 6.31; N, 4.52. MS (m/z): 605 (MH+).
  • Example 53 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine Methyl Ester (C27H30Cl2N2O6)
  • [0561]
    Figure US20030130349A1-20030710-C00154
  • TFA (1.5 ml) was added to a solution of (1S-cis)-N-[[3-[(1,1-Dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester (C[0562] 27H30Cl2N2O6) (290 mg) in CH2Cl2 (1.5 ml) and the mixture was stirred for 3 hr. The solvent was removed in vacuo and the residue was triturated with ether/CHCl3 to give (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester (180 mg) as a colorless powder.
  • [0563] 1H NMR (300 MHz, Acetone-d6), δ 9.75 (1H), 7.71 (2H), 7.4-7.6 (3H), 7.25 (2H), 7.12 (1H), 4.7-4.85 (1H), 3.68 (3H), 3.08 (2H), 2.85 (1H), 2.5-2.6 (1H), 1.6-1.8 (1H), 1.4-1.5 (1H), 1.26 (3H), 1.19 (3H), 0.80 (3H); 13C NMR (75 MHz, Acetone-dQ), δ 176.29, -172.20, 172.11, 162.00, 137.49, 133.12, 131.85, 131.12, 129.71, 128.16, 119.59, 55.98, 53.63, 52.89, 51.37, 46.14, 36.67, 32.47, 22.57, 22.04, 21.41, 20.74; ESMS (m/z) 549 (MH+); Anal. Calcd for C27H30Cl2N2O6.½H2O: C, 58.02; H, 5.55; N, 5.01; Found: C, 58.70; H, 5.53; N, 5.01. MS (m/z): 549 (MH+)
  • Example 54 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C26H28Cl2N2O6)
  • [0564]
    Figure US20030130349A1-20030710-C00155
  • A solution of LiOH (19 mg) in H[0565] 2O (1 ml) was added to a solution of (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester (C27H30Cl2N1O6) (108 mg) in THF (4 ml)/MeOH (1 ml). The mixture was stirred for 2 hr. acidified with 1N HCl (15 ml), and extracted with AcOEt. The extract was washed with brine, dried over MgSO4, and evaporated in vacuo to give (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (80 mg), m.p. 231-233° C.
  • [0566] 1H NMR (300 MHz, Acetone-d6). δ 9.72 (1H), 7.71 (2H) 7.47-7.48 (3H), 7.27 (2H), 7.07 (1H), 4.78-4.85 (1H), 3.15 (2H), 2.85 (1H), 2.5-2.6 (1H), 1.6-1.8 (1H), 1.4-1.5 (1H), 1.26 (3H), 1.19 (3H), 0.81 (3H); 13C NMR (75 MHz, Acetone-d6), δ 177.67, 173.83, 173.62, 163.32, 138.74, 134.64, 133.17, 132.42, 131.10, 129.47, 120.88, 57.29, 54.69, 54.28, 47.47, 37.93, 33.81, 23.91, 23.39, 22.73, 22.08; ESMS (m/z) 535 (MH+), 533 (M−H); Anal. Calcd for C26H28Cl2N2O6.½H2O: C, 57.46; H, 5.34; N, 5.15; Found: C, 57.48; H. 5.38; N, 5.15. MS (m/z): 533 ([M−H]); MS (m/z): 578 (MH+).
  • Example 55 (1S-cis)-N-[[3[(1,1 Dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine
  • [0567]
    Figure US20030130349A1-20030710-C00156
  • LiOH ('177 mg) in H[0568] 2O (10 ml) was added to a mixture of (1S-cis)-N-[(3-[(1,1-Dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl)carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine methyl ester (2.18 g) in THF (10 ml)/MeOH (5 ml) and the mixture was stirred at room temperature for 3 hr, acidified with 1N HCl (20 ml), and extracted with AcOEt. The extract was dried over Na2SO4 and the solvent was removed in vacuo to give (1S-cis)-N-[(3-[(1,1-Dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl)carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine (2.07 g). MS (m/z): 578 (MH+).
  • Example 56 (1S-cis)-4-Amino-N-[[3-[(1,1-, dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-L-phenylalanine Methyl Ester
  • [0569]
    Figure US20030130349A1-20030710-C00157
  • Two methods for the preparation of Example 56 are taught according to Scheme 5a and 5b. [0570]
  • Example 57 (1S-cis)-N-[[3-[(1,1 Dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,5-dichlorobenzoyl)amino]-L-phenylalanine Methyl Ester
  • [0571]
    Figure US20030130349A1-20030710-C00158
  • BOP-Cl (425 mg) and 2,5-dichlorobenzoic acid (319 mg) were added to a mixture of (1S-cis)-4-Amino-N-L[3-[(1,1-dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-L-phenylalanine methyl ester (433 mg) and DIEA (0.7 ml) in CH[0572] 2Cl2 (5 ml). The mixture was stirred for 3 hr at room temperature, acidified with 1N HCl (50 ml) and extracted with CH2Cl2. The extract was dried over Na2SO4 and evaporated in vacuo. The residue was purified by column chromatography on silica gel (eluent; Hexane 50% Hexane/AcOEt) to give (1S-cis)-N-[[3-[(1,1-Dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,5-dichlorobenzoyl)amino]-L-phenylalanine methyl ester (804 mg). MS (m/z): 605 (MH+).
  • Example 58 (1S-cis)-N-[[3-[(1,1-Dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,4,6-trichlorobenzoyl)amino]-L-phenylalanine Methyl Ester
  • [0573]
    Figure US20030130349A1-20030710-C00159
  • The preparation of Example 58 is taught by Scheme 13. [0574]
  • Example 59 (1S-cis)-4-Acetylamino-N-[[3-[(1,1-dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-L-phenylalanine Methyl Ester
  • [0575]
    Figure US20030130349A1-20030710-C00160
  • Acetic anhydride (1 ml) was added to a mixture of (1S-cis)-4-Amino-N-[[3-[(1,1-dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-L-phenylalanine methyl ester (710 mg) and DIEA (4 ml) in CH[0576] 2Cl2 (5 ml). The mixture was stirred for 30 minutes at room temperature and partitioned between sat. NaHCO3 and AcOEt. The organic layer was dried over Na2SO4 and the solvent was removed in-vacuo. The residue was purified by column chromatography on silica gel (eluent; Hexane→EtOH/AcOEt (1:1)) to give (1S—cis)-4-Acetylamino-N-[[3-[(1,1-dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-L-phenylalanine methyl ester (669 mg). MS (m/z): 475 (MH+).
  • Examples 60-153 were prepared in a similar manner, as described in Examples 52-59, and are shown in Table 4. [0577]
    TABLE 4
    Examples 60 through 153:
    Figure US20030130349A1-20030710-C00161
    physico-
    Ex. chemical
    No. R19 R0 R15 Z1 R13 property
    60 H CH3
    Figure US20030130349A1-20030710-C00162
    single bond OH gum MS (m/z): 565 (MH+)
    61 H H
    Figure US20030130349A1-20030710-C00163
    single bond OH M.P.: 168-171° C. MS (m/z): 549 ([M − H])
    62 H CH3 NO2 —OCH2
    Figure US20030130349A1-20030710-C00164
    gum MS (m/z): 581 (MH+)
    63 H H NO2 —OCH2
    Figure US20030130349A1-20030710-C00165
    M.P.: 92-94° C. MS (m/z): 565 ([M − H)
    64 H CH3 OH —OCH2
    Figure US20030130349A1-20030710-C00166
    gum MS (m/z): 552 (MH+)
    65 H H OH —OCH2
    Figure US20030130349A1-20030710-C00167
    MS (m/z): 538 (MH+)
    66 H H
    Figure US20030130349A1-20030710-C00168
    —OCH2
    Figure US20030130349A1-20030710-C00169
    M.P.: 118-121° C. MS (m/z): 696 (MH+)
    67 H H
    Figure US20030130349A1-20030710-C00170
    single bond OH M.P.: 236-238° C. MS (m/z): 538 (MH+)
    68 H H NO2 single OH M.P.: 122-
    bond 125° C.
    MS (m/z):
    407 ([M − H])
    69 H H CH3CO single OH M.P: 105-
    bond 108° C.
    MS (m/z):
    404 ([M − H])
    70 H CH3
    Figure US20030130349A1-20030710-C00171
    —OCH2
    Figure US20030130349A1-20030710-C00172
    gum MS (m/z): 710 (MH+)
    71 H CH3 NO2 single OH gum
    bond MS (m/z):
    423 (MH+)
    72 H CH3 H —NHCO— PhCH2O gum
    MS (m/z):
    511 (MH+)
    73 H H H —NHCO— PhCH2O M.P.: 95-
    97° C.
    MS (m/z):
    495 ([M − H])
    74 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00173
    gum MS (m/z): 487 (MH+)
    75 H H H —NHCO—
    Figure US20030130349A1-20030710-C00174
    M.P.: 151-154° C. MS (m/z): 471 ([M − H])
    76 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00175
    gum MS (m/z): 482 (MH+)
    77 H H H —NHCO—
    Figure US20030130349A1-20030710-C00176
    M.P.: 102-104° C. MS (m/z): 466 ([M − H])
    78 H CH3 H —NHCO— CH3 gum
    MS (m/z):
    419 (MH+)
    79 H H H —NHCO— CH3 M.P. 238-
    240° C.
    MS (m/z):
    403 ([M − H])
    80 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00177
    gum MS (m/z): 509 (MH+)
    81 H H H —NHCO—
    Figure US20030130349A1-20030710-C00178
    M.P.: 195-198° C. MS (m/z): 495 (MH+); MS (m/z): 493 ([M − H])
    82 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00179
    gum MS (m/z): 517 (MH+)
    83 H H H —NHCO—
    Figure US20030130349A1-20030710-C00180
    M.P.: 150-152° C. MS (m/z): 503 (MH+)
    84 H CH3 H —NHCO— Ph gum
    MS (m/z):
    481 (MH+)
    85 H H H —NHCO— Ph M.P.: 145-
    148° C.
    MS (m/z):
    465 ([M − H])
    86 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00181
    gum MS (m/z): 549 (MH+)
    87 H H H —NHCO—
    Figure US20030130349A1-20030710-C00182
    M.P.: 155-158° C. MS (m/z): 533 ([M − H])
    88 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00183
    gum MS (m/z): 585 (MH+)
    89 H H H —NHCO—
    Figure US20030130349A1-20030710-C00184
    M.P.: 190-193° C. MS (m/z): 569 ([[M − H]]
    90 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00185
    gum MS (m/z): 526 (MH+)
    91 H H H —NHCO—
    Figure US20030130349A1-20030710-C00186
    M.P.: 146-149° C. MS (m/z): 512 (MH+)
    92 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00187
    gum MS (m/z): 583 (MH+)
    93 H H H —NHCO—
    Figure US20030130349A1-20030710-C00188
    MS (m/z): 569 ([M − H])
    94 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00189
    gum MS (m/z): 511 (MH+)
    95 H H H —NHCO—
    Figure US20030130349A1-20030710-C00190
    M.P.: 140-143° C. MS (m/z): 497 (MH+)
    96 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00191
    gum MS (m/z): 637 (MH+)
    97 H H H —NHCO—
    Figure US20030130349A1-20030710-C00192
    M.P.: 170-173° C. MS (m/z): 623 ([M − H])
    98 t-Bu CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00193
    gum MS (m/z): 589 (MH+)
    99 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00194
    gum MS (m/z): 533 (MH+)
    100 H H H —NHCO—
    Figure US20030130349A1-20030710-C00195
    M.P.: 165° C. (dec.) MS (m/z): 517 ([M − H])
    101 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00196
    gum MS (m/z): 526 (MH+)
    102 H H H —NHCO—
    Figure US20030130349A1-20030710-C00197
    MS (m/z): 512 (MH+)
    103 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00198
    gum MS (m/z): 617 (MH+)
    104 H H H —NHCO—
    Figure US20030130349A1-20030710-C00199
    M.P.: 148-150° C. MS (m/z): 601 ([M − H])
    105 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00200
    gum MS (m/z): 529 (MH+)
    106 H H H —NHCO—
    Figure US20030130349A1-20030710-C00201
    M.P.: 165-168° C. MS (m/z): 513 ([M − H])
    107 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00202
    gum MS (m/z): 549 (MH+)
    108 H H H —NHCO—
    Figure US20030130349A1-20030710-C00203
    M.P.: 180-183° C. MS (m/z0: 533 ([M − H])
    109 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00204
    gum MS (m/z): 607 (MH+)
    110 H H H —NHCO—
    Figure US20030130349A1-20030710-C00205
    M.P: 162-165° C. MS (m/z): 591 ([M − H])
    111 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00206
    gum MS (m/z): 559, 561 (MH+)
    112 H H H —NHCO—
    Figure US20030130349A1-20030710-C00207
    M.P.: 156-158° C. MS (m/z): 543 ([M − H])
    113 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00208
    gum MS (m/z): 549 (MH+)
    114 H H H —NHCO—
    Figure US20030130349A1-20030710-C00209
    M.P.: 155-157° C. MS (m/z): 533 ([M − H])
    115 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00210
    gum MS (m/z): 557 (MH+)
    116 H H H —NHCO—
    Figure US20030130349A1-20030710-C00211
    M.P.: 162-165° C. MS (m/z): 541 ([M − H])
    117 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00212
    gum MS (m/z): 531 (MH+)
    118 H H H —NHCO—
    Figure US20030130349A1-20030710-C00213
    M.P.: 235-237° C. MS (m/z): 517 ([M − H])
    119 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00214
    gum MS (m/z): 515 (MH+)
    120 H H H —NHCO—
    Figure US20030130349A1-20030710-C00215
    M.P.: 143-145° C. MS (m/z): 499 ([M − H]]
    121 H H H —NHCO— PhCH2 M.P.: 240-242° C. MS (m/z): 479 ([M − H])
    122 H H H —NHCO—
    Figure US20030130349A1-20030710-C00216
    M.P.: 212-214° C. MS (m/z): 563 ([M − H])
    123 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00217
    gum MS (m/z): 531 (MH+)
    124 H H H —NHCO—
    Figure US20030130349A1-20030710-C00218
    M.P.: 141-143° C. MS (m/z): 515 ([M − H])
    125 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00219
    gum MS (m/z): 515 (MH+)
    126 H H H —NHCO—
    Figure US20030130349A1-20030710-C00220
    M.P.: 145-147° C. MS (m/z): 499 ([M − H])
    127 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00221
    gum MS (m/z): 515 (MH+)
    128 H H H —NHCO—
    Figure US20030130349A1-20030710-C00222
    M.P.: 185-188° C. MS (m/z): 499 ([M − H])
    129 t-Bu CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00223
    gum MS (m/z): 553 (MH+)
    130 H H H —NHCO—
    Figure US20030130349A1-20030710-C00224
    M.P.: 158-161° C. MS (m/z): 455 ([M − H])
    131 t-Bu CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00225
    gum MS (m/z): 527 (MH+)
    132 H H H —NHCO—
    Figure US20030130349A1-20030710-C00226
    M.P.: 117-120° C. MS (m/z): 483 ([M − H])
    133 H CH3 H —NHCO— i-Bu gum
    MS (m/z):
    461 (MH+)
    134 H H H —NHCO— i-Bu M.P.: 146-
    148° C.
    MS (m/z):
    445 ([M − H])
    135 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00227
    gum MS (m/z): 569 (MH+)
    136 H H H —NHCO—
    Figure US20030130349A1-20030710-C00228
    M.P.: 160-163° C. MS (m/z): 553 ([M − H])
    137 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00229
    gum MS (m/z): 495 (MH+)
    138 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00230
    gum MS (m/z): 569 (MH+)
    139 H H H —NHCO—
    Figure US20030130349A1-20030710-C00231
    M.P.: 166-169° C.
    140 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00232
    gum MS (m/z): 599 (MH+)
    141 H H H —NHCO— i-BuO M.P.: 125-
    128° C.
    MS (m/z):
    461 ([M − H])
    142 H H H —NHCO—
    Figure US20030130349A1-20030710-C00233
    M.P.: 152-155° C. MS (m/z): 479 ([M − H])
    143 H H H —NHCO—
    Figure US20030130349A1-20030710-C00234
    M.P.: 148-150° C. MS (m/z): 481 ([M − H])
    144 H H H —NHCO—
    Figure US20030130349A1-20030710-C00235
    M.P.: 195-198° C. MS (m/z): 480 ([M − H])
    145 H CH3 H —NHCO—
    Figure US20030130349A1-20030710-C00236
    gum MS (m/z): 536 (MH+)
    146 H H H —NHCO
    Figure US20030130349A1-20030710-C00237
    M.P.: 208-211° C. MS (m/z): 520 ([M − H])
    147 H CH3 H —NHSO2
    Figure US20030130349A1-20030710-C00238
    gum MS (m/z): 585 (MH+)
    148 H H H —NHSO2
    Figure US20030130349A1-20030710-C00239
    M.P.: 224-226° C. MS (m/z): 569 ([M − H])
    149 H CH3 H —NHSO2
    Figure US20030130349A1-20030710-C00240
    gum MS (m/z): 531 (MH+)
    150 H H H —NHSO2
    Figure US20030130349A1-20030710-C00241
    M.P.: 220-223° C. MS (m/z): 515 ([M − H])
    151 H CH3 H —NHCH2
    Figure US20030130349A1-20030710-C00242
    gum MS (m/z): 535 (MH+)
    152 H H H —NHCH2 M.P.: 100-103° C. MS (m/z): 519 ([M − H])
    153 H H H —CONH—
    Figure US20030130349A1-20030710-C00243
    MS (m/z): 533 ([M − H])
  • Example 154 (1S-cis)-N-[[3-(Aminocarbonyl)-2,2,3-trimethylcyclopentyl]carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine Methyl Ester
  • [0578]
    Figure US20030130349A1-20030710-C00244
  • BOP Reagent (674 mg) was added to a mixture of (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine methyl ester (743 mg) and aq. NH[0579] 4OH (0.5 ml) in THF (10 ml). The mixture was stirred for 24 hr. and sat. LiCl (15 ml) was added. The resulting mixture was extracted with AcOEt and the extract was dried over Na2SO4. The solvent was removed in vacuo and the residue was purified by column chromatography on silica gel (eluent; Hexane 90% AcOEt/Hexane) to give (1S-cis)-N-[[3-(Aminocarbonyl)-2,2,3-trimethylcyclopentyl]carbonyl]O-[(2,6-dichlorophenyl)methyl]-L-tyrosine methyl ester (533 mg). MS (m/z): 535 (MH+),
  • Example 155 (1S-cis)-N-[[3-(Aminocarbonyl)-2,2,3-trimethylcyclopentyl]carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine
  • [0580]
    Figure US20030130349A1-20030710-C00245
  • A solution of LiOH (119 mg) in H[0581] 2O (5 ml) was added to a mixture of (1S-cis)-N-[[3-(Aminocarbonyl)-2,2,3-trimethylcyclopentyl]carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine methyl ester (533 mg) in THF (5 ml)/MeOH (3 ml). The mixture was stirred for 3 hr. at room temperature, acidified with 1N HCl (20 ml), and extracted with AcOEt. The extract was dried over Na2SO4 and evaporated in vacuo to give (1S-cis)-N-[[3-(Aminocarbonyl)-2,2,3-tri-methylcyclopentyl]carbonyl]-O-[(2,6-dichlorophenyl)-methyl]-L-tyrosine (466 mg), mp 102-104° C., MS (m/z): 519 ([M−H]).
  • Examples 156-166 were prepared in a similar manner, as described in Examples 154 and 155 and are shown in Table 5. [0582]
    TABLE 5
    Figure US20030130349A1-20030710-C00246
    Examples 156 through 166:
    Ex. physicochemical
    No. R19 R20 R0 R6 property
    156
    Figure US20030130349A1-20030710-C00247
    H H
    Figure US20030130349A1-20030710-C00248
    M.P.: 85-87° C. MS (m/z): 533 ([M − H])
    157 Et H CH3
    Figure US20030130349A1-20030710-C00249
    gum MS (m/z): 563 (MH+)
    158 Et H H
    Figure US20030130349A1-20030710-C00250
    MS (m/z): 550 (MH+)
    159 Me H CH3
    Figure US20030130349A1-20030710-C00251
    gum; 1H NMR (300 MHz, CD3OD), δ 7.3-7.4(3H), 7.14(2H), 6.95(2H), 5.22(2H), 4.71(1H), 3.69(3H), 3.12(1H), 3.00(1H), 2.39(3H), 2.3-2.4(1H), 2.0-2.1(1H), 1.7-1.8(1H), 1.4-1.5(1H), 1.27(3H), 1.18(3H), 0.73(3H).
    160 Me H H
    Figure US20030130349A1-20030710-C00252
    M.P.: 80-83° C. MS (m/z): 533 ([M − H])
    161 Me Me H
    Figure US20030130349A1-20030710-C00253
    1H NMR(300 MHz, CDCl3), δ 7.3-7.4(3H), 7.16(2H), 7.00(2H), 5.25(2H), 4.75(1H), 3.66(3H), 3.22(1H), 3.13(1H), 2.5-2.7(1H), 2.1-2.2(1H), 2.05(3H), 2.03(3H), 1.7-1.8(1H), 1.4-1.6(1H), 1.20(3H), 1.19(3H), 0.73(2H).
    162 MeO H CH3
    Figure US20030130349A1-20030710-C00254
    gum MS (m/z): 565 (MH+)
    163 MeO H H
    Figure US20030130349A1-20030710-C00255
    M.P.: 70-72° C. MS (m/z): 551 ([M − H])
    164 Me H H
    Figure US20030130349A1-20030710-C00256
    MS (m/z): 509 (MH+)
    165 Et H H
    Figure US20030130349A1-20030710-C00257
    MS (m/z): 523 (MH+)
    166 H H H
    Figure US20030130349A1-20030710-C00258
    M.P.: 196-198° C. MS (m/z): 532 ([M − H])
  • Example 167 (1R-cis)-N-[[3-[(1,1-Dimethylethoxy)carbonyl]-1,2,2-trimethylcyclopentyl]carbonyl]-O-(phenylmethyl)-L-tyrosine methyl ester
  • [0583]
    Figure US20030130349A1-20030710-C00259
  • Example 167 was prepared from O-benzyl-L-tyrosine methyl ester hydrochloride and (1R,3S)-3-(tert-butoxycarbonyl)-1,2,2-trimethylcyclopentanecarboxylic acid in a similar manner as described in Example 52. Physicochemical property: gum, MS (m/z): 524 (MH[0584] +).
  • Example 168 (1R-cis)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-C-(phenylmethyl)-L-tyrosine Methyl Ester (C27H33NO6)
  • [0585]
    Figure US20030130349A1-20030710-C00260
  • Example 168 was prepared in a similar manner as described in Example 53. Physicochemical property: gum; MS (m/z): 468 (MH[0586] +); MP 191-192° C. (d).
  • Example 169 (1R-cis)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-O-(phenylmethyl)-L-tyrosine (C26H31NO6)
  • [0587]
    Figure US20030130349A1-20030710-C00261
  • Example 169 was prepared in a similar manner as described in Example 54, MS (m/z): 454 (MH[0588] +).
  • Example 170 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[[N-acetyl-N-(2,6-dichlorophenyl)methyl]amino]-L-phenylalanine Methyl Ester
  • [0589]
    Figure US20030130349A1-20030710-C00262
  • (1S-cis)-N-[[3-[(1,1-Dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[N-[(2,6-dichlorophenyl)methyl]amino]-L-phenylalanine methyl ester (0.29 g), which was derived from 4-[[N-(2,6-dichlorophenyl)methyl]amino]-L-phenylalanine methyl ester in a similar manner as described in Example 52, was dissolved in pyridine (3 ml) and Ac[0590] 2O (2.5 ml) was added. The mixture was stirred overnight, evaporated in vacuo, and extracted with AcOEt. The extract was washed with aq. HCl, brine, sat. NaHCO3, brine, and dried over Na2SO4. The solvent was removed in vacuo to give (1S-cis)-N-[[3-[(1,1-dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[[N-acetyl-N-[(2,6-dichlorophenyl)methyl]amino]-L-phenylalanin methyl ester (0.24 g). The obtained diester was treated in a similar manner as described in Examples 53 and 54 to give (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[[N-acetyl-N-[(2,6-dichlorophenyl)methyl]amino]-L-phenylalanine methyl ester as a colorless solid, mp 241-244° C., MS (m/z): 561 ([M−H]).
  • Example 171 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)methyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine Methyl Ester (C17H33Cl2NO5)
  • [0591]
    Figure US20030130349A1-20030710-C00263
  • NaCNBH[0592] 3 (104 mg) was added to a mixture of O-[(2,6-dichlorophenyl)methyl]-L-tyrosine methyl ester (490 mg), (1R,cis)-3-formyl-1,2,2-trimethylcyclopentanecarboxylic acid, (1R,5S)-4-hydroxy-1,8,8-trimethyl-3-oxabicyclo[3,2,1]-octane-2-one (153 mg), AcOH (0.5 ml), and MeOH (25 ml) under argon. The mixture was stirred for 72 hr. at room temperature. The solvent was removed in vacuo and 10% HCl (20 ml) was added. The resulting mixture was stirred for 2 hr. and extracted with AcOEt. The extract was dried over Na2SO4 and evaporated in vacuo. The residue was purified by column chromatography on silica gel (eluent; 10% AcOEt/Hexane) to give (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)methyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine methyl ester (75 mg), MS (m/z) 522 (MH+)
  • Example 172 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)methyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine (C26H31Cl2NO5)
  • [0593]
    Figure US20030130349A1-20030710-C00264
  • A solution of LiOH (33 mg) in H[0594] 2O (2 ml) was added to a mixture of (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)methyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine methyl ester (C27H33Cl2NO5) (71 mg) in THF (2 ml). The mixture was stirred for 5 hr. at room temperature, neutralized with 1N HCl, and extracted with AcOEt. The extract was dried over Na2SO4 and evaporated in vacuo to give (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)methyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine (40 mg), mp 135-138° C., MS (m/z): 508 (MH+).
  • Example 173 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-N-methyl-O-(phenylmethyl)-L-tyrosine (C27H33NO6)
  • [0595]
    Figure US20030130349A1-20030710-C00265
  • (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-(phenylmethyl)-L-tyrosine (C[0596] 26H31NO6) (52 mg) was added to a suspension of NaH (20.5 mg, 60% in oil) in DMF (5 ml) and the mixture was stirred for 10 min at room temperature. After addition of MeI (43 ml), the mixture was stirred for 14 hr. and quenched with H2O. The resulting mixture was extracted with ether. The extract was evaporated in vacuo and the residue was subjected to saponification using aq. LiOH in THF in a similar manner as described in Example 54. The saponification was carried out for 2 hr. and the mixture was acidified with 1N HCl (10 ml) and extracted with AcOEt. The extract was dried over Na2SO4, evaporated in vacuo, and the residue was purified by column chromatography on silica gel (eluent; 10% MeOH/CH2Cl2) to give (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-N-methyl-O-(phenylmethyl)-L-tyrosine (40 mg), MS (m/z): 466 ([M−H])
  • Example 174 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-[(2,6-dichlorophenyl)methyl]-N-methyl-L-tyrosine (C27H31Cl2NO6)
  • [0597]
    Figure US20030130349A1-20030710-C00266
  • Example 174 was prepared from N-(tert-butoxycarbonyl)-O-[(2,6-dichlorophenyl)methyl]-N-methyl-L-tyrosine and (1S,3R)-3-(tert-butoxycarbonyl)-2,2,3-trimethylcyclopentanecarboxylic acid in a similar manner as described in Example 52, 53 and 54, mp 108-110° C., MS (m/z): 536 (MH[0598] +).
  • Example 175 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosinamide (C26H30Cl2N2O5)
  • [0599]
    Figure US20030130349A1-20030710-C00267
  • NaCN (10 mg) was added to a solution of (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-[2,6-dichlorophenyl)methyl]-L-tyrosine methyl ester (306 mg) in MeOH (10 ml) in a thick walled tube. 15-20 ml of NH[0600] 3 was condensed into the mixture at −78° C. The tube was sealed and warmed to room temperature. After 48 hr. stirring, the mixture was cooled to −78° C. and the sealed tube was opened. The mixture was warmed to room temperature and excess NH3 was removed by bubbling N2. The solvent was removed in vacuo and the residue was purified by column chromatography on silica gel (eluent; AcOEt <10% MeOH/AcOEt) to give (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosinamide (258 mg), mp 120-122° C., MS (m/z): 519 ([M−H]-).
  • Example 176 [1S-[1α(R*),3α]-4-(2,6-Dichlorobenzoyl)-α-[[[3-[(1,1-dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]amino]-γ-oxo-1-piperazinebutanoic Acid Methyl Ester
  • [0601]
    Figure US20030130349A1-20030710-C00268
  • The synthesis of Example 176 is taught by Scheme 17. [0602]
  • Example 177 [1S[1α(R*),3α]]-α-[[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]amino]-4-(2,6-dichiorobenzoyl)-γ-oxo-1-piperazinebutanoic Acid
  • [0603]
    Figure US20030130349A1-20030710-C00269
  • The synthesis of Example 177 is taught by Scheme 17. [0604]
  • Example 178 [1S-[1α((R*),3α]]-4-(2,6-Dichlorobenzoyl)-β-[[[3-[(1,1-dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]amino]-γ-oxo-1-piperazinebutanoic Acid Methyl Ester
  • [0605]
    Figure US20030130349A1-20030710-C00270
  • The synthesis of Example 178 is taught by Scheme 18. [0606]
  • Example 179 [1S-[1α,3α(R*)]-N-[[3-[[(5-Amino-1-carboxypentyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzyl)amino]-L-phenylalanine
  • [0607]
    Figure US20030130349A1-20030710-C00271
  • (1) To a solution of N-(tert-butoxycarbonyl)-4-(2,6-dichlorobenzamido)-L-phenylalanine (9.25 g) in DMF (80-100 ml) was added Merrifield resin (10.0 g, 10.0 meq/g, Novabiochem) and anhydrous potassium fluoride (1.57 g). The reaction mixture was stirred for 1 day at 80° C. and the resulting resin bound amino acid derivative was collected by filtration, washed thoroughly with DMF, 50% aqueous DMF, CH[0608] 3OH, CH2Cl2, CH3OH, and then dried in vacuo to give the resin bound N-(tert-butoxycarbonyl)-4-(2,6-dichlorobenzamido)-L-phenylalanine (0.53 meq/g). Substitution of the amino acid derivative onto the resin was estimated using the picric acid method.
  • (2) To the obtained resin (1.0 g, 0.53 meq/g) was added 50% TFA/CH[0609] 2Cl2 (10-15 ml) and the mixture was stirred for 30 min. The resin was collected by filtration, washed with CH2Cl2, CH3OH, and CH2Cl2. To the resin bound 4-(2,6-dichlorobenzamido)-L-phenylalanine was added a mixture of (1S,3R)-3-(tert-butoxycarbonyl)-2,2,3-trimethylcyclopentanecarboxylic acid (408 mg), 0.5M HBTU-HOBT (3.3 ml) in DMF, DIEA (0.694 ml), DMF (10-15 ml) and the mixture was vortexed for 2 hrs. at room temperature. The resin was collected by filtration, washed with DMF, CH2Cl2, CH3OH, CH2Cl2 to give a resin bound (JS-cis)-N-[[3-[(1,1-dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (0.990 g). To the mixture
  • (3) To the resin (0.390 g, 0.21 meq/g) was added 50% TFA/CH[0610] 2Cl2 (4-5 ml) and the mixture was stirred for 2 hrs. The resin was collected by filtration, washed with CH2Cl21 CH3OH, and CH2Cl2. To the resin bound (1S-cis)-N-[[3-Carboxy-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine was added a mixture of N6-(tert-butoxycarbonyl)-L-lysine trert-butyl ester (0.210 g), 0.5M HBTU-HOBT (1.5 ml) in DMF, DIEA (0.270 ml) and the mixture was vortexed for 2 hrs. The resin was collected by filtration, washed with DMF, CH2Cl2, CH3OH, and CH2Cl2. To the obtained resin was added 50% TFA/CH2Cl2 (4-5 ml) and the mixture was stirred for 30 min. The resin was collected by filtration, washed with CH2Cl2, CH3OH, and CH2Cl2. To the resin was added THF (3 ml), CH3OH (0.9 ml) and 2N LiOH (0.3 ml). The mixture wasd stirred for 15 mins and filtered to a test tube (13×100 mm). The resin was washed with THF/5% CH3OH and the combined filtrate was evaporated. The residue was dilued with H2O (1 ml) and acidified with 1N HCl. The precipitate was centrifuged. The supernatant was decanted, and the pellet was washed with H2O. The residue was lyophilized to give a colorless solid (100 mg).
  • (4) The solid (80 mg) was purified by a Water Delta Prep 3000 system (Waters, Milford, Mass.) equipped with a reversed phase silica C18 column (4.7 cm×30.0 cm), using a linear gradient of increasing acetonitrile in an aqueous solution of triethylammonium phosphate (TEAP) (prepared by diluting phosphoric acid (25 ml) and triethylamine (50 ml) to 6000 ml of deionized water, pH −5.5). The fractions containing desired compound was desalted with 0.1% acetic acid using the above system The collected fractions were lyophilized to give [1S-[1α,3α(R*)]-N-[[3-[[(5-Amino-1-carboxypentyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (Example 179) as a colorless solid (10 mg), ESMS: 661 (M−H])[0611] .
  • Example 180 (1S-cis)-N-[[3-[[(2-Amino-2-oxoethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C28H32Cl2N4O6)
  • [0612]
    Figure US20030130349A1-20030710-C00272
  • The synthesis of Example 180 is taught by Scheme 8, 8-G: wherein R[0613] 8-1=H, R8-2=H, R4=H, R6=4-[(2,6-Dichlorobenzoyl)amino]-phenyl, Stereochemistry=(1S-cis)-L. Accordingly, the synthesis of (1S-cis)-N-[[3-[[(2-Amino-2-oxoethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl) amino]-L-phenylalanine (C28H32Cl2N4O6) is as follows.
  • To 8-F-1 (8F: R[0614] 8-1=H, R8-2=HE, R4=H, R5=CO2CH3, R6=4-[(2,6-dichlorobenzoyl)amino]-phenyl, Stereochemistry=(1S-cis)-L) (1.05 g, 1.7 mmol), dissolved in methanol (30 mL), is added a solution of LiOH2H2O (0.32 g, 7.65 mmol) in water (10 mL), dropwise over 15 minutes. The mixture stirs for 18 hours at room temperature and the pH is then adjusted to ca. 7 by the careful addition of 1N aq. HCl. The majority of the methanol, is removed in vacuo and the pH of the resulting solution is adjusted to ca. 2 with 1N aq. HCl. The resulting flocculent white precipitate is isolated by filtration and dried. The solid is crushed and washed with water (2×10 mL) and dried in vacuo at 50‘C’ to give 0.97 g (97%) of Example 180 (8-G: R8-1=H, R8-2=H, R4=H, R6=4-[(2,6-dichlorobenzoyl)amino]-phenyl) as a white, powdery solid.
  • MP: 203-205° C.; [0615] 1H NMR (300 MHz, DMSO-d6): δ 12.51 (brs, 1H), 10.70 (s, 1H), 7.75 (m, 1H), 7.45-7.57 (3H), 7.33 (m, 1H), 7.20 (m, 2H), 7.11 (brs, 1H), 6.92 (brs, 1H), 4.43 (m, 1H), 3.63 (m, 1H), 3.47 (m, 2H), 3.30 (s, 2H), 3.01 (m, 1H), 2.84 (m, 1H), 2.31 (m, 1H), 1.87 (m, 1H), 1.55 (m, 1H), 1.31 (m, 1H), 1.17 (s, 3H), 1.08 (s, 3H), 0.59 (s, 3H); IR (Mull): 3511, 3325, 3128, 3082, 2868, 1722, 1697, 1664, 1614, 1555, 1537, 1417, 1337, 1246, 799; MS (FAB) m/z (rel. intensity) 591 (M+H, base), 517 (32), 335 (26), 239 (32), 173 (39), 109 (63), 57 (80); HRMS (FAB) calcd for C28H32Cl2N4O6 +H+ 591.1777, found 591.1747.
  • Example 181 (1S-cis)-N-[[3-[[(Carboxymethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C28H31 1C2N3O7)
  • [0616]
    Figure US20030130349A1-20030710-C00273
  • The synthesis of Example 181 is taught by scheme 7, 7-G: wherein R[0617] 7-1=H, R4=H, R5=CO2H, R6=4-[(2,6-Dichlorobenzoyl)amino]phenyl and Stereochemistry=(1S-cis)-L. Accordingly, (1S-cis)-N-[[3-[[(Carboxymethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C28H31Cl2N3O7) is synthesized as follows:
  • (1S-cis)-N-[[3-[[(Carboxymethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester (0.7 g, 1.15 mmol) is dissolved in methanol (12 mL). To this is added a mixture of LiOH.H[0618] 2O (0.243 g, 5.8 mmol), aqueous H2O2 (30%, 2 mL), and H2O (2 mL). After overnight stirring, the reaction mixture is diluted with water (50 mL), and evaporated (room temperature, in vacuo/N2 flow) until the methanol is gone. The aqueous solution is then transferred to a separatory funnel and shaken with diethyl ether (2×20 mL). The aqueous layer is then evaporated, to remove residual diethyl ether, and cooled in an ice water bath. The stirred solution is then brought to pH 3-4 using aqueous HCl (1N). The resultant precipitate is isolated by suction filtration (with water washes) to give the target compound as a white solid (0.4 g, 58% yield)
  • [0619] 1HNMR: (300 MHz, DMSO-d): δ 12.45 (br.s, 1H), 10.6 (s, 1H), 7.74 (m, 2H), 7.57-7.44 (m, 3H), 7.20 (m, 2H), 4.48-4.40 (m, 1H), 3.65 (m, 2H), 2.94 (m, 2H), 2.64 (m, 1H), 2.35 (m, 1H), 1.90 (m, 1H), 1.58 (m, 1H), 1,29 (m, 1H), 1.18 (s, 3H), 1.08 (s, 3H), 0.60 (s, 3H); IR (mull) 3124, 3088, 3078, 1738, 1666, 1628, 1612, 1588, 1563, 1552, 1521, 1429, 1334, 1197, 1170 cm−1; MS (FAB) m/z (rel. intensity) 592 (M+H, 99), 595 (20), 594 (69), 593 (41), 592 (99), 519 (25), 517 (38), 240 (55), 175 (23), 173 (33), 109 (64); HRMS (FAB) m/z calcd for C28H31Cl2N3O7+H+ 592.1617, found 592.1606; Anal. Calcd for C28H31Cl2N3O7: C, 56.76; H, 5.27; N, 7.09; Found: C, 54.92; H, 5.41; N, 6.91; KF Water: 3.05F H2O.
  • Example 182 [1S-[1α,3α(E)-N-[[3-(2-Carboxyethenyl)-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine Disodium Salt (C28H28Cl2N2Na2O6)
  • [0620]
    Figure US20030130349A1-20030710-C00274
  • The synthesis of Example 182 is taught by Scheme 11, 11-G: wherein R[0621] 4=H, R5=CO2H R6=4-[(2,6-Dichlorobenzoyl)amino]-phenyl and Stereochemistry=[1S-[1α,3α(E)]]-L. Accordingly, [1S-[1α,3α(E)]]-N-[[3-(2-Carboxyethenyl)-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine disodium salt (C28H28Cl2Na2N2O6) of 11-G is synthesized as follows:
  • To a solution of 11-F (R[0622] 4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorobenzoyl)amino]-phenyl, Stereochemistry=[1S-[1α,3α(E)]]-L) (0.45 g, 0.78 mmol) in methanol (5 mL) in a flask cooled in an ice water bath is added a solution of LiOH.H2O (0.127 g, 3 mmol) in H2O (5 mL). After two days, the mixture is diluted with water (50 mL), evaporated in vacuo until the methanol is gone and then cooled to 1° C. and brought to pH 2 using 1N HCl. The resultant white precipitate is isolated by suction filtration to give a white solid which is stirred with saturated aqueous NaHCO3 (2 mL) and then transferred to a C-18 reversed phase HPLC column and eluted with a gradient from 0.01% aq NaHCO3 to 100% acetonitrile/0.01% aq NaHCO3. Evaporation in vacuo to give Example 182 as a white solid (0.25 g, 51% yield).
  • [0623] 1H NMR (300 MHz, DMSO-d6) δ 7.51-7.42 (m, 5H), 7.06 (m, 2H), 6.39 (m, 1H), 5.49 (m, 1H), 4.10 (m, 1H) 2.99 (m, 1H), 2.86 (m, 1H), 2.56 (m, 1H), 1.85 (m, 2H), 1.61 (m, 1H), 1.29 (m, 1H), 0.90 (s, 3H), 0.85 (s, 3H), 0.51 (s, 3H); IR (mull) 3393, 3257, 3124, 3035, 1654, 1604, 1562, 1544, 1515, 1431, 1398, 1325, 799, 773, 722 cm−1; MS (FAB) m/z (rel. intensity) 605 (M+H, 44), 629 (9), 627 (14), 608 (8), 607 (30), 606 (14), 605 (44), 585 (14), 583 (21)l, 73 (45), 23 (99); KF Water: 7.09%H2O.
  • Example 183 (1S-cis)-N-[[3-[(Carboxymethoxy)methyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C28H31Cl2N3O7)
  • [0624]
    Figure US20030130349A1-20030710-C00275
  • The synthesis of Example 183 is taught by Scheme 9 and by the narrative accompanying Scheme 9. [0625]
  • Example 184 (1S-cis)-N-[(3-Cyano-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C26H27Cl2N3O4)
  • [0626]
    Figure US20030130349A1-20030710-C00276
  • The synthesis of Example 184 is taught by Scheme 12 under the heading Preparation of Example 184. [0627]
  • Example 185 (1S-cis)-N-[[3-(Carboxymethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine (C28H32Cl2N2O7)
  • [0628]
    Figure US20030130349A1-20030710-C00277
  • The synthesis of Example 185 is taught by scheme 7, 7-G: wherein R[0629] 7-1=H, R4=H, R5=CO2H, R6=4-(2,6-Dichlorophenyl)methoxy]-phenyl and Stereochemistry=(1S-cis)-L. Accordingly, (1S-cis)-N-[[3-[[(Carboxymethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine (C28H32Cl2N2O7) is prepared from 7-G-2 (7-G: R7-1=H, R4=H, R5=CO2CH3, R6=4-[(2,6-Dichlorophenyl)methoxy]phenyl, Stereochemistry=(1S-cis)-L as like as Example 181.
  • [0630] 1H NMR (300 MHz, DMSO-d6) δ 708-6.9 (m, 9H), 5.18 (s, 2H), 4.42 (m, 1H), 3.8-3.6 (m, 2H) 3.02-2.82 (m, 2H), 2.65 (m, 1H), 2.38 (m, 1H), 1.91 (m, 1H), 1.58 (m, 1H), 1.30 (m, 1H), 1.19 (s, 3H), 1.10 (s, 3H), 0.61 (s, 3H); IR (mull) 3409, 1733, 1645, 1612, 1585, 1564, 1511, 1439, 1297, 1239, 1197, 1179, 1018, 786, 770 cm−1; MS (FAB) m/z (rel. intensity) 579 (M+H, 99), 582 (22), 581 (67), 580 (44), 579 (99), 578 (21), 240 (34), 161 (21), 159 (34), 109 (46), 91 (37); HRMS (FAB) calcd for C28H32Cl2N2O+H+ 579.1664, found 579.1667.
  • Example 186 (1R-cis)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl) carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine (C26H29Cl2NO6)
  • [0631]
    Figure US20030130349A1-20030710-C00278
  • Compound III-a (where R[0632] 4=H, R5a=CO2CH3, R6=4-[(2,6-dichlorophenyl)methoxy]phenyl, n=1, Stereochemistry S) and (1R)-camphoric anhydride were heated together in diisopropylethyl amine. The crude mixture was concentrated in vacuo to yield crude I-b (where R1=H, R3=CH3, R4=H, R5a=CO2CH3, R6=4-(2,6-dichlorophenyl)methoxy]phenyl, n=1, Stereochemistry=(1R-cis)-L). I-b was hydrolyzed with LiOH to yield Example 186 (1-a; where R1=H, R3=CH3, R4=H, R5=CO2H, and R6=4-[(2,6-dichlorophenyl)methoxy]phenyl, n=1, Stereochemistry (1R-cis)-L): 13C NMR δ 175.9, 175.4, 173.0, 158.6, 137.3, 133.0, 132.0, 131.3, 130.9, 129.7, 115.5, 65.9, 56,5, 54.2, 53.0, 47,0, 36.8, 33.1, 23.2, 22.8, 22.0, 21.0.
  • Example 187 (1S-cis)-N-[[3-[[(2-Amino-2-oxoethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine (C28H33Cl2N3O6)
  • [0633]
    Figure US20030130349A1-20030710-C00279
  • The synthesis of Example 187 is taught by Scheme 8, 8-G: wherein R[0634] 8-1=H, R8-2=H, R4=H, R6=4-[(2,6-2,6-Dichlorophenyl)methoxy]-phenyl Stereochemistry=(1S-cis)-L. Accordingly, (1S-cis)-N-[[3-[[(2-Amino-2-oxoethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine (C28H33Cl2N3O6) is prepared from 8-F-2 (R8-1=H, R8-2H, R4=H, R5=CO2CH3, R6=4-[(2,6-dichlorophenyl)methoxy]-phenyl, Stereochemistry=(1S-cis)-L) as taught by Scheme 8.
  • [0635] 1H NMR (300 MHz, DMSO-d6) δ 7.74 (m, 2H), 7.55-7.40 (m, 4H,/, 7.15 (m, 3H), 6.94 (m, 3H), 5.16 (s, 2H), 4.41 (m, 1H), 3.75-3.48 (m, 2H), 3.1-2.8 (m, 211H), 2.63 (m, 1H), 2.33 (m, 1H), 1.87 (m, 1H), 1.54 (m, 1H), 1.32 (m, 1H), 1.17 (s, 3H), 1.08 (s, 3H), 0.58 (s, 3H); MS (FAB) m/z (rel. intensity) 578 (M+H, 99), 581 (30), 580 (72), 579 (57), 578 (99), 577 (19), 504 (17), 322 (18), 239 (35), 161 (29), 159 (34); HRMS (FAB) calcd for C28H33Cl2N3O6 +H+ 578.1824, found 578.1836.
  • Example 188 [1S-[1α,3α(R*) 1-N-[[3-(1-Carboxyethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine (C29H34Cl2N2O7)
  • [0636]
    Figure US20030130349A1-20030710-C00280
  • The synthesis for Example 188 is taught by scheme 7, 7-G: wherein R[0637] 7-1=CH3, R4=H, R5=CO2H, R6=4-[(2,6-Dichlorophenyl)methoxy]-phenyl and Stereochemistry [1S-[1α,3α(R*)]-L. Accordingly, [1S-[1α,3α(R*)]]-N-[[3-[[(1-Carboxyethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine (C29H34Cl2N2O7) is prepared from 7-G-3 as taught by Scheme 7, 1H NMR (300 MHz, DMSO-d6) δ 7.71 (m, 1H), 7.54-7.43 (m, 3H), 7.28 (m, 1H), 7.16 (m, 2H), 6.93 (m, 2H), 5.16 (s, 2H), 4.40 (m, 1H), 4.16 (m, 1H), 3.02-2.80 (m, 2H), 2.63 (m, 1H), 2.35 (m, 1H), 1.86 (m, 1H), 1.54 (m, 1H), 1.35-1.23 (m, 4H) 1.14 (s, 3H), 1.08 (s, 3H), 0.59 (s, 3H); IR (mull) 3427, 3031, 1731, 1645, 1612, 1585, 1565, 1512, 1439, 1297, 1239, 1230, 1197, 1179, 1017 cm−1; MS (FAB) m/z (rel. intensity) 593 (M+H, 99), 596 (22), 595 (69), 594 (43), 593 (99), 592 (17), 504 (22), 254 (63), 161 (44), 159 (40), 109 (72)
  • Example 189 (1R-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine (C26H29Cl2NO6)
  • [0638]
    Figure US20030130349A1-20030710-C00281
  • The ester III-a (R[0639] 4=H, R5a=CO2CH3, R6=4-[(2,6-dichlorophenyl)methoxy]phenyl, n=1, Stereochemistry=R) (391 mg, 1 mmol), THF (3 mL), and diisopropylethyl amine (880 μL, 5 mmol) were combined. (1S)-Camphoric anhydride (33-A) (182 mg, 1 mmol) was added and the reaction was heated at reflux for 18 h. The reaction mixture was then cooled and concentrated in vacuo. The material was treated with 1 N HCl (1 mL) and extracted with EtOAc (3×5 mL). The organic portion was dried and concentrated in vacuo to yield 590 mg of crude methyl ester I-a as a mixture of regioisomers and diastereomers. Only characteristic and easily discernable protons are listed for the major product (1R-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine methyl ester (1-a). 1H NMR (CDCl3) δ 0.75 (s, H), 1.00 (s, H), 1.19 (s, H), 3.75 (s, H), 4.84-5.00 (m, H), 5.23 (s, H).
  • The crude methyl ester I-a (560 mg, 1 mmol) was combined with LiOH[0640] 2O (420 mg, 10 mmol) that was dissolved in 6 mL of H2O. After 4.5 h of stirring on the rotovap, 1 N HCl (12 mL) was added to the reaction mixture and a white precipitate was formed. The mixture was-extracted with EtOAc (3×20 mL). The organic portion was dried and concentrated in vacuo to afford 480 mg of crude di-acid Example 189. The material was purified by a Cl8 reverse phase chromatography (on the Delta Prep employing a Delta-PAK C18, 15 μM particle size column that was conditioned with CH3CN/H2O/TFA (40:60:0.1)). The material was eluted with CH3CN/H2O (38:62) to yield 190 mg (36% overall from anhydride) of Example 189: [α]D˜4.5° (c=0.6, EtOH), [α]D[2.8° (c=0.8, EtOH); 1H NMR (DMSO-d6) δ 0.21 (s), 0.59 (s), 0.86 (s), 1.05-1.18 (m), 1.28-1.45 (m), 1.64-1.82 (m), 2.05-2.20 (m), 2.41 (m), 2.57 (m), 2.87 (m), 4.22-4.32 (m), 4.95 (s), 6.73 (m), 6.97 (m), 7.20-7.30 (m), 7.34 (m), 7.61 (m); 13C NMR (DMSO-d6) δ 176.9 (s), 173.4 (s), 172.0 (s), 157.1 (s), 136.0 (s), 131.8 (s), 131.5 (d), 130.3 (s), 130.1 (d), 128.7 (d), 114.3 (d), 64.9 (t), 55.4 (s), 53.5 (d), 52.2 (d), 45.5 (s), 35.9 (t), 32.3 (t), 22.4 (t), 22.0 (q), 21.5 (q), 20,8 (q); IR (mineral oil mull) 3296, 2925, 1712, 1698, 1651, 1512, 1438, 1242, cm−1; MS for C26H29Cl2NO6, m/Z (relative intensity) 523 (M+, 1), 521 (M+, 1), 503 (1), 477 (1), 324 (29), 322 (45), 267 (19), 265 (28), 161 (64), 159 (100). Anal. Calcd for C26H29Cl2NO6: C, 59.78; H, 5.60; Cl, 13.57; N, 2.68. Found: C, 59.41; H, 5.55; Cl, 13.43; N, 2.52. Corrected for 1.14% H2O found by Karl Fisher analysis.
  • Example 190 [1S-[1α(R*),3α]]-N-[[3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-α-methoxymethyl-4-[(2,6-dichlorophenyl)methoxy]benzene-ethanamine
  • [0641]
    Figure US20030130349A1-20030710-C00282
  • The synthesis of Example 190 is taught by Scheme 25. [0642]
  • Example 191 (1S-cis]-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-(hydroxyphenylmethyl)-L-phenylalanine (C26H31NO6)
  • [0643]
    Figure US20030130349A1-20030710-C00283
  • The synthesis of Example 191 is taughtby scheme 2, I-c: wherein R[0644] 2a=—CO2H, R1=Me, R3=R4=H, R5=—CO2H, R6=4-(hydroxyphenylmethyl)-phenyl, X=—C(O)— and Stereochemistry=(1S-cis)-L. Accordingly, (1S-cis]-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-(hydroxyphenylmethyl)-L-phenylalanine (C26H31NO6) is synthesized as follows:
  • To a solution of Example 40 (0.59 g, 1.3 mmol) in absolute EtOH under N[0645] 2 at 0° C. is added NaBH4 (100 mg, 2.7 mmol). The reaction mixture is stirred for 17 h at room temperature diluted with H2O, and quenched with 1 N HCl. The solution is treated with satd NH4Cl and adjusted to approximately pH 3 with 1 N HCl The mixture is extracted with EtOAc. The combined extracts are dried, filtered and concentrated to give a colorless oil (0.60 g). The product is purified by C18 reverse phase chromatography (isocratic 35:65 CH3CN/H2O). The product (0.29 g) is diluted with MeOH/H2O, frozen and lyophilized to give Example 191 as a white powder: TLC Rf=0.18 (650:350:1 CHCl3/acetone/HCO2H); HPLC tR=7.3 min (isocratic 650:350:1 CH3CN/H2O/TFA); [α]D 25+26° (c 0.48, MeOH); IR (mull) 3327, 1709, 1653, 1514, 1242, 1207, 1118, 1017, 950, 700 cm31 1; 1H NMR (CD3OD) δ 7.37-7.14 (9H), 5.73 (1H), 4.90 (2H), 4.66 (1H), 3.20 (1H), 2.96 (1H), 2.70-2.62 (1H), 2.48 (1H), 1.92 (1H), 1.59 (1H), 1.40 (1H), 1.19 (3H), 1.18 (3H), 0.74 (3H); 13C NMR (CD3OD) δ 178.28, 173.84, 144.56, 142.97, 136.31, 128.84, 127.86, 126.79, 126.33, 126.31, 75.37, 56.00, 53.16, 46.19, 39.04, 36.69, 32.33, 22.43, 21.72, 21.04, 20.43; MS (FAB, HR) m/z 454.2234 (calcd [M+H]+454.2229); MS (FAB) m/z 493, 454, 476, 436, 237, 226, 208, 109; Anal. C, 65.54; H, 6.74; N, 3.12 (calcd C, 68.86; H, 6.89; N, 3.09.
  • Example 192 (1S-cis)-N-[[3-(hydroxymethyl)-2,2,3-trimethylcyclopentyl]carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine (C26H31Cl2NO5)
  • [0646]
    Figure US20030130349A1-20030710-C00284
  • The synthesis of Example 192 is taught by Scheme 10 under the heading Preparation of Example 192. [0647]
  • Example 193 (1R-cis)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-O-(2,6-dichlorophenyl)methyl]-D-tyrosine (C26H29Cl2NO6)
  • [0648]
    Figure US20030130349A1-20030710-C00285
  • MeOH (10 mL) was cooled to 0° C. and acetyl chloride (2.14 mL, 30 mmol) was added slowly. After 20 min the t-BOC-D-tyrosine derivative III-a (where R[0649] 4=H, R5a=CO2CH3, R6=4-[(2,6-dichlorophenyl)methoxy]phenyl, n=1, Stereochemistry=R), (2.64 g, 6 mmol) was added. The reaction mixture was stirred at room temperature for 2 h. The mixture was concentrated under vacuum to afford a white solid. The white solid was triturated with Et2O and filtered. The solid was returned to the flask and was combined with THF (15 mL), diisopropylethyl amine (4.4 mL, 25 mmol), and (1R)-camphoric anhydride (1.0 g, 5 mmol). The reaction mixture was heated at reflux for 18 h. The mixture was then cooled and concentrated in vacuo. The material was treated with 1 N HCl (40 mL) and extracted with EtOAc (3×20 mL). The organic portion was dried and concentrated in vacuo to yield 3.30 g of crude methyl ester I-a as a mixture of regioisomers and diastereomers.
  • The crude methyl ester I-a (2.79 g, 5 mmol) was combined with LiOH.H[0650] 2O (2.1 g, 50 mmol) that was dissolved in 30 mL of H2O. After 4.0 h of stirring on the rotovap, 1 N HCl (60 mL) was added to the reaction and a white precipitate formed. The mixture was extracted with EtOAc (3×40 mL). The organic portion was dried and concentrated in vacuo to afford 3.05 g of crude di-acid. The material was chromatographed on 150 g of silica gel eluting with CHCl2/EtOAc/HCO2H (50:50:0.1) to yield pure (1R-cis)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-O-(2,6-dichlorophenyl)methyl]-D-tyrosine (Example 193). This material was dissolved in 20 mL of CH3CN/H2O (1:1). The mixture was concentrated in vacuo until the liquid appeared milky. The solution was frozen and lyophilized to afford Example 193: 1H NMR (DMSO-d6) δ 0.55 (H), 1.13 (H), 1.25 (H), 1.30-1.45 (1H), 1.68-1.86 (1H), 1.95-2.122 (1H), 2.25-2.41 (1H), 2.73, 2.95-3.20, 4.49-4.62, 5.23, 7.01, 7.25, 7.30-7.40, 7.45-7.70; 13C NMR (DMSO-d6) δ 175.3 (s), 174.5 (s), 173.5 (s), 157.3 (s), 136.3 (s), 132.0 (d), 131.7 (s), 130.9 (s), 130.5 (d), 129.0 (d), 114.4 (d), 65.1 (t), 55.3 (s), 53.6 (d), 52.2 (d), 46.3 (s), 35.6 (t), 32.1 (t), 23.2 (q), 22.2 (t), 21.4 (q), 20.6 (q); IR (mineral oil mull) 3436, 2922, 1722, 1707, 1634, 1612, 1512, 1438, 1242, cm−1; MS for C26H29Cl2NO6, m/z (relative intensity) 523 (M+, 0.4), 521 (M+, 0.5), 505 (2), 503 (2), 324 (33), 322 (50), 267 (21), 265 (32), 161 (69), 159 (100). Anal. Calcd for C26H29Cl2NO6: C, 59.78; H, 5.60; Cl, 13.57; N, 268. Found: C, 59.52; H, 5.29; Cl, 13.60; N, 2.52. Corrected for 0.9% H2O found by Karl Fisher analysis.
  • Example 194 [1S[1α(R*),3α]]-α-[[(3-Carboxy-2,2,3-trimethylcyclopentyl) carbonyl]amino]-6-chloro-5-[(2,6-dichlorophenyl)methoxy]-2-pyridinepropanoic Acid
  • [0651]
    Figure US20030130349A1-20030710-C00286
  • The synthesis of Example 194 is taught by Scheme 34. [0652]
  • Example 195 [1S-[1α(R*),3α]]-β-[[(3-Carboxy-2,2,3-trimethylcyclopentyi)carbonyl]amino]-4-(2,6-dichlorobenzoyl)-γ-oxo-1-piperazinebutanoic Acid
  • [0653]
    Figure US20030130349A1-20030710-C00287
  • The synthesis of Example 195 is taught by Scheme 18. [0654]
  • Example 196 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-1-(phenylmethyl)-L-tryptophan (C28H32N2O5)
  • [0655]
    Figure US20030130349A1-20030710-C00288
  • Example 196 was synthesized as described Scheme 24. [0656]
  • Example 197 [1R-cis]-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-(hydroxyphenylmethyl)-L-phenylalanine (C26H31NO6)
  • [0657]
    Figure US20030130349A1-20030710-C00289
  • The synthesis of Example 197 is taught by scheme 2, I-c: wherein R[0658] 2a=—CO2H, R1=R4=H, R3=Me, R5=—CO2H, R6=4-(hydroxyphenylmethyl)-phenyl, X=—C(O)— and Stereochemistry=(1R-cis)-L. Accordingly, [1R-cis]-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-(hydroxyphenylmethyl)-L-phenylalanine (C26H31NO6) is prepared from 32-B and (1R)-camphoric anhydride by the procedures taught by Scheme 2 and by the synthesis of Examples 40, 186, and 191: TLC silica gel Rf=0.31 (900:100:1 CHCl3/MeOH/HCO2H); HPLC tR=5.6 min (isocratic 650:350:1 CH3CN/H2O/TFA); [α]D 25+24° (c 0.38, MeOH); UV (MeOH) λmax (ε) 223 (14200), 254 (425 sh), 258 (493), 263 (481), 268 (364 sh), 272 (257 sh); IR (mull) 3379, 1712, 1642, 1514, 1417, 1377, 1342, 1274, 1179, 1133, 1017, 700 cm−1; 1H NMR (CD3OD) δ 7.35-7.15 (m 9H), 5.73 (1H), 4.91 (3H), 4.72 (1H), 3.26 (1H), 2.99 (1H), 2.72 (1H), 2.44-2.30 (1H), 2.15-2.00 (1H), 1.82-1.67 (1H), 1.40-1.27 (1H), 1.05 (3H), 1.03 (3H), 0.54 (3H), 0.53 (3H); 13C NMR (CD3OD) δ 176.29, 176.24, 173.56, 144.56, 143.17, 136.26, 136.23, 128.82, 127.88, 126.85, 126.39, 126.31, 75.33, 55.77, 53.34, 52.47, 46.20, 36.09, 32.03, 22.11, 21.98, 20.17, 20.07; MS (HR FAB) m/z 454.2230 (calcd [M+H]+454.2229); MS (FAB) m/z 454, 436, 237, 226, 208, 109; H2O (Karl Fischer) 1.44W; Anal. C, 67.68; H, 6.98; N, 3.06 (calcd adjusted for H2O: C, 68.86; H, 6.89; N, 3.09).
  • Example 198 (1S-cis)-N-[[(3-methoxymethyl)-2,2,3-trimethylcyclopentyl]carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-tyrosine (C27H33Cl2NO5)
  • [0659]
    Figure US20030130349A1-20030710-C00290
  • The synthesis of Example 198 is taught by Scheme 10 under the heading Preparation of Example 198. [0660]
  • Example 199 (1R-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-O-[(2,6-dichlorophenyl)methyl]-D-tyrosine (C26H29Cl2NO6)
  • [0661]
    Figure US20030130349A1-20030710-C00291
  • In the same manner as the synthesis of 189, 199 was synthesized and purified (0.5 g, 49% overall yield): [α][0662] D=−31°.
  • Example 200 [1R-[1α,3α(S*)]]-N-[[3-[[[1-Carboxy-2-[4-[(2,6-dichlorophenyl)methoxy]phenyl]ethyl]-amino]carbonyl]-1,2,2-trimethylcyclopentyl]carbonyl]-L-proline 1,1-dimethylethyl Ester, Monosodium Salt (C35H43C2N2NaO7)
  • [0663]
    Figure US20030130349A1-20030710-C00292
  • The sythesis of Example 200 is taught by Scheme 21. [0664]
  • Example 201 [1S-[1α(R*),3α]]-Z-[[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]amino]-4-[(2,6-dichlorophenyl)methoxy]-benzenebutanoic Acid (C27H31Cl2NO6)
  • [0665]
    Figure US20030130349A1-20030710-C00293
  • The sythesis of Example 201 is taught by scheme 2, I-c: wherein R[0666] 2a=—CO2H, R1=Me, R3=R4=H, R5=—CH2CO2H, R6=4-[(2,6-Dichlorophenyl)methoxy]-phenyl-, n=1, Stereochemistry=[1S-[1α(R*),3α]]. Accordingly, [1S-[1α(R*),3α]]-β-[[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]amino]-4-[(2,6-dichlorophenyl)methoxy]-benzenebutanoic acid (C27H31Cl2NO6) is prepared from 30-B and 15-D as taught by Scheme 2. TLC: Rf=0.22 (50:50.0.2 hexanes/ELOAc/HCO2H); [α]D+3 (MeOH); IR 3320, 3057, 3026, 2953, 2924, 2868, 2855, 1703, 1641, 1612, 1585, 1565, 1511, 1465, 1439, 1403, 1378, 1297, 1283, 1241, 1197, 1179, 1158, 1017, 768 cm−1; 1H NMR δ 0.71 (3H), 1.22 (3H), 1.23 (3H), 1.41-1.53 (1H), 1.67-1.86 (1H), 2.26-2.60 (4H), 2.71-2.87 (2H), 2.96-3.08 (1H), 4.46-4.62 (1H); 5.26 (2H), 5.43 (1H), 6.98 (2H), 7.13 (2H), 7.23-7.28 (1H), 7.37 (2H); 13C NMR δ 20.22, 21.66, 22.29, 22.91, 31.91, 37.19, 39.64, 46.44, 48.57, 54.26, 56.48, 65.14, 115.09, 128.37, 129.62, 130.23, 130.33, 131.98, 136.90, 157.72, 171.75, 178.54, 182.19; MS (FAB) m/z 536, 495, 464, 449, 431, 418, 386, 353, 336, 287, 236; H2O (Karl Fischer) 0.70%; Anal. C, 60.08; H, 5.86; Cl, 13.05; N, 2.70 (calcd corrected for H2O: C, 60.03; H, 5.86; Cl 13.13; N, 2.59).
  • Example 202 (1S-cis)-1-[(3,4-Dichlorophenyl)methyl]-N-[[3-[(1,1-dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-L-histidine (C27H35Cl2N3O5)
  • [0667]
    Figure US20030130349A1-20030710-C00294
  • To (1S-cis)-N-([3-[(1,1-dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-1-[(3,4-dichlorophenyl)methyl]-L-histidine methyl ester (0.5 g, 0.88 mmol) in H[0668] 2O (10 mL) was added LiOH.H2O (10 equiv, 8.8 mmol, 0.37 g) in cold H2O (10 mL). Stirred reaction overnight. Lowered pH of reaction solution to 5 with 1N HCl. Filtered resulting precipitate, washed precipitate with H2O and dried precipitate under hi vacuum conditions to yield 0.46 g (95%) of (1S-cis)-1-[(3,4-Dichlorophenyl)methyl]-N-[[[3-[(1,1-dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-L-histidine Example 202 (I-c; where R2a=(1,1-dimethylethoxy)carbonyl, R1=CH3, R3=H, R4=H, R5=CO2H, and R6=1-(3,4-dichlorophenyl)methyl]-4-imidazolyl, n=1, Stereochemistry=(1S-cis)-L): mp 234-235° C.
  • Example 203 (1S-cis)-N-[[(3-Methoxymethoxymethyl)-2,2,3-trimethylcyclopentyl]carbonyl]-O-[(2,6-dichlorophenyl)methyl]-L-r-yrosine (C27H33Cl2NO5)
  • [0669]
    Figure US20030130349A1-20030710-C00295
  • The synthesis of Example 203 is taught by Scheme 10 under the heading Preparation of Example 203. [0670]
  • Example 204 [1R-[1α(S*)3α]]-β-[[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]amino]-4-[(2,6-dichlorophenyl)methoxy]-benzenebutanoic Acid (C27H31Cl2NO6)
  • [0671]
    Figure US20030130349A1-20030710-C00296
  • The synthesis of Example 204 is taught by scheme 2, I-c: wherein R[0672] 2a=—CO2H, R1=R4=H, R3=Me, R5-CH2CO2H, R6=4-[(2,6-Dichlorophenyl)methoxy]phenyl-, n=1, Stereochemistry=[1R-[1α(S*),3α]]. Accordingly, [1R-[1α(S*),3α]]-β-[[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]amino]-4-[(2,6-dichlorophenyl)methoxy]benzenebutanoic acid (C27H31Cl2NO6) is obtained from 30-B and (1S)-camphoric anhydride (33-A) by the procedures taught by Scheme 1: mp 106-108° C.; TLC Rf=0.24 (250:250:1 hexanes/EtOAc/HCO2H) [a]D=+13 (MeOH); IR 3144, 3057, 3030, 2954, 2924, 2870, 2855, 1706, 1027, 1612, 1585, 1565, 1511, 1466, 1439, 1377, 1299, 1241, 1197, 1178, 1017, 1000, 778, 768 cm−1; 1H NMR δ 0.70 (3H), 1.16 (3H), 1.16 (3H), 1.30 (3H), 1.43-1.61 (1H), 1.77-1.97 (1H), 2.09-2.32 (2H), 2.36-2.49 (1H), 2.57-2.68 (1H), 2.69-2.83 (2H), 2.90-3.02 (1H), 4.44-4.59 (1H), 5.25 (2H), 5.73 (1H), 6.97 (2H), 7.11 (2H), 7.22-7.26 (3H), 7.37 (2H); 13C NMR δ 20.88, 21.16, 22.04, 23.15, 32.57, 37.76, 39.59, 47.39, 48.16, 52.44, 55.73, 65.17, 86.13, 115.02, 128.37, 129.56, 130.34, 132.00, 136.89, 157.72, 174.53, 178.42, 181.12; MS (FAB) m/z 536, 518, 490, 378, 354, 336, 294, 159, 137, 109, 88, 69, 55; H2O (Karl Fischer) 0.52%; Anal. C, 60.07; H, 5.84; Cl, 13.03; N, 2.71 (calcd corrected for H2O: C, 60.14; H, 5.85; Cl, 13.15; N, 2.60),
  • Example 205 (1S-cis)-O-[(2,6-Dichlorophenyl)methyl]-N-[[3-(methoxycarbonyl)-2,2,3-trimethylcyclopentyl]carbonyl]-L-tyrosine (C27H31Cl2NO5)
  • [0673]
    Figure US20030130349A1-20030710-C00297
  • Example 205 is taught by Scheme 20, 20-D: wherein R[0674] 4=H, R5=CO2H, R6=4-[(2,6-Dichlorophenyl)methoxy]-phenyl and Stereochemistry=[1S-cis]-L.
  • To a solution of the diester 20-D (R[0675] 4=H, R5=CO2CH3; R6=4-[(2,6-Dichlorophenyl)methoxy]phenyl, Stereochemistry=(1S-cis)-L (0.32 g, 0.58 mmol) in methanol (5 mL) is added a solution of LiOH.H2O (0.31 g, 7.4 mmol) in H2O (5 mL). After overnight stirring, the mixture is diluted with water (50 mL) and evaporated in vacuo until the methanol is gone. The aqueous solution is cooled in an ice bath and brought to pH 4 using 1N HCl, resulting in a white precipitate which is chromatographed on silica gel (10% methanol/chloroform) to give a white solid (0.25 g, 80% yield).
  • [0676] 1H NMR (D6DMSO) 7.85 (m, 1H), 7.58-7.44 (m, 3H), 7.19 (m, 2H), 6.96 (m, 2H), 5.18 (s, 2H), 4.43 (m, 1H), 3.59 (s, 3H), 3.03-2.81 (m, 2H), 2.69 (m, 1H), 2.40 (m, 1H), 1.91 (m, 1H), 1.56 (m, 1H), 1.39 (m, 1H), 1.16 (s, 3H), 1.31 (s, 3H), 0.59 (s, 3H); IP (mull) 1727, 1639, 1612, 1585, 1565, 1511, 1439, 1298, 1241, 1197, 1179, 1160, 1119, 779, 768; MS (FAB) m/z (rel. intensity) 536 (M+H, 99), 538 (66), 537 (39), 536 (99), 322 (22), 197 (27), 169 (21), 159 (29), 137 (31), 109 (66), 107 (22).
  • Example 206 (1S-cis)-4-[(2,6-Dichlorobenzoyl)amino]-N-[2,2,3-trimethyl-3-[[(phenylmethoxy)amino]-carbonyl]cyclopentyl]carbonyl]-L-phenylalanine Methyl Ester (C34H37Cl2N3O6)
  • [0677]
    Figure US20030130349A1-20030710-C00298
  • The synthesis of Example 206 is taught by Scheme 23 under the heading Preparation 23-C. [0678]
  • Example 207 (1S-cis)-N-[[3-[[(2-Cyanoethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine Methyl Ester
  • [0679]
    Figure US20030130349A1-20030710-C00299
  • The synthesis of Example 207 is taught by Scheme 22 under the heading Preparation of 22-C. [0680]
  • Example 208 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[[(2-chloro-3-pyridinyl)carbonyl]amino]-L-phenylalanine (C25H28ClN3O6)
  • [0681]
    Figure US20030130349A1-20030710-C00300
  • The synthesis of Example 208 is taught by Scheme 2, I-c: wherein R[0682] 2a=—CO2H, R1=Me, R3=R4=H, R5=—CO2H, R6=4-[[(2-chloro-3-pyridyl)carbonyl]amino]phenyl, n=1, Stereochemistry=(1S-cis)-L. Accordingly, (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl3-4-[[(2-chloro-3-pyridinyl)carbonyl]amino]-L-phenylalanine (C25H28ClN3O6) is prepared from 31-C, 15-D and 2-chloro-3-nicotinic acid by the procedures taught by Scheme 2: mp 172-174° C.; TLC silica gel Rf=0.17 (750:250:3 EtOAc/hexanes/HCO2H); IR (mineral oil mull) 3410, 3289, 3193, 3124, 3050, 2955, 2924, 2868, 2855, 1714, 1655, 1606, 1582, 1537, 1516, 1461, 1415, 1401, 1377, 1329, 1279, 1257, 1242, 1207, 1187, 1152 cm−1; 1H NMR δ 0.80 (3H), 1.13-1.28 (1H), 1.16 (3H), 1.19 (3H), 1.38-1.50 (1H), 1.67-1.83 (1H), 2.08-2.23 (1H), 2.45-2.58 (2H), 3.04-3.24 (2H), 4.75-4.87 (1H), 5.96 (1H), 7.12 (2H), 7.32 (1H), 7.56 (2H), 7.96 (1H), 8.43 (1H), 9.32 (1H); MS (FAB) m/z 504, 502, 484, 468, 456, 371, 320, 302, 274, 140, 109; Anal. C, 57.54; H, 5.84; Cl, 6.79; N, 8.05 (calcd corrected for H2O. C, 57.55; H, 5.83; Cl, 6.79; N, 8.08).
  • Example 209 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[[(2,6-dichloro-3-pyridinyl)carbonyl]amino]-L-phenylalanine (C25H27Cl2N3O6)
  • [0683]
    Figure US20030130349A1-20030710-C00301
  • The synthesis of Example 209 is taught by Scheme 2, I-c: wherein R[0684] 2a=—CO2H, R1=Me, R3=R4=H, R5=—CO2H, R6=4-[[(2,6-dichloro-3-pyridyl)carbonyl]amino]phenyl, n=1, Stereochemistry=(1S-cis)-L. Accordingly, (1S-cis)-N-((3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[[(2,6-dichloro-3-pyridinyl)carbonyl]amino]-L-phenylalanine (C25H27Cl2N3O6) is prepared from 31-C, 15-D and 2,6-dichloro-3-nicotinic acid as taught by Scheme 2 and by the synthesis of Example 208: mp 246-247° C.; TLC silica gel Rf=0.28 (750:250:2 EtOAc/hexanes/HCO2H); IR (mineral oil mull) 3292, 3196, 3125, 3059, 2954, 2923, 2855, 1712, 1656, 1607, 1575, 1544, 1515, 1461, 1426, 1414, 1377, 1343, 1329, 1272, 1244, 1206, 1186, 1160, 1144 cm−1; 1H NMR δ 0.65 (3H), 1.10 (3H), 1.15 (3H), 1.24-1.37 (1H), 1.43-1.62 (1H), 1.83-1.97 (1H), 2.26-2.45 (1H), 2.67 (1H), 2.82-3.04 (2H), 3.30 (3H), 4.38-4.49 (1H), 7.21 (2 H), 7.55 (2H), 7.71 (1H), 7.80 (1H), 8.14 (1H), 10.63 (1H); MS (FAB) m/z 538, 536, 538, 518, 490, 371, 354, 336, 281, 200, 174, 137, 109.
  • Example 210 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[[(2-methoxy-6-chloropyridin-3-yl)carbonyl]-amino]-L-phenylalanine
  • [0685]
    Figure US20030130349A1-20030710-C00302
  • The synthesis of Example 210 is taught by Scheme 2, I-c: wherein R[0686] 2a=—CO2H, R1=Me, R3=R4=H, R5=—CO2H, R6=4-[[(6-chloro-2-methoxy-3-pyridyl)carbonyl]amino]phenyl, n=1, Stereochemistry=(1S-cis)-L. Accordingly, Example 210 is synthesized as follows:
  • A solution of the dimethyl ester of Example 209 (0.098 g, 0.18 mmol) in 4:1 THF/MeOH is reacted at room temperature under Ar with an aqueous solution of LiOH:H[0687] 2O (0.042 g, 1.0 mmol). The reaction mixture is stirred for 2.5 h. It is diluted with H2O, acidified with aq HCl, and extracted with EtOAc. The combined extracts are washed with brine, dried, filtered and concentrated to an oil that is purified by silica gel flash chromatography (650:350:4 EtOAc/hexanes/HCO2H). The product is azeotroped thrice from toluene, diluted with CH3CN/H2O, and lyophilized to give 0.043 g (0.081 mmol, 45%) of Example 210 as a white solid: mp 128-131 C; TLC silica gel Rf=0.13 (500:500:2 EtOAc/hexanes/HCO2H); 1H NMR δ 0.64 (3H), 1.10 (3H), 1.15 (3H), 1.23-1.38 (1H), 1.44-1.53 (m, 1H), 1.78-1.97 (m, 1H), 2.66 (t, 2H, J=9.4), 2.78-3.07 (m, 2H), 3.67-3.84 (1H), 3.96 (3H), 4.37-4.46 (1H), 7.19 (2H), 7.57 (2H), 7,78 (2H), 8.05 (1H), 10.18 (1H); MS (FAB) m/z 534, 532, 514, 486, 371, 360, 350, 332, 304, 275, 190, 127, 109.
  • Example 211 [1S-[1α,3α(R*)]]-N-[[[3-[[(1-Carboxyethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine
  • [0688]
    Figure US20030130349A1-20030710-C00303
  • The synthesis of Example 211 is taught by Scheme 19 under the heading Preparation of Example 211. [0689]
  • Example 212 [1S-[1α,3α((S*) 1-N-[3-[[2-(Aminocarbonyl)-1-pyrrolidinyl]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[[(2,6-dichlorobenzoyl)amino]-L-phenylalanine Monolithium Salt
  • [0690]
    Figure US20030130349A1-20030710-C00304
  • The synthesis of Example 212 is taught by Scheme 19 under the heading Preparation of Example 212. [0691]
  • Example 213 [1S-[1α,3α(R*)]]-N-[3-[[2-(Aminocarbonyl)-1-pyrrolidinyl]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine Monosodium Salt
  • [0692]
    Figure US20030130349A1-20030710-C00305
  • The synthesis of Example 213 is taught by Scheme 19 under the heading Preparation of Example 213. [0693]
  • Example 214 (1S-cis)-N-[[3-[[(2-Carboxyethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C29H33Cl2N3O7)
  • [0694]
    Figure US20030130349A1-20030710-C00306
  • The synthesis for Example 214 is taught by Scheme 19 under the heading Preparation of Example 214. [0695]
  • Example 215 [1S-[1α,3α(R*)]]-N-[3-[[2-[(Methylamino)carbonyl]-1-pyrrolidinyl]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine Monosodium Salt
  • [0696]
    Figure US20030130349A1-20030710-C00307
  • The synthesis for Example 215 is taught by Scheme 19 under the heading Preparation of Example 215. [0697]
  • Example 216 (1S-cis)-N-[[3-[[(2-Cyanoethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine
  • [0698]
    Figure US20030130349A1-20030710-C00308
  • The synthesis of Example 216 is taught by Scheme 22 under the heading Preparation of Example 216 [0699]
  • Example 217 (1S-cis)-4-((2,6-Dichlorobenzoyl)amino]-N-[[2,2,3-trimethyl-3-[[(phenylmethoxy)amino]-carbonyl]cyclopentyl]carbonyl]-L-phenylalanine (C33H35Cl2N3O6)
  • [0700]
    Figure US20030130349A1-20030710-C00309
  • The synthesis for Example 217 is taught by Scheme 23 under the heading Preparation of Example 217. [0701]
  • Example 218 (1S-cis)-4-[(2,6-} Dichlorobenzoyl)amino]-N-[[3-[(hydroxyamino)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-L-phenylalanine Methyl Ester
  • [0702]
    Figure US20030130349A1-20030710-C00310
  • The synthesis for Example 218 is taught by Scheme 23 under the heading Preparation of Example 218. [0703]
  • Example 219 (1S-cis)-4-[(2,6-Dichlorobenzoyl)amino]-N-[[3-[(hydroxyamino)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-L-phenylalanine
  • [0704]
    Figure US20030130349A1-20030710-C00311
  • The synthesis for Example 219 is taught by Scheme 22 under the heading Preparation of Example 219. [0705]
  • Example 220 (1S-cis)-N-[[(3-Carboxy-2,2,3-trimethylcyclopentyl]carbonyl]-4-[[[(1,1′-biphenyl)-4-yl]amino]-carbonyl]-L-phenylalanine
  • [0706]
    Figure US20030130349A1-20030710-C00312
  • The synthesis for Example 220 is taught by Schemes 35 and 2 under the heading Preparation of Example 220. [0707]
  • Example 221 (1S-cis)-[[(3-Carboxy-2,2,3-trimethylcyclopentyl]carbonyl]-4-[[(4-chlorophenyl)amino]carbonyl]-L-phenylalanine
  • [0708]
    Figure US20030130349A1-20030710-C00313
  • The synthesis for Example 221 is taught by Schemes 35 and 2 under the heading Preparation of Example 221 [0709]
  • Example 222 (1S-cis)-[[(3-Carboxy-2,2,3-trimethylcyclopentyl]carbonyl]-4-[[(2-trifluoromethylphenyl)amino]carbonyl]-L-phenylalanine
  • [0710]
    Figure US20030130349A1-20030710-C00314
  • The synthesis for Example 222 is taught by Schemes 35 and 2 under the heading Preparation of Example 222. [0711]
  • Example 223 (1S-cis)-[[(3-Carboxy-2,2,3-trimethylcyclopentyl]carbonyl]-4-[[(2,4,6-trichlorophenyl)amino]-carbonyl]-L-phenylalanine
  • [0712]
    Figure US20030130349A1-20030710-C00315
  • The synthesis for Example 223 is taught by Schemes 35 and 2 under the heading Preparation of Example 223. [0713]
  • Example 224 [1S-[160 (R*),3α]]-4-[[[(1-Carboxy-3-methylbutyl]amino]carbonyl]-N-[[(3-carboxy-2,2,3-trimethylcyclopentyl]-carbonyl]-L-phenylalanine
  • [0714]
    Figure US20030130349A1-20030710-C00316
  • The synthesis for Example 224 is taught by Schemes 35 and 2 under the heading Preparation of Example 224. [0715]
  • Example 225 (1S-cis)-4-[(2,6-Dichlorobenzoyl)amino]-N-[[2,2,3-trimethyl-3-[(4-morpholinyl)carbonyl]-cyclopentyl]carbonyl]-L-phenylalanine
  • [0716]
    Figure US20030130349A1-20030710-C00317
  • The preparation of Example 225 is taught by Scheme 27. [0717]
  • Example 226 (1S-cis)-4-[(2,6-Dichlorobenzoyl)amino]-N-[2,2,3-trimethyl-3-[(4-morpholinyl)carbonyl]-cyclopentyl]carbonyl]-L-phenylalanine Methyl Ester
  • [0718]
    Figure US20030130349A1-20030710-C00318
  • The preparation of Example 226 is taught by Scheme 27. [0719]
  • Example 227 (1S-cis)-N-[[3-[(1,1-Dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[[[(2,6-dichlorophenyl)amino]carbonyl]amino]-L-phenylalanine Methyl Ester
  • [0720]
    Figure US20030130349A1-20030710-C00319
  • The synthesis of Example 227 is taught by Scheme 26. [0721]
  • Example 228 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[[[(2,6-dichlorophenyl)amino]carbonyl]amino]-L-phenylalanine Methyl Ester
  • [0722]
    Figure US20030130349A1-20030710-C00320
  • The synthesis of Example 228 is taught by Scheme 26. [0723]
  • Example 229 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[[[(2,6-dichlorophenyl)amino]carbonyl]amino]-L-phenylalanine
  • [0724]
    Figure US20030130349A1-20030710-C00321
  • The synthesis of Example 229 is taught by Scheme 26 [0725]
  • Example 230 (1S-cis)-N-[[3-[(1,1-Dimethylethoxy)carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2-chloro-5-trifluoromethylbenzoyl)amino]-L-phenylalanine Methyl Ester
  • [0726]
    Figure US20030130349A1-20030710-C00322
  • Example is 230 synthesized in the same manner as Example 54. [0727]
  • Example 231 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2-chloro-5-trifluoromethylbenzoyl)amino]-L-phenylalanine Methyl Ester
  • [0728]
    Figure US20030130349A1-20030710-C00323
  • Example 231 is synthesized in the same manner as Example 54. [0729]
  • Example 232 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2-chloro-5-trifluoromethylbenzoyl)amino]-L-phenylalanine
  • [0730]
    Figure US20030130349A1-20030710-C00324
  • Example 232 is synthesized in the same manner as Example 54. [0731]
  • Example 233 (1S-cis)-N-[[3-[[(2-Amino-2-oxoethyl)amino]carbonyl]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine Methyl Ester
  • [0732]
    Figure US20030130349A1-20030710-C00325
  • The synthesis for Example 233 is taught by Scheme 8 under the heading Preparation of 8-F. [0733]
  • Example 234 [S-[1α(R) 3α]]-2-[[[3-Carboxy-2,2,3-trimethylcyclopentyl]carbonyl]amino]-6-[(2,6-dichlorobenzoyl)amino] Hexanoic Acid
  • [0734]
    Figure US20030130349A1-20030710-C00326
  • Example 234 was prepared according to Scheme 2. Physical properties as follows: [0735] 1H NMR (300 MHz, CD3OD) δ 7.36 (3H), 4.29 (1H), 3.24 (2H) 2.71 (1H), 2.45 (1H), 2.04 (1H), 1.71 (8H), 1.23 (3H), 1.13 (3H), 0.75 (3H); 13C NMR (75 MHz, CD3OD) δ 176.74, 173.06, 164.47, 137.20, 131.64, 130.84, 128.05, 55.91, 52.87, 46.16, 38.50, 32.31, 30.44, 28.43, 22.64, 22.32, 21.92, 21.07, 20.57; MS (FAB) m/z (rel. intensity) 501 (M+, 22), 504 (20), 503 (32), 502 (26), 501 (22), 109 (17), 73 (99), 69 (25), 57 (27), 55 (23); MS (ES−) for C23H30Cl2N2O6 m/z 499.3 (M−H); Anal. Calcd for C23H30Cl2N2O6.0.25H2O: C, 54.60; H. 6.07; N, 5.53. Found: C, 54.58; H, 6.14; N, 5.45.
  • Example 235 [1S-[1α(R*),3α]]-2-[[[3-Carboxy-2,2,3-trimethylcyclopentyl]carbonyl]amino]-5-[(2,6-dichlorobenzoyl)amino] Pentanoic Acid
  • [0736]
    Figure US20030130349A1-20030710-C00327
  • Example 235 was prepared according to Scheme 2. Physical properties as follows: [0737] 1H NMR (300 MHz, CD3OD) δ 7.38 (3H), 4.41 (1H), 3.40 (2H) 2.83 (1H), 2.53 (1H), 2.08 (1H), 1.79 (3H), 1.47 (1H), 1.30 (3H), 1.24 (3H), 0.83 (3H); 13C NMR (75 MHz, CD3OD) δ 178.23, 174.24, 173.83, 165.84, 136.07, 131.79, 130.67, 127.80, 56.06, 52.92, 52.10, 38.84, 32.35, 28.57, 25.43, 22.33, 21.61, 20.95, 20.50; MS (FAB) m/z (rel. intensity) 487 (M+, 99), 490 (14), 489 (67), 488 (25), 487 (99), 305 (19), 242 (16), 175 (12), 173 (18); Anal. Calcd for C22H28Cl2N2O6: C, 54.22; H, 5.79; N, 5.75. Found: C, 53.91; H, 5.93; N, 5.43.
  • Example 236 [1S-[1α,3α(1S*,3R*)]]-N-[[3-[[[(3-Carboxy-1,2,2-trimethylcyclopentyl)amino]carbonyl]-amino]-2,2,3-trimethylcyclopentyl]carbonyl]-4-[(2,6-dichlorobenzoyl)-amino]-L-phenylalanine
  • [0738]
    Figure US20030130349A1-20030710-C00328
  • The synthesis of Example 236 is taught by Schemes 28 and 29. [0739]
  • Example 237 (1S-cis)-N-[(3-Carboxy-2,2,3-triwethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine (C26H28Cl2N2O6)
  • [0740]
    Figure US20030130349A1-20030710-C00329
  • The synthesis for Example 237 is taught by Scheme 37 under the heading Preparation of Example 237. [0741]
  • Example 238 (1S-trans)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine (C26H28Cl2N2O6)
  • [0742]
    Figure US20030130349A1-20030710-C00330
  • The synthesis for Example 238 is taught by Scheme 37 under the heading Preparation of Example 238. [0743]
  • Example 239 (1S-trans)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C26H28Cl2N2O6)
  • [0744]
    Figure US20030130349A1-20030710-C00331
  • The synthesis for Example 239 is taught by Scheme 37 under the heading Preparation of Example 239. [0745]
  • Example 240 (1R-trans)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C26H28Cl2N2O6)
  • [0746]
    Figure US20030130349A1-20030710-C00332
  • The synthesis of Example 240 is taught by Scheme 37 under the heading Preparation of Example 240. [0747]
  • Example 241 (1R-trans)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine (C26H28Cl2N2O6)
  • [0748]
    Figure US20030130349A1-20030710-C00333
  • The synthesis of Example 241 is taught by Scheme 37 under the heading Preparation of Example 241. [0749]
  • Example 242 (1R-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine (C26H28Cl2N2O6)
  • [0750]
    Figure US20030130349A1-20030710-C00334
  • The synthesis of Example 242 is taught by Scheme 37 under the heading Preparation of Example 242. [0751]
  • Example 243 (1R-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C26H28Cl2N2O6)
  • [0752]
    Figure US20030130349A1-20030710-C00335
  • The synthesis of Example 243 is taught by Scheme 37 under the heading Preparation of Example 243. [0753]
  • Example 244 (1S-cis)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C26H28Cl2N2O6)
  • [0754]
    Figure US20030130349A1-20030710-C00336
  • The synthesis of Example 244 is taught by Scheme 37 under the heading Preparation of Example 244. [0755]
  • Example 245 (1S—cis)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine (C26H28Cl2N2O6)
  • [0756]
    Figure US20030130349A1-20030710-C00337
  • The synthesis of Example 245 is taught by Scheme 37 under the heading Preparation of Example 245. [0757]
  • Example 246 (1S-trans)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine (C26H28Cl2N2O6)
  • [0758]
    Figure US20030130349A1-20030710-C00338
  • The synthesis of Example 246 is taught by Scheme 37 under the heading Preparation of Example 246. [0759]
  • Example 247 (1S-trans)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl.)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C26H28Cl2N2O6)
  • [0760]
    Figure US20030130349A1-20030710-C00339
  • The synthesis of Example 247 is taught by Scheme 37 under the heading Preparation of Example 247 [0761]
  • Example 248 (1R-cis)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C26H28Cl2N2O6)
  • [0762]
    Figure US20030130349A1-20030710-C00340
  • The synthesis of Example 248 is taught by Scheme 37 under the heading Preparation of Example 248. [0763]
  • Example 249 (1R-cis)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine (C26H28Cl2N2O6)
  • [0764]
    Figure US20030130349A1-20030710-C00341
  • The synthesis of Example 249 is taught by Scheme 37 under the heading Preparation of Example 249. [0765]
  • Example 250 (1R-trans)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C26H28Cl2N2O6)
  • [0766]
    Figure US20030130349A1-20030710-C00342
  • The synthesis of Example 250 is taught by Scheme 37 under the heading Preparation of Example 250. [0767]
  • Example 251 (1R-trans)-N-[(3-Carboxy-1,2,2-trimethylcyclopentyl)carbonyl]-4-[(2,6-dichlorobenzoyl)amino]-D-phenylalanine (C26H28Cl2N2O6)
  • [0768]
    Figure US20030130349A1-20030710-C00343
  • The synthesis of Example 251 is taught by Scheme 37 under the heading Preparation of Example 251. [0769]
  • Example 252 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-3-bromo-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine (C26H28Cl2N2O6)
  • [0770]
    Figure US20030130349A1-20030710-C00344
  • The synthesis for Example 252 is taught by Scheme 2 (Method B) and is explained under the heading Preparation of Example 252. [0771]
  • Example 253 (1S-cis)-N-[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-4-[[2-chloro-4-[(1,1-dimethyl)ethyl]benzoyl]amino]-L-phenylalanine (C30H37Cl1N2O6)
  • [0772]
    Figure US20030130349A1-20030710-C00345
  • The synthesis of Example 253 is taught by Scheme 38 under the heading Preparation of Example 253. [0773]
  • Example 254 [1S-[1α(R*),3α]]-α-[[(3-Carboxy-2,2,3-trimethylcyclopentyl)carbonyl]amino-]-3-[(2,6-dichlorophenyl)methoxy]-6-pyridinepropanoic Acid (C25H28Cl2N2O6)
  • [0774]
    Figure US20030130349A1-20030710-C00346
  • The synthesis of Example 254 is taught by Scheme 39 under the heading Preparation of Example 254. [0775]
  • REFERENCE EXAMPLES (R. EXAMPLES) R. Example 1 (1R)-1,8,8-Trimethyl-3-oxabicyclo[3.2.1]octane-2,4-dione [(1R)-camphoric Anhydride]
  • [0776]
    Figure US20030130349A1-20030710-C00347
  • Method A: [0777]
  • A mixture of (1R-cis)-1,2,2-Trimethylcyclopentane-1,3-dicarboxylic acid) [(1R,3S)-camphoric acid] (40.0 g), AcCl (23.5 g), and Ac[0778] 2O (81.6 g) was heated under reflux for 3 hr. The mixture was concentrated in vacuo, dissolved in CHCl31 washed with sat. NaHCO3, brine, and dried over Na2SO4. The solvent was removed in vacuo and the residue was triturated with i-Pr2O to give (1R)-1,8,8-Trimethyl-3-oxabicyclo[3.2.1]octane-2,4-dione [(1R)-camphoric anhydride (34.0 g) as crystals.
  • Method B: [0779]
  • DIEA (8.0 g) was added to a suspension of (1R,3S)-camphoric acid (5.0 g) and BOP Reagent (11.1 g) in THF (50 ml) at room temperature. The mixture was stirred for 4 hr. and the solvent was removed in vacuo. The residue was extracted with AcOEt and the extract was washed with 5Q HCl, sat. NaHCO[0780] 3, and sat. LiCl, and dried over Na2SO4. The solvent was removed in vacuo and the residue was purified by flash chromatography on silica gel (eluent; CHCl3) to give (1R)-camphoric anhydride (4.1 g) as a colorless powder.
  • R. Example 2 (1R-cis)-1,2,2-Trimethylcyclopentane-1,3-dicarboxylic acid 3-(1,1-dimethylethyl) Ester
  • [0781]
    Figure US20030130349A1-20030710-C00348
  • To a solution of (1R)-camphoric anhydride (0.18 g) in THF (2 ml) was added 1M t-BuOK in THF (1 ml) at −78° C. under N[0782] 2 and the mixture was stirred for 10 hr. at the same temperature. The mixture was concentrated in vacuo, dissolved in H2O, and extracted with AcOEt. The aqueous layer was acidified with 1N HCl to pH 3 and extracted with CH2Cl2. The extract was washed with brine and dried over Na2SO4. The solvent was removed in vacuo to give (1R-cis)-1,2,2-Trimethylcyclopentan-1,3-dicarboxylic acid 3-dimethylethyl ester (0.24 g).
  • R. Example 3 (1R-cis)-3-Formyl-1,2,2-trimethylcyclopentanecarboxylic Acid
  • [0783]
    Figure US20030130349A1-20030710-C00349
  • A mixture of (1R)-camphoric anhydride (1.76 g), N,O-dimethylhydroxylamine hydrochloride (1.13 g), DIEA (8.4 ml) in THF (15 ml) was heated for 3 hr. at 85° C. in a sealed tube. After cooling, the tube was opened and 1N HCl (75 ml) was added. The resulting mixture was extracted with AcOEt. [0784]
  • The extract was dried over Na[0785] 2SO4 and the solvent was removed in vacuo. The residue was purified by column chromatography on silica gel to give (1R,3S)-3-(N-methoxy-N-methylcarbamoyl)-1,2,2-trimethylcyclopentanecarboxylic acid (800 mg).
  • To a solution of the obtained compound (215 mg) in THF (5 ml) was added a 1M solution of LiAlH[0786] 4 in THF (1.5 ml) at −78° C. The mixture was stirred for 1 hr. at −78° C., warmed to 0° C. and quenched with 1N HCl. The resulting mixture was extracted with AcOEt. The extract was dried over Na2SO4 and the solvent was removed in vacuo to give a 3:1 mixture of (1R-cis)-3-Formyl-1,2,2-trimethylcyclopentanecarboxylic acid (aldehyde form) and (1R,5S)-4-hydroxy-1,8,8-trimethyl-3-oxabicyclo [32.1]-octane-2-one (acetal form) (153 mg). Both forms are exchangeable with each other in solution.
  • R. Example 4 O-(Cyclohexylmethyl)-L-tyrosine Methyl Ester, Hydrochloride Salt
  • [0787]
    Figure US20030130349A1-20030710-C00350
  • To a mixture of N-(tert-butoxycarbonyl)-L-tyrosine methyl ester (356 mg), K[0788] 2CO3 (830 mg), n-Bu4NI (89 mg) in DMF (5 ml) was added cyclohexylmethyl bromide (202 , l) and the mixture was stirred for 1 day at room temperature. After addition of brine (40 ml), the resulting mixture was extracted with AcOEt. The extract was dried over Na2SO4 and the solvent was removed in vacuo. The residue was purified by column chromatography on silica gel (eluent; 9:1, Hexane/AcOEt→1:1, Hexane/AcOEt) to give N-(tert-butoxycarbonyl)-O-cyclohexylmethyl-L-tyrosine methyl ester (470 mg). The obtained compound (347 mg) was dissolved in 3N HCl/AcOt (5 ml) and the mixture was stirred overnight. The solvent and excess HCl was removed in vacuo to give O-(Cyclohexylmethyl)-L-tyrosine methyl ester, hydrochloride salt.
  • R. Example 5 1-[(2,4-Dichlorophenyl)methyl]-L-histidine Methyl Ester, Dihydrochloride Salt
  • [0789]
    Figure US20030130349A1-20030710-C00351
  • N-(tert-butoxycarbonyl)-L-histidine methyl ester (1.0 g) was added to a solution of 2,4-dichiorobenzyl chloride (0.73 g) and Et[0790] 3N (0.27 g) in benzene (10 ml) and the mixture was heated under reflux for 1 hr. The mixture was cooled and the precipitate was removed by filtration. The filtrate was washed with H2O, dried over Na2SO4, and the solvent was removed in vacuo. The residue was purified by column chromatography on silica gel (eluent; 95:5, CHCl3/MeOH) to give N-(tert-butoxycarbonyl)-1-[(2,4-dichlorophenyl)methyl]-L-histidine methyl ester. The obtained compound was treated in a similar manner as described in R. Example 4 to give 1-[(2,4-Dichlorophenyl)methyl]-L-histidine methyl ester, dihydrochloride salt.
  • R. Example 6 1-[(2,6-Dichlorophenyl)methyl]-L-histidine methyl ester, dihydrochloride salt.
  • [0791]
    Figure US20030130349A1-20030710-C00352
  • L-histidine (3.13 g) was added to a solution of NaNH[0792] 2 in liquid ammonia prepared from Na (0.93 g) and FeCl3 (catalytic amount) in liquid ammonia (50 ml) After 15 min., a solution of 2,6-dichlorobenzylchloride (3.96 g) in THF (5 ml) was added and the mixture was stirred for 3 hr. The reaction mixture was quenched with H2O and ether. The pH of the aqueous layer was adjusted to pH 8 with 5% HCl followed by cooling. The resulting precipitate was collected by filtration, washed with H2O, and dried to give 1-[(2,6-dichlorophenyl)methyl]-L-histidine (3.58 g). The obtained compound (0.80 g) was dissolved in MeOH (30 ml) and HCl gas was bubbled for 10 min. at 0° C. The reaction mixture was stirred for 15 hr. at room temperature and the solvent was removed in vacuo to give 1-[(2,6-Dichlorophenyl)methyl]-L-histidine methyl ester, dihydrochloride salt.
  • R. Example 7 3-Nitro-L-tyrosine Methyl Ester, Hydrochloride Salt
  • [0793]
    Figure US20030130349A1-20030710-C00353
  • To a solution of N-(tert-butoxycarbonyl)-L-tyrosine methyl ester (5.64 g) in THF (10 ml) was added NH[0794] 4NO3 (3 g) and concentrated HNO3 (3 ml). After 30 sec., the reaction mixture turned to dark red with reflux occurring. The reaction mixture was quenched with solid NaHCO3 and H2O, and extracted with AcOEt. The extract was dried over Na2SO4 and the solvent was removed in vacuo. The residue was purified by column chromatography on silica gel (eluent; Hexane→1:1, Hexane/AcOEt) to give N-(tert-butoxycarbonyl)-3-nitro-L-tyrosine methyl ester (3.42 g). The obtained compound was treated in a similar manner as described in R. Example 4 to give 3-Nitro-L-tyrosine methyl ester, hydrochloride salt.
  • R. Example 8 3-[(2,6-Dichlorobenzoyl)amino]-L-tyrosine Methyl Ester, Hydrochloride Salt
  • [0795]
    Figure US20030130349A1-20030710-C00354
  • A mixture of N-(tert-butoxycarbonyl)-3-nitro-L-tyrosine methyl ester (2.0 g) and 10% Pd-C (1.0 g) in MeOH (10 ml) was subjected to catalytic hydrogenolysis at atmospheric pressure. The catalyst was filtered off and the filtrate was evaporated in vacuo. The residue was triturated with ether to give 3-amino-N-(tert-butoxycarbonyl)-L-tyrosine methyl ester (1.5 g) as a solid. To a solution of the obtained compound (1.0 g) in CH[0796] 2Cl2 (10 ml) was added 2,6-dichlorobenzoyl chloride (0.74 g) and DIEA (1.1 g) and the mixture was stirred at room temperature overnight. The solvent was removed in vacuo and the residue was purified by column chromatography on silica gel (eluent; 2:1, Hexane/AcOEt) to give N-(tert-butoxycarbonyl)-3-(2,6-dichlorobenzamido)-L-tyrosine methyl ester (1.2 g). The obtained compound was treated in a similar manner as described in R. Example 4 to give 3-[(2,6-Dichlorobenzoyl)amino]-L-tyrosine methyl ester, hydrochloride salt.
  • R. Example 9 4-[[(2,6-Dichlorophenyl)methyl]amino]-L-phenylalanine Methyl Ester, Hydrochloride Salt
  • [0797]
    Figure US20030130349A1-20030710-C00355
  • To a mixture of 4-amino-N-(tert-butoxycarbonyl)-L-phenylalanine methyl ester (0.59 g) and 2,6-dichlorobenzaldehyde (0.35 g) in MeOH (9 ml) was added NaCNBH[0798] 3 (0.38 g), AcOH (1 ml), and molecular sieves 4A (catalytic amount) and the mixture was stirred overnight at room temperature.
  • The reaction mixture was quenched with brine and 1N HCl. The solvent was removed in vacuo and the residue was extracted with AcOEt. The extract was washed with 1N HCl, brine, dried over MgSO[0799] 4, and the solvent was removed in vacuo. The residue was purified by column chromatography on silica gel (eluent; 10% EtOH/CH2Cl2) to give N-(tert-butoxycarbonyl)-4-[(2,6-dichlorophenyl)methyl]amino]-L-phenylalanine (0.36 g) as a colorless solid. The obtained compound (0.5 g) was dissolved in MeOH (5 ml) and HCl gas was bubbled for 5 min. at 0° C. The mixture was stirred for 2 hr. and the solvent was removed in vacuo to give 4-[[(2,6-Dichlorophenyl)methyl]amino]-L-phenylalanine methyl ester, hydrochloride salt.
  • R. Example 10 (S)-α-Amino-2-naphthalenepropanoic Acid Phenylmethyl Ester, 4-methylphenylsulfonic Acid Salt
  • [0800]
    Figure US20030130349A1-20030710-C00356
  • A mixture of (S)-2-amino-3-(2-naphthyl)-propanoic acid (0.40 g), benzyl alcohol (2 ml), and p-toluenesulfonic acid monohydrate (0.42 g) in toluene (5 ml) was heated for 6 hr under reflux. [0801]
  • The reaction mixture was cooled and diluted with ether (10 ml)/hexane (10 ml). The resulting precipitate was collected by filtration and recrystallized from EtOH/ether to give (S)-α-Amino-2-naphthalenepropanoic acid phenylmethyl ester, 4-methylphenylsulfonic acid salt (0.73 g), mp 174-176° C. [0802]
  • R. Example 11 4-[[(2,6-Dichlorophenyl)amino]carbonyl]-L-phenylalanine Methyl Ester, Hydrochloride Salt
  • [0803]
    Figure US20030130349A1-20030710-C00357
  • A solution of 4-bromo-N-(tert-butoxycarbonyl)-L-phenylalanine (359 mg) in THF was added to a THF solution of n-BuLi (2.7 ml of 1.6 M solution in hexane) at −78° C. The mixture was stirred for 2 hr. at room temperature, quenched with 1N HCl (15 ml), and extracted with AcOEt. The extract was dried over Na[0804] 2SO4 and the solvent was removed in vacuo. The residue was purified by column chromatography on silica gel (eluent: Hexane AcOEt) to give N-(tert-butoxycarbonyl)-4-[(2,6-dichlorophenyl)-carbamoyl]-L-phenylalanine (95 mg). The obtained compound was treated in a similar manner as described in R. Example 4 to give 4-[[(2,6-Dichlorophenyl)amino]carbonyl]-L-phenylalanine methyl ester, hydrochloride salt.
  • R. Examples 12-46 were prepared in a similar manner as described in R. Examples 4-11, and are shown in the Tables 6-8. [0805]
    TABLE 6
    Figure US20030130349A1-20030710-C00358
    R. Examples 12 through 26:
    R. Ex.
    No. * R15 Z1 R13 R18
    12 S H single t-BuO— H
    bond
    13 S H —OCH2
    Figure US20030130349A1-20030710-C00359
    H
    14 S H —OCH2
    Figure US20030130349A1-20030710-C00360
    H
    15 S H —OCH2
    Figure US20030130349A1-20030710-C00361
    H
    16 S H —OCH2
    Figure US20030130349A1-20030710-C00362
    H
    17 S I —OCH2
    Figure US20030130349A1-20030710-C00363
    I
    18 S H —OCH2
    Figure US20030130349A1-20030710-C00364
    H
    19 S PhCH2O —OCH2 Ph H
    20 S H —OCH2
    Figure US20030130349A1-20030710-C00365
    H
    21 S H —OCH2 Ph H
    22 R H —OCH2 Ph H
    23 S NO2 —OCH2
    Figure US20030130349A1-20030710-C00366
    H
    24 S OH —OCH2
    Figure US20030130349A1-20030710-C00367
    H
    25 S
    Figure US20030130349A1-20030710-C00368
    —OCH2
    Figure US20030130349A1-20030710-C00369
    H
    26 S
    Figure US20030130349A1-20030710-C00370
    single Bond OH H
  • [0806]
    TABLE 7
    Figure US20030130349A1-20030710-C00371
    R. Examples 27 through 35:
    R. Ex.
    No. Z2 R16
    27 single bond Ph3C—
    28 CH2
    Figure US20030130349A1-20030710-C00372
    29 CH2 Ph
    30 CH2
    Figure US20030130349A1-20030710-C00373
    31 CH2 PhCH2O
    32 CH2
    Figure US20030130349A1-20030710-C00374
    33 CH2
    Figure US20030130349A1-20030710-C00375
    34 CH2
    Figure US20030130349A1-20030710-C00376
    35 CH2
    Figure US20030130349A1-20030710-C00377
  • [0807]
    TABLE 8
    Figure US20030130349A1-20030710-C00378
    R. Examples 36 through 46:
    R. Ex.
    No. * R6
    36 S
    Figure US20030130349A1-20030710-C00379
    37 S
    Figure US20030130349A1-20030710-C00380
    38 S
    Figure US20030130349A1-20030710-C00381
    39 S
    Figure US20030130349A1-20030710-C00382
    40 S Ph
    41 S
    Figure US20030130349A1-20030710-C00383
    42 S
    Figure US20030130349A1-20030710-C00384
    43 S
    Figure US20030130349A1-20030710-C00385
    44 S
    Figure US20030130349A1-20030710-C00386
    45 S
    Figure US20030130349A1-20030710-C00387
    46 R
    Figure US20030130349A1-20030710-C00388
  • R. Example 47 (1S-cis)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic Acid 3-methyl ester (C11H18O4), 36-B
  • [0808]
    Figure US20030130349A1-20030710-C00389
  • Reference Example 47 was prepared according to Scheme 35: 36-B where R[0809] 36-1 is CH3, and the stereochemistry is (1S-cis) as follows:
  • The preparation follows that of Preparation 10-A using (1S,3R)-(−)-Camphoric acid as the starting material. Physical properties as follows: [0810] 1H NMR (CDCl3) δ 11.45 (1H), 3.69 (3H), 2.82 (1H), 2.54 (1H), 2.24 (1H), 1.82 (1H), 1.52 (1H), 1.27 (3H), 1.25 (3H), 0.86 (3H); Anal: Calcd. for C11H18O4: C, 61.66; H, 8.47; Found: C, 61.60; H, 8.30.
  • R. Example 48 (1S-cis)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic Acid 1-(1,1-dimethylethyl)-3-methyl Diester (C15H26O4) 36-C
  • [0811]
    Figure US20030130349A1-20030710-C00390
  • Reference Example 48 was prepared according to Scheme 36: 36-C where R[0812] 36-1 is CH3, R36-2 is t-Bu, and the stereochemistry is (1S-cis) as follows:
  • To a solution of (1S-cis)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic acid 3-methyl ester (9.69 g), 36-B, in 3 mL of CH[0813] 2Cl2 and 80 mL of cyclohexane was added t-Butyl-2,2,2-trichloroacetimidate (16.18 mL) and 35 μL of BF3.Et2O. The reaction was stirred at ambient temperature for 18 hours and then filtered. The filtrate was purified by flash chromatography silica gel eluting with AcOEt-hexane to obtain (1S-cis)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic acid 1-(1,1-dimethylethyl)-3-methyl diester (12.89 g), 36-C. Physical properties as follows: m.p.=35-37° C.; 1H NMR (CDCl3) δ 3.68 (3H), 2.79 (1H), 2.53 (1H), 2.17 (1H), 1.76 (1H), 1.24 (3H), 1.17 (3H), 0.81 (3H); Anal: Calcd. for C15H26O4: C, 66.64; H, 9.67; Found: C, 66.61; H, 9.64.
  • R. Example 49 (1S-cis)-1,2,2-Trimethylcyclopentane-1,3-dicarboxylic Acid 1-(1,1-dimethylethyl) Ester (C14H24O4), 36-D
  • [0814]
    Figure US20030130349A1-20030710-C00391
  • Reference Example 49 was prepared according to Scheme 36 in the Preparation of 36-D where R[0815] 36-2 is t-Bu and the stereochemistry is (1S-cis) as follows:
  • The preparation follows that of Preparation 15-D using 36-C, (1S-cis)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic acid 1-(1,1-dimethylethyl)-3-methyl diester, as starting material. Physical properties as follows: m.p.=95.7-97.5° C.; [0816] 1H NMR (CDCl3) δ 2.82 (1H), 2.53 (1H), 2.13 (1H), 1.82 (1H), 1.45 (10H), 1.29 (3H), 1.17 (3H), 0.88 (3H); Anal: Calcd. for C15H26O4: C, 65.60; H. 9.44; Found: C, 65.60; H, 9.44.
  • R. Example 50 (1S-trans)-[3-(Phenylmethoxy)carbonyl]-1,2,2-trimethylcyclopentane-1-carboxylic acid 1-(1,1-dimethylethyl) ester (C21H30O4), 36-E-1
  • [0817]
    Figure US20030130349A1-20030710-C00392
  • Reference Example 50 was prepared according to Scheme 36: Preparation of 36-E-1 where R[0818] 36-1 is —CH2C6H5, R36-2 is t-Bu, and the stereochemistry is (1S-trans) as follows:
  • To a solution of (1S-cis)-1,2,2-Trimethylcyclopentane-1,3-dicarboxylic acid 1-(1,1-dimethylethyl) ester 36-D (20.0 g) in 300 mL of CH[0819] 3CN was added benzyl bromide (16.01 g) and DIEA (12.1 g). The reaction was stirred at ambient temperature for 22 hours, concentrated in vacuo, and diluted with CH2Cl2. The solution was consecutively washed with water, 10% HCl, water, and brine. The organic layer was dried (Na2SO4) and concentrated in vacuo. The concentrate was purified by chromatography on silica gel eluting with AcOEt-hexane to obtain (1S-cis)-[3-(Phenylmethoxy)carbonyl]-1,2,2-trimethylcyclopentane-1-carboxylic acid 1-(1,1-dimethylethyl) ester (18.94 g)
  • Physical properties as follows: [0820] 1H NMR (CDCl3) δ 7.34 (5H), 5.12 (2H), 2.82 (1H), 2.51 (1H), 2.19 (1H), 1.82 (1H), 1.44 (10H), 1.23 (3H), 1.15 (3H), 0.78 (3H); MS (EI) m/z (rel. intensity) 0 (M+, 0), 181 (64), 180 (64), 179 (17), 166 (22), 155 (16), 153 (15), 109 (18), 92 (15), 91 (99), 57 (40); Anal. Calcd for C21H30O4: C, 72.80; H, 8.73; Found: C, 70.76; H, 8.38; N, 0.36.
  • The product of the above reaction was combined with THF (44 mL) and NaH (634 mg) and refluxed for 0.5 hour then cooled, diluted with saturated NaHCO[0821] 3, and extracted with AcOEt. The organic layer was concentrated to obtain a mixture of starting material and (1S-trans)-[3-(Phenylmethoxy)carbonyl]-1,2,2-trimethylcyclopentane-1-carboxylic acid 1-(1,1-dimethylethyl) ester. These were separated by HPLC on an (R, R)Whelk-O, 5×25 cm column, eluting with 2% isopropanol in hexane at 50 mL/minute. The detector monitored 215 nm. The first peak at 19 minutes contained (1S-trans)-[1-(Phenylmethoxy)carbonyl]-1, 2, 2-trimethylcyclopentane-1-carboxylic acid 1-(1,1-dimethylethyl) ester 36-E-1 (3.88 g). Physical properties as follows: 1H NMR (CDCl3) δ 7.34 (5H) 5.12 (2H), 3.02 (1H), 2.18 (2H), 1.98 (1H), 1.58 (1H), 1.44 (9H), 1.08 (3H), 1.06 (3H), 0.79 (3H); MS (FAB) m/z 347 (MH+), 501, 292, 291, 245, 183, 109, 92, 91, 57, 41; HPLC 0.46×25 cm (R, R)Whelk-O column eluted at 0.5 mL/minute with 2 isopropanol in hexane, monitoring 215 nm, RT 13.65 minutes.
  • R. Example 51 (1R-trans)-[3-(Phenylmethoxy)carbonyl]-1,2,2-trimethylcyclopentane-1-carboxylic Acid 1-(1,1-dimethylethyl) Ester (C21H30O4), 36-E-2
  • [0822]
    Figure US20030130349A1-20030710-C00393
  • Reference Example 51 was prepared according to Scheme 36: Preparation of 36-E-2 where R[0823] 36-1 is —CH2C6H5, R36-2 is t-Bu, and the stereochemistry is (1R-tranis) as follows:
  • The preparation follows that of Preparation 36-E-1 using 15-D, (1R-cis)-1,2,2-Trimethylcyclopentane-1,3-dicarboxylic acid 1-(1,1-dimethylethyl) ester, as starting material. Physical properties as follows: [0824] 1H NMR (CDCl3) δ 7.35 (5H), 5.12 (2H), 3.02 (1H), 2.13 (2H), 1.98 (1H), 1.58 (1H), 1.44 (9H), 1.08 (3H), 1.05 (3H), 0.78 (3H); HPLC 0.46×25 cm (R, R)Whelk-O column eluted at 1.0 mL/minute with 2 isopropanol in hexane, monitoring 254 nm, RT=5.54 minutes
  • R. Example 52 (1S-trans)-[3-(Phenylmethoxy)carbonyl]-1,2,2-trimethylcyclopentane-1-carboxylic Acid (C17H22O4), 36-F-1
  • [0825]
    Figure US20030130349A1-20030710-C00394
  • Reference Example 52 was prepared according to Scheme 36: Preparation of 36-F-1 where R[0826] 36-1 is —CH2C6H5, and the stereochemistry is (1S-trans) as follows:
  • A sample of 36-E-1, (1S-trans)-[3-(Phenylmethoxy)carbonyl]-1,2,2-trimethylcyclopentane-1-carboxylic acid 1-(1,1-dimethylethyl) ester (2.95 g), was dissolved in TFA at 0° C. and then allowed to come to ambient temperature. The solution was stirred for 19 hours then concentrated, redissolved in t-butyl methyl ether and washed with a saturated solution of NaHCO[0827] 3. The organic layer was concentrated in vacuo to obtain (1S-trans)-[3-(phenylmethoxy)carbonyl]-1,2,2-trimethylcyclopentane-1-carboxylic acid 36-F-1 (2.44 g).
  • Physical properties as follows: [0828] 1H NMR (CDCl3) δ 7.34 (, 5H), 5.13 (, 2H), 3.08 (, 1H), 2.16 (, 2H), 2.02 (, 1H), 1.64 (, 1H), 1.16 (, 3H), 1.10 (, 3H), 0.83 (, 3H); Anal. Calcd for C17H22O4: C, 70.32; H, 7.64. Found: C, 70.94; H, 7.63; N, 0.04.
  • R. Example 53 (1R-trans)-[3-(Phenylmethoxy) carbonyl]-1,2,2-trimethylcyclopentane-1-carboxylic Acid (C17H22O4), 36-F-2
  • [0829]
    Figure US20030130349A1-20030710-C00395
  • Reference Example 53 was prepared according to Scheme 36: Preparation of 36-F-2 where R[0830] 36-1 is —CH2C6H5, and the stereochemistry is (1R-trans) as follows:
  • The preparation follows that of Preparation 36-F-1 using 36-E-2, (1R-trans)-[3-(Phenylmethoxy)carbonyl]l-1,2,2-trimethylcyclopentane-1-carboxylic acid 1-(1, 1-dimethylethyl) ester, as starting material. Physical properties as follows: [0831]
  • MS (EI) m/z (rel. intensity) 290 (M[0832] +, 5), 183 (15), 182 (9), 164 (8), 153 (22), 136 (10), 109 (22), 92 (24), 91 (99), 65 (9), 55 (9); Anal. Calcd for C17H22O4: C, 70.32; H, 7.64. Found: C, 70.66; H, 7.74; N, 0.22.
  • R. Example 54 (1S-trans)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic Acid 1-(1,1-dimethylethyl) Ester (C14H24O4), 36-G-1
  • [0833]
    Figure US20030130349A1-20030710-C00396
  • Reference Example 54 was prepared according to Scheme 36: Preparation of 36-G-1 where R[0834] 36-2 is t-Bu and the stereochemistry is (1S-trans) as follows:
  • A sample of-36-E-1, (1S-trans)-[3-(Phenylmethoxy)carbonyl]-1,2,2-trimethylcyclopentane-1-carboxylic acid 1-(1,1-dimethylethyl) ester (3.27 g), was dissolved in EtOH (100 mL) and added 5% palladium on carbon (1.0 g) and cyclochexene (50 mL). The mixture was refluxed for 4 hours and then stirred at ambient temperature for 18 hours. The reaction was then filtered and the filtrate concentrated in vacuo to obtain 36-G-1, (1S-trans)-1,2,2-trimethylcyclopentane-1-carboxylic acid 1-(1,1-dimethylethyl) ester (2.45 g). Physical properties as follows: [0835] 1H NMR (CDCl3) δ 3.05 (1H), 2.11 (2H), 1.99 (1H), 1.60 (1H), 1.45 (9H), 1.11 (3H), 1.10 (3H), 0.89 (3H); MS (FAB) m/z (rel. intensity) 257 (MH+, 43), 411 (23), 257 (43), 201 (99), 183 (20), 177 (14), 155 (26), 109 (36), 57 (81), 41 (22), 29 (15); Anal. Calcd for C14H24O4: C, 65.60; H. 9.44. Found: C, 65.62; H, 9.42; N, 0.03.
  • R. Example 55 (1R-trans)-1,2,2-trimethylcyclopentane-1,3-dicarboxylic Acid 1-(1,1-dimethylethyl) Ester (C14H24O4), 36-G-2
  • [0836]
    Figure US20030130349A1-20030710-C00397
  • Reference Example 55 was prepared according to Scheme 36: Preparation of 36-G-2 where R[0837] 36-2 is t-Bu and the stereochemistry is (1R-trans) as follows:
  • The preparation follows that of Preparation 36-G-1 using 36-E-2, (1R-trans)[3-(Phenylmethoxy)carbonyl]-1,2,2-trimethylcyclopentane-1-carboxylic acid 1-(1, 1-dimethylethyl) ester, as starting material. Physical properties as follows: [0838] 1H NMR (CDCl3) δ 3.03 (, 1H), 2.13 (, 2H), 1.98 (, 1H), 1.59 (, 1H), 1.45 (, 9H), 1.11 (, 3H), 1.10 (, 3H), 0.89 (, 3H); MS (FAB) m/z (rel. intensity) 257 (MH+, 35), 411 (19), 279 (14), 257 (35), 201 (99), 183 (18), 155 (26), 1.09 (35), 57 (74), 41 (19), 29 (12); Anal. Calcd for C14H24O4: C, 65.60; H, 9.44. Found: C, 65.56; H, 9.44; N, 0.18.
  • R. Example 56 3-Bromo-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine Methyl Ester Hydrochloride Salt (C17H15BrCl2N2O3.HCl)
  • [0839]
    Figure US20030130349A1-20030710-C00398
  • Reference Example 56 was prepared as follows: [0840]
  • To a solution of 4-[(2,6-Dichlorobenzoyl)amino]-L-phenylalanine methyl ester hydrochloride salt, 37-B-1, (205.7 mg) in acetic acid (5 mL) was added an excess of bromine (5.55 g) in acetic acid (5 mL) and iron powder (416.2 mg). The reaction was stirred at ambient temperature for 3 hours. The reaction concentrated in vacuo and the remaining acetic acid removed as an azeotrope with toluene. The crude material was diluted with water, made basic with saturated sodium bicarbonate and extracted with AcOEt. The extract was purified by flash chromatography on silica gel eluting with methanol-methylene chloride. The purified material was dissolved in methanol saturated with hydrogen chloride and concentrated in vacuo and then crystallized from methanol with AcOEt to obtain 3-Bromo-4-[(2,6-dichlorobenzoyl)amino]-L-phenylalanine methyl ester hydrochloride salt (540.4 mg). Physical properties as follows: m.p. 200-205; [0841] 1H NMR (CD3OD) δ 7.76 (1H), 7.64 (1H), 7.46 (3H), 7.34 (1H), 4.38 (1H), 3.85 (3H), 3.30 (1H), 3.15 (1H); HRMS (FAB) Calcd for C17H15BRCL2N2O3+H, 444.9722, found 444.9724.
  • R. Example 57 N-[(1,1-Dimethylethoxy)carbonyl]-3-iodo-L-alanine Methyl Ester (C9H16INO4).
  • [0842]
    Figure US20030130349A1-20030710-C00399
  • The preparation of Reference Example 57 is taught by Scheme 39 under the heading Preparation of Reference Example 57. [0843]
  • R. Example 58
  • [0844]
    Figure US20030130349A1-20030710-C00400
  • The aminoester product of Reference Example 58 is useful as a synthetic intermediate (for example, reagent 37-B of Scheme 37). Reference Example 58 is prepared as-follows: [0845]
  • To a cold (0-5° C.) solution of anhydrous methanolic HCl was added 100 g of L-4-nitrophenylalanine (Advanced ChemTech) portionwise over 15 min. The mechanically stirred mixture was heated to gentle reflux for 48 h. The mixture was allowed to cool and then filtered through a sintered glass filter funnel, washing the collected solids with hot MeOH until only insoluble residues remained. The filtrate was concentrated in vacuo to afford the methyl ester (120 g) as waxy off white solid which was used without further purification. [0846]
  • To a suspension of methyl ester described above (87 g, 0.33 mole) in CH[0847] 2Cl2 (1500 mL) at ambient temperature was added di-t-butyldicarbonate (109 g, 0.50 mole) followed by the dropwise addition of Et3N (51 mL, 0.37 mole). After 15 min additional Et3N (40 mL, 0.29 mol) was added to maintain a slightly basic mixture (ca. pH 8). The reaction mixture was stirred 18 h and additional CH2Cl2 (1400 mL) and Et3N (15 mL, 0.11 mol) were added. After an additional 2 h the reaction mixture was quenched by the slow addition of MeOH (100 mL), stirred for 1 h and then partitioned between CH2Cl2 and cold 10% aqueous KHSO4. The organic layer was washed with saturated NaHCO3 and brine, dried (Na2SO4), filtered and concentrated in vacuo. Flash chromatography of the residue using hexane and a gradient of a 1:1 mixture of EtOAc/CH2Cl2 (25-33%) afforded the Boc-methyl ester (69 g) as a white solid. Physical properties as follows: 1H NMR (300 MHz; CDCl3) δ 8.16 (2H), 7.31 (2H), 5.04 (1H), 4.63 (1H), 3.73 (3H), 3.18 (2H), 1.41 (9H); MS (ES+) for C15H20N2O6 m/z 325.2 (M+H)+.
  • Palladium on carbon (100% w/w, 1.25 g) was added to a Parr hydrogenation flask under a N[0848] 2 atmosphere and carefully wetted with 100 mL of MeOH/THF (1:1). A solution of the Boc-methyl ester described above (25 g, 77 mmol) in 400 mL of MeOH/THF (1:1) was added and the mixture shaken on a hydrogenation apparatus under a hydrogen atmosphere (20 psi) for 1 h at ambient temperature. The reaction mixture was filtered through a pad of Celite and the solids washed several times with MeOH. The combined filtrates were concentrated in vacuo to afford the 4-aminophenylalanyl derivative (22.7 g) which was used without further purification. Physical properties as follows: 1H NMR (300 MHz, CDCl3) δ 6.89 (2H), 6.61 (2H), 4.96 (1H), 4.50 (1H), 3.69 (3H), 2.95 (2H), 1.41 (9H); MS (ES+) for C15H22N2O4 m/z 295.2 (M+H)+.
  • A cold (0-5° C.) solution of 2,6-dichlorobenzoyl chloride (11.1 mL, 77.5 mmol) in 125 mL of THF was treated dropwise with a solution of the 4-aminophenylalanyl derivative described above (22.7 g, 77.1 mmol) and Et[0849] 3N (16 mL, 115 mmol) in 125 mL of THF. The reaction mixture was allowed to warm to temperature and stir an additional 18 h. The mixture was diluted with EtOAc (2 L) and then washed with 1N HCl, H2O, 1N NaOH and brine. The organic extract was dried (Na2SO4), filtered, and concentrated in vacuo to give the crude product as a pale yellow solid. This material was recrystallized from acetone/hexanes (ca. 1:1) to afford the amide (30.8 g) as a crystalline solid. Physical properties as follows: mp 192.2-193.1° C.; IR (mull) 3305, 1747, 1736, 1690, 1665, 1609, 1548, 1512, 1433, 1414, 1325, 1277, 1219, 1171, 781 cm−1; 1H NMR (300 MHz; CDCl3) δ 7.57 (2H), 7.34 (4H) 7.14 (2H), 4.98 (1H), 4.60 (1H), 3.74 (3H), 3.11 (2H), 1.42 (9H); MS (ES+) for C22H24Cl2N2O5 m/z 467.0 (M+H)+.
  • To 650 mL of anhydrous 4M HCl in dioxane at ambient temperature was added the amide described above (30.6 g, 65.5 mmol) portionwise and the resulting mixture was stirred until all the solids had dissolved (ca. 1 h). Volatiles were removed in vacuo to give a light yellow solid which was partitioned between water (500 mL) and ether (1 L). The water layer was separated and concentrated in vacuo to approximately 200 mL. The aqueous solution was then frozen and lyophilized to afford the aminoester product Reference Example 58 (25.6 g) as a light yellow solid. Physical properties as follows: [α][0850] 25 D=+5 (c 1, MeOH); IR (mull) 3244, 3186, 3112, 1747, 1660, 1604, 1562, 1539, 1516, 1431, 1416, 1327, 1273, 1243, 799 cm−1; 1H NMR (300 MHz; CD3OD) δ 7.69 (2H) 7.45 (3H), 7.29 (2H), 4.34 (1H), 3.83 (3H), 3.21 (2H); 13C NMR (300 MHz; CD3OD) δ 169.0, 163.9, 137.8, 136.08, 131.8, 131.0, 130.3, 129.7, 127.9, 120.6, 53.8, 52.3, 35.4; MS (ES+) for C17H16Cl2N2O3 m/z 367.1 (M+H)+.
  • Biological Assays [0851]
  • Jurkat-Endothelial Cell Adhesion Assay: [0852]
  • The following assay established the activity of the present compounds in inhibiting β[0853] 1-mediated cell adhesion in a representative in vitro system. This assay measures the adhesive interactions of a T-cell line, Jurkat, known to express the α4β1 integrin, to endothelial monolayers in the presence of test compounds. The test compounds were added in increasing concentrations to T-cells and then the T-cell compound mixture was added to IL-1 stimulated endothelial cell monolayers. The plates were incubated, washed and the percentage of attached cells was quantitated. The present assay directly demonstrates the cell adhesion inhibitory activity and adhesion modulatory activity of the compounds. Human umbilical vein endothelial cells were purchased from Clonetics (San Diego, Calif.) at passage number 2. The cells were grown on 0.5% porcine skin gelatin pre-coated flasks (Sigma, St. Louis Mo.) in EGM-UV media (Clonetics, San Diego, Calif.) supplemented with 10% fetal bovine serum. Cells are refed every 2-3 days reaching confluence by day 4 to 6. The cells are monitored for factor VIII antigen and results show that at passage 12, the cells are positive for this antigen. The endothelial cells are not used following passage 6. The T-cell line Jurkat was obtained from American Type Tissue Culture Collection (Rockville, Md.) and the cells were cultured in RPMI containing 10% fetal calf serum. The cells were washed twice in Hank's Balanced Salt Solution (HBSS) and resuspended in Dulbecco's Minimal Eagle's Media (DMEM) containing 2.5 mg/ml Human Serum Albumin (HSA). Jurkat cells (1×106 cells/ml) were stained with 10 ng/ml BCECF-AM (Molecular Probes, Eugene, Oreg.)) in HESS without phenol red. The cells were loaded with BCECF for 60 minutes in the dark at 37° C., washed 2 times, and resusupended in DMEM-HSA— solution.
  • Confluent endothelial monolayers, grown in 96-well tissue culture plates, were stimulated for 4 hr. at 37° C. with 0.1 ng/ml (˜50 U/ml) recombinant IL-1 (Amgen, Thousand Oaks, Calif.). Following this incubation, the monolayers were washed twice with HESS and 0.1 ml of DMEM-HSA solution was added. Jurkat cells (5×10[0854] 5 cells) were combined with the appropriate concentration of the test compound and 0.1 ml of the Jurkat cell-compound mixture was added to the endothelial cell monolayers. Generally, 100, 20, 5 and 1.25 μM compound concentrations were tested. These concentrations are adjusted downward for analogs found or thought to be more potent. The plates were placed on ice for 5 minutes to allow for Jurkat cell settling and the plates were incubated at 37° C. for 20 minutes. Following this incubation, the monolayers were washed twice with PBS containing 1 mM calcium chloride and 1 mM magnesium chloride and the plates were read using a Millipore Cytofluor 2300 (Marlboro, Mass.). Fluorescence in each well was measured as Arbitrary Fluorescence Units and percent adhesion in the absence of compound was adjusted to 100% and the % adhesion in the presence of compound was calculated. Monolayers were also fixed in 3% paraformaldehyde and evaluated microscopically to verify the adhesion. This procedure is a modification of a previously published method (Cardarelli et al., J. Diol. Cheem. 269:18668-18673 (1994)).
  • Jurkat-CS-1 Assay [0855]
  • The CS-1 derived peptide, CLHPGEILDVPST, and the scrambled control peptide, CLHGPIELVSDPT, were synthesized on a Beckman 990 synthesizer using t-Boc methodology. The peptides were immobilized onto microtiter plates using the heterobifunctional crosslinker 3-(2-pyridyldithio)propionic acid N-hydroxysuccinimide ester (SPDP) as reported by Pierschbacher et al., [0856] Proc. Natl. Acad. USA, 801224-1227 (1983). Microtiter plates were coated with 20 μg/ml HSA for 2 hr. at room temperature, washed once with PBS and derivatized with 10 μg/ml SPDP for 1 hr. After washing, 100 μl of a 100 μg/ml cysteine containing-peptide solution which had been recently dissolved was added to the wells and allowed to crosslink to the plates overnight at 4° C. Unbound peptide was removed from plates by washing with PBS. To block non-reacted sites, the plates are coated with 100 μl of a 2.5 mg/ml BSA solution in PBS for 1 hr. at 37° C. 100 μl of Jurkat cells (2.5×106 cells/ml) in DMEM plus BSA (2.5 mg/ml) was mixed with an appropriate concentration of the compound to be tested and the mixture was added to peptide coated dishes and incubated for 1 hr. at 37° C. Generally 100, 20, 5 and 1.25 μM concentrations of the compound were tested. The concentrations of the compound were adjusted downward for compounds thought or found to be more potent.
  • Following this incubation the plates were washed once with PBS and the attached cells were fixed with 3% paraformaldehyde in PBS and stained with 0.56 toluidine blue in 3.7% formaldehyde. The cells were stained overnight at room temperature and the optical density at 590 nm of toluidine blue stained cells was determined using a vertical pathway spectrophotometer to quantitate attachment (VMAX Kinetic Microplate Reader, Molecular Devices, Menlo Park, Calif.). This procedure is-a modification of a previously published method (Cardarelli et al, [0857] J. Biol. Chem., 269:18668-18673 (1994) and Cardarelli et al, Proc. Natl. Acad. Sci. USA, 83:2647-2651 (1986)).
  • The preferred compounds are those which have low IC[0858] 50 values in the Jurkat EC assay or the Jurkat-CS-1 assay described above or which have at least moderate activity in both assays. All of the compounds of the present invention have an activity of less than 50 μM in the Jurkat CS-1 assay or less than 500 μM in the Jurkat EC assay. Compounds with activity in the Jurkat CS-1 assay preferably have IC50 values of less than 1 μM, more preferably less than 0.5 μM, most preferably less than or equal to 0.08 μM. Compounds with activity in the Jurkat EC assay preferably have IC50 values of less than 10 μM, more preferably less than 5 μM, most preferably less than or equal to 0.8 μM.
  • In the Jurkat EC Assay, IC[0859] 50 value ranges (μM) are depicted by A, B, and C and in the Jurkat CS-1 Assay, IC50 value ranges are depicted by D, E, and F. These ranges are as follows:
  • Jurkat EC: 5≦A<10, 0.8<B<5, and C≦0.8 [0860]
  • Jurkat CS-1: 0.5≦D<1, 0.08<E<0.5, and F≦0.08 [0861]
  • The following chart illustrates the IC[0862] 50 values for selected compounds of the present invention in the Jurkat EC Assay and the Jurkat CS-1 Assay, The ranges are so described above.
    ADDITIONAL IN VITRO BIOLOGICAL DATA
    Example
    No. Jurkat EC Jurkat CS-1
    2 D
    4 B E
    10 B E
    12 C F
    13 A D
    14 A
    16 A
    24 B E
    26 B D
    28 B E
    29 A D
    31 A
    36 A D
    38 B E
    46 B
    53 B D
    54 C F
    61 B E
    62 A
    63 B F
    65 C E
    75 B
    77 B E
    79 A
    81 C E
    83 C F
    85 A E
    86 D
    87 C F
    89 B E
    91 B F
    92 C F
    93 C F
    95 B E
    96 A
    97 C F
    100 C F
    102 C F
    103 C F
    104 C F
    105 B E
    106 C F
    108 C F
    110 C F
    112 C F
    113 D
    114 C E
    116 C F
    118 B D
    120 B E
    121 B D
    122 B
    124 B E
    126 B E
    128 B E
    130 B E
    132 B E
    134 B
    136 A D
    137 E
    141 B D
    142 B E
    143 D
    144 B E
    146 B E
    148 C E
    150 B D
    152 C F
    153 B E
    155 A D
    161 B
    163 D
    166 C F
    170 A
    179 A F
    180 C F
    181 C F
    182 C F
    183 C F
    184 C F
    185 B E
    186 B E
    187 B E
    188 B E
    189 B
    194 B E
    208 C E
    209 C F
    210 C F
    211 C F
    212 A E
    213 B E
    214 C F
    215 C E
    216 C F
    217 C F
    219 C F
    220 B E
    221 A D
    222 C E
    223 C F
    225 C F
    231 C D
    232 C F
    236 C E
    253 B E
    254 B E
  • Activity of Camphoric Acids in Dextran Pleurisy Model [0863]
  • Certain compounds of the present invention were tested in a Dextran® pleurisy model. [0864]
  • Rationale for Developing an α[0865] 4β1 Integrin Antagonist to Treat Inflammatory Diseases
  • VLA-4, a member of the β1 integrin family of adhesion molecules, is thought to play a critical role in several types of inflammatory disease processes by promoting leukocyte adhesion to vascular cell adhesion molecule (VCAM-1) and the CS-1 domain of fibronectin in extracellular tissue matrix (Elices M J, Osborn L, Takada Y, Crouse C, Luhowskyj S, Hemler M, Lobb R R. [0866] VCAM-1 on activated endothelium interacts with the leukocyte integrin VLA-4 at a site distinct from the VLA-4-fibronectin binding site. Cell; 60: 577-584, 1990, Humphries M J, Akiyama S K, Komoriya A, Olden K, Yamada K M. Identification of an alternatively-spliced site in human plasma fibronectin that mediates cell type-specific adhesion. J Cell Biol; 103: 2637-2647, 1986, Wayner E A, Garcia-Pardo A, Humphries M J, McDonald J A, Carter W G. Identification and characterization of the T lymphocyte adhesion receptor for an alternative cell attachment domain (CS-1) in plasma fibronectin. J Cell Biol; 109: 1321-1330, 1989, Guan J-L, Hynes R O. Lymlphoid cells recognize an alternatively-sipliced segment of fibronectin via the integrin α 4β1. Cell; 60: 53-61 1990) Of the cell types expressing VLA-4, the major emphasis has been on eosinophils, lymphocytes, and monocytes. Validation of the role of VLA-4 has relied predominantly on the use of anti-VLA-4 antibodies which have been shown to suppress delayed-type hypersensitivity responses (Issekutz T B. Dual inhibition of VLA-4 and LFA-1 maximally inhibits cutaneous delaved-type hypersensitivity-induced inflammation. Am J Pathol; 143: 1286-1293, 1993, Scheynius A, Camp R L, Puré E. Reduced contact sensitivity reactions in mice treated with monoclonal antibodies to leukocyte function-associated molecule-1 and intercellular adhesion molecule-1. J Immunol; 150: 655-663, 1993, Ferguson T A, Kupper T S. Antigen-independent processes in antigen-specific immunity. J Immunol; 150: 1172-1182, 1993, Chisholm P L, Williams C A, Lobb R R. Monoclonal antibodies to the integrin α-4 subunit inhibit the murine contact hypersensitivity response. Eur J Immunol; 23: 682-688, 1993, Elices M J, Tamraz S, Tollefson V, Vollger L W. The integrin VLA-4 mediates leukocyte recruitment to skin inflammatory sites in vivo. Clin Exp Rheumatol; 11 (Suppl 8) S77-80), 1993, experimental allergic encephalomyelitis (Yednock T A, Cannon C, Fritz L C, Sanchez-Madrid F, Steinman L M, Karin N. Prevention of experimental autoimmune encephalomyelitis by antibodies against α 4β1 integrin. Nature; 356: 63-66, 1992, Canella B, Raine C S. The VCAM-1/VLA-4 pathway is involved in chronic lesion expression in multiple sclerosis (MS). J Neuropathol Exp Neurol; 52: 311, 1993), HIV-induced encephalitis (Sasseville V G, Newman W, Brodie S J, Hesterberg P, Pauley D, Ringler D J. Monocyte adhesion to endothelium in simian immunodeficiency virus-induced AIDS encephalitis is mediated by vascular cell adhesion molecule-1/α4β1 integrin reactions. Am J Pathol; 144: 27-40, 1994), pulmonary inflammation and airway hyperreactvity in asthma (Abraham W M, Sielczak M W, Ahmed A, Cortes A, Lauredo I T, Kim J. Pepinsky, B, et al. α1-integrins mediate antigen-induced late bronchial responses and prolonged airway hyperresponsiveness in sheep. J Clin Invest; 93: 776-787, 1994, Pretolani M, Ruffié C, Roberto LapaeSilva J. Joseph D, Lobb R R, Vargaftig B B. Antibody to very late activation antigen 4 prevents antigen-induced bronchial hyperreactivity and cellular infiltration in the guinea-pig airways. J Exp Med; 180: 795-805, 1994), experimental models of autoimmune-mediated diabetes (Yang X-D, Karin N, Tisch R, Steinman L, McDevitt H O. Inhibition of insulitis and prevention of diabetes in non-obese diabetic mice by blocking L-selectin and very late antigen 4 adhesion receptors. Proc Natl Acad Sci USA; 90: 10494-10498, 1993, Burkly L C, Jakubowski A, Hattori M. Protection against adoptive transfer of autoimmune diabetes medicated through very late antigen-4 integrin. Diabetes; 43: 529-534, 1994), and experimental colitis (Podolsky D K, Lobb R, King N, Benjamin C D, Pepinsky B, Sehgal P, et al. Attentuation of colitis in the cotton-top Tamarin by anti-α4 integrin monoclonal antibody. J Clin Invest; 92: 372-380, 1993). Since eosinophils represent a major component of the inflammatory cell influx in asthmatic lung tissue we developed a simple acute inflammatory model of VLA-4 integrin-dependent eosinophil infiltration which could be used to identify VLA-4 antagonists; such compounds would be of potential value in the treatment of asthma as well as other diseases in which VLA-4 played a role.
  • Materials and Methods [0867]
  • Animals, Housing and Viral Testing: [0868]
  • C57BL/6 mice (Jackson, Bar Harbor, Me.;), 6-8 weeks old, weighing 20-25 g were used throughout All mice were acclimated for at least 7-14 days after arrival and maintained under controlled temperature (20-22° C.) and a 12 hr daily light cycle (6.00 A.M.-6.00 P.M.). Mice were housed in laminar flow racks and checked biweekly for viral infections (mouse hepatitis virus, minute virus of mice, rodent orphan parvovirus, Sendai) with kits obtained from Oreganon Teknika (Durham, N.C.) using established enzyme-linked immunoabsorbent assays. Mice testing positive for any of the above were omitted from the study. All mice were fed standard laboratory chow (Upjohn Lab Rodent Irradiated Mouse Chow, #5011-3, PMI Feeds, St. Louis, Mo.) and acidified drinking water (pH 5.0) ad libitum. [0869]
  • Induction of Inflammation by Intrapleural Injection of Dextran: [0870]
  • Intrapleural injections were made using a 27G needle cut to 3-4 mm and blunted by filing. Injections were made by inserting the needle between the mid-intercostal ribs on the right side of the thoracic cavity. [0871]
  • Dextran (MW 5-40×10[0872] 6, St Louis, Mo.) was injected as a 10% solution in saline in a volume of 100 μl/mouse. Care was taken to avoid bleeding at the site of injection at which the intercostal muscles were cut to facilitate smooth insertion of the needle.
  • Quantitation of Pleural Inflammatory Leukocyte Responses: [0873]
  • Pleural leukocytes were collected as follows: 4 h post-induction, pleural inflammatory exudate was removed by washing with 2×1.0 ml Ca[0874] ++/Mg++ free HBSS (Gibco, Grand Island, N.Y.) containing 45 mg EDTA/100 ml HBSS, 4° C. Total leukocyte counts were made by hemocytometer following erythrocyte lysis in 2% acetic acid in PBS buffer; exudate leukocyte pellets were resuspended in serum for cytospin preparations and stained (Diff Quik, Baxter Healthcare, McGraw Park, Ill.) for differential leukocyte counts (neutrophils, eosinophils, and mononuclear leukocytes). The pleural cavities of mice receiving either no intrapleural injection, or saline were washed and the cells counted in the same way to estimate baseline or saline-induced pleural leukocyte counts respectively
  • Administration of Drugs: [0875]
  • All drugs were dissolved in PBS and the pH adjusted to 7.5 with NaOH. Each drug was administered intravenously through the retroorbital sinus at hourly intervals (0-3 h) starting from time “0” as indicated. Mice were carefully monitored for side effects; none were noted for the series of compounds reported herein. [0876]
  • The following camphoric acid analogues were tested for their inhibitory effects on dextran-induced leukocyte infiltration: Examples 4, 12, 54, 63, 166, 93, 180, 181, 183, 184, 217, Camphoric acid or PBS (saline) was administered iv. as a control. Inhibition of eosinophll infiltration, which was suppressed by anti-alpha-4 Mab (PS/2, 50%), was used as a readout of VLA-4 antagonist activity of the camphoric acids tested. Data for neutrophils are also reported. [0877]
  • Results: [0878]
  • Dextran Pleural Leukocyte Response. [0879]
  • The total pleural leukocyte counts were 255×10[0880] 4 (+/−16 SEM) cells in the normal pleural cavity; of the normal pleural leukocyte population, all cells were mononuclear (a similar response was observed following intrapleural saline injection). Four hours after intrapleural injection of dextran total pleural leukocyte counts increased to 719×104 (+/−67 SEM) and comprised 36.8×104 (+/−4.1 SEM) eosinophils, 292×104 (+/−25 SEM) neutrophils and 391×104 (+/−48 SEM) mononuclear leukocytes.
  • Inhibition of neutrophils and eosinophil (%Δ) are depicted by A, B and C according to the following ranges: A≦−50, −50<B≦−10, −10<C≦0 [0881]
  • Inhibition of Dextran Pleural Leukocyte Infiltration by Camphoric Acids. [0882]
    %Δ Pleural Leukocytes
    Treatment Dose (mg/kg) # Neutro Eos
    Ex. 12  50 × 4 iv B A
    50 × 2 iv B A
    Ex. 54  50 × 4 iv B A
    Ex. 4  25 × 2 iv
    50 × 2 iv B A
    Ex. 63  25 × 2 iv B B
    50 × 2 iv B B
    Ex. 166 25 × 2 iv
    50 × 2 iv C B
    Ex. 93  25 × 2 iv
    50 × 2 iv C B
    Ex. 181 25 × 2 iv B B
    50 × 2 iv B B
    Ex. 180 25 × 2 iv B B
    50 × 2 iv B B
    Ex. 183 25 × 2 iv B B
    50 × 2 iv B B
    Ex. 184 25 × 2 iv B B
    50 × 2 iv B A
    Ex. 217 25 × 2 iv
    50 × 2 iv C B
  • p<0.05 * Drug treated vs camphoric acid or PBS treated control. [0883]

Claims (15)

What is claimed is:
1. A compound of the formula (I):
Figure US20030130349A1-20030710-C00401
wherein
n is an integer of 0 or 1;
R1 is a hydrogen atom or methyl group,
R2 is a group of the formula: —CN, —COOH, —(C1-6 alkylene)OH, —CH2O(C1-6 alkyl), —(C1-6 alkylene)COOH, —CH2O(C1-6 alkylene)O(C1-6 alkyl) —CH2O(C1-6 alkylene)COOH, —(C2-7 alkenylene)COOH, —CO(C1-6 alkylene)COOH, —CO(C2-7 alkenylene)COOH, —CO(C1-6 alkylene)O(C1-6 alkyl) —CO(C1-6 alkylene)CO(C1-6 alkyl), —CONH(C1-6 alkyl), —CONHO(C1-6 alkyl), —CONH(C1-6 alkylene)COOH, —CONH2, —CONH(C3-7 cycloalkyl),
Figure US20030130349A1-20030710-C00402
—CONHOCH2Ph, —CONH(C1-6 alkylene)CN, —COO(C1-6 alkyl), —CH2O(C1-6 alkylene)CONH2, —CONH(C1-6 alkylene)CONH2, —CONHOH, —NHCOOCH2Ph,
Figure US20030130349A1-20030710-C00403
R3 is a hydrogen atom or a methyl group;
X is a methylene group or a group of the formula: —CO—,
R4 is a hydrogen atom or a C1-6 alkyl group;
R5 is a group of the formula: —COOH or an ester or an amide thereof, —(C1-6 alkylene)COOH or an ester or an amide thereof, —(C1-7 alkylene)O(C1-6 alkyl), —(C1-7 alkylene)OH, —COO(C1-6 alkyl), —CONH(C1-6 alkyl), or —CONH2;
R6 is a substituent of the formula:
Figure US20030130349A1-20030710-C00404
wherein,
R7, which occurs one or more times and which may be the same or different in each occurrence, is —H, —OH, —NO2, —NH2, —C1-C5 alkyl, —F, —Cl, —Br, —I, —COOH, —COO(C1-6 alkyl), —O(C1-C8 alkyl),
—CONH(C1-6 alkylene)COOH, —OCH2(C3-C7 cycloalkyl) or a substituent of the formula
Figure US20030130349A1-20030710-C00405
R8, which occurs one or more times and which may be the same or different in each occurrence, is —H, —OH, —NH2, —NO2, —C1-C7 alkyl, —F, —Cl, —Br, —I, —CF3, phenyl, or —O(C1-6 alkyl);
R9 is selected from a group of the formula: —H, —C1-C5 alkyl, —C3-C7 cycloalkyl, —(—C1-C6 alkylene)aryl, aryl, or a substituent of the formula:
Figure US20030130349A1-20030710-C00406
R10, which occurs one or more times and which may be the same or different in each occurrence, is —H, —F, —Cl, —Br, or —I, —NO2, —C1-6 alkyl or —O(C1-6 alkyl);
R11 is selected from a group of the formula:
Figure US20030130349A1-20030710-C00407
R12, which occurs one or more times and which may be the same or different in each occurrence, is —H, —CF3, —OCF3, —F, —Br, —Cl, or —I;
with the proviso that R1 and R3 must be different and also with the proviso that when R2 or R6 is a moiety of the formula —COOH or contains a moiety of the formula —COOH, then a pharmaceutically acceptable ester or a pharmaceutically acceptable amide thereof are included, and also with the proviso that
when R7 is the formula
Figure US20030130349A1-20030710-C00408
R9 is other than hydrogen,
and also with the proviso that when R6 is the formula
Figure US20030130349A1-20030710-C00409
or a pharmaceutically acceptable salt thereof.
2. The compound according to claim 1, which is a compound of the formula (1-1):
Figure US20030130349A1-20030710-C00410
wherein n, R1 through R6 and X are as defined above.
3. The compound according to claim 1, which is a compound of the formula (1-2):
Figure US20030130349A1-20030710-C00411
wherein n, R1 through R4, R6 and X are as defined above and R5 is a group of the formula: —COOH, —(C1-6 alkylene)COOH, —(C1-7 alkylene)O(C1-6 alkyl) —(C1-7 alkylene)OH, —COO(C1-6 alkyl)-CONH(C1-6 alkyl), or —CONH2.
4. The compound according to claim 1, wherein R6 is
Figure US20030130349A1-20030710-C00412
wherein
Y is a hydrogen atom or a chlorine atom.
5. The compound according to claim 1, wherein R2 is a group of the formula: is a group of the formula: —COOH or an ester or an amide thereof, —CONHCH2COOH, —CONHOCH2Ph or —CONHCH2CONH2.
6. The compound according to claim 1, wherein said compound is selected from the group consisting of Examples 4, 24, 26, 28, 29, 31, 36, 38, 77, 130, 146, 194, 208, 209, 210, 212, 213, 215, and 225.
7. The compound according to claim 6, wherein said compound is selected from the group consisting of Examples 4, 24, 26, 28, 38, 77, 130, 146, 194, 208, 209, 210, 213, 215, and 225.
8. The compound according to claim 7, wherein said compound is selected from the group consisting of Examples 208, 209, 210, 215, and 225.
9. The compound according to claim 1, wherein said compound is selected from the group consisting of Examples 4, 24, 26, 28, 29, 36, 38, 77, 130, 194, 208, 209, 210, 212, 213, 215, and 225.
10. The compound according to claim 9, wherein said compound is selected from the group consisting of Examples 4, 24, 28, 38, 77, 130, 194, 208, 209, 210, 212, 213, 215, and 225.
11. The compound according to claim 10, wherein said compound is selected from the group consisting of Examples 209, 210, and 225.
12. The compound according to claim 1, wherein said compound is (1S cis)-N-[3-carboxy-2,2,3-trimethylcyclopentyl)carbonyl]-1-[(3,4-dichlorophenyl)methyl]-L-histidine.
13. A pharmaceutical composition comprising:
a therapeutically effective amount of the compound as set forth in claims 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12; and
a pharmaceutically acceptable carrier or diluent.
14. A method for treating or preventing (4 μl adhesion mediated conditions in a human which comprises administering to a patient an effective amount of the compound according to claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12.
15. A method according to claim 14, wherein said condition is selected from the group consisting of rheumatoid arthritis, asthma, allergy conditions, allograft rejection, psoriasis, eczema, contact dermatitis and other skin inflammatory diseases, inflammatory and immunoinflammatory conditions including ophthalmic inflammatory conditions, inflammatory bowel diseases, atherosclerosis, and ulcerative colitis.
US10/193,137 1997-06-23 2002-07-12 Inhibitors of α4β1 mediated cell adhesion Expired - Fee Related US6596752B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/193,137 US6596752B1 (en) 1997-06-23 2002-07-12 Inhibitors of α4β1 mediated cell adhesion

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US5051597P 1997-06-23 1997-06-23
US09/102,584 US6482849B1 (en) 1997-06-23 1998-06-23 Inhibitors of α4β1 mediated cell adhesion
US10/193,137 US6596752B1 (en) 1997-06-23 2002-07-12 Inhibitors of α4β1 mediated cell adhesion

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/102,584 Division US6482849B1 (en) 1997-06-23 1998-06-23 Inhibitors of α4β1 mediated cell adhesion

Publications (2)

Publication Number Publication Date
US20030130349A1 true US20030130349A1 (en) 2003-07-10
US6596752B1 US6596752B1 (en) 2003-07-22

Family

ID=21965686

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/102,584 Expired - Fee Related US6482849B1 (en) 1997-06-23 1998-06-23 Inhibitors of α4β1 mediated cell adhesion
US10/193,137 Expired - Fee Related US6596752B1 (en) 1997-06-23 2002-07-12 Inhibitors of α4β1 mediated cell adhesion

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/102,584 Expired - Fee Related US6482849B1 (en) 1997-06-23 1998-06-23 Inhibitors of α4β1 mediated cell adhesion

Country Status (10)

Country Link
US (2) US6482849B1 (en)
EP (1) EP0991619B1 (en)
JP (1) JP2001517246A (en)
AT (1) ATE249421T1 (en)
AU (1) AU8163398A (en)
DE (1) DE69818049T2 (en)
DK (1) DK0991619T3 (en)
ES (1) ES2206953T3 (en)
PT (1) PT991619E (en)
WO (1) WO1998058902A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009002964A1 (en) * 2007-06-26 2008-12-31 Lexicon Pharmaceuticals, Inc. Methods of treating serotonin-mediated diseases and disorders
WO2010062829A1 (en) * 2008-11-28 2010-06-03 Lexicon Pharmaceuticals, Inc. Tryptophan hydroxylase inhibitors for treating osteoporosis
US7875622B2 (en) 2007-07-11 2011-01-25 Lexicon Pharmaceuticals, Inc. Methods and compositions for treating pulmonary hypertension and related diseases and disorders
EP2510941A2 (en) 2007-02-20 2012-10-17 Merrimack Pharmaceuticals, Inc. Methods of treating multiple sclerosis by administration of alpha-fetoprotein in combination with an integrin antagonist
WO2018134836A1 (en) * 2017-01-23 2018-07-26 Synven Shree Life Sciences Pvt. Ltd. A novel, feasible and cost effective process for the manufacture of d – penicillamine
US11427530B2 (en) * 2018-02-09 2022-08-30 Vistagen Therapeutics, Inc. Synthesis of 4-chlorokynurenines and intermediates

Families Citing this family (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6093696A (en) * 1997-05-30 2000-07-25 Celltech Therapeutics, Limited Tyrosine derivatives
US6583139B1 (en) 1997-07-31 2003-06-24 Eugene D. Thorsett Compounds which inhibit leukocyte adhesion mediated by VLA-4
US6291453B1 (en) 1997-07-31 2001-09-18 Athena Neurosciences, Inc. 4-amino-phenylalanine type compounds which inhibit leukocyte adhesion mediated by VLA-4
US6492421B1 (en) 1997-07-31 2002-12-10 Athena Neurosciences, Inc. Substituted phenylalanine type compounds which inhibit leukocyte adhesion mediated by VLA-4
US6489300B1 (en) 1997-07-31 2002-12-03 Eugene D. Thorsett Carbamyloxy compounds which inhibit leukocyte adhesion mediated by VLA-4
US6939855B2 (en) 1997-07-31 2005-09-06 Elan Pharmaceuticals, Inc. Anti-inflammatory compositions and method
US6559127B1 (en) 1997-07-31 2003-05-06 Athena Neurosciences, Inc. Compounds which inhibit leukocyte adhesion mediated by VLA-4
US6423688B1 (en) 1997-07-31 2002-07-23 Athena Neurosciences, Inc. Dipeptide and related compounds which inhibit leukocyte adhesion mediated by VLA-4
US7030114B1 (en) 1997-07-31 2006-04-18 Elan Pharmaceuticals, Inc. Compounds which inhibit leukocyte adhesion mediated by VLA-4
US6362341B1 (en) 1997-07-31 2002-03-26 Athena Neurosciences, Inc. Benzyl compounds which inhibit leukocyte adhesion mediated by VLA-4
MY153569A (en) 1998-01-20 2015-02-27 Mitsubishi Tanabe Pharma Corp Inhibitors of ?4 mediated cell adhesion
US6329372B1 (en) 1998-01-27 2001-12-11 Celltech Therapeutics Limited Phenylalanine derivatives
EP1056714B1 (en) 1998-02-26 2004-08-11 Celltech Therapeutics Limited Phenylalanine derivatives as inhibitors of alpha4 integrins
US6521626B1 (en) 1998-03-24 2003-02-18 Celltech R&D Limited Thiocarboxamide derivatives
GB9811159D0 (en) 1998-05-22 1998-07-22 Celltech Therapeutics Ltd Chemical compounds
GB9811969D0 (en) 1998-06-03 1998-07-29 Celltech Therapeutics Ltd Chemical compounds
GB9812088D0 (en) * 1998-06-05 1998-08-05 Celltech Therapeutics Ltd Chemical compounds
GB9814414D0 (en) 1998-07-03 1998-09-02 Celltech Therapeutics Ltd Chemical compounds
GB9821061D0 (en) 1998-09-28 1998-11-18 Celltech Therapeutics Ltd Chemical compounds
GB9821222D0 (en) 1998-09-30 1998-11-25 Celltech Therapeutics Ltd Chemical compounds
GB9825652D0 (en) 1998-11-23 1999-01-13 Celltech Therapeutics Ltd Chemical compounds
GB9826174D0 (en) 1998-11-30 1999-01-20 Celltech Therapeutics Ltd Chemical compounds
WO2000037429A2 (en) * 1998-12-22 2000-06-29 Tanabe Seiyaku Co., Ltd. INHIBITORS OF α4β1 MEDIATED CELL ADHESION
US6407066B1 (en) 1999-01-26 2002-06-18 Elan Pharmaceuticals, Inc. Pyroglutamic acid derivatives and related compounds which inhibit leukocyte adhesion mediated by VLA-4
EP1154993B1 (en) * 1999-02-18 2004-10-06 F. Hoffmann-La Roche Ag Thioamide derivatives
DE60021368T2 (en) * 1999-04-19 2006-07-27 Lexicon Pharmaceuticals (New Jersey), Inc. PPAR (GAMMA) AGONISTS FOR THE TREATMENT OF TYPE II DIABETES
US6518283B1 (en) 1999-05-28 2003-02-11 Celltech R&D Limited Squaric acid derivatives
SK287075B6 (en) * 1999-08-13 2009-11-05 Biogen Idec Ma Inc. Cell adhesion inhibitors and pharmaceutical compositions comprising them
US6534513B1 (en) 1999-09-29 2003-03-18 Celltech R&D Limited Phenylalkanoic acid derivatives
US6455539B2 (en) 1999-12-23 2002-09-24 Celltech R&D Limited Squaric acid derivates
OA12126A (en) 1999-12-28 2006-05-05 Pfizer Prod Inc Non-peptidyl inhibitors of VLA-4 dependent cell binding useful in treating inflammatory, autoimmune,and respiratory diseases.
WO2001047868A1 (en) 1999-12-28 2001-07-05 Ajinomoto Co., Inc. Novel phenylalanine derivatives
NZ520299A (en) * 2000-02-03 2004-05-28 Eisai Co Ltd Integrin expression inhibitors containing sulphonamide derivatives
AU2001248553A1 (en) 2000-04-17 2001-10-30 Celltech R And D Limited Enamine derivatives as cell adhesion molecules
US6545013B2 (en) 2000-05-30 2003-04-08 Celltech R&D Limited 2,7-naphthyridine derivatives
US6403608B1 (en) 2000-05-30 2002-06-11 Celltech R&D, Ltd. 3-Substituted isoquinolin-1-yl derivatives
US6740654B2 (en) 2000-07-07 2004-05-25 Celltech R & D Limited Squaric acid derivatives
AU2001275724A1 (en) 2000-08-02 2002-02-13 Celltech R&D Limited 3-substituted isoquinolin-1-yl derivatives
MY129000A (en) 2000-08-31 2007-03-30 Tanabe Seiyaku Co INHIBITORS OF a4 MEDIATED CELL ADHESION
ES2200617B1 (en) 2001-01-19 2005-05-01 Almirall Prodesfarma, S.A. DERIVATIVES OF UREA AS ANTAGONISTS OF INTEGRINAS ALPHA 4.
MY140707A (en) * 2002-02-28 2010-01-15 Mitsubishi Tanabe Pharma Corp Process for preparing a phenylalanine derivative and intermediates thereof
US7247725B2 (en) 2002-10-30 2007-07-24 Merck & Co., Inc. Gamma-aminoamide modulators of chemokine receptor activity
US7678798B2 (en) 2004-04-13 2010-03-16 Incyte Corporation Piperazinylpiperidine derivatives as chemokine receptor antagonists
JP2007532681A (en) * 2004-04-16 2007-11-15 ジェネンテック・インコーポレーテッド Methods for increasing B cell depletion
US8267905B2 (en) * 2006-05-01 2012-09-18 Neurosystec Corporation Apparatus and method for delivery of therapeutic and other types of agents
US7803148B2 (en) 2006-06-09 2010-09-28 Neurosystec Corporation Flow-induced delivery from a drug mass
EP2056793A4 (en) * 2006-07-31 2011-08-17 Neurosystec Corp Free base gacyclidine nanoparticles
PL2288715T3 (en) 2008-04-11 2015-03-31 Merrimack Pharmaceuticals Inc Human serum albumin linkers and conjugates thereof
CN102282168A (en) 2008-11-18 2011-12-14 梅里麦克制药股份有限公司 Human serum albumin linkers and conjugates thereof
BRPI0920936A2 (en) 2008-11-26 2020-08-11 Pfizer Inc 3-aminocyclopentanecarboxamides as modulators of chemokine receptors
WO2010079413A2 (en) * 2009-01-09 2010-07-15 Orchid Research Laboratories Ltd. Dipeptidyl peptidase iv inhibitors
CN102307875A (en) 2009-02-09 2012-01-04 苏伯俭股份有限公司 Pyrrolopyrimidinyl axl kinase inhibitors
WO2011094890A1 (en) * 2010-02-02 2011-08-11 Argusina Inc. Phenylalanine derivatives and their use as non-peptide glp-1 receptor modulators
EP2769223A1 (en) 2011-10-17 2014-08-27 Westfälische Wilhelms-Universität Münster Assessment of pml risk and methods based thereon
US20150202287A1 (en) 2012-08-30 2015-07-23 Merrimack Pharmaceuticals, Inc. Combination therapies comprising anti-erbb3 agents
US20150045435A1 (en) * 2013-08-06 2015-02-12 Indiana University Research And Technology Corporation Compounds and methods for treating diabetes
CN112969504B (en) 2018-10-30 2024-04-09 吉利德科学公司 Compounds for inhibiting alpha 4 beta 7 integrin
SG11202103484RA (en) 2018-10-30 2021-05-28 Gilead Sciences Inc Quinoline derivatives as alpha4beta7 integrin inhibitors
AU2019373244B2 (en) 2018-10-30 2022-05-26 Gilead Sciences, Inc. Imidazopyridine derivatives as alpha4beta7 integrin inhibitors
CN112996786A (en) 2018-10-30 2021-06-18 吉利德科学公司 Compounds for inhibiting alpha 4 beta 7 integrins
CN110305034B (en) * 2019-06-24 2022-06-07 佛山市顺德区香江精细化工实业有限公司 Composition containing camphoric acid derivative and application thereof in cosmetics
KR20220047323A (en) 2019-08-14 2022-04-15 길리애드 사이언시즈, 인코포레이티드 Compounds for Inhibiting Alpha 4 Beta 7 Integrin

Family Cites Families (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4589881A (en) 1982-08-04 1986-05-20 La Jolla Cancer Research Foundation Polypeptide
US4578079A (en) 1982-08-04 1986-03-25 La Jolla Cancer Research Foundation Tetrapeptide
US4517686A (en) 1982-08-04 1985-05-21 La Jolla Cancer Research Foundation Polypeptide
US4792525A (en) 1982-08-04 1988-12-20 La Jolla Cancer Research Foundation Tetrapeptide
US4614517A (en) 1982-08-04 1986-09-30 La Jolla Cancer Research Foundation Tetrapeptide
US4661111A (en) 1982-08-04 1987-04-28 La Jolla Cancer Research Foundation Polypeptide
JPS6354321A (en) 1985-03-27 1988-03-08 Ajinomoto Co Inc Blood sugar lowering agent
US4683291A (en) 1985-10-28 1987-07-28 Scripps Clinic And Research Foundation Platelet binding inhibitors
US6221367B1 (en) * 1992-06-15 2001-04-24 Emisphere Technologies, Inc. Active agent transport systems
US6306840B1 (en) * 1995-01-23 2001-10-23 Biogen, Inc. Cell adhesion inhibitors
WO1998053818A1 (en) 1997-05-29 1998-12-03 Merck & Co., Inc. Sulfonamides as cell adhesion inhibitors
ATE318841T1 (en) 1997-05-29 2006-03-15 Merck & Co Inc BIARYLALKANEOUS ACIDS USED AS CELL ADHESION INHIBITORS
JP2002512625A (en) 1997-05-29 2002-04-23 メルク エンド カンパニー インコーポレーテッド Heterocyclic amide compounds as cell adhesion inhibitors
US6093696A (en) 1997-05-30 2000-07-25 Celltech Therapeutics, Limited Tyrosine derivatives
PL338373A1 (en) 1997-07-31 2000-10-23 Elan Pharm Inc Dipeptide and its affinite compounds inhibiting adhesion of leucocytes occurring through mediation of vla-4
AR013384A1 (en) 1997-07-31 2000-12-27 Athena Neurosciences Inc SUBSTITUTED TYPE OF PHENYLALANINE COMPOUNDS INHIBITING LEUKOCYTE ADHESION INTERMEDIATED BY VLA-4
BR9812118A (en) 1997-07-31 2000-07-18 Elan Pharm Inc 4-Amino-phenylalanine-type compounds that inhibit vla-4-mediated leukocyte adhesion
JP2001512137A (en) 1997-07-31 2001-08-21 エラン・ファーマシューティカルズ・インコーポレーテッド Dipeptide compounds that inhibit leukocyte adhesion mediated by VLA-4
BR9811569A (en) 1997-07-31 2000-09-19 Elan Pharm Inc Compounds that inhibit vla-4-mediated leukocyte adhesion
AR016133A1 (en) 1997-07-31 2001-06-20 Wyeth Corp CARBAMILOXI COMPOUND INHIBITING THE ADHESION OF LEUKOCYTES THROUGH VLA-4, COMPOUNDS THAT ARE DRUGS OF THESE COMPOUNDS, PHARMACEUTICAL COMPOSITION, METHOD FOR SETTING VLA-4 TO A BIOLOGICAL SAMPLE, METHOD FOR THE TREATMENT OF A TREATMENT
KR20010022413A (en) 1997-07-31 2001-03-15 진 엠. 듀발 Benzyl compounds which inhibit leukocyte adhesion mediated by VLA-4
BR9811594A (en) 1997-07-31 2000-09-05 Elan Pharm Inc Sulphonylated dipeptide compounds that inhibit vla-4 mediated leukocyte adhesion
RU2220950C2 (en) 1997-08-22 2004-01-10 Ф.Хоффманн-Ля Рош Аг N-alkanoylphenylalanine derivatives
EP1005446B1 (en) 1997-08-22 2004-02-25 F. Hoffmann-La Roche Ag N-aroylphenylalanine derivatives
AU1463499A (en) 1997-11-21 1999-06-15 Merck & Co., Inc. Substituted pyrrole derivatives as cell adhesion inhibitors
AU751950B2 (en) 1997-11-24 2002-09-05 Merck & Co., Inc. Substituted beta-alanine derivatives as cell adhesion inhibitors
CA2309338A1 (en) 1997-11-24 1999-06-03 Merck & Co., Inc. Cyclic amino acid derivatives as cell adhesion inhibitors
US6197794B1 (en) 1998-01-08 2001-03-06 Celltech Therapeutics Limited Phenylalanine derivatives
US6329372B1 (en) 1998-01-27 2001-12-11 Celltech Therapeutics Limited Phenylalanine derivatives
EP1056714B1 (en) 1998-02-26 2004-08-11 Celltech Therapeutics Limited Phenylalanine derivatives as inhibitors of alpha4 integrins
US6521626B1 (en) 1998-03-24 2003-02-18 Celltech R&D Limited Thiocarboxamide derivatives
GB9811159D0 (en) 1998-05-22 1998-07-22 Celltech Therapeutics Ltd Chemical compounds
GB9812088D0 (en) 1998-06-05 1998-08-05 Celltech Therapeutics Ltd Chemical compounds
WO1999064395A1 (en) 1998-06-11 1999-12-16 Merck & Co., Inc. Heterocyclic amide compounds as cell adhesion inhibitors

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2510941A2 (en) 2007-02-20 2012-10-17 Merrimack Pharmaceuticals, Inc. Methods of treating multiple sclerosis by administration of alpha-fetoprotein in combination with an integrin antagonist
WO2009002964A1 (en) * 2007-06-26 2008-12-31 Lexicon Pharmaceuticals, Inc. Methods of treating serotonin-mediated diseases and disorders
US20090005381A1 (en) * 2007-06-26 2009-01-01 Philip Manton Brown Methods of treating serotonin-mediated diseases and disorders
US7875622B2 (en) 2007-07-11 2011-01-25 Lexicon Pharmaceuticals, Inc. Methods and compositions for treating pulmonary hypertension and related diseases and disorders
US8410121B2 (en) 2007-07-11 2013-04-02 Lexicon Pharmaceuticals, Inc. Methods of treating pulmonary hypertension
WO2010062829A1 (en) * 2008-11-28 2010-06-03 Lexicon Pharmaceuticals, Inc. Tryptophan hydroxylase inhibitors for treating osteoporosis
WO2018134836A1 (en) * 2017-01-23 2018-07-26 Synven Shree Life Sciences Pvt. Ltd. A novel, feasible and cost effective process for the manufacture of d – penicillamine
US11427530B2 (en) * 2018-02-09 2022-08-30 Vistagen Therapeutics, Inc. Synthesis of 4-chlorokynurenines and intermediates

Also Published As

Publication number Publication date
ES2206953T3 (en) 2004-05-16
DK0991619T3 (en) 2003-12-29
US6596752B1 (en) 2003-07-22
EP0991619A1 (en) 2000-04-12
PT991619E (en) 2004-02-27
DE69818049D1 (en) 2003-10-16
US6482849B1 (en) 2002-11-19
ATE249421T1 (en) 2003-09-15
WO1998058902A1 (en) 1998-12-30
JP2001517246A (en) 2001-10-02
EP0991619B1 (en) 2003-09-10
DE69818049T2 (en) 2004-07-15
AU8163398A (en) 1999-01-04

Similar Documents

Publication Publication Date Title
US6482849B1 (en) Inhibitors of α4β1 mediated cell adhesion
AU764553B2 (en) Inhibitors of alpha 4 beta 1 mediated cell adhesion
EP1049662B1 (en) Inhibitors of alpha4 mediated cell adhesion
US6420418B1 (en) Heterocycle amides as cell adhesion inhibitors
EP1144365B1 (en) Inhibitors of alpha-4 beta-1 mediated cell adhesion
US7265146B2 (en) Integrin receptor inhibitors
BG65755B1 (en) Cell adhesion inhibitors
PL191082B1 (en) Cell adhesion inhibitors, process for their preparation and use
WO2001068586A2 (en) α4β1 AND α4β7 INTEGRIN INHIBITORS
JP2003503350A (en) VLA-4 inhibitor compounds
CA2140931A1 (en) Non-peptidic surrogates of the ldv sequence and their use in the treatment of inflammation, autoimmune disease and tumour progression
KR20010034317A (en) VLA-4 Antagonists
US6685617B1 (en) Inhibitors of α4β1 mediated cell adhesion
US20030232868A1 (en) Cyclic carboxylic acids as integrin antagonists
IE910083A1 (en) Cycloalkyl-substituted glutaramide diuretic agents
PL201421B1 (en) Spiroimidazolidine derivatives, their preparation, their use and pharmaceutical preparations comprising them
GB2369357A (en) Aliphatic, cyclic amino carboxylic acids as integrin antagonists
US20020091142A1 (en) Alpha4beta1 and alpha4beta7 integrin inhibitors
AU3006401A (en) Vla-4 integrin antagonists

Legal Events

Date Code Title Description
REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20070722