WO1993001159A1 - Fused tricyclic compounds, pharmaceutical compositions containing them and their use in therapy - Google Patents

Fused tricyclic compounds, pharmaceutical compositions containing them and their use in therapy Download PDF

Info

Publication number
WO1993001159A1
WO1993001159A1 PCT/GB1992/001212 GB9201212W WO9301159A1 WO 1993001159 A1 WO1993001159 A1 WO 1993001159A1 GB 9201212 W GB9201212 W GB 9201212W WO 9301159 A1 WO9301159 A1 WO 9301159A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
phenyl
compound
coor
formula
Prior art date
Application number
PCT/GB1992/001212
Other languages
French (fr)
Inventor
Walfred Spencer Saari
Monique Bodil Van Niel
Brian John Williams
Original Assignee
Merck Sharp & Dohme Limited
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
Priority claimed from GB919114886A external-priority patent/GB9114886D0/en
Priority claimed from GB929208141A external-priority patent/GB9208141D0/en
Application filed by Merck Sharp & Dohme Limited filed Critical Merck Sharp & Dohme Limited
Priority to US08/175,432 priority Critical patent/US5495047A/en
Publication of WO1993001159A1 publication Critical patent/WO1993001159A1/en

Links

Classifications

    • 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
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C211/00Compounds containing amino groups bound to a carbon skeleton
    • C07C211/01Compounds containing amino groups bound to a carbon skeleton having amino groups bound to acyclic carbon atoms
    • C07C211/26Compounds containing amino groups bound to a carbon skeleton having amino groups bound to acyclic carbon atoms of an unsaturated carbon skeleton containing at least one six-membered aromatic ring
    • C07C211/29Compounds containing amino groups bound to a carbon skeleton having amino groups bound to acyclic carbon atoms of an unsaturated carbon skeleton containing at least one six-membered aromatic ring the carbon skeleton being further substituted by halogen atoms or by nitro or nitroso groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C217/00Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton
    • C07C217/02Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having etherified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C217/48Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having etherified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being unsaturated and containing rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C217/00Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton
    • C07C217/54Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having etherified hydroxy groups bound to carbon atoms of at least one six-membered aromatic ring and amino groups bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton
    • C07C217/64Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having etherified hydroxy groups bound to carbon atoms of at least one six-membered aromatic ring and amino groups bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton with amino groups linked to the six-membered aromatic ring, or to the condensed ring system containing that ring, by carbon chains further substituted by singly-bound oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/02Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C229/04Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C229/06Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated having only one amino and one carboxyl group bound to the carbon skeleton
    • C07C229/10Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated having only one amino and one carboxyl group bound to the carbon skeleton the nitrogen atom of the amino group being further bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings
    • C07C229/14Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated having only one amino and one carboxyl group bound to the carbon skeleton the nitrogen atom of the amino group being further bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings to carbon atoms of carbon skeletons containing rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/02Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C229/34Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton containing six-membered aromatic rings
    • C07C229/36Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton containing six-membered aromatic rings with at least one amino group and one carboxyl group bound to the same carbon atom of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/01Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C233/16Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms
    • C07C233/17Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom
    • C07C233/18Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom having the carbon atom of the carboxamide group bound to a hydrogen atom or to a carbon atom of an acyclic saturated carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/01Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C233/16Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms
    • C07C233/17Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom
    • C07C233/22Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom having the carbon atom of the carboxamide group bound to an acyclic carbon atom of a carbon skeleton containing six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/01Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C233/45Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups
    • C07C233/46Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom
    • C07C233/47Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom having the carbon atom of the carboxamide group bound to a hydrogen atom or to a carbon atom of an acyclic saturated carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/64Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings
    • C07C233/67Carboxylic 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 singly-bound oxygen atoms
    • C07C233/68Carboxylic 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 singly-bound oxygen atoms with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom
    • C07C233/73Carboxylic 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 singly-bound oxygen atoms 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
    • 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
    • C07C237/04Carboxylic 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 the carbon skeleton being acyclic and saturated
    • C07C237/06Carboxylic 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 the carbon skeleton being acyclic and saturated having the nitrogen atoms of the carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • 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
    • C07C237/04Carboxylic 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 the carbon skeleton being acyclic and saturated
    • C07C237/08Carboxylic 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 the carbon skeleton being acyclic and saturated having the nitrogen atom of at least one of the carboxamide groups bound to an acyclic carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms
    • 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
    • C07C237/20Carboxylic 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 the carbon skeleton containing six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C255/00Carboxylic acid nitriles
    • C07C255/01Carboxylic acid nitriles having cyano groups bound to acyclic carbon atoms
    • C07C255/11Carboxylic acid nitriles having cyano groups bound to acyclic carbon atoms containing cyano groups and singly-bound oxygen atoms bound to the same saturated acyclic carbon skeleton
    • C07C255/13Carboxylic acid nitriles having cyano groups bound to acyclic carbon atoms containing cyano groups and singly-bound oxygen atoms bound to the same saturated acyclic carbon skeleton containing cyano groups and etherified hydroxy groups bound to the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C255/00Carboxylic acid nitriles
    • C07C255/49Carboxylic acid nitriles having cyano groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton
    • C07C255/54Carboxylic acid nitriles having cyano groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton containing cyano groups and etherified hydroxy groups bound to the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/06Esters of carbamic acids
    • C07C271/08Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms
    • C07C271/10Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C271/16Esters 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 singly-bound oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/06Esters of carbamic acids
    • C07C271/08Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms
    • C07C271/10Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C271/20Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms to carbon atoms of hydrocarbon radicals substituted by nitrogen atoms not being part of nitro or nitroso groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • 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
    • C07C275/00Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups
    • C07C275/04Derivatives 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 acyclic carbon atoms
    • C07C275/06Derivatives 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 acyclic carbon atoms of an acyclic and saturated carbon skeleton
    • C07C275/10Derivatives 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 acyclic carbon atoms of an acyclic and saturated carbon skeleton being further substituted by singly-bound oxygen atoms
    • 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
    • C07C275/04Derivatives 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 acyclic carbon atoms
    • C07C275/20Derivatives 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 acyclic carbon atoms of an unsaturated carbon skeleton
    • C07C275/24Derivatives 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 acyclic carbon atoms of an unsaturated carbon skeleton containing 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
    • 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
    • C07C311/01Sulfonamides having sulfur atoms of sulfonamide groups bound to acyclic carbon atoms
    • C07C311/02Sulfonamides having sulfur atoms of sulfonamide groups bound to acyclic carbon atoms of an acyclic saturated carbon skeleton
    • C07C311/03Sulfonamides having sulfur atoms of sulfonamide groups bound to acyclic carbon atoms of an acyclic saturated carbon skeleton having the nitrogen atoms of the sulfonamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C311/04Sulfonamides having sulfur atoms of sulfonamide groups bound to acyclic carbon atoms of an acyclic saturated carbon skeleton having the nitrogen atoms of the sulfonamide groups bound to hydrogen atoms or to acyclic carbon atoms to acyclic carbon atoms of hydrocarbon radicals substituted by singly-bound oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/18Heterocyclic 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 one double bond between ring members or between a ring member and a non-ring member
    • C07D207/22Heterocyclic 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 one double bond between ring members or between a ring member and a non-ring member 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
    • C07D207/24Oxygen or sulfur atoms
    • C07D207/262-Pyrrolidones
    • C07D207/2732-Pyrrolidones 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 other ring carbon atoms
    • C07D207/277Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D207/282-Pyrrolidone-5- carboxylic acids; Functional derivatives thereof, e.g. esters, nitriles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/06Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having one or two double bonds between ring members or between ring members and non-ring members
    • C07D241/08Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having one or two double bonds between ring members or between ring members and non-ring members with oxygen atoms directly attached to ring carbon atoms
    • 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
    • C07D295/04Heterocyclic 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 with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/08Heterocyclic 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 with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly bound oxygen or sulfur atoms
    • C07D295/084Heterocyclic 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 with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly bound oxygen or sulfur atoms with the ring nitrogen atoms and the oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings
    • C07D295/092Heterocyclic 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 with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly bound oxygen or sulfur atoms with the ring nitrogen atoms and the oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings with aromatic radicals attached to the chain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06026Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atom, i.e. Gly or Ala
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/02Systems containing only non-condensed rings with a three-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2603/00Systems containing at least three condensed rings
    • C07C2603/02Ortho- or ortho- and peri-condensed systems
    • C07C2603/04Ortho- or ortho- and peri-condensed systems containing three rings
    • C07C2603/06Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members
    • C07C2603/10Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members containing five-membered rings
    • C07C2603/12Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members containing five-membered rings only one five-membered ring
    • C07C2603/18Fluorenes; Hydrogenated fluorenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2603/00Systems containing at least three condensed rings
    • C07C2603/02Ortho- or ortho- and peri-condensed systems
    • C07C2603/04Ortho- or ortho- and peri-condensed systems containing three rings
    • C07C2603/30Ortho- or ortho- and peri-condensed systems containing three rings containing seven-membered rings
    • C07C2603/32Dibenzocycloheptenes; Hydrogenated dibenzocycloheptenes

Definitions

  • the tachykinins are a group of naturally-occurring peptides found widely distributed throughout mammalian tissues, both within the central nervous system and in the peripheral nervous and circulatory systems.
  • the structures of three known mammalian tachykinins are as follows:
  • Substance P is believed inter alia to be involved in the neurotransmission of pain sensations
  • substance P is believed to be involved in the inflammatory response in diseases such as rheumatoid arthritis and
  • osteoarthritis [O'Byrne et al in Arthritis and Rheumatism (1990) 33 1023-8].
  • Other disease areas where tachykinin antagonists are believed to be useful are allergic conditions [Hamelet et al Can. J. Pharmacol. Physiol. (1988) 66 1361-7], immunoregulation [Lotz et al Science
  • Substance P may also play a role in
  • demyelinating diseases such as multiple sclerosis and amyotrophic lateral sclerosis [J. Luber-Narod et. al., poster to be presented at C.I.N.P. XVIIIth Congress, 28th June-2nd July, 1992, in press], and in disorders of bladder function such as bladder detrusor hyper-reflexia (Lancet. 16th May, 1992, 1239).
  • tachykinins have utility in the following disorders:
  • dysthymic disorders chronic obstructive airways disease
  • hypersensitivity disorders such as poison ivy
  • vasospastic diseases such as angina and Reynauld's disease
  • fibrosing and collagen diseases such as scleroderma and eosinophillic fascioliasis
  • reflex sympathetic dystrophy such as shoulder/hand syndrome
  • addiction disorders such as alcoholism
  • stress related somatic disorders neuropathy, neuralgia
  • disorders related to immune enhancement or suppression such as systemic lupus erythmatosis
  • opthalmic disease such as conjuctivitis, vernal conjunctivitis, and the like
  • cutaneous diseases such as contact dermatitis, atropic dermatitis, urticaria, and other eczematoid dermatitis (European patent application no. 0 394 989).
  • peptide derivatives are likely to be of limited utility as therapeutic agents. It is for this reason that non-peptide tachykinin receptor antagonists are sought.
  • this invention provides a class of potent non-peptide tachykinin receptor antagonists.
  • the compounds of the present invention do not suffer from the shortcomings, in terms of metabolic instability, of known peptide-based tachykinin receptor antagonists.
  • the present invention provides a compound of formula (I), or a salt or prodrug thereof:
  • Z represents O, S or NR 8 , where R 8 represents H or C 1-6 alkyl;
  • R 1 and R 2 independently represent H; C 1-6 alkyl optionally substituted by hydroxy, cyano, COR a , COOR a , CONR a R b , COC 1-4 alkylNR a R b , CONR a C 1-4 alkylCONR a R b or
  • R a and R b each independently represent H, C 1-6 alkyl, phenyl (optionally substituted by one or more of C 1-6 alkyl, C 1-6 alkoxy, halo and trifluoromethyl) or phenyl (C 1-4 alkyl) (optionally substituted in the phenyl ring by one or more of C 1-6 alkyl, C 1-6 alkoxy, halo and trifluoromethyl)); phenyl (C 1-4 alkyl), (optionally substituted by one or more of C 1-6 alkyl, C 1-6 alkoxy, halo and trifluoromethyl in the phenyl ring); C 2-6 alkylene; COC 1-6 alkylhalo; COR a ; COOR a ; CONHR a ;
  • R 3 represents H or C 1-6 alkyl
  • R 4 represents H, C 1-6 alkyl or phenyl
  • R 5 represents (CH 2 ) qpheny1, wherein q is 0, 1, 2 or 3 which may optionally be substituted in the phenyl ring by one or more of C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, halo, cyano, nitro, trifluoromethyl,
  • each R 7 and R 8 independently represents C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, halo, cyano, nitro, trifluoromethyl, trimethylsilyl, SR c , SOR c , SO 2 R c , OR c , NR c R d , NR c COR d , NR c COOR d , COOR c or CONR c R d , where R c and R d are as above defined; and
  • n and n independently represent 0, 1, 2 , 3 or
  • suitable alkyl groups include methyl, ethyl, n- or iso-propyl, n-, sec-, iso- or tert-butyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl, and cycloalkyl- alkyl groups such as cyclopropylmethyl;
  • suitable alkenyl groups include vinyl and allyl; and suitable alkynyl groups include propargyl.
  • halo as used herein includes fluoro, chloro, bromo and iodo, especially chloro and fluoro.
  • W 1 is a bond, O, S or a group NR 6 , where R 6 is H or
  • R 1 and R 2 each independently represent H, C 1-6 alkyl, phenyl(C 1-4 alkyl), COR 10 COOR 10 or CONHR 10 , where R 10 is C 1-6 alkyl or phenyl;
  • R 4 represents H, C 1-6 alkyl or phenyl (optionally substituted by one or more of C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, halo, cyano, nitro,
  • R 5 represents (CH 2 ) q phenyl, wherein q is 0, 1, 2 or 3 which may optionally be substituted in the phenyl ring by one or more of C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, halo, cyano, nitro, trifluoromethyl, SCH 3 ,
  • any phenyl (including benzyl) ring in the definitions of R 1 or R 2 (including R 10 ) above may be optionally substituted by one or more of C 1-6 alkyl, C 1-6 alkoxy, halo or trifluoromethyl.
  • Q is 10,11-dihydro-5H-dibenzo[a,d] cyclohepten-5-yl, 5H-dibenzo[a,d]cyclohepten-5-yl or 9-fluorenyl.
  • X and Y each represent H.
  • Z represents oxa.
  • C 1-6 alkyl especially methyl
  • Suitable values for the group R 3 include H and methyl, preferably H.
  • R 4 represents H.
  • R 5 represents (CH 2 ) q phenyl where q is
  • R 5 represents a
  • R 5 represents 3,5-dimethylphenyl or 3,5-bistrifluoromethylphenyl.
  • n and n each represent O.
  • R 1 , R 2 and Z are as defined for formula (I) above;
  • Q 1 represents 10,11-dihydro-5H-dibenzo[a,d] cyclohepten-5-yl, 5H-dibenzo[a,d]cylcohepten-5-yl or 9-fluorenyl.
  • R 20 and R 21 each independently represent H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, halo, cyano, nitro, trifluoromethyl, trimethylsilyl, SR c , SOR c , SO 2 R c , OR c , NR c R d , NR c COR d , NR c COOR d , COOR c or CONR c R d , where R c and R d are as above defined;
  • R 20 and R 21 are other than hydrogen and are located in the 3- and 5-positions. Most preferably R 20 and R 21 each represent methyl or trifluoromethyl.
  • Suitable pharmaceutically acceptable salts of the compounds of this invention include acid addition salts which may, for example, be formed by mixing a solution of the compound according to the invention with a solution of a pharmaceutically
  • Salts of amine groups may also comprise quaternary ammonium salts in which the amino nitrogen atom carries a suitable organic group such as an alkyl, alkenyl, alkynyl or aralkyl moiety.
  • a suitable organic group such as an alkyl, alkenyl, alkynyl or aralkyl moiety.
  • suitable pharmaceutically acceptable salts thereof may include metal salts such as alkali metal salts, e.g. sodium or potassium salts; and alkaline earth metal salts, e.g. calcium or magnesium salts.
  • the present invention includes within its scope prodrugs of the compounds of formula (I) above.
  • prodrugs will be functional derivatives of the compounds of formula (I) which are readily
  • the compounds according to the invention may exist both as enantiomers and as diastereomers. It is to be understood that all such isomers and mixtures thereof are encompassed within the scope of the present
  • compositions comprising one or more compounds of this invention in association with a pharmaceutically
  • compositions are in unit dosage forms such as tablets, pills, capsules, powders, granules, sterile parenteral solutions or suspensions, or suppositories, for oral, parenteral or rectal administration.
  • a pharmaceutical carrier e.g. conventional
  • tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium
  • a solid preformulation composition containing a homogeneous mixture of a compound of the present invention, or a non- toxic pharmaceutically acceptable salt thereof.
  • preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
  • This solid preformulation composition is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 500 mg of the active ingredient of the present invention.
  • the tablets or pills of the novel composition can be coated or otherwise compounded to provide a dosage form affording the
  • the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose
  • compositions of the present invention may be incorporated for administration orally or by injection include aqueous solutions, suitably flavoured syrups, aqueous or oil suspensions, and flavoured emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
  • Suitable dispersing or suspending agents for aqueous suspensions include synthetic and natural gums such as tragacanth, acacia, alginate, dextran, sodium carboxymethylcellulose, methylcellulose, polyvinylpyrrolidone or gelatin.
  • the present invention further provides a process for preparing a pharmaceutical composition comprising a compound of formula (I), which process comprises bringing a compound of formula (I) into
  • the compounds of the present invention are of value in the treatment of a wide variety of clinical conditions which are characterised by the presence of an excess of tachykinin, in particular substance P,
  • disorders of the central nervous system such as anxiety, depression, psychosis and schizophrenia
  • neurodegenerative disorders such as dementia, including senile dementia of the Alzheimer type, Alzheimer's disease and Down's syndrome;
  • demyelinating diseases such as MS and ALS and other neuropathological disorders such as peripheral neuropathological disorders
  • neuropathy for example, diabetic or chemotherapy-induced neuropathy, and postherpetic and other neuralgias;
  • respiratory diseases such as chronic obstrucutive airways disease, bronchopneumonia, bronchospasm and asthma;
  • inflammatory diseases such as inflammatory bowel disease, psoriasis, fibrositis, osteoarthritis and rheumatoid arthritis; allergies such as eczema and rhinitis;
  • hypersensitivity disorders such as poison ivy; ophthalmic diseases such as conjunctivitis, vernal conjunctivitis, and the like; cutaneous diseases such as contact
  • dermatitis atropic dermatitis, urticaria, and other eczematoid dermatitis
  • addiction disorders such as alcoholism
  • stress related somatic disorders such as shoulder/hand syndrome
  • dysthymic disorders adverse immunological reactions such as rejection of transplanted tissues and disorders related to immune enhancement or suppression such as systemic lupus erythematosis
  • gastrointestinal (GI) disorders and diseases of the GI tract such as disorders associated with the neuronal control of viscera such as ulcerative colitis, Crohn's disease and incontinence
  • disorders of bladder function such as bladder detrusor hyper-reflexia
  • fibrosing and collagen diseases such as scleroderma and eosinophilic fascioliasis
  • disorders of blood flow caused by vasodilation and vasospastic diseases such as angina, migraine and Reynaud's disease
  • pain or nociception for example, that attributable to or associated with any of the foregoing conditions
  • neurodegenerative disorders such as senile dementia of the Alzheimer type, Alzheimer's disease and Down's syndrome; respiratory diseases such as bronchospasm and asthma; inflammatory diseases such as inflammatory bowel disease, osteoarthritis and rheumatoid arthritis; adverse immunological reactions such as rejection of transplanted tissues; gastrointestinal (GI) disorders and diseases of the GI tract such as disorders associated with the neuronal control of viscera such as ulcerative colitis, Crohn's disease and incontinence; disorders of blood flow caused by vasodilation; and pain or nociception, for example, that attributable to or associated with any of the foregoing conditions or the transmission of pain in migraine.
  • GI gastrointestinal
  • the compounds of formula (I) are particularly useful in the treatment of pain or nociception and/or inflammation and disorders associated therewith such as, for example, neuropathy, such as diabetic and
  • the present invention further provides a compound of formula (I) for use in the manufacture of a medicament for the treatment of physiological disorders associated with an excess of tachykinins, especially substance P.
  • the present invention also provides a method for the the treatment or prevention of
  • tachykinin reducing amount of a compound or composition of this invention.
  • a suitable dosage level is about 0.001 to 50 mg/kg per day, such as 0.001 to 25 mg/kg per day, preferably about 0.005 to 10 mg/kg per day, and especially about 0.005 to 5 mg/kg per day.
  • the compounds may be administered on a regimen of 1 to 4 times per day, preferably once daily.
  • reaction is conveniently carried out in a suitable organic solvent, such as an ether, for example, tetrahydrofuran.
  • a suitable organic solvent such as an ether, for example, tetrahydrofuran.
  • Suitable bases of use in the reaction include alkali or alkaline earth metal hydrides, for example, sodium hydride.
  • the compounds of formula (I) prepared to the above described process may, if necessary or desired be converted to other compounds of formula (I).
  • reaction is conveniently effected in a suitable organic solvent, such as an ether, for example, diethyl ether or tetrahydrofuran.
  • a suitable organic solvent such as an ether, for example, diethyl ether or tetrahydrofuran.
  • Suitable reducing agents of use in the reaction include borane and metal hydrides, such as lithium aluminium hydride.
  • the reaction is conveniently effected in a suitable organic solvent, such as an ether, for example, tetrahydrofuran.
  • the reaction is conveniently effected by heating a solution of the compound of formula (III) in concentrated hydrochloric acid at reflux.
  • R 40 and R 41 both represent H (IVA), by decarboxylation.
  • the reaction is conveniently effected by heating the compound of formula (IVA) in a concentrated mineral acid, such as concentrated hydrochloric acid, to a temperature of about 90-120oC, such as about 100oC.
  • a concentrated mineral acid such as concentrated hydrochloric acid
  • Lawesson's reagent or phosphorus pentasulphide in a suitable solvent e.g. pyridine
  • a suitable solvent e.g. pyridine
  • Suitable reducing agents include metal hydrides, such as lithium aluminium hydride.
  • reaction is conveniently effected in a suitable organic solvent, such as an ether, for example, tetrahydrofuran, suitably at elevated temperature, such as the reflux temperature of the solvent.
  • a suitable organic solvent such as an ether, for example, tetrahydrofuran
  • R 3 is as defined for formula (I), by reaction with a compound of formula Q-Hal wherein Hal is halo, such as bromo, chloro or iodo, in the presence of a base.
  • Suitable bases of use in the reaction include metal hydroxides, for example, sodium hydroxide.
  • the reaction is conveniently effected in a mixture of water and a suitable organic solvent, such as a hydrocarbon, for example, toluene, in the presence of a phase transfer catalyst, such as benzyltrimethyl ammonium chloride.
  • Compounds of formula (IVA) may be prepared from the corresponding compounds of formula (IV) wherein R 40 and R 41 each represnt alkyl (IVB), by saponification.
  • the saponification is conveniently effected using an alkali metal hydroxide, such as sodium
  • aqueous solvent such as aqueous alcohol, for example, aqueous methanol
  • elevated temperature e.g. the reflux temperature of the chosen solvent.
  • compounds of formula (I) wherein one or both of R 1 and R 2 represent, for example, an acyl or a benzyl group may be converted to compounds of formula (I) wherein one or both of R 1 and R 2 represent H by, for example, hydrolysis or catalytic hydrogenation.
  • Suitable reagents and conditions are decribed in the accompanying examples, or will be readily apparent to one skilled in the art of organic chemistry.
  • novel compounds may be prepared in racemic form, or individual enantiomers may be prepared either by enantiospecific synthesis or by resolution.
  • the novel compounds may, for example, be resolved into their component enantiomers by standard techniques, such as the formation of diastereomeric pairs by salt formation with an optically active acid, such as (-)-di-p-toluoyl-d-tartaric acid and/or (+)-di-p-toluoyl-1-tartaric acid followed by fractional crystallization and regeneration of the free base.
  • the novel compounds may also be resolved by formation of diastereomeric esters or amides, followed by chromatographic separation and removal of the chiral auxiliary.
  • protecting groups may be removed at a convenient
  • Example 1f (0.55g, Example 1f) in 2ml dry CH 2 Cl 2 was treated with trifluoroacetic acid (2ml) and stirred for 20 minutes. Solvent and other volatiles were removed in vacuo . The residue was dissolved in ethyl acetate, washed with aqueous Na 2 CO 3 , dried (MgSO 4 ) and solvent removed in vacuo. The residue was dissolved in ethanol and treated with oxalic acid to give the title compound, mp 175-177°C (ethanol-diethyl ether).
  • Example 3 The product of Example 3 was acetylated in an analogous manner to that described in Example 2 to give the title compound, mp 109-111°C.
  • compositions according to the invention are provided.
  • the resulting granulation is sieved, dried and blended with the remainder of the corn starch and the magnesium stearate. The resulting granulation is then compressed into tablets containing
  • the sodium phosphate, citric acid monohydrate and sodium chloride are dissolved in a portion of the water.
  • the compound of formula (I) is dissolved or suspended in the solution and made up to volume.
  • the white soft paraffin is heated until molten.
  • the liquid paraffin and emulsifying wax are incorporated and stirred until dissolved.
  • the compound of formula (I) is added and stirring continued until dispersed. The mixture is then cooled until solid.
  • the cDNA for the human NK1R was cloned into the expression vector pCDM9 which was derived from pCDM8 (INVITROGEN) by inserting the
  • ampicillin resistance gene (nucleotide 1973 to 2964 from BLUESCRIPT SK+ (trademark, STRATAGENE, La Jolla, CA, USA)) into the Sac II site.
  • Transfection of 20 ug of the plasmid DNA into 10 million COS cells was achieved by electroporation in 800 ⁇ l of transfection buffer (135 mM NaCl, 1.2 mM CaCl 2 , 1.2 mM MgCl 2 , 2.4 mM K 2 HPO 4 , 0.6 mM KH 2 PO 4 , 10 mM glucose, 10 mM N-2-hydroxyethyl-piperazine-N'-2-ethane sulphonic acid (HEPES) pH 7.4) at 260 V and 950 ⁇ F using the IBI GENEZAPPER (trademark IBI, New Haven, CT, USA).
  • the cells were incubated in 10% fetal calf serum, 2 mM glutamine, 100U/ml penicillin-streptomycin, and 90% DMEM media (GIBCO, Grand Island, NY, USA) in 5% CO 2 at 37oC for three days before the binding assay.
  • the cDNA was subcloned into the vector pRcCMV (INVITROGEN).
  • Transfection of 20 ug of the plasmid DNA into CHO cells was achieved by electroporation in 800 ⁇ l of transfection buffer supplemented with 0.625 mg/ml Herring sperm DNA at 300 V and 950 ⁇ F using the IBI
  • GENEZAPPER (IBI). The transfected cells were incubated in CHO media [10% fetal calf serum, 100 U/ml penicillin-streptomycin, 2 mM glutamine, 1/500 hypoxanthine-thymidine (ATCC), 90% IMDM media (JRH BIOSCIENCES,
  • the binding assay of human NKIR expressed in either COS or CHO cells is based on the use of 125 I-substance P ( 125 I-SP, from DU PONT, Boston, MA) as a radioactively labeled ligand which competes with unlabeled substance P or any other ligand for binding to the human NKIR.
  • 125 I-substance P 125 I-SP, from DU PONT, Boston, MA
  • Monolayer cell cultures of COS or CHO were dissociated by the non-enzymatic solution (SPECIALTY MEDIA, Lavellette, NJ) and resuspended in appropriate volume of the binding buffer (50 mM Tris pH 7.5, 5 mM MnCl 2 , 150 mM NaCl, 0.04 mg/ml bacitracin, 0.004 mg/ml leupeptin, 0.2 mg/ml BSA, 0.01 mM phosphoramidon) such that 200 ⁇ l of the cell suspension would give rise to about 10,000 cpm of specific 125 I-SP binding (approximately 50,000 to 200,000 cells).
  • the binding buffer 50 mM Tris pH 7.5, 5 mM MnCl 2 , 150 mM NaCl, 0.04 mg/ml bacitracin, 0.004 mg/ml leupeptin, 0.2 mg/ml BSA, 0.01 mM phosphoramidon
  • the binding assay 200 ul of cells were added to a tube containing 20 ul of 1.5 to 2.5 nM of 125 I-SP and 20 ⁇ l of unlabeled substance P or any other test compound. The tubes were incubated at 4oC or at room temperature for 1 hour with gentle shaking. The bound radioactivity was separated from unbound radioactivity by GF/C filter (BRANDEL, Gaithersburg, MD) which was prewetted with 0.1% polyethylenimine. The filter was washed with 3 ml of wash buffer (50 mM Tris pH 7.5, 5 mM MnCl 2 , 150 mM NaCl) three times and its radioactivity was determined by gamma counter.
  • wash buffer 50 mM Tris pH 7.5, 5 mM MnCl 2 , 150 mM NaCl
  • the activation of phospholiphase C by NK1R may also be measured in CHO cells expressing the human NK1R by determining the accumulation of inositol monophosphate which is a degradation product of IP 3 .
  • CHO cells are seeded in 12-well plate at 250,000 cells per well. After incubating in CHO media for 4 days, cells are loaded with 5 ⁇ Ci of 3 H-myoinositol in 1 ml of media per well by overnight incubation. The extracellular radioactivity is removed by washing with phosphate buffered saline. LiCl is added to the well at final concentration of 10 mM with or without the test compound, and incubation is continued at 37oC for 15 min.
  • Substance P is added to the well at final concentration of 0.3nM to activate the human NK1R. After 30 min of incubation at 37oC, the medium is removed and 0.1 N HCl is added. Each well is sonicated at 4oC and extracted with CHCl 3 /methanol (1:1). The aqueous phase is applied to a 1 ml Dowex AG 1X8 ion exchange column. The column is washed with 0.1 N formic acid followed by 0.025 M ammonium formate-0.1 N formic acid. The inositol monophosphate is eluted with 0.2 M ammonium formate-0.1 N formic acid and quantitated by beta counter.

Abstract

Compounds of formula (I), and salts and prodrugs thereof, wherein Q represents a group (a) where W represents a bond, O, S, -CH2CH2-, -CH=CH- or NR6; and one or both of the phenyl rings may be replaced by a heteroaryl moiety; X and Y each represent H or X and Y together are =O; Z represents O, S or NR?8; R1 and R2¿ independently represent H; optionally substituted C¿1-6? alkyl, optionally substituted phenyl(C0-4alkyl), C2-6 alkylene; COC1-6alkylhalo; COR?a; COORa; CONHRa¿; COC¿1-4?alkylNR?aRb¿; or CONRaC1-4alkylCONRaRb; R3 represents H or C¿1-6?alkyl; R?4¿ represents H, C¿1-6? alkyl or phenyl; R?5¿ represents optionally substituted (CH¿2?)qphenyl; are tachykinin antagonists, useful for treating pain.

Description

FUSED TRICYCLIC COMPOUNDS, PHARMACEUTICAL COMPOSITIONS CONTAINING THEM MID THEIR USE IN THERAPY This invention relates to a class of tricyclic compounds, which are useful as tachykinin receptor antagonists.
The tachykinins are a group of naturally-occurring peptides found widely distributed throughout mammalian tissues, both within the central nervous system and in the peripheral nervous and circulatory systems. The structures of three known mammalian tachykinins are as follows:
Substance P:
Arg-Pro-Lys-Pro-Gln-Gln-Phe-Phe-Gly-Leu-Met-NH2
Neurokinin A:
His-Lys-Thr-Asp-Ser-Phe-Val-Gly-Leu-Met-NH2
Neurokinin B:
Asp-Met-His-Asp-Phe-Phe-Val-Gly-Leu-Met-NH2
Substance P is believed inter alia to be involved in the neurotransmission of pain sensations
[Otsuka et al, "Role of Substance P as a Sensory
Transmitter in Spinal Cord and Sympathetic Ganglia" in 1982 Substance P in the Nervous System, Ciba Foundation Symposium 91, 13-34 (published by Pitman) and Otsuka and Yanagisawa, "Does Substance P Act as a Pain Transmitter?" TIPS (Dec. 1987) 8 506-510], specifically in the
transmission of pain in migraine (B.E.B. Sandberg et al, J. Med Chem, (1982) 25 1009) and in arthritis [Levine et al, in Science (1984) 226 547-549]. These peptides have also been implicated in gastrointestinal (GI) disorders and diseases of the GI tract such as inflammatory bowel disease [Mantyh et al in Neuroscience (1988) 25 (3) 817- 37 and D. Regoli in "Trends in Cluster Headache" Ed. Sicuteri et al Elsevier Scientific Publishers, Amsterdam
(1987) page 85)]. It is also hypothesised that there is a neurogenic mechanism for arthritis in which substance P may play a role [Kidd et al "A Neurogenic Mechanism for Symmetrical Arthritis" in The Lancet, 11 November 1989 and Grönblad et al "Neuropeptides in Synovium of Patients with Rheumatoid Arthritis and Osteoarthritis" in J.
Rheumatol. (1988) 15(12) 1807-10]. Therefore, substance P is believed to be involved in the inflammatory response in diseases such as rheumatoid arthritis and
osteoarthritis [O'Byrne et al in Arthritis and Rheumatism (1990) 33 1023-8]. Other disease areas where tachykinin antagonists are believed to be useful are allergic conditions [Hamelet et al Can. J. Pharmacol. Physiol. (1988) 66 1361-7], immunoregulation [Lotz et al Science
(1988) 241 1218-21 and Kimball et al, J. Immunol. (1988) 141 (10) 3564-9], vasodilation, bronchospasm, reflex or neuronal control of the viscera [Mantyh et al, PNAS
(1988) 85 3235-9] and, possibly by arresting or slowing β-amyloid-mediated neurodegenerative changes [Yankner et al, Science (1990) 250, 279-82], in senile dementia of the Alzheimer type, Alzheimer's disease and Down's
Syndrome. Substance P may also play a role in
demyelinating diseases such as multiple sclerosis and amyotrophic lateral sclerosis [J. Luber-Narod et. al., poster to be presented at C.I.N.P. XVIIIth Congress, 28th June-2nd July, 1992, in press], and in disorders of bladder function such as bladder detrusor hyper-reflexia (Lancet. 16th May, 1992, 1239).
It has furthermore been suggested that tachykinins have utility in the following disorders:
depression, dysthymic disorders, chronic obstructive airways disease, hypersensitivity disorders such as poison ivy, vasospastic diseases such as angina and Reynauld's disease, fibrosing and collagen diseases such as scleroderma and eosinophillic fascioliasis, reflex sympathetic dystrophy such as shoulder/hand syndrome, addiction disorders such as alcoholism, stress related somatic disorders, neuropathy, neuralgia, disorders related to immune enhancement or suppression such as systemic lupus erythmatosis (European patent application no. 0 436 334), opthalmic disease such as conjuctivitis, vernal conjunctivitis, and the like, and cutaneous diseases such as contact dermatitis, atropic dermatitis, urticaria, and other eczematoid dermatitis (European patent application no. 0 394 989).
In view of their metabolic instability, peptide derivatives are likely to be of limited utility as therapeutic agents. It is for this reason that non-peptide tachykinin receptor antagonists are sought.
In essence, this invention provides a class of potent non-peptide tachykinin receptor antagonists. By virtue of their non-peptide nature, the compounds of the present invention do not suffer from the shortcomings, in terms of metabolic instability, of known peptide-based tachykinin receptor antagonists.
The present invention provides a compound of formula (I), or a salt or prodrug thereof:
Figure imgf000005_0001
wherein Q represents a group
Figure imgf000006_0001
where W represents a bond, O, S, -CH2CH2-, -CH=CH- or a group NR6, where R6 is H or C1-6alkyl and one or both of the phenyl rings may be replaced by a heteroaryl moiety;
X and Y each represent H or X and Y together form a group =O;
Z represents O, S or NR8, where R8 represents H or C1-6alkyl;
R1 and R2 independently represent H; C1-6 alkyl optionally substituted by hydroxy, cyano, CORa, COORa, CONRaRb, COC1-4alkylNRaRb, CONRaC1-4alkylCONRaRb or
NRaRb, (where Ra and Rb each independently represent H, C1-6 alkyl, phenyl (optionally substituted by one or more of C1-6alkyl, C1-6alkoxy, halo and trifluoromethyl) or phenyl (C1-4alkyl) (optionally substituted in the phenyl ring by one or more of C1-6 alkyl, C1-6 alkoxy, halo and trifluoromethyl)); phenyl (C1-4 alkyl), (optionally substituted by one or more of C1-6 alkyl, C1-6 alkoxy, halo and trifluoromethyl in the phenyl ring); C2-6 alkylene; COC1-6alkylhalo; CORa; COORa; CONHRa;
COC1-4alkylNRaRb; or CONRaC1_4alkylCONRaRb; (where Ra and Rb are as previously defined) or R1 and R2 together form a chain (CH2)p where p is 4 or 5 and where one non-terminal methylene group may optionally be replaced by an oxygen atom or a group NRx, where Rx is H or C1-6 alkyl;
R3 represents H or C1-6alkyl; R4 represents H, C1-6 alkyl or phenyl
(optionally substituted by one or more of C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, halo, cyano, nitro,
trifluoromethyl, trimethylsilyl, SRc, SORc, SO2Rc, ORc, NRcRd, NRcCORd, NRcCOORd, COORc or CONRcRd, where Rc and Rd each independently represent H, C1-6 alkyl, phenyl or trifluoromethyl);
R5 represents (CH2) qpheny1, wherein q is 0, 1, 2 or 3 which may optionally be substituted in the phenyl ring by one or more of C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, halo, cyano, nitro, trifluoromethyl,
trimethylsilyl, SRc, SORc, SO2Rc, ORc, NRcRd, NRcCORd, NRcCOORd, COORc or CONRcRd, where Rc and Rd are as above defined;
each R7 and R8 independently represents C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, halo, cyano, nitro, trifluoromethyl, trimethylsilyl, SRc, SORc, SO2Rc, ORc, NRcRd, NRcCORd, NRcCOORd, COORc or CONRcRd, where Rc and Rd are as above defined; and
m and n independently represent 0, 1, 2 , 3 or
4.
The alkyl, alkenyl and alkynyl groups referred to with respect to any of the above formulae may
represent straight, branched or cyclic groups. Thus, for example, suitable alkyl groups include methyl, ethyl, n- or iso-propyl, n-, sec-, iso- or tert-butyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl, and cycloalkyl- alkyl groups such as cyclopropylmethyl; suitable alkenyl groups include vinyl and allyl; and suitable alkynyl groups include propargyl.
The term "halo" as used herein includes fluoro, chloro, bromo and iodo, especially chloro and fluoro. When one or both of the phenyl rings of Q is/are replaced by a heteroaryl moiety, this will
suitably be a pyridyl moiety.
A particular subgroup of compounds according to formula (I) is represented by compounds wherein Q
represents a group
Figure imgf000008_0001
where W1 is a bond, O, S or a group NR6, where R6 is H or
C1-6 alkyl;
R1 and R2 each independently represent H, C1-6 alkyl, phenyl(C1-4 alkyl), COR10 COOR10 or CONHR10, where R10 is C1-6 alkyl or phenyl;
R4 represents H, C1-6 alkyl or phenyl (optionally substituted by one or more of C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, halo, cyano, nitro,
trifluoromethyl, SCH3, SOCH3, SO2CH3, ORc, NRcRd,
NRcCORd, NRcCOORd, COORc or CONRcRd (where Rc and Rd are as above defined); and
R5 represents (CH2)qphenyl, wherein q is 0, 1, 2 or 3 which may optionally be substituted in the phenyl ring by one or more of C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, halo, cyano, nitro, trifluoromethyl, SCH3,
SOCH3, SO2CH3, ORc, NRcRd, NRcCORd, NRcCOORd, COORc or CONRcRd (where Rc and Rd are as above defined);
wherein any phenyl (including benzyl) ring in the definitions of R1 or R2 (including R10) above may be optionally substituted by one or more of C1-6 alkyl, C1-6 alkoxy, halo or trifluoromethyl. In the compounds of the invention, W suitably represents a bond- CH2CH2 or HC=CH.
Preferably Q is 10,11-dihydro-5H-dibenzo[a,d] cyclohepten-5-yl, 5H-dibenzo[a,d]cyclohepten-5-yl or 9-fluorenyl.
Preferably X and Y each represent H.
Preferably Z represents oxa.
Suitable values for the groups R1 and R
include H, C1-6 alkyl, especially methyl; C1-6alkyl substituted by, for example, cyano, hydroxy, NH2,
CO2C1-6alkyl and CONH2.
Suitable values for the group R3 include H and methyl, preferably H.
Preferably R4 represents H.
Suitably R5 represents (CH2)qphenyl where q is
0, 1 or 2 and the phenyl is substituted. Suitable phenyl substituents include methyl, methoxy, nitro, cyano, halo and trifluoromethyl. Preferably R5 represents a
substituted phenyl group. More preferably R5 represents 3,5-dimethylphenyl or 3,5-bistrifluoromethylphenyl.
Preferably m and n each represent O.
A preferred sub-group of compounds according to the invention is represented by formula (Ia)
Figure imgf000009_0001
wherein R1, R2 and Z are as defined for formula (I) above; Q1 represents 10,11-dihydro-5H-dibenzo[a,d] cyclohepten-5-yl, 5H-dibenzo[a,d]cylcohepten-5-yl or 9-fluorenyl.
R20 and R21 each independently represent H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, halo, cyano, nitro, trifluoromethyl, trimethylsilyl, SRc, SORc, SO2Rc, ORc, NRcRd, NRcCORd, NRcCOORd, COORc or CONRcRd, where Rc and Rd are as above defined;
and salts and prodrugs thereof.
Particularly preferred are compounds of formula
(la) wherein R20 and R21 are other than hydrogen and are located in the 3- and 5-positions. Most preferably R20 and R21 each represent methyl or trifluoromethyl.
For use in medicine, the salts of the compounds of formula I will be non-toxic pharmaceutically
acceptable salts. Other salts may, however, be useful in the preparation of the compounds according to the
invention or of their non-toxic pharmaceutically
acceptable salts. Suitable pharmaceutically acceptable salts of the compounds of this invention include acid addition salts which may, for example, be formed by mixing a solution of the compound according to the invention with a solution of a pharmaceutically
acceptable acid such as hydrochloric acid, oxalic acid, fumaric acid, p-toluenesulphonic acid, maleic acid, succinic acid, acetic acid, citric acid, tartaric acid, carbonic acid or phosphoric acid. Salts of amine groups may also comprise quaternary ammonium salts in which the amino nitrogen atom carries a suitable organic group such as an alkyl, alkenyl, alkynyl or aralkyl moiety. Thus, for example, when both R1 and R2 are other than hydrogen, the nitrogen atom to which they are attached may be further substituted to give a quaternary ammonium salt. Furthermore, where the compounds of the invention carry an acidic moiety, suitable pharmaceutically acceptable salts thereof may include metal salts such as alkali metal salts, e.g. sodium or potassium salts; and alkaline earth metal salts, e.g. calcium or magnesium salts.
The present invention includes within its scope prodrugs of the compounds of formula (I) above. In general, such prodrugs will be functional derivatives of the compounds of formula (I) which are readily
convertible in vivo into the required compound of formula (I). Conventional procedures for the selection and preparation of suitable prodrug derivatives are
described, for example, in "Design of Prodrugs", ed. H. Bundgaard, Elsevier, 1985.
The compounds according to the invention may exist both as enantiomers and as diastereomers. It is to be understood that all such isomers and mixtures thereof are encompassed within the scope of the present
invention.
The invention also provides pharmaceutical compositions comprising one or more compounds of this invention in association with a pharmaceutically
acceptable carrier. Preferably these compositions are in unit dosage forms such as tablets, pills, capsules, powders, granules, sterile parenteral solutions or suspensions, or suppositories, for oral, parenteral or rectal administration. For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical carrier, e.g. conventional
tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium
stearate, dicalcium phosphate or gums, and other
pharmaceutical diluents, e.g. water, to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention, or a non- toxic pharmaceutically acceptable salt thereof. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid preformulation composition is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 500 mg of the active ingredient of the present invention. The tablets or pills of the novel composition can be coated or otherwise compounded to provide a dosage form affording the
advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose
acetate.
The liquid forms in which the novel
compositions of the present invention may be incorporated for administration orally or by injection include aqueous solutions, suitably flavoured syrups, aqueous or oil suspensions, and flavoured emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil or peanut oil, as well as elixirs and similar pharmaceutical vehicles. Suitable dispersing or suspending agents for aqueous suspensions include synthetic and natural gums such as tragacanth, acacia, alginate, dextran, sodium carboxymethylcellulose, methylcellulose, polyvinylpyrrolidone or gelatin.
The present invention further provides a process for preparing a pharmaceutical composition comprising a compound of formula (I), which process comprises bringing a compound of formula (I) into
association with a pharmaceutically acceptable carrier or excipient.
The compounds of the present invention are of value in the treatment of a wide variety of clinical conditions which are characterised by the presence of an excess of tachykinin, in particular substance P,
activity. These may include disorders of the central nervous system such as anxiety, depression, psychosis and schizophrenia; neurodegenerative disorders such as dementia, including senile dementia of the Alzheimer type, Alzheimer's disease and Down's syndrome;
demyelinating diseases such as MS and ALS and other neuropathological disorders such as peripheral
neuropathy, for example, diabetic or chemotherapy-induced neuropathy, and postherpetic and other neuralgias;
respiratory diseases such as chronic obstrucutive airways disease, bronchopneumonia, bronchospasm and asthma;
inflammatory diseases such as inflammatory bowel disease, psoriasis, fibrositis, osteoarthritis and rheumatoid arthritis; allergies such as eczema and rhinitis;
hypersensitivity disorders such as poison ivy; ophthalmic diseases such as conjunctivitis, vernal conjunctivitis, and the like; cutaneous diseases such as contact
dermatitis, atropic dermatitis, urticaria, and other eczematoid dermatitis; addiction disorders such as alcoholism; stress related somatic disorders; reflex sympathetic dystrophy such as shoulder/hand syndrome; dysthymic disorders; adverse immunological reactions such as rejection of transplanted tissues and disorders related to immune enhancement or suppression such as systemic lupus erythematosis; gastrointestinal (GI) disorders and diseases of the GI tract such as disorders associated with the neuronal control of viscera such as ulcerative colitis, Crohn's disease and incontinence; disorders of bladder function such as bladder detrusor hyper-reflexia; fibrosing and collagen diseases such as scleroderma and eosinophilic fascioliasis; disorders of blood flow caused by vasodilation and vasospastic diseases such as angina, migraine and Reynaud's disease; and pain or nociception, for example, that attributable to or associated with any of the foregoing conditions, especially the transmission of pain in migraine. For example, the compounds of formula (I) may suitably be used in the treatment of disorders of the central nervous system such as anxiety, psychosis and schizophrenia;
neurodegenerative disorders such as senile dementia of the Alzheimer type, Alzheimer's disease and Down's syndrome; respiratory diseases such as bronchospasm and asthma; inflammatory diseases such as inflammatory bowel disease, osteoarthritis and rheumatoid arthritis; adverse immunological reactions such as rejection of transplanted tissues; gastrointestinal (GI) disorders and diseases of the GI tract such as disorders associated with the neuronal control of viscera such as ulcerative colitis, Crohn's disease and incontinence; disorders of blood flow caused by vasodilation; and pain or nociception, for example, that attributable to or associated with any of the foregoing conditions or the transmission of pain in migraine.
The compounds of formula (I) are particularly useful in the treatment of pain or nociception and/or inflammation and disorders associated therewith such as, for example, neuropathy, such as diabetic and
chemotherapy-induced neuropathy, postherpetic and other neuralgias, asthma, osteroarthritis, rheumatoid arthritis and migraine.
The present invention further provides a compound of formula (I) for use in the manufacture of a medicament for the treatment of physiological disorders associated with an excess of tachykinins, especially substance P. The present invention also provides a method for the the treatment or prevention of
physiological disorders associated with an excess of tachykinins, especially substance P, which method
comprises administration to a patient in need thereof of a tachykinin reducing amount of a compound or composition of this invention.
In the treatment of conditions involving actions of tachykinins released physiologically in response to noxious or other stimuli, a suitable dosage level is about 0.001 to 50 mg/kg per day, such as 0.001 to 25 mg/kg per day, preferably about 0.005 to 10 mg/kg per day, and especially about 0.005 to 5 mg/kg per day. The compounds may be administered on a regimen of 1 to 4 times per day, preferably once daily.
The compounds according to the invention wherein Z is O or S may be prepared by reaction of a compound of formula (II)
Figure imgf000016_0001
wherein Q, R1, R2, R3 , X and Y, are defined as for formula (I) and Z is O or S, with a compound of formula HalCHR4R5, where R4 and R5 are as defined for formula (I) and Hal is halo, such as bromo, chloro or iodo, in the presence of a base.
The reaction is conveniently carried out in a suitable organic solvent, such as an ether, for example, tetrahydrofuran.
Suitable bases of use in the reaction include alkali or alkaline earth metal hydrides, for example, sodium hydride.
The compounds of formula (I) prepared to the above described process may, if necessary or desired be converted to other compounds of formula (I). Thus, for eaxmple, the compounds of the invention wherein Z is a group NR8 and X and Y together represent =O may be prepared from the compounds of formula (II) wherein Z is O and X and Y together represent =0 by reaction with a compound of formula HNR8CHR4R5 in the presence of a coupling agent, such as dicyclohexylcarbodiimide.
The reaction is conveniently effected in a suitable organic solvent, such as an ether, for example, diethyl ether or tetrahydrofuran.
The compounds according to the invention wherein Z is NR8 and X and Y are hydrogen may be prepared from the corresponding compounds of formula (I) wherein X and Y together represent =0, by reduction.
Suitable reducing agents of use in the reaction include borane and metal hydrides, such as lithium aluminium hydride. The reaction is conveniently effected in a suitable organic solvent, such as an ether, for example, tetrahydrofuran.
Compounds of formula (II) wherein Z is O and X and Y together represent a group =O may be prepared, for example, from intermediates of formula (III)
Figure imgf000017_0001
wherein Q and R3 are as above defined and Ph represents phenyl, by hydrolysis.
The reaction is conveniently effected by heating a solution of the compound of formula (III) in concentrated hydrochloric acid at reflux.
Alternatively, compounds of formula (II) wherein Z is O, X and Y are =O and R3 is H may be prepared from intermediates of formula (IVA)
Figure imgf000018_0001
wherein Q, R1 and R2 are as above defined, and R40 and R41 both represent H (IVA), by decarboxylation.
The reaction is conveniently effected by heating the compound of formula (IVA) in a concentrated mineral acid, such as concentrated hydrochloric acid, to a temperature of about 90-120ºC, such as about 100ºC.
Compounds of formula (II) wherein Z is O and X and Y are =O may also be prepared by conventional
procedures for the preparation of amino acids which are well documented and are described, for example, in
Chemistry and Biochemistry of the Amino Acids, ed. G. C. Barrett, Chapman and Hall, 1985.
Compounds of formula (II) wherein Z is S may be prepared from the corresponding compounds of formula (II) wherein Z is 0 by treating the latter compound with
Lawesson's reagent or phosphorus pentasulphide in a suitable solvent, e.g. pyridine, at ambient or elevated temperature, suitably at the reflux temperature of the chosen solvent.
Compounds of formula (II) wherein X and Y represent H may be prepared from the corresponding compounds of formula (II) wherein X and Y together represent =O, by reduction.
Suitable reducing agents include metal hydrides, such as lithium aluminium hydride. The
reaction is conveniently effected in a suitable organic solvent, such as an ether, for example, tetrahydrofuran, suitably at elevated temperature, such as the reflux temperature of the solvent.
Intermediates of formula (III) may be prepared from compounds of formula (V)
Figure imgf000019_0001
wherein R3 is as defined for formula (I), by reaction with a compound of formula Q-Hal wherein Hal is halo, such as bromo, chloro or iodo, in the presence of a base.
Suitable bases of use in the reaction include metal hydroxides, for example, sodium hydroxide. The reaction is conveniently effected in a mixture of water and a suitable organic solvent, such as a hydrocarbon, for example, toluene, in the presence of a phase transfer catalyst, such as benzyltrimethyl ammonium chloride.
Compounds of formula (V) are commercially available or may be prepared by procedures readily apparent to one skilled in the art.
Compounds of formula (IVA) may be prepared from the corresponding compounds of formula (IV) wherein R40 and R41 each represnt alkyl (IVB), by saponification.
The saponification is conveniently effected using an alkali metal hydroxide, such as sodium
hydroxide, in an aqueous solvent, such as aqueous alcohol, for example, aqueous methanol, preferably at elevated temperature, e.g. the reflux temperature of the chosen solvent. Compounds of formula (IVB) may be prepared by reaction of a compound of formula Q-Hal as above defined with a malonate derivative of formula
R41O2CCH(NR1R2)CO2R40, which malonate derivatives are commercially available or may be prepared from
commercially avaible compounds by conventional means known to those skilled in the art.
Compounds of formula Q-Hal are commercially available or may be prepared by conventional procedures known to those skilled in the art.
Compounds of formula (I) may also be prepared from other compounds of formula (I). Thus, for example, compounds of formula (I) wherein one or both of R1 and R2 represent hydrogen may be reacted with an optionally substituted alkylating or an acylating agent to produce compounds of formula (I) wherein one or both of R1 and R2 represent an optionally substituted alkyl or an acyl group. Suitable procedures are described in the
accompanying examples, or will be readily apparent to one skilled in the art.
Conversely, compounds of formula (I) wherein one or both of R1 and R2 represent, for example, an acyl or a benzyl group, may be converted to compounds of formula (I) wherein one or both of R1 and R2 represent H by, for example, hydrolysis or catalytic hydrogenation. Suitable reagents and conditions are decribed in the accompanying examples, or will be readily apparent to one skilled in the art of organic chemistry.
Intermediates of formula (II) are novel compounds. Intermediates of formula (II) and the
preparation thereof represent further aspects of the present invention.
Where the above-described process for the preparation of the compounds according to the invention gives rise to mixtures of stereoisomers these isomers may, if desired, be separated, suitably by conventional techniques such as preparative chromatography.
The novel compounds may be prepared in racemic form, or individual enantiomers may be prepared either by enantiospecific synthesis or by resolution. The novel compounds may, for example, be resolved into their component enantiomers by standard techniques, such as the formation of diastereomeric pairs by salt formation with an optically active acid, such as (-)-di-p-toluoyl-d-tartaric acid and/or (+)-di-p-toluoyl-1-tartaric acid followed by fractional crystallization and regeneration of the free base. The novel compounds may also be resolved by formation of diastereomeric esters or amides, followed by chromatographic separation and removal of the chiral auxiliary.
During any of the above synthetic sequences it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups, such as those described in Protective Groups in Organic Chemistry, ed. J.F.W. McOmie, Plenum Press, 1973; and T.W. Greene and P.G.M. Wutts, Protective Groups in Organic Synthesis. John Wiley & Sons, 1991. The
protecting groups may be removed at a convenient
subsequent stage using methods known from the art.
The following Examples illustrate the preparation of compounds according to the invention. EXAMPLE 1: 1-(5H-Dibenzo[a,d]cyclohepten-5-yl)-2-(3,5- dimethylbenzyloxy)ethylamine, oxalate salt
a) Through a cooled (-15°C) solution of dibenzosuberenol (20g) and calcium chloride (20g) in toluene (750ml) was bubbled hydrogen chloride gas for 15 minutes. The solution was warmed to room temperature over 1 hour and hydrogen chloride gas bubbled through for a further 20 minutes. After 16 hours the solution was filtered and the filtrate evaporated in υacuo to leave 5-chloro-5H-dibenzo[a,d]cycloheptene as a yellow crystalline solid. 1H NMR (360MHz, CDCl3) δ 6.25 (1H, bs),
7.14 (2H, s), 7.39-7.45 (8H, m).
b) To a stirred suspension of diethyl acetamidomalonate (21.2g) in tetrahydrofuran (150ml) was slowly added sodium hydride (3.4g, 80% dispersion in oil) under an atmosphere of nitrogen. After lb to this solution was added a solution of 5-chloro-5H-dibenzo[a,d]cycloheptene (20.3g, Example 1a) in tetrahydrofuran (100ml). The solution was heated to reflux for 18h, cooled to room temperature and to this was added a saturated solution of NH4Cl and ethyl acetate. The organic phase was dried (MgSO4), concentrated in vacuo and the residue chromatographed on silica (eluting with ethyl acetate:petroleum ether bp 60-80 ° C ( 1 :4 ) . This gave 2-diethyl( 5H- dibenzo[a,d]cyclohepten-5-yl)-2-acetamidomalonate as a yellow solid. 1H NMR (360MHz, CDCl3) δ 1.02-1.04 (6H, m), 1.6 (3H, s), 3.82-3.91 (2H, m), 4.0-4.13 (2H, m), 5.78 (1H, s), 6.39 (1H, s),
6.79 (2H, s), 7.21-7.36 (6H, m), 7.54-7.56 (2H, m). c) A mixture of diethyl 2-(5H-dibenzo[a,d]cyclohepten-5-yl)-2-acetamidomalonate (20.2g, Example 1b) and sodium hydroxide (4g) in methanol (75ml) and water (75ml) was heated to reflux for 8h. To the cooled solution was added 1M-HCl until pH = 6. The solution was concentrated in vacuo and acidified to pH = 2 by addition of 1M-HCl. The solid which formed was collected by filtration and heated together with 6M-HCl for 3h at 100°C. The cooled solution was filtered and the residue dissolved in hot water, refiltered and to the filtrate was added aqueous ammonia until pH 7-8. The solid which formed on cooling to room temperature was collected by filtration and dried in vacuo to give 5H-dibenzo[a,d]cyclohepten-5-ylglycine. 1H NMR (360MHz, d4 MeOH), δ 4.08-4.12 (1H, m), 4.35-4.39 (1H, m), 7.0-7.11 (2H, m), 7.24-7.30 (3H, m), 7.34-7.45 (5H, m).
d) 5-H-Dibenzo[a,d]cyclohepten-5-ylglycine (4.8g, Example
1c) was added in portions to a solution of lithium aluminium hydride (54ml of 1.0M solution) in 30ml dry THF at 0°C. The reaction was allowed to stand at room temperature overnight, quenched with 2N sodium hydroxide and poured through Celite. The filtrate was washed with water, dried over MgSO4 and solvent removed in vacuo to leave a residue which was induced to crystallised in ethyl acetate or dichloromethane to give 5H-dibenzo[a,d]cycloheptene glycinol. 1H NMR (360MHz, d4 MeOH) δ 3.05-3.17 (2H, m), 3.29-3.33 (1H, m), 3.89-3.94 (1H, m), 6.90-6.97 (2H, m), 7.23-7.38 (8H, m). e) A mixture of 5H-dibenzo[a,d]cycloheptene glycinol (1.13g, Example 1d) and di-tert-butyldicarbonate (1.0g) in 10ml CH2Cl2 was stirred at room temperature for 1.5h. Solvent was removed in vacuo to give N-tert-butoxycarbonyl (5H-dibenzo[a,d]cycloheptene)glycinol which could be induced to crystallise in diethyl ether.1H NMR (360MHz, CDCl3) δ 1.24 (9H, s), 2.43 (1H, brs), 3.1-3.3 (1H, m), 3.3-3.5 (1H, m), 4.1-4.2 (1H, m), 4.2-4.4 (1H, m), 4.4-4.6 (1H, m), 6.9-7.1 (2H, m), 7.2-7.4 (8H, m).
f) To a solution of N-tert-butoxycarbonyl (5H-dibenzo[a,d]cycloheptene) glycinol (1.49g, Example le) and 3,5-dimethylbenzyl bromide (0.92g) in 4ml DMF-THF (1:1) under nitrogen at 0°C was added sodium hydride (130mg, 80% dispersion). The reaction was stirred at room temperature for 2h then partitioned between aqueous NH4Cl-ethyl acetate. The organic phase was dried (MgSO4) and solvent removed in vacuo. The residue was chromatographed on silica (5% ethyl acetate-petroleum ether eluant) to give N-tert-butoxycarbonyl(5H-dibenzo[a,d]cycloheptene)glycinol-3,5-dimethylbenzyl ether, mp 116-118°C (petroleum ether).1H NMR (360MHz, CDCl3) 1.15
(9H, s), 2.36 (6H, s), 2.90-2.93 (1H, m), 3.18-3.21 (1H, m), 4.16-4.37 (4H, m), 4.79-4.82 (1H, m), 6.88-6.94 (4H, m), 7.05-7.08 (1H, m), 7.21-7.32 (8H, m).
g) A solution of N-tert-butoxycarbonyl (5H-dibenzo[a,d]cycloheρtene)glycinol-3,5-dimethylbenzyl ether
(0.55g, Example 1f) in 2ml dry CH2Cl2 was treated with trifluoroacetic acid (2ml) and stirred for 20 minutes. Solvent and other volatiles were removed in vacuo . The residue was dissolved in ethyl acetate, washed with aqueous Na2CO3, dried (MgSO4) and solvent removed in vacuo. The residue was dissolved in ethanol and treated with oxalic acid to give the title compound, mp 175-177°C (ethanol-diethyl ether). 1H NMR (360MHz, DMSO-d6) 2.27 (6H, s), 2.85-2.87 (1H, m), 3.17-3.19 (1H, m), 3.58-3.40 (1H, m), 4.15-4.40 (3H, m), 6.88 (2H, s), 6.92 (1H, s), 6.99-7.08 (2H, m), 7.28-7.48 (8H, m).
EXAMPLE 2: N-Acetamido-1-(5H-dibenzo[a,d]cyclohepten-5-yl)-2-(3,5-dimethylbenzyloxy)ethylamine
A solution of the product of Example 1 (0.35g) and acetic anhydride (0.178ml) in 3ml pyridine was allowed to stand overnight. The reaction was partitioned between ethyl acetate and 1N HCl. The organic phase was dried (MgSO4) and solvent removed in vacuo . The residue was chromatographed (20% ethyl acetate-petroleum ether) to give the title compound, mp 115-117°C (ethyl acetate-petroleum ether). 1H NMR (360MHz, CDCl3) 1.54 (6H, s), 2.34 (3H, s), 2.92-2.94 (1H, s), 3.19-3.23
(1H, m), 4.20-4.31 (3H, m), 4.65-4.8 (1H, m), 5.5-5.6 (1H, m), 6.90-6.96 (4H, m), 7.01-7.05 (1H, m), 7.21-7.34 (8H, m). Found: C, 81.3; H, 7.3; N, 3.2: Calculated for C28H29NO2 C, 81.72; H, 7.10; N, 3.40%.
EXAMPLE 3: 1-(10.11-Dihydro-5H-dibenzo[a,d]cyclohepten-5- yl]-2-(3,5-dimethylbenzyloxy)ethylamine, oxalate salt 5-Chlorodibenzosuberane was treated in an analogous manner to that described in Example 1 to yield the title compound, mp 197-200°C.1H NMR (360MHz,DMSO-d6) δ 2.27 (6H, m), 2.8-3.1 (1H, br m), 3.15-3.25 (1H, m), 3.3-3.5 (3H, m), 4.1-4.2 (2H, m), 4.3-4.35 (1H, m), 4.4-4.5 (1H, m), 6.93 (3H, s),
7.1-7.3 (8H, m). Found: C, 71.83; H, 6.77; N, 2.97; Calculated for
C25H29NO·C2H2O4C, 72.14; H, 6.95; N' 3.12%.
EXAMPLE 4: N-Acetamido-1-(10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5-yl)-2-(3,5-dimethylbenzyloxy)ethylamine
The product of Example 3 was acetylated in an analogous manner to that described in Example 2 to give the title compound, mp 109-111°C. 1H NMR (360MHz, CDCl3) δ 1.60 (3H, s), 2.35 (6H, s), 2.84-2.91 (2H, m), 3.37 (2H, s), 3.46-3.68
(2H, m), 4.14-4.17 (1H, m), 4.35-4.44 (2H, m), 4.84-4.90 (1H, m),
5.66-5.71 (1H, m), 6.98-7.25 (11H, m). Found: C, 71.83; H, 6.77;
N, 2.97; Calculated for C25H29NO.C2H2O4C, 72.14; H, 6.95;
N, 3.12%.
EXAMPLE 5: N.N-Dimethyl-1-(10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5-yl)-2-(3,5-dimethylbenzyloxy)ethylamine, oxalate salt
To a solution of the product of Example 3 (320mg) in 2ml methanol at 0°C was added acetic acid (0.25ml) followed by sodium borohydride (0.1g) and aqueous formaldehyde (0.167ml of 37 wt %). Solvent was removed after 3 days and the residue purified by chromatography (1:1 ethyl acetate-petroleum ether eluant). The purified free base was dissolved in diethyl ether and oxalic acid in methanol was added. Solvent was removed to give the title compound, mp 68-69°C (ethyl acetate-petroleum ether).1H NMR (360MHz, DMSO-d6) 2.24 (6H, s), 2.49 (6H, s), 2.8-3.0 (2H, m), 3.32-3.50 (4H, m), 3.8-4.4 (4H, br m), 6.82 (2H, s), 6.88 (1H, s), 7.0-7.3 (8H, m). Found: C, 69.34; H, 6.71; N, 2.64: Calculated for C28H33NO.1.5 (C2H2O4) C, 69.64; H, 6.78; N, 2.62%.
EXAMPLE 6: 1-(10,11-Dihydro-5H-dibenzo[a,d]cyclohepten-5-yl)-2-(3,5-bistrifluoromethylbenzyloxy)ethyl amine, oxalate salt
Prepared in an analogous manner to that described for the product of Example 3. mp 175-176°C. 1H NMR (360MHz, DMSO-d6) 2.8-3.0 (2H, m), 3.3-3.6 (3H, m), 4.1-4.3 (2H, m), 4.5- 4.7 (2H, m), 7.0-7.4 (8H, m), 8.0-8.1 (3H, m).
EXAMPLE 7: N-Carbomethoxymethyl-1-(10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5-yl)-2-(3,5- bistrifluoromethylbenzyloxy)ethylamine, oxalate salt
A mixture of the title product of Example 6, methyl bromoacetate (one equivalent) and triethylamine (one equivalent) were refluxed in THF for 22h. The reaction was partitioned between diethyl ether-water and the organic phase separated and dried (MgSO4). Solvent was removed in vacuo and the residue redissolved in ethanol. Oxalic acid was added, solvent removed in vacuo, and the residue recrystallised to give the title compound, mp 160-165°C (ethyl acetate-petroleum ether).1H NMR (360MHz, DMSO-d6) 2.8-3.0 (2H, m), 3.2 (2H, s), 3.4-3.5 (6H, m), 3.6-3.65 (1H, m), 3.9-3.95 (1H, m), 4.5 (3H, s), 7.00-7.2 (8H, m), 7.9 (3H, s).
EXAMPLE 8: N-Carbamoylmethyl-1-(10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5-yl)-2-(3,5-bistrifluoromethylbenzyloxy)ethylamine, oxalate salt
A solution of the product of Example 6 in 5ml methanol saturated with ammonia was sealed in a reaction vessel and left at 0°C for 48h. Solvent was removed in vacuo, and the residue dissolved in ethanol. Oxalic acid and water were added to give the title compound, mp 120-122°C. 1H NMR (360MHz, DMSO-d6) 2.8-3.0 (2H, m), 3.3-3.7 (6H, m), 4.2-4.6 (4H, m), 7.00-7.4
(8H, m), 7.9 (1H, s), 7.95 (2H, s).,
EXAMPLE 9: 3,5-Dimethylbenzyl-2-(9-fluoroenyl)glycinate
a) A solution of N-(diphenylmethylene)glycine ethyl ester (5g) in tetrahydrofuran (15ml) was added dropwise to a cooled solution of lithium diisopropylamide (1 mol eq) in tetrahydrofuran (20ml) and hexane (11ml) at -70°C. After 1h at -70°C, a solution of 9-bromofluorene (4.58g) in tetrahydrofuran (15ml) was added. After stirring the solution for 1h at -70°C and then at room temperature (16h), the solution was diluted with saturated ammonium chloride solution (500ml) and diethyl ether (500ml). The organic phase was washed with saturated brine and dried (MgSO4). After removal of the solvent in vacuo the residue was purified on a silica column to give ethyl N-(diphenylmethylene)-(9-fluoroenyl)glycinate, 2.2g.
b) The product of part (a) (2.2g) was heated at reflux for 16h with 5.5M-hydrochloric acid (30ml). The cooled solution was washed with diethyl ether and the aqueous phase evaporated to dryness. The residue was recrystallised from acetone-water to give 2-(9-fluorenyl)glycine hydrochloride, 0.7g.
c) To a solution of the product of part (b) (0.7g) and sodium carbonate (0.95g) in dioxan (5ml) and water (10ml) was added di-t-butyldicarbonate (0.664g). After stirring the solution for 16h at room temperature, water (50ml) and diethyl ether (60ml) were added. To the aqueous phase was added solid citric acid to pH 3 and the product extracted into ethyl acetate (3 × 30ml).
The combined organic phases were washed with water, saturated brine, dried (MgSO4) and evaporated to dryness to give N-t-butoxycarbonyl-2-(9-fluorenyl)glycine 0.75g.
d) To a solution of the product of part (c) (0.750g) in methanol (10ml) was added a solution of cesium carbonate
(348mg) in water (5ml). The solvent was removed by evaporation and the residue dried by repeated evaporation from dimethylformamide (3 × 30ml). To the residue dissolved in dimethylformamide (50ml) was added 3,5-dimethylbenzyl bromide (0.636g) and the solution stirred at room temperature for 16h. The solvent was removed in vacuo and the residue partitioned between dichloromethane and water. The organic phase was washed with saturated brine, dried (MgSO4) and evaporated in vacuo . The residue was chromatographed on silica gel eluting with mixtures of ethyl acetate in petroleum ether bp 60-80°C to give 3,5-dimethylbenzyl-N-t-butoxycarbonyl-2-(9-fluorenyl)glycinate. Trifluoroacetic acid (3ml) was added to the ester (0.2g) and after 40 minutes the solvent was removed by evaporation and a solution of 4-toluenesulfonic acid (84mg) in ethanol (1ml) added to give 3,5-dimethylbenzyl-2-(9-fluorenyl)glycinate, 4-toluenesulphonic acid salt, mp 150-151°C. Found: C, 70.19; H, 5.74; N, 2.61. C24H23NO2.C7H8SO3 requires C, 70.30; H, 5.90; N, 2.64%. m/z (FAB+) = 358 (M+H). EXAMPLE 10 : 8 ,5 -Dimethylbenzyl N-acetyl-2-(9 -fluorenyl)glycinate
3 , 5-Dime thylbenzyl N-t-butoxycarbo nyl-2-(9-fluorenyl)glycinate (Example 9d, 0.5g) was dissolved in trifluoroacetic acid (10ml) for 40 minutes then evaporated in vacuo. To a solution of the residue in pyridine (10ml) was added acetic anhydride (1ml) for 16h. The solvent was removed in υacuo and the residue crystallised from diethyl ether/petroleum ether bp 60-80°C to give the title compound mp 162-164°C. Found: C, 78.19; H, 6.26; N, 3.45. C26H25NO3 requires C,. 78.17; H, 6.30; N, 3.50%. m/z (CI+) = 400 (M+H), (CD = 398 (M- H). The following examples illustrate pharmaceutical
compositions according to the invention.
EXAMPLE 11A Tablets containing 1-25mg of compound
Amount mg
Compound of formula (I) 1.0 2.0 25.0 Microcrystalline cellulose 20.0 20.0 20.0 Modified food corn starch 20.0 20.0 20.0 Lactose 58.5 57.5 34.5
Magnesium Stearate 0.5 0.5 0.5
EXAMPLE 11B Tablets containing 26-100mg of compound
Amount mg
Compound of formula (I) 26.0 50.0 100.0
Microcrystalline cellulose 80.0 80.0 80.0
Modified food corn starch 80.0 80.0 80.0
Lactose 213.5 189.5 139.5
Magnesium Stearate 0.5 0.5 0.5 The compound of formula (I), cellulose, lactose and a portion of the corn starch are mixed and granulated with
10% corn starch paste. The resulting granulation is sieved, dried and blended with the remainder of the corn starch and the magnesium stearate. The resulting granulation is then compressed into tablets containing
1.0mg, 2.0mg, 25.0mg, 26.0mg, 50.0mg and 100mg of the active compound per tablet.
EXAMPLE 12 Parenteral injection
Amount mg
Compound of formula (I) 1 to 100mg
Citric Acid Monohydrate 0.75mg
Sodium Phosphate 4.5mg
Sodium Chloride 9mg Water for injection to 1ml
The sodium phosphate, citric acid monohydrate and sodium chloride are dissolved in a portion of the water. The compound of formula (I) is dissolved or suspended in the solution and made up to volume.
EXAMPLE 13 Topical formulation
Amount mg
Compound of formula (I) 1-10g
Emulsifying Wax 30g
Liquid paraffin 20g
White Soft Paraffin to 100g
The white soft paraffin is heated until molten. The liquid paraffin and emulsifying wax are incorporated and stirred until dissolved. The compound of formula (I) is added and stirring continued until dispersed. The mixture is then cooled until solid.
SUBSTANCE P ANTAGONISM ASSAY
A. Receptor Expression in Monkey Kidney Cell Line (COS)
To express the cloned human neurokinin-1- receptor (NK1R) transiently in COS, the cDNA for the human NK1R was cloned into the expression vector pCDM9 which was derived from pCDM8 (INVITROGEN) by inserting the
ampicillin resistance gene (nucleotide 1973 to 2964 from BLUESCRIPT SK+ (trademark, STRATAGENE, La Jolla, CA, USA)) into the Sac II site. Transfection of 20 ug of the plasmid DNA into 10 million COS cells was achieved by electroporation in 800 μl of transfection buffer (135 mM NaCl, 1.2 mM CaCl2, 1.2 mM MgCl2, 2.4 mM K2HPO4, 0.6 mM KH2PO4, 10 mM glucose, 10 mM N-2-hydroxyethyl-piperazine-N'-2-ethane sulphonic acid (HEPES) pH 7.4) at 260 V and 950 μF using the IBI GENEZAPPER (trademark IBI, New Haven, CT, USA). The cells were incubated in 10% fetal calf serum, 2 mM glutamine, 100U/ml penicillin-streptomycin, and 90% DMEM media (GIBCO, Grand Island, NY, USA) in 5% CO2 at 37ºC for three days before the binding assay.
B. Stable Expression in Chinese Hamster Ovarian Cell Line
To establish a stable cell line expressing cloned human NKIR, the cDNA was subcloned into the vector pRcCMV (INVITROGEN). Transfection of 20 ug of the plasmid DNA into CHO cells was achieved by electroporation in 800 μl of transfection buffer supplemented with 0.625 mg/ml Herring sperm DNA at 300 V and 950 μF using the IBI
GENEZAPPER (IBI). The transfected cells were incubated in CHO media [10% fetal calf serum, 100 U/ml penicillin-streptomycin, 2 mM glutamine, 1/500 hypoxanthine-thymidine (ATCC), 90% IMDM media (JRH BIOSCIENCES,
Lenexa, KS, USA), 0.7 mg/ml G418 (GIBCO)] in 5% CO2 at 37ºC until colonies were visible. Each colony was separated and propagated. The cell clone with the highest number of human NKIR was selected for subsequent applications such as drug screening. C. Assay Protocol using COS or CHO
The binding assay of human NKIR expressed in either COS or CHO cells is based on the use of 125I-substance P (125I-SP, from DU PONT, Boston, MA) as a radioactively labeled ligand which competes with unlabeled substance P or any other ligand for binding to the human NKIR.
Monolayer cell cultures of COS or CHO were dissociated by the non-enzymatic solution (SPECIALTY MEDIA, Lavellette, NJ) and resuspended in appropriate volume of the binding buffer (50 mM Tris pH 7.5, 5 mM MnCl2, 150 mM NaCl, 0.04 mg/ml bacitracin, 0.004 mg/ml leupeptin, 0.2 mg/ml BSA, 0.01 mM phosphoramidon) such that 200 μl of the cell suspension would give rise to about 10,000 cpm of specific 125I-SP binding (approximately 50,000 to 200,000 cells). In the binding assay, 200 ul of cells were added to a tube containing 20 ul of 1.5 to 2.5 nM of 125I-SP and 20 μl of unlabeled substance P or any other test compound. The tubes were incubated at 4ºC or at room temperature for 1 hour with gentle shaking. The bound radioactivity was separated from unbound radioactivity by GF/C filter (BRANDEL, Gaithersburg, MD) which was prewetted with 0.1% polyethylenimine. The filter was washed with 3 ml of wash buffer (50 mM Tris pH 7.5, 5 mM MnCl2, 150 mM NaCl) three times and its radioactivity was determined by gamma counter.
The activation of phospholiphase C by NK1R may also be measured in CHO cells expressing the human NK1R by determining the accumulation of inositol monophosphate which is a degradation product of IP3. CHO cells are seeded in 12-well plate at 250,000 cells per well. After incubating in CHO media for 4 days, cells are loaded with 5μCi of 3H-myoinositol in 1 ml of media per well by overnight incubation. The extracellular radioactivity is removed by washing with phosphate buffered saline. LiCl is added to the well at final concentration of 10 mM with or without the test compound, and incubation is continued at 37ºC for 15 min. Substance P is added to the well at final concentration of 0.3nM to activate the human NK1R. After 30 min of incubation at 37ºC, the medium is removed and 0.1 N HCl is added. Each well is sonicated at 4ºC and extracted with CHCl3/methanol (1:1). The aqueous phase is applied to a 1 ml Dowex AG 1X8 ion exchange column. The column is washed with 0.1 N formic acid followed by 0.025 M ammonium formate-0.1 N formic acid. The inositol monophosphate is eluted with 0.2 M ammonium formate-0.1 N formic acid and quantitated by beta counter.
The data in Table 1 were obtained for compounds of formula (I):
TABLE 1
SUBSTANCE P ANTAGONISM RESULTS
Compound of EX # IC50 @ NK1R (nM)
1 80
2 70
3 100, 190
4 150
5 320
6 250
7 42% @ 1μM
8 60
9 300
10 300

Claims

CLAIMS:
1. A compound of formula (I), or a salt or prodrug thereof:
Figure imgf000036_0002
wherein
Q represents a group
Figure imgf000036_0001
where W represents a bond, O, S, -CH2CH2-, -CH=CH- or a group NR6, where R6 is H or C1-6alkyl and one or both of the phenyl rings may be replaced by a heteroaryl moiety;
X and Y each represent H or X and Y together form a group =0;
Z represents O, S or NR8, where R8 represents H or C1-6alkyl;
R1 and R2 independently represent H; C1-6 alkyl optionally substituted by hydroxy, cyano, CORa, COORa, CONRaRb, COC1-4alkylNRaRb, CONRaC1-4alkylCONRaRb or
NRaRb, (where Ra and Rb each independently represent H, C1-6 alkyl, phenyl (optionally substituted by one or more of C1-6alkyl, C1-6alkoxy, halo and trifluoromethyl) or phenyl (C1-4alkyl) (optionally substituted in the phenyl ring by one or more of C1-6 alkyl, C1-6 alkoxy, halo and trifluoromethyl)); phenyl (C1-4 alkyl), (optionally substituted by one or more of C1-6 alkyl, C1-6 alkoxy, halo and trifluoromethyl in the phenyl ring); C2-6 alkylene; COC1-6alkylhalo; CORa; COORa; CONHRa;
COC1-4alkylNRaRb; or CONRaC1-4alkylCONRaRb; (where Ra and Rb are as previously defined) or R1 and R2 together form a chain (CH2)p where p is 4 or 5 and where one nonterminal methylene group may optionally be replaced by an oxygen atom or a group NRc, where Rx is H or C1-6 alkyl;
R3 represents H or C1-6alkyl;
R4 represents H, C1-6 alkyl or phenyl (optionally substituted by one or more of C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, halo, cyano, nitro, trifluoromethyl, trimethylsilyl, SRc, SORc, SO2Rc, ORc, NRcRd, NRcCORd, NRcCOORd, COORc or CONRcRd, where Rc and Rd each
independently represent H, C1-6 alkyl, phenyl or
trifluoromethyl);
R5 represents (CH2)qphenyl, wherein q is 0, 1, 2 or 3 which may optionally be substituted in the phenyl ring by one or more of C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, halo, cyano, nitro, trifluoromethyl, trimethylsilyl, SRc, SORc, SO2Rc, ORc, NRcRd, NRcCORd, NRcCOORd, COORc or CONRcRd, where Rc and Rd are as above defined;
each R7 and R8 independently represents C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, halo, cyano, nitro,
trifluoromethyl, trimethylsilyl, SRc, SORc, SO2Rc, ORc, NRcRd, NRcCORd, NRcCOORd, COORc or CONRcRd, where Rc and Rd are as above defined; and
m and n independently represent 0, 1, 2, 3 or 4.
2. A compound as claimed in claim 1 wherein Q represents a group
Figure imgf000038_0001
where W1 is a bond, O, S or a group NR6, where R6 is H or C1-6 alkyl;
R1 and R2 each independently represent H, C1-6 alkyl, phenyl(C1-4 alkyl), COR10, COOR10 or CONHR10, where R10 is C1-6 alkyl or phenyl;
R4 represents H, C1-6 alkyl or phenyl (optionally substituted by one or more of C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, halo, cyano, nitro, trifluoromethyl, SCH3, SOCH3, SO2CH3, ORc, NRcRd, NRcCORd, NRcCOORd, COORc or CONRcRd (where Rc and Rd are as above defined); and
R5 represents (CH2)qphenyl, wherein q is 0, 1, 2 or 3 which may optionally be substituted by one or more of C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, halo, cyano, nitro, trifluoromethyl, SCH3, SOCH3, SO2CH3, ORc, NRcRd, NRcCORd, NRcCOORd, COORc or CONRcRd (where Rc and Rd are as above defined);
wherein any phenyl (including benzyl) ring in the definitions of R1 or R2 (including R10) above may be optionally substituted by one or more of C1-6 alkyl, C1-6 alkoxy, halo or trifluoromethyl.
3. A compound as claimed in claim 1 or claim 2 wherein W represents a bond.
4. A compound as claimed in claim 1 wherein W represents CH2CH2 or HC=CH.
5. A compound as claimed in any one of claims 1 to 4 wherein X and Y each represent H and Z represents 0.
6. A compound as claimed in any one of claims 1 to 5 wherein R4 is H and R5 is substituted phenyl.
7. A compound as claimed in claim 1 selected from: 1-(5H-dibenzo[a,d]cyclohepten-5-yl)-2-(3,5-dimethylbenzyloxy)ethylamine;
N-acetamido-1-(5H-dibenzo[a,d]cyclohepten-5-yl)-2-(3,5-dimethylbenzyloxy)ethylamine;
1-(10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5-yl)-2-(3,5-dimethylbenzyloxy)ethylamine;
N-acetamido-1-(10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5-yl)-2-(3,5-dimethylbenzyloxy)ethylamine;
N,N-dimethyl-1-(10,11-dihydro-5H-dibenzo[a,d]cyclohepten- 5-yl)-2-(3,5-dimethylbenzyloxy)ethylamine;
1-(10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5-yl)-2-(3,5-bistrifluoromethylbenzyloxy)ethylamine;
N-carbomethoxymethyl-1-(10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5-yl)-2-(3,5-bistrifluoromethylbenzyloxy)ethylamine;
N-carbamoylmethyl-1-(10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5-yl)-2-(3,5-bistrifluoromethylbenzyloxy)ethylamine;
3,5-dimethylbenzyl-2-(9-fluorenyl)glycinate;
3,5-dimethylbenzyl N-acetyl-2-(9-fluorenyl)glycinate; and salts and prodrugs thereof.
8. A compound as claimed in any one of the
preceding claims for use in therapy.
9. A process for the preparation of a compound as claimed in any one of claims 1 to 7 which process comprises reaction of a compound of formula (II):
Figure imgf000040_0001
wherein Q, R1, R2, R3 , X and Y are as defined for formula (I) and Z is O or S, with a compound of formula HalCHR4R5 where R4 and R5 are as defined for formula (I) and Hal is halo, in the presence of a base, and, if necessary or desired, converting the compound of formula (I) so prepared to another compound of formula (I), or a salt or prodrug thereof.
10. A pharmaceutical composition comprising a compound as claimed in any of claims 1 to 7 in
association with a pharmaceutically acceptable carrier or excipient.
11. A method for the treatment or prevention of a physiological disorder associated with an excess of tachykinins, which method comprises administration to a patient in need thereof of a tachykinin-reducing amount of a compound according to claim 1.
12. A method according to claim 11 for the
treatment or prevention of pain or inflammation.
13. A method according to claim 11 for the treatment or prevention of migraine.
14. A method according to claim 11 for the treatment or prevention of postherpetic neuralgia.
PCT/GB1992/001212 1991-07-10 1992-07-03 Fused tricyclic compounds, pharmaceutical compositions containing them and their use in therapy WO1993001159A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US08/175,432 US5495047A (en) 1991-07-10 1992-07-03 Fused tricyclic compounds, pharmaceutical compositions containing them and their use in therapy

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GB919114886A GB9114886D0 (en) 1991-07-10 1991-07-10 Therapeutic agents
GB9114886.6 1991-07-10
GB9208141.3 1992-04-14
GB929208141A GB9208141D0 (en) 1992-04-14 1992-04-14 Therapeutic agents

Publications (1)

Publication Number Publication Date
WO1993001159A1 true WO1993001159A1 (en) 1993-01-21

Family

ID=26299217

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB1992/001212 WO1993001159A1 (en) 1991-07-10 1992-07-03 Fused tricyclic compounds, pharmaceutical compositions containing them and their use in therapy

Country Status (2)

Country Link
US (1) US5495047A (en)
WO (1) WO1993001159A1 (en)

Cited By (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994016697A1 (en) 1993-01-19 1994-08-04 Rhone-Poulenc Rorer S.A. Synergising association having an antagonist effect on nk1 and nk2 receptors
EP0680749A2 (en) * 1994-05-05 1995-11-08 L'oreal Cosmetic composition comprising an antagonist of substance P
FR2719476A1 (en) * 1994-05-05 1995-11-10 Oreal Cosmetics for application to sensitive skins
EP0717998A1 (en) 1994-12-19 1996-06-26 L'oreal Use of substance P antagonist for the treatment of pruritis, eye or eyelid pain or disesthesies
EP0722736A1 (en) 1994-12-19 1996-07-24 L'oreal Topical composition containing an antagonist of substance P
EP0722722A1 (en) 1994-12-19 1996-07-24 L'oreal Use of an antagonist of substance P for the treatment of cutaneous erythema of neurogenic origin
EP0747345A2 (en) * 1995-06-06 1996-12-11 Bristol-Myers Squibb Company Ethylamido fluorenes and improved method of making same
EP0769300A2 (en) 1995-10-20 1997-04-23 Pfizer Inc. Antiemetic composition containing an NK-1 receptor antagonists
EP0774250A1 (en) 1995-11-20 1997-05-21 L'oreal Use of a TNF alpha antagonist against skin redness of neurogenic origin
US5658943A (en) * 1995-01-05 1997-08-19 Warner-Lambert Company Phenylalanine derivatives as endothelin antagonists
US6203803B1 (en) 1994-12-14 2001-03-20 Societe L'oreal S.A. Use of a substance P antagonist in a cosmetic composition, and the composition thus obtained
WO2003040125A1 (en) * 2001-11-09 2003-05-15 Novartis Ag Piperazine derivatives having sst1 antagonistic activity
US6579885B2 (en) 1999-11-03 2003-06-17 Albany Molecular Research, Inc. Aryl and heteroaryl substituted tetrahydroisoquinolines and use thereof
US7084152B2 (en) 2000-07-11 2006-08-01 Amr Technology, Inc. 4-phenyl substituted tetrahydroisoquinolines therapeutic use thereof
WO2006123182A2 (en) 2005-05-17 2006-11-23 Merck Sharp & Dohme Limited Cyclohexyl sulphones for treatment of cancer
US7163949B1 (en) 1999-11-03 2007-01-16 Amr Technology, Inc. 4-phenyl substituted tetrahydroisoquinolines and use thereof
WO2007011820A2 (en) 2005-07-15 2007-01-25 Amr Technology, Inc. Aryl-and heteroaryl-substituted tetrahydrobenzazepines and use thereof to block reuptake of norepinephrine, dopamine, and serotonin
WO2007041052A2 (en) 2005-09-29 2007-04-12 Merck & Co., Inc. Acylated spiropiperidine derivatives as melanocortin-4 receptor modulators
WO2007093827A1 (en) 2006-02-15 2007-08-23 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Thiophene and thiazole substituted trifluoroethanone derivatives as histone deacetylase (hdac) inhibitors
WO2008090117A1 (en) 2007-01-24 2008-07-31 Glaxo Group Limited Pharmaceutical compositions comprising 3, 5-diamin0-6- (2, 3-dichl0phenyl) -l, 2, 4-triazine or r (-) -2, 4-diamino-5- (2, 3-dichlorophenyl) -6-fluoromethyl pyrimidine and an nk1
WO2008120653A1 (en) 2007-04-02 2008-10-09 Banyu Pharmaceutical Co., Ltd. Indoledione derivative
WO2009002495A1 (en) 2007-06-27 2008-12-31 Merck & Co., Inc. 4-carboxybenzylamino derivatives as histone deacetylase inhibitors
WO2009111354A2 (en) 2008-03-03 2009-09-11 Tiger Pharmatech Tyrosine kinase inhibitors
WO2010000073A1 (en) 2008-07-03 2010-01-07 Neuraxon, Inc. Benzoxazines, benzothiazines, and related compounds having nos inhibitory activity
WO2010114780A1 (en) 2009-04-01 2010-10-07 Merck Sharp & Dohme Corp. Inhibitors of akt activity
WO2010132442A1 (en) 2009-05-12 2010-11-18 Albany Molecular Reserch, Inc. 7-([1,2,4,]triazolo[1,5,-a]pyridin-6-yl)-4-(3,4-dichlorophenyl)-1,2,3,4- tetrahydroisoquinoline and use thereof
WO2010132487A1 (en) 2009-05-12 2010-11-18 Bristol-Myers Squibb Company CRYSTALLINE FORMS OF (S)-7-([1,2,4]TRIAZOLO[1,5-a]PYRIDIN-6-YL)-4-(3,4-DICHLOROHPHENYL)-1,2,3,4-TETRAHYDROISOQUINOLINE AND USE THEREOF
WO2011046771A1 (en) 2009-10-14 2011-04-21 Schering Corporation SUBSTITUTED PIPERIDINES THAT INCREASE p53 ACTIVITY AND THE USES THEREOF
EP2336120A1 (en) 2007-01-10 2011-06-22 Istituto di ricerche di Biologia Molecolare P. Angeletti S.R.L. Combinations containing amide substituted indazoles as poly(ADP-ribose)polymerase (PARP) inhibitors
WO2011163330A1 (en) 2010-06-24 2011-12-29 Merck Sharp & Dohme Corp. Novel heterocyclic compounds as erk inhibitors
WO2012018754A2 (en) 2010-08-02 2012-02-09 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF CATENIN (CADHERIN-ASSOCIATED PROTEIN), BETA 1 (CTNNB1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2012027236A1 (en) 2010-08-23 2012-03-01 Schering Corporation NOVEL PYRAZOLO[1,5-a]PYRIMIDINE DERIVATIVES AS mTOR INHIBITORS
WO2012030685A2 (en) 2010-09-01 2012-03-08 Schering Corporation Indazole derivatives useful as erk inhibitors
WO2012036997A1 (en) 2010-09-16 2012-03-22 Schering Corporation Fused pyrazole derivatives as novel erk inhibitors
WO2012087772A1 (en) 2010-12-21 2012-06-28 Schering Corporation Indazole derivatives useful as erk inhibitors
WO2012145471A1 (en) 2011-04-21 2012-10-26 Merck Sharp & Dohme Corp. Insulin-like growth factor-1 receptor inhibitors
WO2013063214A1 (en) 2011-10-27 2013-05-02 Merck Sharp & Dohme Corp. Novel compounds that are erk inhibitors
WO2013165816A2 (en) 2012-05-02 2013-11-07 Merck Sharp & Dohme Corp. SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS
EP2698157A1 (en) 2006-09-22 2014-02-19 Merck Sharp & Dohme Corp. Method of treatment using fatty acid synthesis inhibitors
WO2014052563A2 (en) 2012-09-28 2014-04-03 Merck Sharp & Dohme Corp. Novel compounds that are erk inhibitors
WO2014085216A1 (en) 2012-11-28 2014-06-05 Merck Sharp & Dohme Corp. Compositions and methods for treating cancer
WO2014100065A1 (en) 2012-12-20 2014-06-26 Merck Sharp & Dohme Corp. Substituted imidazopyridines as hdm2 inhibitors
WO2014120748A1 (en) 2013-01-30 2014-08-07 Merck Sharp & Dohme Corp. 2,6,7,8 substituted purines as hdm2 inhibitors
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
US9034899B2 (en) 2009-05-12 2015-05-19 Albany Molecular Research, Inc. Aryl, heteroaryl, and heterocycle substituted tetrahydroisoquinolines and use thereof
US9085531B2 (en) 2004-07-15 2015-07-21 Albany Molecular Research, Inc. Aryl- and heteroaryl-substituted tetrahydroisoquinolines and use thereof to block reuptake of norepinephrine, dopamine, and serotonin
US9156812B2 (en) 2008-06-04 2015-10-13 Bristol-Myers Squibb Company Crystalline form of 6-[(4S)-2-methyl-4-(2-naphthyl)-1,2,3,4-tetrahydroisoquinolin-7-yl]pyridazin-3-amine
EP3327125A1 (en) 2010-10-29 2018-05-30 Sirna Therapeutics, Inc. Rna interference mediated inhibition of gene expression using short interfering nucleic acids (sina)
WO2019094311A1 (en) 2017-11-08 2019-05-16 Merck Sharp & Dohme Corp. Prmt5 inhibitors
WO2020033284A1 (en) 2018-08-07 2020-02-13 Merck Sharp & Dohme Corp. Prmt5 inhibitors
WO2020033282A1 (en) 2018-08-07 2020-02-13 Merck Sharp & Dohme Corp. Prmt5 inhibitors
US11096950B2 (en) 2006-11-01 2021-08-24 Barbara Brooke Jennings Compounds, methods, and treatments for abnormal signaling pathways for prenatal and postnatal development
EP4079856A1 (en) 2010-08-17 2022-10-26 Sirna Therapeutics, Inc. Rna interference mediated inhibition of hepatitis b virus (hbv) gene expression using short interfering nucleic acid (sina)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2001500850A (en) 1996-08-22 2001-01-23 ワーナー―ランバート・コンパニー Non-peptide bombesin receptor antagonist
US6573244B1 (en) * 1999-05-17 2003-06-03 The United States Of America As Represented By The Secretary Of The Army Previns as specific inhibitors and therapeutic agents for Botulinum toxin B and Tetanus neurotoxins
US6395763B1 (en) 1999-06-25 2002-05-28 Vertex Pharmaceuticals Incorporated Prodrugs of carbamate inhibitors of IMPDH

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2035535A1 (en) * 1969-07-18 1971-01-28 Centre Europeen de Recherches Mau vernay CERM, Riom, Puy de Dome (Frankreich) Square bracket on 3 alkoxy 2 (diaryl) methyl square bracket on propylamine, its manufacture and use
GB2054588A (en) * 1979-07-11 1981-02-18 Synthese Soc Ind Prod Amino-ether oxides and pharmaceutical formulations thereof
EP0394989A2 (en) * 1989-04-28 1990-10-31 Fujisawa Pharmaceutical Co., Ltd. Peptide compounds, process for preparation thereof and pharmaceutical composition comprising the same

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2035535A1 (en) * 1969-07-18 1971-01-28 Centre Europeen de Recherches Mau vernay CERM, Riom, Puy de Dome (Frankreich) Square bracket on 3 alkoxy 2 (diaryl) methyl square bracket on propylamine, its manufacture and use
GB2054588A (en) * 1979-07-11 1981-02-18 Synthese Soc Ind Prod Amino-ether oxides and pharmaceutical formulations thereof
EP0394989A2 (en) * 1989-04-28 1990-10-31 Fujisawa Pharmaceutical Co., Ltd. Peptide compounds, process for preparation thereof and pharmaceutical composition comprising the same

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CHEMICAL ABSTRACTS, vol. 64, 1966, Columbus, Ohio, US; abstract no. 8294b, *

Cited By (71)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994016697A1 (en) 1993-01-19 1994-08-04 Rhone-Poulenc Rorer S.A. Synergising association having an antagonist effect on nk1 and nk2 receptors
EP0680749A2 (en) * 1994-05-05 1995-11-08 L'oreal Cosmetic composition comprising an antagonist of substance P
FR2719476A1 (en) * 1994-05-05 1995-11-10 Oreal Cosmetics for application to sensitive skins
EP0680749A3 (en) * 1994-05-05 1996-09-11 Oreal Cosmetic composition comprising an antagonist of substance P.
US6333042B1 (en) 1994-05-05 2001-12-25 Societe L'oreal S.A. Use of a substance P antagonist in a cosmetic composition, and the composition thus obtained
US6235291B1 (en) 1994-05-05 2001-05-22 Societe L'oreal S.A. Use of a substance P antagonist in a cosmetic composition, and the composition thus obtained
US6203803B1 (en) 1994-12-14 2001-03-20 Societe L'oreal S.A. Use of a substance P antagonist in a cosmetic composition, and the composition thus obtained
EP0717998A1 (en) 1994-12-19 1996-06-26 L'oreal Use of substance P antagonist for the treatment of pruritis, eye or eyelid pain or disesthesies
EP0722736A1 (en) 1994-12-19 1996-07-24 L'oreal Topical composition containing an antagonist of substance P
EP0722722A1 (en) 1994-12-19 1996-07-24 L'oreal Use of an antagonist of substance P for the treatment of cutaneous erythema of neurogenic origin
EP0722722B2 (en) 1994-12-19 2003-03-05 L'oreal Use of an antagonist of substance P for the treatment of cutaneous erythema of neurogenic origin
US5658943A (en) * 1995-01-05 1997-08-19 Warner-Lambert Company Phenylalanine derivatives as endothelin antagonists
AU697037B2 (en) * 1995-06-06 1998-09-24 Bristol-Myers Squibb Company Ethylamido fluorenes and improved method of making same
EP0747345A3 (en) * 1995-06-06 1997-07-30 Bristol Myers Squibb Co Ethylamido fluorenes and improved method of making same
EP0747345A2 (en) * 1995-06-06 1996-12-11 Bristol-Myers Squibb Company Ethylamido fluorenes and improved method of making same
EP0769300A2 (en) 1995-10-20 1997-04-23 Pfizer Inc. Antiemetic composition containing an NK-1 receptor antagonists
EP0774250A1 (en) 1995-11-20 1997-05-21 L'oreal Use of a TNF alpha antagonist against skin redness of neurogenic origin
US6579885B2 (en) 1999-11-03 2003-06-17 Albany Molecular Research, Inc. Aryl and heteroaryl substituted tetrahydroisoquinolines and use thereof
US7163949B1 (en) 1999-11-03 2007-01-16 Amr Technology, Inc. 4-phenyl substituted tetrahydroisoquinolines and use thereof
US7084152B2 (en) 2000-07-11 2006-08-01 Amr Technology, Inc. 4-phenyl substituted tetrahydroisoquinolines therapeutic use thereof
US7271168B2 (en) 2001-11-09 2007-09-18 Novartis Ag Piperazine derivatives having SST1 antagonistic activity
WO2003040125A1 (en) * 2001-11-09 2003-05-15 Novartis Ag Piperazine derivatives having sst1 antagonistic activity
US9085531B2 (en) 2004-07-15 2015-07-21 Albany Molecular Research, Inc. Aryl- and heteroaryl-substituted tetrahydroisoquinolines and use thereof to block reuptake of norepinephrine, dopamine, and serotonin
US9499531B2 (en) 2004-07-15 2016-11-22 Albany Molecular Research, Inc. Aryl- and heteroaryl-substituted tetrahydroisoquinolines and use thereof to block reuptake of norepinephrine, dopamine, and serotonin
WO2006123182A2 (en) 2005-05-17 2006-11-23 Merck Sharp & Dohme Limited Cyclohexyl sulphones for treatment of cancer
WO2007011820A2 (en) 2005-07-15 2007-01-25 Amr Technology, Inc. Aryl-and heteroaryl-substituted tetrahydrobenzazepines and use thereof to block reuptake of norepinephrine, dopamine, and serotonin
US9403776B2 (en) 2005-07-15 2016-08-02 Albany Molecular Research, Inc. Aryl- and heteroaryl-substituted tetrahydrobenzazepines and use thereof to block reuptake of norepinephrine, dopamine, and serotonin
WO2007041052A2 (en) 2005-09-29 2007-04-12 Merck & Co., Inc. Acylated spiropiperidine derivatives as melanocortin-4 receptor modulators
WO2007093827A1 (en) 2006-02-15 2007-08-23 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Thiophene and thiazole substituted trifluoroethanone derivatives as histone deacetylase (hdac) inhibitors
EP2698157A1 (en) 2006-09-22 2014-02-19 Merck Sharp & Dohme Corp. Method of treatment using fatty acid synthesis inhibitors
EP2946778A1 (en) 2006-09-22 2015-11-25 Merck Sharp & Dohme Corp. Method of treatment using fatty acid synthesis inhibitors
US11096950B2 (en) 2006-11-01 2021-08-24 Barbara Brooke Jennings Compounds, methods, and treatments for abnormal signaling pathways for prenatal and postnatal development
EP2805945A1 (en) 2007-01-10 2014-11-26 MSD Italia S.r.l. Amide substituted indazoles as poly(ADP-ribose)polymerase (PARP) inhibitors
EP2336120A1 (en) 2007-01-10 2011-06-22 Istituto di ricerche di Biologia Molecolare P. Angeletti S.R.L. Combinations containing amide substituted indazoles as poly(ADP-ribose)polymerase (PARP) inhibitors
WO2008090117A1 (en) 2007-01-24 2008-07-31 Glaxo Group Limited Pharmaceutical compositions comprising 3, 5-diamin0-6- (2, 3-dichl0phenyl) -l, 2, 4-triazine or r (-) -2, 4-diamino-5- (2, 3-dichlorophenyl) -6-fluoromethyl pyrimidine and an nk1
WO2008120653A1 (en) 2007-04-02 2008-10-09 Banyu Pharmaceutical Co., Ltd. Indoledione derivative
EP3103791A1 (en) 2007-06-27 2016-12-14 Merck Sharp & Dohme Corp. 4-carboxybenzylamino derivatives as histone deacetylase inhibitors
WO2009002495A1 (en) 2007-06-27 2008-12-31 Merck & Co., Inc. 4-carboxybenzylamino derivatives as histone deacetylase inhibitors
WO2009111354A2 (en) 2008-03-03 2009-09-11 Tiger Pharmatech Tyrosine kinase inhibitors
US9498476B2 (en) 2008-06-04 2016-11-22 Albany Molecular Research, Inc. Crystalline form of 6-[(4S)-2-methyl-4-(2-naphthyl)-1,2,3,4-tetrahydroisoquinolin-7-yl]pyridazin-3-amine
US9156812B2 (en) 2008-06-04 2015-10-13 Bristol-Myers Squibb Company Crystalline form of 6-[(4S)-2-methyl-4-(2-naphthyl)-1,2,3,4-tetrahydroisoquinolin-7-yl]pyridazin-3-amine
WO2010000073A1 (en) 2008-07-03 2010-01-07 Neuraxon, Inc. Benzoxazines, benzothiazines, and related compounds having nos inhibitory activity
WO2010114780A1 (en) 2009-04-01 2010-10-07 Merck Sharp & Dohme Corp. Inhibitors of akt activity
WO2010132487A1 (en) 2009-05-12 2010-11-18 Bristol-Myers Squibb Company CRYSTALLINE FORMS OF (S)-7-([1,2,4]TRIAZOLO[1,5-a]PYRIDIN-6-YL)-4-(3,4-DICHLOROHPHENYL)-1,2,3,4-TETRAHYDROISOQUINOLINE AND USE THEREOF
WO2010132442A1 (en) 2009-05-12 2010-11-18 Albany Molecular Reserch, Inc. 7-([1,2,4,]triazolo[1,5,-a]pyridin-6-yl)-4-(3,4-dichlorophenyl)-1,2,3,4- tetrahydroisoquinoline and use thereof
US9604960B2 (en) 2009-05-12 2017-03-28 Albany Molecular Research, Inc. Aryl, heteroaryl, and heterocycle substituted tetrahydroisoquinolines and use thereof
US9034899B2 (en) 2009-05-12 2015-05-19 Albany Molecular Research, Inc. Aryl, heteroaryl, and heterocycle substituted tetrahydroisoquinolines and use thereof
US9173879B2 (en) 2009-05-12 2015-11-03 Bristol-Myers Squibb Company Crystalline forms of (S)-7-([1,2,4]triazolo[1,5-a ]pyridin-6-yl)-4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydroisoquinoline and use thereof
WO2011046771A1 (en) 2009-10-14 2011-04-21 Schering Corporation SUBSTITUTED PIPERIDINES THAT INCREASE p53 ACTIVITY AND THE USES THEREOF
WO2011163330A1 (en) 2010-06-24 2011-12-29 Merck Sharp & Dohme Corp. Novel heterocyclic compounds as erk inhibitors
EP3330377A1 (en) 2010-08-02 2018-06-06 Sirna Therapeutics, Inc. Rna interference mediated inhibition of catenin (cadherin-associated protein), beta 1 (ctnnb1) gene expression using short interfering nucleic acid (sina)
WO2012018754A2 (en) 2010-08-02 2012-02-09 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF CATENIN (CADHERIN-ASSOCIATED PROTEIN), BETA 1 (CTNNB1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
EP4079856A1 (en) 2010-08-17 2022-10-26 Sirna Therapeutics, Inc. Rna interference mediated inhibition of hepatitis b virus (hbv) gene expression using short interfering nucleic acid (sina)
WO2012027236A1 (en) 2010-08-23 2012-03-01 Schering Corporation NOVEL PYRAZOLO[1,5-a]PYRIMIDINE DERIVATIVES AS mTOR INHIBITORS
WO2012030685A2 (en) 2010-09-01 2012-03-08 Schering Corporation Indazole derivatives useful as erk inhibitors
WO2012036997A1 (en) 2010-09-16 2012-03-22 Schering Corporation Fused pyrazole derivatives as novel erk inhibitors
EP3327125A1 (en) 2010-10-29 2018-05-30 Sirna Therapeutics, Inc. Rna interference mediated inhibition of gene expression using short interfering nucleic acids (sina)
EP3766975A1 (en) 2010-10-29 2021-01-20 Sirna Therapeutics, Inc. Rna interference mediated inhibition of gene expression using short interfering nucleic acid (sina)
WO2012087772A1 (en) 2010-12-21 2012-06-28 Schering Corporation Indazole derivatives useful as erk inhibitors
WO2012145471A1 (en) 2011-04-21 2012-10-26 Merck Sharp & Dohme Corp. Insulin-like growth factor-1 receptor inhibitors
WO2013063214A1 (en) 2011-10-27 2013-05-02 Merck Sharp & Dohme Corp. Novel compounds that are erk inhibitors
WO2013165816A2 (en) 2012-05-02 2013-11-07 Merck Sharp & Dohme Corp. SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS
EP3919620A1 (en) 2012-05-02 2021-12-08 Sirna Therapeutics, Inc. Short interfering nucleic acid (sina) compositions
WO2014052563A2 (en) 2012-09-28 2014-04-03 Merck Sharp & Dohme Corp. Novel compounds that are erk inhibitors
WO2014085216A1 (en) 2012-11-28 2014-06-05 Merck Sharp & Dohme Corp. Compositions and methods for treating cancer
WO2014100065A1 (en) 2012-12-20 2014-06-26 Merck Sharp & Dohme Corp. Substituted imidazopyridines as hdm2 inhibitors
WO2014120748A1 (en) 2013-01-30 2014-08-07 Merck Sharp & Dohme Corp. 2,6,7,8 substituted purines as hdm2 inhibitors
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
WO2019094311A1 (en) 2017-11-08 2019-05-16 Merck Sharp & Dohme Corp. Prmt5 inhibitors
WO2020033282A1 (en) 2018-08-07 2020-02-13 Merck Sharp & Dohme Corp. Prmt5 inhibitors
WO2020033284A1 (en) 2018-08-07 2020-02-13 Merck Sharp & Dohme Corp. Prmt5 inhibitors

Also Published As

Publication number Publication date
US5495047A (en) 1996-02-27

Similar Documents

Publication Publication Date Title
US5495047A (en) Fused tricyclic compounds, pharmaceutical compositions containing them and their use in therapy
US5610183A (en) Phenylglycine derivatives pharmaceutical compositions containing them and their use in therapy
US5629347A (en) Aromatic compounds, pharmaceutical compositions containing them and their use in therapy
US5334606A (en) Oxazolidindione substituted indole derivatives
US5472978A (en) Aromatic compounds, pharmaceutical compositions containing them and their use in therapy
US5328927A (en) Hetercyclic compounds, processes for their preparation and pharmaceutical compositions containing them
US5633281A (en) 3,3-diphenyl prop-2-yl amino acid derivatives and their use as tachykinin antagonists
EP0520555B1 (en) Azabicyclic compounds, pharmaceutical compositions containing them and their use in therapy
US5633266A (en) Azacyclic compounds compositions containing them and their use as tachykinin antagonists
US5256671A (en) Azabicyclic compounds, pharmaceutical compositions containing them and their use in therapy
US5459270A (en) Azacyclic compounds, processes for their preparation and pharmaceutical compositions containing them
US5674889A (en) Aromatic compounds, compositions containing them and their use in therapy
US5554633A (en) Substituted amines as tachykinin receptor antagonists
US5668153A (en) Piperidine derivatives and their use as therapeutic agents
GB2271774A (en) Piperazine derivatives
GB2266529A (en) Tetrahydroisoquinoline derivatives
AU3642995A (en) Substituted aryl piperazines as neurokinin antagonists
GB2269170A (en) Azatricyclic tachykinin antagonists

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): CA JP US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IT LU MC NL SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i

Free format text: IN PAT.BUL.03/93 UNDER INID (51) "IPC" DELETE "A61K 31/135, 31/165, 31/44, 31/35, 31/38"

WWE Wipo information: entry into national phase

Ref document number: 08175432

Country of ref document: US

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: CA