WO2001081312A2 - Method of treatment using phenyl and biaryl derivatives as prostaglandin e inhibitors and compounds useful therefore - Google Patents

Method of treatment using phenyl and biaryl derivatives as prostaglandin e inhibitors and compounds useful therefore Download PDF

Info

Publication number
WO2001081312A2
WO2001081312A2 PCT/CA2001/000563 CA0100563W WO0181312A2 WO 2001081312 A2 WO2001081312 A2 WO 2001081312A2 CA 0100563 W CA0100563 W CA 0100563W WO 0181312 A2 WO0181312 A2 WO 0181312A2
Authority
WO
WIPO (PCT)
Prior art keywords
prostaglandin
compound
pain
substituted
mediated disease
Prior art date
Application number
PCT/CA2001/000563
Other languages
French (fr)
Other versions
WO2001081312A3 (en
Inventor
Michel Gallant
Nicholas Lachance
Marc Labelle
Robert Zamboni
Helene Juteau
Yves Gareau
Patrick Lacombe
Original Assignee
Merck Frosst Canada & Co.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merck Frosst Canada & Co. filed Critical Merck Frosst Canada & Co.
Priority to EP01927526A priority Critical patent/EP1278734A2/en
Priority to JP2001578407A priority patent/JP2003531194A/en
Priority to AU2001254555A priority patent/AU2001254555A1/en
Priority to CA002405170A priority patent/CA2405170A1/en
Publication of WO2001081312A2 publication Critical patent/WO2001081312A2/en
Publication of WO2001081312A3 publication Critical patent/WO2001081312A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/62Oxygen or sulfur atoms
    • C07D213/63One oxygen atom
    • C07D213/64One oxygen atom attached in position 2 or 6
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C57/00Unsaturated compounds having carboxyl groups bound to acyclic carbon atoms
    • C07C57/30Unsaturated compounds having carboxyl groups bound to acyclic carbon atoms containing six-membered aromatic rings
    • C07C57/38Unsaturated compounds having carboxyl groups bound to acyclic carbon atoms containing six-membered aromatic rings polycyclic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C59/00Compounds having carboxyl groups bound to acyclic carbon atoms and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups
    • C07C59/40Unsaturated compounds
    • C07C59/58Unsaturated compounds containing ether groups, groups, groups, or groups
    • C07C59/64Unsaturated compounds containing ether groups, groups, groups, or groups containing six-membered aromatic rings
    • C07C59/66Unsaturated compounds containing ether groups, groups, groups, or groups containing six-membered aromatic rings the non-carboxylic part of the ether containing six-membered aromatic rings
    • C07C59/68Unsaturated compounds containing ether groups, groups, groups, or groups containing six-membered aromatic rings the non-carboxylic part of the ether containing six-membered aromatic rings the oxygen atom of the ether group being bound to a non-condensed six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/54Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/55Acids; Esters
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/06Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to the ring carbon atoms
    • C07D333/24Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/26Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with 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
    • C07D333/30Hetero atoms other than halogen
    • C07D333/34Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • This invention relates to methods for treating prostaglandin mediated diseases. More particularly, the compounds are antagonists of the pain and inflammatory effects of E-type prostaglandins. Additionally preferred compounds are included.
  • Two review articles describe the characterization and therapeutic relevance of the prostanoid receptors as well as the most commonly used selective agonists and antagonists: Eicosanoids: From Biotechnology to Therapeutic Applications, Folco, Samuelsson, Maclouf, and Nelo eds, Plenum Press, New York, 1996, chap. 14, 137-154 and Journal of Lipid Mediators and Cell Signalling, 1996, 14, 83-87.
  • PGE2 Prostaglandin E2
  • selective prostaglandin ligands, agonists or antagonists have anti- inflammatory, antipyretic and analgesic properties similar to a conventional non- steroidal anti-inflammatory drug, and in addition, inhibit hormone-induced uterine contractions and have anti-cancer effects.
  • These compounds have a diminished ability to induce some of the mechanism-based side effects of NSALDs which are indiscriminate cyclooxygenase inhibitors.
  • the compounds have a reduced potential for gastrointestinal toxicity, a reduced potential for renal side effects, a reduced effect on bleeding times and a lessened ability to induce asthma attacks in aspirin-sensitive asthmatic subjects.
  • a method of treating or preventing a prostaglandin E mediated disease comprises administering to a mammalian patient in need of such treatment or prevention a compound of formula I:
  • R is a group Ar as defined hereinafter;
  • Rl is hydrogen, hydroxy, Ci-6 " alkyl, Ci-6alkoxy, X(CH2)pAr, or a methylenedioxy group attached to two adjacent ring carbon atoms;
  • R2 is -(CH 2 ) x C(O)N(R4)S(O) y R5, -(CH 2 ) ⁇ S(O) y N(R4)C(O)R5, -(CH 2 ) ⁇ C(O)N(R4)C(O)R5, -(CH2) x S(O) y N(R4)S(O) y R5, -(CH2) ⁇ C ⁇ 2 4 , or tetrazol-5-yl optionally substituted by C ⁇ _6alkyl;
  • R3 is X(CH2)pAr or X(CH2)pR 4 or a group of formula (a):
  • Ar is a group of formula (b) or (c):
  • Ar is naphthyl, indolyl, pyridyl, thienyl, furyl, oxazolidinyl, oxazolyl, thiazolyl, isothiazolyl, pyrazolyl, thriazolyl, tetrazolyl, imidazolyl, imidazolidinyl, thiazolidinyl, isoxazolyl, oxadiazolyl, thiadiazolyl, mo ⁇ holinyl, piperidinyl, piperazinyl, pyrrolyl, or pyrimidinyl, all of which may be unsubstituted or substituted by one or more R ⁇ or R8 groups;
  • each B is independently -CH2- or -O-;
  • R4 is hydrogen or Ci-6alkyl
  • R is hydrogen or Ci-ioalkyl or Ar, both of which may be unsubstituted or substituted by one or two Cl, F, Br, hydroxy, XC ⁇ _5alkyl, C ⁇ _5alkyl, NO2, tetrazol-5- yl optionally substituted by Ci-6alkyl, or R5 is N(R4)2;
  • R6 is hydrogen, RlO, CO2R 11 , C ⁇ 2C(Rl0) 2 O(CO)XRH, P ⁇ 3(RH)2, SO2NRHRIO, NRHSO2R 10 , CONRHSO2R 10 , SO3RH, S(O) q Rl l, S(O) q N(Rl l)C(O)Rl0, S(O) q N(RH)S(O) q Rl0, C(O)N(RH)C(O)R10, N(Rl l)C(O)Rl0, N(Rll)2, N(RH)C(0)NRH , P(O)(ORll)Rll, CN, - C ⁇ 2(CH2)mC(O)N(R4) 2 , C(R10) 2 N(RH)2, C(0)N(R4) 2 , OR4 or tetrazolyl optionally substituted by C 1-6 alkyl;
  • R7 and R9 are independently hydrogen, RlO, OH, Ci-8 alkoxy, S(O)qRl0, N(R4)2, Br, F, I, Cl, CF3, NO2, NHCOR4, Rl2c ⁇ 2RU, -X-R13-Y, -X(CR4) p OR4, S(CH2) p C ⁇ 2H, (CH2)pX-R 13 , -X(CH2) p CONRl lS ⁇ 2R 1 0, (CH2)pXCONRl lSO2R 10 or X(CH2) p R 6 wherein each methylene group within - X(CH2)qR6 may be unsubstituted or substituted by one or two -(CH2)pAr groups; R8 is hydrogen, RlO, OH, Ci -5alkoxy, S(O) q Rl0, N(R4) 2 , Br, F, I, Cl or NHCOR4 wherein the Ci-5 alkoxy may be unsubstituted or substituted
  • RlO is hydrogen, Ar, Ci-ioalkyl, C2-10alke n yl > C2-10alkynyl, all of which may be unsubstituted or substituted by one or more OH,.CH2OH, N(R4)2 or halogen; or RlO is N(R4) 2 ;
  • RU is independently hydrogen, Ci-ioalkyl, C2-10alkenyl or C2-8alkynyl, all of which may be unsubstituted or substituted by one or more OH, N(R4)2, CO2R1 , halogen or XCi-5alkyl; or RU is (CH2)pAr;
  • Rl2 is divalent Ar, Ci-ioalkylene, Ci-ioalkylidene, C2-10 a lk en yl ene > C2-10 alkynylene, all of which may be unsubstituted or substituted by one or more of OH, CH2OH, N(R4)2 or halogen;
  • Rl3 is a bond, Ci-ioalkylene, Ci-ioalkenylene, Ci-ioalkylidene, Ci-ioalkynylene, all of which may be linear or branched, or phenylene, all of which may be unsubstituted or substituted by one or more OH, N(R4)2, COOH or halogen;
  • Rl4 is hydrogen, Ci-6 alkyl, C2-6 alkenyl or C2-7 alkynyl;
  • X is (CH2)p, O, NR4 or S(O) p ;
  • Y is CH3 or X(CH2) p Ar
  • a method of treating or preventing a prostaglandin E mediated disease comprises administering to a mammalian patient in need of such treatment or prevention a compound of formula I:
  • R is a group Ar as defined hereinafter;
  • Rl is hydrogen, hydroxy, Ci-6alkyl, C ⁇ _6alkoxy, X(CH2)pAr, or a methylenedioxy group attached to two adjacent ring carbon atoms;
  • R2 is -(CH2) x C(O)N(R4)S(O) y R5, -(CH2) ⁇ S(O) y N(R4)C(O)R5, -(CH 2 ) x C(O)N(R4)C(O)R5, -(CH 2 ) ⁇ S(O) y N(R4)S(O) y R5, -(CH2) ⁇ C02R4, or tetrazol-5-yl optionally substituted by Ci-6alkyl;
  • R3 is X(CH2) p Ar or X(CH2) p R 4 or a group of formula (a):
  • Ar is a group of formula (b) or (c):
  • Ar is naphthyl, indolyl, pyridyl, thienyl, furyl, oxazolidinyl, oxazolyl, thiazolyl, isothiazolyl, pyrazolyl, thriazolyl, tetrazolyl, imidazolyl, imidazolidinyl, thiazolidinyl, isoxazolyl, oxadiazolyl, thiadiazolyl, mo ⁇ holinyl, piperidinyl, piperazinyl, pyrrolyl, or pyrimidinyl, all of which may be unsubstituted or substituted by one or more R7 or R8 groups;
  • each B is independently -CH2- or -O-;
  • R4 is hydrogen or Ci- ⁇ alkyl
  • R5 is hydrogen or Ci-ioalkyl or Ar, both of which may be unsubstituted or substituted by one or two Cl, F, Br, hydroxy, XCi-5alkyl, Ci-5alkyl, NO2, tetrazol-5- yl optionally substituted by Ci-6alkyl, or R5 is N(R4)2;
  • R6 is hydrogen, RlO, CO2RU, C ⁇ 2C(RlO) 2 O(CO)XRH, P ⁇ 3(RH)2, SO2NRHRIO, NRHSO2RIO, CONRHSO2RIO, SO3RH, S(O) q Rl l, S(O) q N(Rl l)C(O)Rl0, S(O) q N(RH)S(O) q RlO, C(O)N(RH)C(O)Rl0, N(Rl l)C(O)Rl0, N(Rll)2, N(RH)C(0)NRH, P(O)(ORl l)Rl l, CN, - C ⁇ 2(CH2) m C(O)N(R4) 2 , C(RlO) 2 N(RH)2, C(O)N(R4) 2I OR4 or tetrazolyl optionally substituted by Cl-6 alkyl;
  • R7 and R9 are independently hydrogen, RlO, OH, C1 -8 alkoxy, S(O)qRl0, N(R4)2, Br, F, I, Cl, CF3, NO2, NHCOR4, Rl2c ⁇ 2RU, -X-R13-Y, -X(CR4) p OR4, S(CH 2 ) p C ⁇ 2H, (CH2) p X-Rl3, -X(CH2) p CONRl lSO2Rl°, (CH2) p XCONRl lS ⁇ 2Rl° or X(CH2) p R 6 wherein each methylene group within - X(CH2)qR6 may be unsubstituted or substituted by one or two -(CH2)pAr groups; R8 is hydrogen, RlO, OH, Ci-Salkoxy, S(O) q Rl0, N(R4) 2 , Br, F, I, Cl or NHCOR4 wherein the Ci-5 alkoxy may be
  • RlO is hydrogen, Ar, Ci-ioalkyl, C2-10 a lkenyl, C2-10 a lkynyl, all of which may be unsubstituted or substituted by one or more OH,.CH2OH, N(R4)2 or halogen; or RlO is N(R4) 2 ;
  • R 1 is independently hydrogen, Ci-ioalkyl, C2-10 a lkenyl or C2-8 a lkynyl, all of which may be unsubstituted or substituted by one or more OH, N(R4)2, CO2R14, halogen or XCi-5alkyl; or Rl 1 is (CH2)pAr;
  • Rl2 is divalent Ar, Ci-ioalkylene, C -ioalkylidene, C2-10 a lkenylene, C2-10 alkynylene, all of which may be unsubstituted or substituted by one or more of OH, CH2OH, N(R4)2 or halogen;
  • Rl3 is a bond, Ci-ioalkylene, Ci-ioalkenylene, Ci-ioalkylidene, Ci-ioalkynylene, all of which may be linear or branched, or phenylene, all of which may be unsubstituted or substituted by one or more OH, N(R4)2, COOH or halogen;
  • Rl4 is hydrogen, Ci-6 alkyl, C2-6 alkenyl or C2-7 alkynyl;
  • X is (CH2) p , O, NR4 or S(O) p ;
  • Y is CH3 or X(CH2)pAr
  • An embodiment of the invention that is of particular interest relates to a method of treating or preventing a prostaglandin mediated disease comprising administering to a mammalian patient in need of such treatment a compound of formula I in an amount which is effective for treating or preventing a prostaglandin mediated disease, wherein the prostaglandin mediated disease is selected from the group consisting of:
  • coagulation disorders selected from hypoprothrombinemia, haemophilia and other bleeding problems;
  • kidney disease (14) kidney disease; (15) thrombosis;
  • Another embodiment of the invention is a method of treating or preventing a prostaglandin mediated disease comprising administering to a mammalian patient in need of such treatment a compound of formula I in an amount which is effective for treating or preventing a prostaglandin mediated disease, wherein the prostaglandin mediated disease is selected from the group consisting of: pain, fever or inflammation associated with rheumatic fever, influenza or other viral infections, common cold, low back and neck pain, skeletal pain, post-partum pain, dysmenorrhea, headache, migraine, toothache, sprains and strains, myositis, neuralgia, synovitis, arthritis, including rheumatoid arthritis, degenerative joint diseases (osteoarthritis), gout and ankylosing spondylitis, bursitis, burns including radiation and corrosive chemical injuries, sunburns, pain following surgical and dental procedures as well as immune and autoimmune diseases.
  • the prostaglandin mediated disease is selected from the group
  • Another embodiment of the invention is a method of treating or preventing a prostaglandin mediated disease comprising administering to a mammalian patient in need of such treatment a compound of formula I in an amount which is effective for treating or preventing a prostaglandin mediated disease, wherein the prostaglandin mediated disease is pain, fever or inflammation associated with dysmenorrhea.
  • Another embodiment of the invention is a method of treating or preventing a prostaglandin mediated disease comprising administering to a mammalian patient in need of such treatment a compound of formula I in an amount which is effective for treating or preventing a prostaglandin mediated disease, wherein the compound is co-administered with other agents or ingredients.
  • Another embodiment of the invention is a method of treating or preventing a prostaglandin mediated disease comprising administering to a mammalian patient in need of such treatment a compound of formula I in an amount which is effective for treating or preventing a prostaglandin mediated disease, wherein the compound is co-administered with another agent or ingredient selected from the group consisting of:
  • an analgesic selected from acetaminophen, phenacetin, aspirin, a narcotic;
  • a decongestant selected from phenylephrine, phenylpropanolamine, pseudophedrine, oxymetazoline, ephinephrine, naphazoline, xylometazoline, propylhexedrine, or levo-desoxyephedrine;
  • an antiitussive selected from codeine, hydrocodone, caramiphen, carbetapentane and dextrametho ⁇ han;
  • prostaglandin ligand selected from misoprostol, enprostil, rioprostil, ornoprostol and rosaprostol; a diuretic; and
  • COX-2 inhibitors are disclosed in U.S. Patent Nos. 5,474,995; 5,633,272; and 5,466,823; and in WO 96/25405, WO 97/38986, WO 98/03484, WO 97/14691, and WO 95/0051.
  • Another embodiment of the invention is a method of treating or preventing a prostaglandin mediated disease comprising administering to a mammalian patient in need of such treatment a compound of formula I in an amount which is effective for treating or preventing a prostaglandin mediated disease, wherein the compound is co-administered with a cyclooxygenase-2 selective nonsteroidal anti- inflammatory drug or a conventional nonsteroidal anti-inflammatory drug.
  • Another embodiment of the invention is a method of treating or preventing a prostaglandin mediated disease comprising administering to a mammalian patient in need of such treatment a compound of formula I in an amount which is effective for treating or preventing a prostaglandin mediated disease, wherein the compound is co-administered with a conventional nonsteroidal anti-inflammatory drug selected from the group consisting of: aspirin, ibuprofen, naproxen, and ketoprofen.
  • a conventional nonsteroidal anti-inflammatory drug selected from the group consisting of: aspirin, ibuprofen, naproxen, and ketoprofen.
  • Another embodiment of the invention is a method of treating or preventing a prostaglandin mediated disease comprising administering to a mammalian patient in need of such treatment a compound of formula I in an amount which is effective for treating or preventing a prostaglandin mediated disease, wherein the compound is co-administered with a cyclooxygenase-2 selective nonsteroidal anti- inflammatory drug selected from rofecoxib and celecoxib.
  • a cyclooxygenase-2 selective nonsteroidal anti- inflammatory drug selected from rofecoxib and celecoxib.
  • Additional preferred species for use in treating prostaglandin mediated diseases or conditions include the following:
  • AT-BN 2,2 -azobisisobutyronitrile
  • Bn benzyl
  • DL3AL diisobutyl aluminum hydride
  • DIPHOS 1 ,2-bis(diphenylphosphino)ethane
  • EDCI l-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
  • HBBS Hanks balanced salt solution
  • HEPES N-[2-hydroxyethyl]piperazine-N'-[2-ethanesulfonic acid]
  • KHMDS potassium hexamethyldisilazane
  • LPS lipopolysaccharide
  • m-CPBA metachloroperbenzoic acid
  • PCC pyridinium chlorochromate
  • Ph phenyl
  • PPTS pyridinium p-toluenesulfonate
  • pTSA p-toluenesulfonic acid
  • r.t. room temperature
  • rac. racemic
  • THF tetrahydrofuran
  • alkyl, alkenyl, and alkynyl mean linear, branched, and cyclic structures and combinations thereof.
  • alkyl includes “cycloalkyl” and “lower alkyl” and extends to cover carbon fragments having up to 20 carbon atoms.
  • alkyl groups include octyl, nonyl, undecyl, dodecyl, tridecyl, tetradecyl, pentadecyl, eicosyl, 3,7- diethyl-2,2-dimethyl-4-propylnonyl, and the like.
  • Lower alkyl includes “lower cycloalkyl” and means alkyl groups of from 1 to 7 carbon atoms. Examples of lower alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, s- and t-butyl, pentyl, hexyl, heptyl, and the like.
  • Cycloalkyl includes “lower cycloalkyl” and means a hydrocarbon, containing one or more rings of from 3 to 12 carbon atoms, with the hydrocarbon having up to a total of 20 carbon atoms.
  • Examples of cycloalkyl groups are cyclopropyl, cyclopentyl, cyclo-heptyl, aldamantyl, cyclododecylmethyl, 2-ethyl-l- bicyclo[4.4.0]decyl, and the like.
  • Lower cycloalkyl means a hydrocarbon containing one or more rings of from 3 to 7 carbon atoms, with the hydrocarbon having up to a total of 7 carbon atoms.
  • lower cycloalkyl groups are cyclopropyl, cyclopropylmethyl, cyclobutyl, 2-cyclopentylethyl, cycloheptyl, bicyclo[2.2.1]hept-2-yl, and the like.
  • alkenyl includes “cycloalkenyl” and “lower alkenyl” and means alkenyl groups of 2 to 20 carbon atoms. Examples of alkenyl groups include allyl, 5-decen-l-yl, 2-dodecen-l-yl, and the like.
  • “Lower alkenyl” includes “lower cycloalkenyl” and means alkenyl groups of 2 to 7 carbon atoms. Examples of lower alkenyl groups include vinyl, allyl, isopropenyl, pentenyl, hexenyl, heptenyl, 1-propenyl, 2-butenyl, 2-methyl-2-butenyl, and the like.
  • “Cycloalkenyl” includes “lower cycloalkenyl” and means alkenyl groups of 3 to 20 carbon atoms, which include a ring of 3 to 12 carbon atoms, and in which the alkenyl double bond may be located anywhere in the structure.
  • cycloalkenyl groups are cyclopropen-1-yl, cyclohexen-3-yl, 2-vinyladamant-l-yl, 5- methylene-dodec-1-yl, and the like.
  • “Lower cycloalkenyl” means alkenyl groups of 3 to 7 carbon atoms, which include a ring of 3 to 7 carbon atoms and in which the double bond may be located anywhere in the structure.
  • lower cycloalkenyl groups are cyclopropen-1-yl, cyclohexen-3-yl, 2-cyclopentylethen-l-yl, and the like.
  • alkynyl includes “cycloalkynyl” and “lower alkynyl” and means alkynyl groups of 2 to 20 carbon atoms. Examples of alkynyl groups are ethynyl, 2-pentadecyn-l-yl, 1-eicosyn-l-yl, and the like.
  • “Lower alkynyl” includes “lower cycloalkynyl” and means alkynyl groups of 2 to 7 carbon atoms. Examples of lower alkynyl groups include ethynyl, propargyl, 3 -methyl- 1-pentynyl, 2-heptynyl and the like.
  • “Cycloalkynyl” includes “lower cycloalkynyl” and means alkynyl groups of 5 to 20 carbon atoms, which include a ring of 3 to 20 carbon atoms.
  • the alkynyl triple bond may be located anywhere in the group, with the proviso that if it is within a ring, such a ring must be of 10 members or greater.
  • cycloalkynyl examples include cyclododecyn-3-yl, 3-cyclohexyl-l-propyn-l-yl, and the like.
  • “Lower cycloalkynyl” means alkynyl groups of 5 to 7 carbon atoms which include a ring of 3 to 5 carbon atoms.
  • Examples of lower cycloalkynyl are cyclopropylethynyl, 3-(cyclobutyl)-l-propynyl, and the like.
  • Halogen includes F, Cl, Br, and I.
  • Some of the compounds described herein contain one or more asymmetric centers and may thus give rise to diastereomers and optical isomers.
  • the present invention is meant to comprehend such possible diastereomers as well as their racemic and resolved, enantiomerically pure forms and pharmaceutically acceptable salts thereof.
  • compositions of the present invention comprise a compound of Formula I as an active ingredient or a pharmaceutically acceptable salt, thereof, and may also contain a pharmaceutically acceptable carrier and optionally other therapeutic ingredients.
  • pharmaceutically acceptable salts refers to salts prepared from pharmaceutically acceptable non-toxic bases including inorganic bases and organic bases. Salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc, and the like. Particularly preferred are the ammonium, calcium, magnesium, potassium, and sodium salts.
  • Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N-dibenzylethylenediamine, diethylamine, 2- diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N- ethyl-mo ⁇ holine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, mo ⁇ holine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like.
  • basic ion exchange resins
  • salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids.
  • acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic acid, and the like.
  • Particularly preferred are citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, and tartaric acids.
  • prophylactic or therapeutic dose of a compound of Formula I will, of course, vary with the nature and the severity of the condition to be treated and with the particular compound of Formula I and its route of administration. It will also vary according to a variety of factors including the age, weight, general health, sex, diet, time of administration, rate of excretion, drug combination and response of the individual patient. In general, the daily dose from about 0.001 mg to about 100 mg per kg body weight of a mammal, preferably 0.01 mg to about 10 mg per kg. On the other hand, it may be necessary to use dosages outside these limits in some cases.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • a formulation intended for the oral administration of humans may contain from 0.5 mg to 5 g of active agent compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95 percent of the total composition.
  • Dosage unit forms will generally contain between from about 1 mg to about 2 g of an active ingredient, typically 25 mg, 50 mg, 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 800 mg, or 1000 mg.
  • compounds of formula I may be administered orally, topically, parenterally, by inhalation spray or rectally in dosage unit formulations containing conventional non- toxic pharmaceutically acceptable carriers, adjuvants and vehicles.
  • parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques.
  • the compound of the invention is effective in the treatment of humans.
  • compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavouring agents, colouring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example, magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and abso ⁇ tion in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. They may also be coated by the technique described in the U.S. Patent 4,256,108; 4,166,452; and 4,265,874 to form osmotic therapeutic tablets for control release.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredients is mixed with water-miscible solvents such as propylene glycol, PEGs and ethanol, or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water-miscible solvents such as propylene glycol, PEGs and ethanol
  • an oil medium for example peanut oil, liquid paraffin, or olive oil.
  • Aqueous suspensions contain the active material in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monoole
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl, p-hydroxybenzoate, one or more colouring agents, one or more flavouring agents, and one or more sweetening agents, such as sucrose, saccharin or aspartame.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavouring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent e.g., kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol,
  • the pharmaceutical compositions of the invention may also be in the form of an oil-in-water emulsion.
  • the oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures of these.
  • Suitable emulsifying agents may be naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening and flavouring agents.
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavouring and colouring agents.
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1,3-butane diol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. Cosolvents such as ethanol, propylene glycol or polyethylene glycols may also be used.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this pu ⁇ ose any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • compositions may also be administered in the form of suppositories for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ambient temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • a suitable non-irritating excipient which is solid at ambient temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient which is solid at ambient temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials are cocoa butter and polyethylene glycols.
  • creams, ointments, gels, solutions or suspensions, etc., containing the compound of formula I are employed.
  • Topical formulations may generally be comprised of a pharmaceutical carrier, cosolvent, emulsifier, penetration enhancer, preservative system, and emollient.
  • prostaglandin receptors The ability of the compounds of formula I to interact with prostaglandin receptors makes them useful for preventing or reversing undesirable symptoms caused by prostaglandins in a mammalian, especially human subject.
  • This mimicking or antagonism of the actions of prostaglandins indicates that the compounds and pharmaceutical compositions thereof are useful to treat, prevent, or ameliorate in mammals and especially in humans: Pain, fever and inflammation of a variety of conditions including rheumatic fever, symptoms associated with influenza or other viral infections, common cold, low back and neck pain, skeletal pain, post- partum pain, dysmenorrhea, headache, migraine, toothache, sprains and strains, myositis, neuralgia, synovitis, arthritis, including rheumatoid arthritis, degenerative joint diseases (osteoarthritis), gout and ankylosing spondylitis, bursitis, burns including radiation and corrosive chemical injuries, sunburns, pain following surgical and dental procedures as well
  • Such a compound may inhibit cellular neoplastic transformations and metastic tumor growth and hence can be used in the treatment of cancer.
  • Compounds of formula I may also be of use in the treatment and/or prevention prostaglandin-mediated proliferation disorders such as may occur in diabetic retinopathy and tumor angiogenesis.
  • Compounds of formula I will also inhibit prostanoid-induced smooth muscle contraction by antagonizing contractile prostanoids or mimicking relaxing prostanoids and hence may be use in the treatment of dysmenorrhea, premature labor, asthma and eosinophil related disorders.
  • a compound of formula I will prove useful as an alternative to conventional nonsteroidal anti-inflammatory drugs (NS AID'S) particularly where such non-steroidal anti-inflammatory drugs may be contraindicated such as in patients with peptic ulcers, gastritis, regional enteritis, ulcerative colitis, diverticulitis or with a recurrent history of gastrointestinal lesions; GI bleeding, coagulation disorders including anemia such as hypoprothrombinemia, haemophilia or other bleeding problems; kidney disease; thrombosis, occlusive vascular diseases; those prior to surgery or taking anti- coagulants.
  • Compounds of formula I will also be useful as a cytoprotective agent for patients under chemotherapy.
  • the invention encompasses pharmaceutical compositions for treating prostaglandin E2 mediated diseases as defined above comprising a non-toxic therapeutically effective amount of the compound of formula I as defined above and one or more ingredients such as another pain reliever including acetaminophen or phenacetin; a COX-2 selective inhibiting agent; a conventional NSAID; a potentiator including caffeine; an H2-antagonist, aluminum or magnesium hydroxide, simethicone, a decongestant including phenylephrine, phenylpropanolamine, pseudophedrine, oxymetazoline, ephinephrine, naphazoline, xylometazoline, propylhexedrine, or levo-desoxyephedrine; an antiitussive including codeine, hydro
  • the invention encompasses a method of treating prostaglandin E2 mediated diseases comprising: administration to a patient in need of such treatment a non-toxic therapeutically effective amount of the compound of formula I, optionally co- administered with one or more of such ingredients as listed immediately above.
  • the acid group (AC(O)B) is introduce by Horner- Emmons (or Wittig) condensation. Suzuki' s coupling between the boronic acid 11 and the aryl dihalide 1 led to the aldehyde 12 under previously described conditions.
  • the acid unit is introduce by a Homer-Emmons reaction to afford the diaryl halide 13 followed by cross coupling with the boronic acid 1 to give the desired ester 9.
  • coupling between 12 and 2 followed by a Homer-Emmons reaction on the resulting aldehyde 14, can also afford the ester 9.
  • the compounds were purified by flash chromatography on silica gel, recrystallization and/or swish (suspension in a solvent followed by filtration of the solid); The course of reactions was followed by thin layer chromatography
  • Step 4 3-r2-(3-Bromophenyl)phenyllpropanoic acid:
  • Step 5 Methyl 3-f 2-r3-(4.4.5.5-tetramethyl-1.3.2-dioxaborolan-2-yl)phenyllphenyll propanoate: After esterification of the previous acid (1.72 g, 5.64 mmol) in ether
  • Step 7 Methyl 3-(2-(3-r5-chloro-2-(phenylmethoxy)-3-pyridyl1phenyl)phenyl) propanoate:
  • Step 8 3-(2-(3-r5-Chloro-2-(phenyImethoxy)-3-pyridyl1phenyl)phenyl)propanoic acid :
  • Step 1 Ethyl (E)-3-(2- ( 3-r2-(PH ⁇ NYLM ⁇ THOXY)PH ⁇ NYLlPH ⁇ NYL)PH ⁇ NYL) PROP-2-ENOIC ACID (7)
  • Step 1 Ethyl (E)-3-(2-
  • Step 2 (E)-3-(2- ⁇ 3-r2-(Phenylmethoxy)phenyl]phenyl ⁇ phenyl)prop-2-enoic acid :
  • Step 2 2- ⁇ 5-r2-(Phenylmethoxy)phenyl1-3-pyridyllbenzaldehyde:
  • step 1 a mixture of the previous bromide (507 mg, 1.93 mmol, stepl), 2-(phenylmethoxy)benzeneboronic acid (638 mg, 2.80 mmol), 2M Na 2 CO 3 (3.5 mL) and (Ph 3 P) 4 Pd (102 mg, 0.09 mmol) in DME (10 mL) was heated to 80 °C for 6 h. Purification by flash chromatography (To EtOAc, 9:1) provided the desired material as a yellow oil (642 mg, 91%).
  • Step 3 (E)- 3-(2-
  • step 2 a mixture of the previous aldehyde (640 mg, 1.75 mmol, step 2), triethyl phosphonoacetate (420 uL, 2.12 mmol) and NaH (2.7 mmol) in Tol (6 mL) was stirred at it for 6 h.
  • the crude material was not purified.
  • the crude ester was hydrolyzed in THF:MeOH:2N LiOH (6 mL:3 mL:3 mL).
  • Step 2 l-Bromo-4-r3-(3-methylphenoxy)propoxylbenzene :
  • Step 3 7ert-butyl 3-(2-f4-r3-(3-methylphenoxy)propoxy1phenyl jphenyl) propanoate
  • Step 4 Tert-butyl 2-methyl-3-(2- ⁇ 4-r3-(3-methylphenoxy)propoxy1phenyl) phenyl )propanoate :
  • a solution of the previous ester (2.76 g, 6.17 mmol) in THF (10 mL) was added to a solution of lithium N-isopropylcyclohexylamide (56 mL; 0.14 M; 7.8 mmol) in THF at -78 °C then 30 min later, iodomethane (1.3 mL, 21 mmol).
  • the solution was warmed slowly (1.5 h) at 0 °C, then poured in IN HCl and extracted with EtOAc (2x).
  • Step 5 2-Methyl-3-(2- ⁇ 4-r3-(3-methylphenoxy)propoxy1phenyl ⁇ phenyl)propanoic acid:
  • Step 2 (3-r3-(3-Bromophenoxy)propoxy]phenyl ⁇ methan-l-ol :
  • Step 4 7ert-butyl 3-[2-(3- ⁇ 3-r3-(hydroxymethyl)phenoxy1propoxy)phenyl) phenyl] propanoate :
  • Step 5 7ert-butyl 3-(2-(3-r3-(3-formylphenoxy)propoxy1phenyl
  • Step 6 Tert-butyl (E)-3-(2-r3-(3- ⁇ 3-r2-(7-chloro-2- ⁇ uinolyl)vinyl1phenoxyl propoxy) phenyll phenyl) propanoate : To a suspension of (7-chloroquinolin-2-yl)methyl triphenylphosponium bromide (1.71 g; 3.3 mmol) in 15 mL THF at -78°C was added dropwise t-BuOK (3.6 mL; 1.0 M; 3.6mmol). The mixture was stirred at -78°C for 15 min, at 0 °C for 30 min then was cooled down to -78 °C.
  • Step 7 (E)-3-(2-r3-(3-f 3-r2-(7-chloro-2-quinolyl)vinyl1phenoxy ⁇ propoxy) phenyll phenyl ⁇ propanoic acid :
  • Step 8 (E)-3-(2-r3-(3- ⁇ 3-r2-(7-chloro(2-quinolyl))vinyl1phenoxylpropoxy) phenyllphenyl ) -N-(2-thienylsulfonyl)propanamide:
  • Step 1 (E)-3-(2-(3-r2-(7-CHLORO(2-OUINOLYL))VINYL1PHENYL)PHENYL)-N-(2- THIE ⁇ YLSULFO ⁇ YL)PROP-2-E ⁇ AMIDE (103)
  • Step 1 (E)-2-
  • Step 2 Ethyl (E)-3-(2- ⁇ 3-r2-(7-chloro-2-quinolyl)vinyl1phenyl)phenyl)prop-2- enoate:
  • Step 3 (E)-3-(2-f 3-r2-(7-chloro-2-quinolyl)vinyl1phenyl
  • 1,4-dioxane was added 4 mL of NaOH (10 M). The mixture was stirred at 90°C for 12h, cooled down and acidified using HCl 10%. The residual precipitate ( HCl salt of the quinoline) was filtered, washed with ⁇ t 2 O and dried in vacuo to yield the title compound (3.5 g; 97%) which was used without further purification.
  • 1H NMR (dmso-d ⁇ ) ⁇ 6.49 (IH, d), 7.28 (IH, d), 7.4-8.0 (14H, m), 8.38 (IH, d).
  • Step 4 (E)-3-(2-(3-r2-(7-chloro(2-quinolyl))vinvnphenyl ⁇ phenyl)-N-(2-thienyl sulfonyl) prop-2-enamide:
  • the compounds of Formula I can be tested using the following assays to determine their prostanoid antagonist or agonist activity in vitro and in vivo and their selectivity.
  • the prostaglandin receptors investigated were DP, EP 1 ; EP 2 , EP 3 , EP 4 , FP, IP and TP.
  • HEK 293(ebna) cell line Prostanoid receptor cDNAs corresponding to full length coding sequences were subcloned into the appropriate sites of mammalian expression vectors and transfected into HEK 293(ebna) cells.
  • HEK 293(ebna) cells expressing the individual cDNAs were grown under selection and individual colonies were isolated after 2-3 weeks of growth using the cloning ring method and subsequently expanded into clonal cell lines.
  • HEK 293(ebna) cells are maintained in culture, harvested and membranes are prepared by differential centrifugation, following lysis of the cells in the presence of protease inhibitors, for use in receptor binding assays.
  • Prostanoid receptor binding assays are performed in 10 mM MES/KOH (pH 6.0) (EPs, FP and TP) or 10 mM HEPES/KOH (pH 7.4) (DP and IP), containing 1 mM EDTA, 10 mM divalent cation and the appropriate radioligand.
  • the reaction is initiated by addition of membrane protein.
  • Ligands are added in dimethylsulfoxide which is kept constant at 1 % (v/v) in all incubations.
  • Non-specific binding is determined in the presence of 1 ⁇ M of the corresponding non-radioactive prostanoid. Incubations are conducted for 60 min at room temperature or 30 °C and terminated by rapid filtration. Specific binding is calculated by subtracting non specific binding from total binding. The residual specific binding at each ligand concentration is calculated and expressed as a function of ligand concentration in order to construct sigmoidal concentration- response curves for determination of ligand affinity.
  • Prostanoid receptor agonist and antagonist assays Whole cell second messenger assays measuring stimulation (EP 2 , EP , DP and
  • IP in HEK 293(ebna) cells) or inhibition (EP 3 in human erythroleukemia (HEL) cells) of intracellular cAMP accumulation or mobilization of intracellular calcium (EPi, FP and TP in HEK 293(ebna) cells stably transfected with apo-aequorin) are performed to determine whether receptor ligands are agonists or antagonists.
  • cAMP assays cells are harvested and resuspended in HBSS containing 25 mM HEPES, pH 7.4.
  • Incubations contain 100 ⁇ M RO-20174 (phosphodiesterase type IN inhibitor, available from Biomol) and, in the case of the EP 3 inhibition assay only, 15 ⁇ M forskolin to stimulate cAMP production. Samples are incubated at 37°C for 10 min, the reaction is terminated and cAMP levels are then measured.
  • For calcium mobilization assays cells are charged with the co-factors reduced glutathione and coelenterazine, harvested and resuspended in Ham's F12 medium. Calcium mobilization is measured by monitoring luminescence provoked by calcium binding to the intracellular photoprotein aequorin. Ligands are added in dimethylsulfoxide which is kept constant at 1 % (v/v) in all incubations.
  • second messenger responses are expressed as a function of ligand concentration and both EC 50 values and the maximum response as compared to a prostanoid standard are calculated.
  • the ability of a ligand to inhibit an agonist response is determined by Schild analysis and both K B and slope values are calculated.
  • mice Female Lewis rats (body weight -146-170 g) were weighed, ear marked, and assigned to groups (a negative control group in which arthritis was not induced, a vehicle control group, a positive control group administered indomethacin at a total daily dose of 1 mg/kg and four groups administered with a test compound at total daily doses of 0.10-3.0 mg/kg) such that the body weights were equivalent within each group.
  • Six groups of 10 rats each were injected into a hind paw with 0.5 mg of Mycobacte ⁇ um butyricum in 0.1 mL of light mineral oil (adjuvant), and a negative control group of 10 rats was not injected with adjuvant.
  • Body weights, contralateral paw volumes (determined by mercury displacement plethysmography) and lateral radiographs (obtained under Ketamine and Xylazine anesthesia) were determined before (day -1) and 21 days following adjuvant injection, and primary paw volumes were determined before (day -1) and on days 4 and 21 following adjuvant injection.
  • the rats were anesthetized with an intramuscular injection of 0.03 - 0.1 mL of a combination of Ketamine (87 mg/kg) and Xylazine (13 mg/kg) for radiographs and injection of adjuvant.
  • radiographs were made of both hind paws on day 0 and day 21 using the Faxitron (45 kVp, 30 seconds) and Kodak X-OMAT TL film, and were developed in an automatic processor. Radiographs were evaluated for changes in the soft and hard tissues by an investigator who was blinded to experimental treatment. The following radiographic changes were graded numerically according to severity: increased soft issue volume (0-4), narrowing or widening of joint spaces (0-5) subchondral erosion (0-3), periosteal reaction (0-4), osteolysis (0-4) subluxation (0-3), and degenerative joint changes (0-3). Specific criteria were used to establish the numerical grade of severity for each radiographic change. The maximum possible score per foot was 26.
  • test compound at total daily doses of 0.1, 0.3, 1, and 3 mg/kg/day, indomethacin at a total daily dose of 1 mg/kg/day, or vehicle (0.5% methocel in sterile water) were administered per os b.i.d. beginning post injection of adjuvant and continuing for 21 days.
  • the compounds were prepared weekly, refrigerated in the dark until used, and vortex mixed immediately prior to administration.

Landscapes

  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Neurology (AREA)
  • Rheumatology (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Pulmonology (AREA)
  • Pain & Pain Management (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Reproductive Health (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Vascular Medicine (AREA)
  • Cardiology (AREA)
  • Dermatology (AREA)
  • Oncology (AREA)
  • Immunology (AREA)
  • Endocrinology (AREA)
  • Psychiatry (AREA)
  • Hospice & Palliative Care (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

This invention encompasses a method for the treatment or prevention of prostaglandin mediated diseases comprising administering to a mammalian patient a compound of formula (I) in an amount that is effective to treat or prevent said prostaglandin mediated disease. Novel compounds are also disclosed.

Description

TITLE OF THE INVENTION
METHOD OF TREATMENT USLNG PHENYL AND BIARYL DERIVATIVES AS
PROSTAGLANDIN E INHIBITORS AND COMPOUNDS USEFUL THEREFORE
BACKGROUND OF THE INVENTION
This invention relates to methods for treating prostaglandin mediated diseases. More particularly, the compounds are antagonists of the pain and inflammatory effects of E-type prostaglandins. Additionally preferred compounds are included. Two review articles describe the characterization and therapeutic relevance of the prostanoid receptors as well as the most commonly used selective agonists and antagonists: Eicosanoids: From Biotechnology to Therapeutic Applications, Folco, Samuelsson, Maclouf, and Nelo eds, Plenum Press, New York, 1996, chap. 14, 137-154 and Journal of Lipid Mediators and Cell Signalling, 1996, 14, 83-87. An article from The British Journal of Pharmacology (1994, 112, 735-
740) suggests that Prostaglandin E2 (PGE2) exerts allodynia through the EPi receptor subtype and hyperalgesia through EP2 and EP3 receptors in the mouse spinal cord.
Thus, selective prostaglandin ligands, agonists or antagonists, depending on which prostaglandin E receptor subtype is being considered, have anti- inflammatory, antipyretic and analgesic properties similar to a conventional non- steroidal anti-inflammatory drug, and in addition, inhibit hormone-induced uterine contractions and have anti-cancer effects. These compounds have a diminished ability to induce some of the mechanism-based side effects of NSALDs which are indiscriminate cyclooxygenase inhibitors. In particular, the compounds have a reduced potential for gastrointestinal toxicity, a reduced potential for renal side effects, a reduced effect on bleeding times and a lessened ability to induce asthma attacks in aspirin-sensitive asthmatic subjects.
In The American Physiological Society (1994, 267, R289-R-294), studies suggest that PGE2-induced hyperthermia in the rat is mediated predominantly through the EPl receptor. World patent applications WO 96/06822 (March 7, 1996), WO 96/11902 (April 25, 1996) and EP 752421-A1 (January 08, 1997) disclose compounds as being useful in the treatment of prostaglandin mediated diseases. SUMMARY OF THE INVENTION
A method of treating or preventing a prostaglandin E mediated disease is described which comprises administering to a mammalian patient in need of such treatment or prevention a compound of formula I:
Figure imgf000003_0001
wherein:
R is a group Ar as defined hereinafter;
Rl is hydrogen, hydroxy, Ci-6"alkyl, Ci-6alkoxy, X(CH2)pAr, or a methylenedioxy group attached to two adjacent ring carbon atoms;
R2 is -(CH2)xC(O)N(R4)S(O)yR5, -(CH2)χS(O)yN(R4)C(O)R5, -(CH2)χC(O)N(R4)C(O)R5, -(CH2)xS(O)yN(R4)S(O)yR5, -(CH2)χCθ2 4, or tetrazol-5-yl optionally substituted by Cι_6alkyl;
R3 is X(CH2)pAr or X(CH2)pR4 or a group of formula (a):
Figure imgf000003_0002
(a)
Ar is a group of formula (b) or (c):
Figure imgf000004_0001
or Ar is naphthyl, indolyl, pyridyl, thienyl, furyl, oxazolidinyl, oxazolyl, thiazolyl, isothiazolyl, pyrazolyl, thriazolyl, tetrazolyl, imidazolyl, imidazolidinyl, thiazolidinyl, isoxazolyl, oxadiazolyl, thiadiazolyl, moφholinyl, piperidinyl, piperazinyl, pyrrolyl, or pyrimidinyl, all of which may be unsubstituted or substituted by one or more R~ϊ or R8 groups;
A is C=O or (C(R4)2)m;
each B is independently -CH2- or -O-;
R4 is hydrogen or Ci-6alkyl;
R is hydrogen or Ci-ioalkyl or Ar, both of which may be unsubstituted or substituted by one or two Cl, F, Br, hydroxy, XCι_5alkyl, Cι_5alkyl, NO2, tetrazol-5- yl optionally substituted by Ci-6alkyl, or R5 is N(R4)2;
R6 is hydrogen, RlO, CO2R11, Cθ2C(Rl0)2O(CO)XRH, Pθ3(RH)2, SO2NRHRIO, NRHSO2R10, CONRHSO2R10, SO3RH, S(O)qRl l, S(O)qN(Rl l)C(O)Rl0, S(O)qN(RH)S(O)qRl0, C(O)N(RH)C(O)R10, N(Rl l)C(O)Rl0, N(Rll)2, N(RH)C(0)NRH , P(O)(ORll)Rll, CN, - Cθ2(CH2)mC(O)N(R4)2, C(R10)2N(RH)2, C(0)N(R4)2, OR4 or tetrazolyl optionally substituted by C 1-6 alkyl;
R7 and R9 are independently hydrogen, RlO, OH, Ci-8 alkoxy, S(O)qRl0, N(R4)2, Br, F, I, Cl, CF3, NO2, NHCOR4, Rl2cθ2RU, -X-R13-Y, -X(CR4)pOR4, S(CH2)pCθ2H, (CH2)pX-R13, -X(CH2)pCONRl lSθ2R10, (CH2)pXCONRl lSO2R10 or X(CH2)pR6 wherein each methylene group within - X(CH2)qR6 may be unsubstituted or substituted by one or two -(CH2)pAr groups; R8 is hydrogen, RlO, OH, Ci -5alkoxy, S(O)qRl0, N(R4)2, Br, F, I, Cl or NHCOR4 wherein the Ci-5 alkoxy may be unsubstituted or substituted by OH, methoxy or halogen;
RlO is hydrogen, Ar, Ci-ioalkyl, C2-10alkenyl> C2-10alkynyl, all of which may be unsubstituted or substituted by one or more OH,.CH2OH, N(R4)2 or halogen; or RlO is N(R4)2;
RU is independently hydrogen, Ci-ioalkyl, C2-10alkenyl or C2-8alkynyl, all of which may be unsubstituted or substituted by one or more OH, N(R4)2, CO2R1 , halogen or XCi-5alkyl; or RU is (CH2)pAr;
Rl2 is divalent Ar, Ci-ioalkylene, Ci-ioalkylidene, C2-10alkenylene> C2-10 alkynylene, all of which may be unsubstituted or substituted by one or more of OH, CH2OH, N(R4)2 or halogen;
Rl3 is a bond, Ci-ioalkylene, Ci-ioalkenylene, Ci-ioalkylidene, Ci-ioalkynylene, all of which may be linear or branched, or phenylene, all of which may be unsubstituted or substituted by one or more OH, N(R4)2, COOH or halogen;
Rl4 is hydrogen, Ci-6 alkyl, C2-6 alkenyl or C2-7 alkynyl;
X is (CH2)p, O, NR4 or S(O)p;
Y is CH3 or X(CH2)pAr;
q is zero, one or two; p is an integer from 0 to 6; m is 1, 2 or 3; n is 1 to 4; x is 0 to 4; y is 1 or 2; the dotted line signifies the optional presence of a bond such that it represents a single or double bond. DF.T ATT F.D DESCRIPTION
In one aspect of the invention, a method of treating or preventing a prostaglandin E mediated disease is described which comprises administering to a mammalian patient in need of such treatment or prevention a compound of formula I:
Figure imgf000006_0001
wherein:
R is a group Ar as defined hereinafter;
Rl is hydrogen, hydroxy, Ci-6alkyl, Cι_6alkoxy, X(CH2)pAr, or a methylenedioxy group attached to two adjacent ring carbon atoms;
R2 is -(CH2)xC(O)N(R4)S(O)yR5, -(CH2)χS(O)yN(R4)C(O)R5, -(CH2)xC(O)N(R4)C(O)R5, -(CH2)χS(O)yN(R4)S(O)yR5, -(CH2)χC02R4, or tetrazol-5-yl optionally substituted by Ci-6alkyl;
R3 is X(CH2)pAr or X(CH2)pR4 or a group of formula (a):
Figure imgf000006_0002
(a)
Ar is a group of formula (b) or (c):
Figure imgf000007_0001
or Ar is naphthyl, indolyl, pyridyl, thienyl, furyl, oxazolidinyl, oxazolyl, thiazolyl, isothiazolyl, pyrazolyl, thriazolyl, tetrazolyl, imidazolyl, imidazolidinyl, thiazolidinyl, isoxazolyl, oxadiazolyl, thiadiazolyl, moφholinyl, piperidinyl, piperazinyl, pyrrolyl, or pyrimidinyl, all of which may be unsubstituted or substituted by one or more R7 or R8 groups;
Figure imgf000007_0002
each B is independently -CH2- or -O-;
R4 is hydrogen or Ci-βalkyl;
R5 is hydrogen or Ci-ioalkyl or Ar, both of which may be unsubstituted or substituted by one or two Cl, F, Br, hydroxy, XCi-5alkyl, Ci-5alkyl, NO2, tetrazol-5- yl optionally substituted by Ci-6alkyl, or R5 is N(R4)2;
R6 is hydrogen, RlO, CO2RU, Cθ2C(RlO)2O(CO)XRH, Pθ3(RH)2, SO2NRHRIO, NRHSO2RIO, CONRHSO2RIO, SO3RH, S(O)qRl l, S(O)qN(Rl l)C(O)Rl0, S(O)qN(RH)S(O)qRlO, C(O)N(RH)C(O)Rl0, N(Rl l)C(O)Rl0, N(Rll)2, N(RH)C(0)NRH, P(O)(ORl l)Rl l, CN, - Cθ2(CH2)mC(O)N(R4)2, C(RlO)2N(RH)2, C(O)N(R4)2I OR4 or tetrazolyl optionally substituted by Cl-6 alkyl;
R7 and R9 are independently hydrogen, RlO, OH, C1 -8 alkoxy, S(O)qRl0, N(R4)2, Br, F, I, Cl, CF3, NO2, NHCOR4, Rl2cθ2RU, -X-R13-Y, -X(CR4)pOR4, S(CH2)pCθ2H, (CH2)pX-Rl3, -X(CH2)pCONRl lSO2Rl°, (CH2)pXCONRl lSθ2Rl° or X(CH2)pR6 wherein each methylene group within - X(CH2)qR6 may be unsubstituted or substituted by one or two -(CH2)pAr groups; R8 is hydrogen, RlO, OH, Ci-Salkoxy, S(O)qRl0, N(R4)2, Br, F, I, Cl or NHCOR4 wherein the Ci-5 alkoxy may be unsubstituted or substituted by OH, methoxy or halogen;
RlO is hydrogen, Ar, Ci-ioalkyl, C2-10alkenyl, C2-10alkynyl, all of which may be unsubstituted or substituted by one or more OH,.CH2OH, N(R4)2 or halogen; or RlO is N(R4)2;
R 1 is independently hydrogen, Ci-ioalkyl, C2-10alkenyl or C2-8alkynyl, all of which may be unsubstituted or substituted by one or more OH, N(R4)2, CO2R14, halogen or XCi-5alkyl; or Rl 1 is (CH2)pAr;
Rl2 is divalent Ar, Ci-ioalkylene, C -ioalkylidene, C2-10alkenylene, C2-10 alkynylene, all of which may be unsubstituted or substituted by one or more of OH, CH2OH, N(R4)2 or halogen;
Rl3 is a bond, Ci-ioalkylene, Ci-ioalkenylene, Ci-ioalkylidene, Ci-ioalkynylene, all of which may be linear or branched, or phenylene, all of which may be unsubstituted or substituted by one or more OH, N(R4)2, COOH or halogen;
Rl4 is hydrogen, Ci-6 alkyl, C2-6 alkenyl or C2-7 alkynyl;
X is (CH2)p, O, NR4 or S(O)p;
Y is CH3 or X(CH2)pAr;
q is zero, one or two; p is an integer from 0 to 6; m is 1, 2 or 3; n is 1 to 4; x is 0 to 4; y is 1 or 2; the dotted line signifies the optional presence of a bond such that it represents a single or double bond. An embodiment of the invention that is of particular interest relates to a method of treating or preventing a prostaglandin mediated disease comprising administering to a mammalian patient in need of such treatment a compound of formula I in an amount which is effective for treating or preventing a prostaglandin mediated disease, wherein the prostaglandin mediated disease is selected from the group consisting of:
(1) pain, fever or inflammation associated with rheumatic fever, influenza or other viral infections, common cold, low back and neck pain, skeletal pain, post-partum pain, dysmenorrhea, headache, migraine, toothache, sprains and strains, myositis, neuralgia, synovitis, arthritis, including rheumatoid arthritis, degenerative joint diseases (osteoarthritis), gout and ankylosing spondylitis, bursitis, burns including radiation and corrosive chemical injuries, sunburns, pain following surgical and dental procedures as well as immune and autoimmune diseases;
(2) cellular neoplastic transformations or metastic tumor growth; (3) diabetic retinopathy and tumor angiogenesis;
(4) prostanoid-induced smooth muscle contraction associated with dysmenorrhea, premature labor, asthma or eosinophil related disorders;
(5) Alzheimer's disease;
(6) glaucoma; (7) bone loss;
(8) osteoporosis;
(9) promotion of bone formation;
(10) Paget's disease;
(11) cytoprotection in peptic ulcers, gastritis, regional enteritis, ulcerative colitis, diverticulitis or other gastrointestinal lesions;
(12) GI bleeding and patients undergoing chemotherapy;
(13) coagulation disorders selected from hypoprothrombinemia, haemophilia and other bleeding problems;
(14) kidney disease; (15) thrombosis;
(16) occlusive vascular disease;
(17) presurgery; and
(18) anti-coagulation.
Another embodiment of the invention is a method of treating or preventing a prostaglandin mediated disease comprising administering to a mammalian patient in need of such treatment a compound of formula I in an amount which is effective for treating or preventing a prostaglandin mediated disease, wherein the prostaglandin mediated disease is selected from the group consisting of: pain, fever or inflammation associated with rheumatic fever, influenza or other viral infections, common cold, low back and neck pain, skeletal pain, post-partum pain, dysmenorrhea, headache, migraine, toothache, sprains and strains, myositis, neuralgia, synovitis, arthritis, including rheumatoid arthritis, degenerative joint diseases (osteoarthritis), gout and ankylosing spondylitis, bursitis, burns including radiation and corrosive chemical injuries, sunburns, pain following surgical and dental procedures as well as immune and autoimmune diseases.
Another embodiment of the invention is a method of treating or preventing a prostaglandin mediated disease comprising administering to a mammalian patient in need of such treatment a compound of formula I in an amount which is effective for treating or preventing a prostaglandin mediated disease, wherein the prostaglandin mediated disease is pain, fever or inflammation associated with dysmenorrhea.
Another embodiment of the invention is a method of treating or preventing a prostaglandin mediated disease comprising administering to a mammalian patient in need of such treatment a compound of formula I in an amount which is effective for treating or preventing a prostaglandin mediated disease, wherein the compound is co-administered with other agents or ingredients.
Another embodiment of the invention is a method of treating or preventing a prostaglandin mediated disease comprising administering to a mammalian patient in need of such treatment a compound of formula I in an amount which is effective for treating or preventing a prostaglandin mediated disease, wherein the compound is co-administered with another agent or ingredient selected from the group consisting of:
(1) an analgesic selected from acetaminophen, phenacetin, aspirin, a narcotic; (2) a cyclooxygenase-2 selective nonsteroidal anti-inflammatory drug or a conventional nonsteroidal anti -inflammatory drug;
(3) caffeine;
(4) an H2-antagonist;
(5) aluminum or magnesium hydroxide; (6) simethicone; (7) a decongestant selected from phenylephrine, phenylpropanolamine, pseudophedrine, oxymetazoline, ephinephrine, naphazoline, xylometazoline, propylhexedrine, or levo-desoxyephedrine;
(8) an antiitussive selected from codeine, hydrocodone, caramiphen, carbetapentane and dextramethoφhan;
(9) another prostaglandin ligand selected from misoprostol, enprostil, rioprostil, ornoprostol and rosaprostol; a diuretic; and
(10) a sedating or non-sedating antihistamine. Examples of COX-2 inhibitors are disclosed in U.S. Patent Nos. 5,474,995; 5,633,272; and 5,466,823; and in WO 96/25405, WO 97/38986, WO 98/03484, WO 97/14691, and WO 95/0051. Another embodiment of the invention is a method of treating or preventing a prostaglandin mediated disease comprising administering to a mammalian patient in need of such treatment a compound of formula I in an amount which is effective for treating or preventing a prostaglandin mediated disease, wherein the compound is co-administered with a cyclooxygenase-2 selective nonsteroidal anti- inflammatory drug or a conventional nonsteroidal anti-inflammatory drug.
Another embodiment of the invention is a method of treating or preventing a prostaglandin mediated disease comprising administering to a mammalian patient in need of such treatment a compound of formula I in an amount which is effective for treating or preventing a prostaglandin mediated disease, wherein the compound is co-administered with a conventional nonsteroidal anti-inflammatory drug selected from the group consisting of: aspirin, ibuprofen, naproxen, and ketoprofen.
Another embodiment of the invention is a method of treating or preventing a prostaglandin mediated disease comprising administering to a mammalian patient in need of such treatment a compound of formula I in an amount which is effective for treating or preventing a prostaglandin mediated disease, wherein the compound is co-administered with a cyclooxygenase-2 selective nonsteroidal anti- inflammatory drug selected from rofecoxib and celecoxib. Preferred subsets of compounds and species that are useful for the methods described herein are set forth in WO96/30358, incoφorated by reference.
Additional preferred species for use in treating prostaglandin mediated diseases or conditions include the following:
Figure imgf000012_0001
Figure imgf000013_0001
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0002
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000018_0002
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
Figure imgf000021_0001
53
Representative compounds having a sulfonamide group are shown below.
Figure imgf000021_0002
Figure imgf000022_0001
Figure imgf000023_0001
Figure imgf000024_0001
Figure imgf000025_0001
Figure imgf000026_0001
Figure imgf000027_0002
Qn = 7-chloro-quinol-2-yl 2-Ph-Et-SMe = 2-phenylethylthiomethyl 3-(3-Me-Ph-oxy)Pr-oxy = 3-(3-methylphenoxy)propyl-l-oxy
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
Definitions :
The following abbreviations have the indicated meanings:
Ac = acetyl
AT-BN = 2,2 -azobisisobutyronitrile Bn = benzyl
DL3AL = diisobutyl aluminum hydride
DIPHOS = 1 ,2-bis(diphenylphosphino)ethane
DMAP = 4-(dimethylamino)pyridine
DMF = N,N-dimethylformamide DMSO = dimethyl sulfoxide
EDCI = l-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
Et3N = triethylamine
EtOAc = ethyl acetate
HBBS = Hanks balanced salt solution HEPES = N-[2-hydroxyethyl]piperazine-N'-[2-ethanesulfonic acid]
Hex = hexanes
KHMDS = potassium hexamethyldisilazane
LDA = lithium diisopropylamide
LPS = lipopolysaccharide m-CPBA = metachloroperbenzoic acid
MES = 2-[N-moφholino]ethanesulfonic acid
Ms = methanesulfonyl = mesyl
MsO = methanesulfonate = mesylate
NBS = N-bromosuccinimide NSAID = non-steroidal anti-inflammatory drug
PCC = pyridinium chlorochromate
PDC = pyridinium dichromate
Ph = phenyl
PPTS = pyridinium p-toluenesulfonate pTSA = p-toluenesulfonic acid r.t. = room temperature rac. = racemic
TLC = thin layer chromatography
Tf = trifluoromethanesulfonyl = triflyl TfO = trifluoromethanesulfonate = triflate THF = tetrahydrofuran
TLC = thin layer chromatography
Ts = p-toluenesulfonyl = tosyl
TsO = p-toluenesύlfonate = tosylate C3H5 = allyl
Alkyl group abbreviations
Me = methyl
Et = ethyl n-Pr = normal propyl i-Pr = isopropyl n-Bu = normal butyl i-Bu = isobutyl s-Bu = secondary butyl t-Bu = tertiary butyl c-Pr = cyclopropyl c-Bu = cyclobutyl c-Pen = cyclopentyl c-Hex = cyclohexyl The terms alkyl, alkenyl, and alkynyl mean linear, branched, and cyclic structures and combinations thereof.
The term "alkyl" includes "cycloalkyl" and "lower alkyl" and extends to cover carbon fragments having up to 20 carbon atoms. Examples of alkyl groups include octyl, nonyl, undecyl, dodecyl, tridecyl, tetradecyl, pentadecyl, eicosyl, 3,7- diethyl-2,2-dimethyl-4-propylnonyl, and the like.
"Lower alkyl" includes "lower cycloalkyl" and means alkyl groups of from 1 to 7 carbon atoms. Examples of lower alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, s- and t-butyl, pentyl, hexyl, heptyl, and the like.
"Cycloalkyl" includes "lower cycloalkyl" and means a hydrocarbon, containing one or more rings of from 3 to 12 carbon atoms, with the hydrocarbon having up to a total of 20 carbon atoms. Examples of cycloalkyl groups are cyclopropyl, cyclopentyl, cyclo-heptyl, aldamantyl, cyclododecylmethyl, 2-ethyl-l- bicyclo[4.4.0]decyl, and the like.
"Lower cycloalkyl" means a hydrocarbon containing one or more rings of from 3 to 7 carbon atoms, with the hydrocarbon having up to a total of 7 carbon atoms. Examples of lower cycloalkyl groups are cyclopropyl, cyclopropylmethyl, cyclobutyl, 2-cyclopentylethyl, cycloheptyl, bicyclo[2.2.1]hept-2-yl, and the like.
The term "alkenyl" includes "cycloalkenyl" and "lower alkenyl" and means alkenyl groups of 2 to 20 carbon atoms. Examples of alkenyl groups include allyl, 5-decen-l-yl, 2-dodecen-l-yl, and the like.
"Lower alkenyl" includes "lower cycloalkenyl" and means alkenyl groups of 2 to 7 carbon atoms. Examples of lower alkenyl groups include vinyl, allyl, isopropenyl, pentenyl, hexenyl, heptenyl, 1-propenyl, 2-butenyl, 2-methyl-2-butenyl, and the like. "Cycloalkenyl" includes "lower cycloalkenyl" and means alkenyl groups of 3 to 20 carbon atoms, which include a ring of 3 to 12 carbon atoms, and in which the alkenyl double bond may be located anywhere in the structure. Examples of cycloalkenyl groups are cyclopropen-1-yl, cyclohexen-3-yl, 2-vinyladamant-l-yl, 5- methylene-dodec-1-yl, and the like. "Lower cycloalkenyl" means alkenyl groups of 3 to 7 carbon atoms, which include a ring of 3 to 7 carbon atoms and in which the double bond may be located anywhere in the structure. Examples of lower cycloalkenyl groups are cyclopropen-1-yl, cyclohexen-3-yl, 2-cyclopentylethen-l-yl, and the like.
The term "alkynyl" includes "cycloalkynyl" and "lower alkynyl" and means alkynyl groups of 2 to 20 carbon atoms. Examples of alkynyl groups are ethynyl, 2-pentadecyn-l-yl, 1-eicosyn-l-yl, and the like.
"Lower alkynyl" includes "lower cycloalkynyl" and means alkynyl groups of 2 to 7 carbon atoms. Examples of lower alkynyl groups include ethynyl, propargyl, 3 -methyl- 1-pentynyl, 2-heptynyl and the like. "Cycloalkynyl" includes "lower cycloalkynyl" and means alkynyl groups of 5 to 20 carbon atoms, which include a ring of 3 to 20 carbon atoms. The alkynyl triple bond may be located anywhere in the group, with the proviso that if it is within a ring, such a ring must be of 10 members or greater. Examples of cycloalkynyl are cyclododecyn-3-yl, 3-cyclohexyl-l-propyn-l-yl, and the like. "Lower cycloalkynyl" means alkynyl groups of 5 to 7 carbon atoms which include a ring of 3 to 5 carbon atoms. Examples of lower cycloalkynyl are cyclopropylethynyl, 3-(cyclobutyl)-l-propynyl, and the like. Halogen includes F, Cl, Br, and I.
It is intended that the definition of any substituent in a particular molecule be independent of its definition elsewhere in the molecule. Optical Isomers - Diastereomers - Geometric Isomers
Some of the compounds described herein contain one or more asymmetric centers and may thus give rise to diastereomers and optical isomers. The present invention is meant to comprehend such possible diastereomers as well as their racemic and resolved, enantiomerically pure forms and pharmaceutically acceptable salts thereof.
Some of the compounds described herein contain olefinic double bonds, and unless specified otherwise, are meant to include both E and Z geometric isomers.
Salts
The pharmaceutical compositions of the present invention comprise a compound of Formula I as an active ingredient or a pharmaceutically acceptable salt, thereof, and may also contain a pharmaceutically acceptable carrier and optionally other therapeutic ingredients. The term "pharmaceutically acceptable salts" refers to salts prepared from pharmaceutically acceptable non-toxic bases including inorganic bases and organic bases. Salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc, and the like. Particularly preferred are the ammonium, calcium, magnesium, potassium, and sodium salts. Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N-dibenzylethylenediamine, diethylamine, 2- diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N- ethyl-moφholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, moφholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like.
When the compound of the present invention is basic, salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids. Such acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic acid, and the like. Particularly preferred are citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, and tartaric acids.
It will be understood that in the discussion of methods of treatment which follows, references to the compounds of Formula I are meant to also include the pharmaceutically acceptable salts.
Dose Ranges
The magnitude of prophylactic or therapeutic dose of a compound of Formula I will, of course, vary with the nature and the severity of the condition to be treated and with the particular compound of Formula I and its route of administration. It will also vary according to a variety of factors including the age, weight, general health, sex, diet, time of administration, rate of excretion, drug combination and response of the individual patient. In general, the daily dose from about 0.001 mg to about 100 mg per kg body weight of a mammal, preferably 0.01 mg to about 10 mg per kg. On the other hand, it may be necessary to use dosages outside these limits in some cases.
The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. For example, a formulation intended for the oral administration of humans may contain from 0.5 mg to 5 g of active agent compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95 percent of the total composition. Dosage unit forms will generally contain between from about 1 mg to about 2 g of an active ingredient, typically 25 mg, 50 mg, 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 800 mg, or 1000 mg.
Pharmaceutical Compositions
For the treatment of any of the prostanoid mediated diseases compounds of formula I may be administered orally, topically, parenterally, by inhalation spray or rectally in dosage unit formulations containing conventional non- toxic pharmaceutically acceptable carriers, adjuvants and vehicles. The term parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques. In addition to the treatment of warm-blooded animals such as mice, rats, horses, cattle, sheep, dogs, cats, etc., the compound of the invention is effective in the treatment of humans.
The pharmaceutical compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavouring agents, colouring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example, magnesium stearate, stearic acid or talc. The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absoφtion in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. They may also be coated by the technique described in the U.S. Patent 4,256,108; 4,166,452; and 4,265,874 to form osmotic therapeutic tablets for control release.
Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredients is mixed with water-miscible solvents such as propylene glycol, PEGs and ethanol, or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
Aqueous suspensions contain the active material in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl, p-hydroxybenzoate, one or more colouring agents, one or more flavouring agents, and one or more sweetening agents, such as sucrose, saccharin or aspartame. Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavouring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavouring and colouring agents, may also be present.
The pharmaceutical compositions of the invention may also be in the form of an oil-in-water emulsion. The oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures of these. Suitable emulsifying agents may be naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening and flavouring agents. Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavouring and colouring agents. The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1,3-butane diol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. Cosolvents such as ethanol, propylene glycol or polyethylene glycols may also be used. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this puφose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.
Compounds of formula I may also be administered in the form of suppositories for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ambient temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials are cocoa butter and polyethylene glycols. For topical use, creams, ointments, gels, solutions or suspensions, etc., containing the compound of formula I are employed. (For puφoses of this application, topical application shall include mouth washes and gargles.) Topical formulations may generally be comprised of a pharmaceutical carrier, cosolvent, emulsifier, penetration enhancer, preservative system, and emollient.
Utilities
The ability of the compounds of formula I to interact with prostaglandin receptors makes them useful for preventing or reversing undesirable symptoms caused by prostaglandins in a mammalian, especially human subject. This mimicking or antagonism of the actions of prostaglandins indicates that the compounds and pharmaceutical compositions thereof are useful to treat, prevent, or ameliorate in mammals and especially in humans: Pain, fever and inflammation of a variety of conditions including rheumatic fever, symptoms associated with influenza or other viral infections, common cold, low back and neck pain, skeletal pain, post- partum pain, dysmenorrhea, headache, migraine, toothache, sprains and strains, myositis, neuralgia, synovitis, arthritis, including rheumatoid arthritis, degenerative joint diseases (osteoarthritis), gout and ankylosing spondylitis, bursitis, burns including radiation and corrosive chemical injuries, sunburns, pain following surgical and dental procedures as well as immune and autoimmune diseases. In addition, such a compound may inhibit cellular neoplastic transformations and metastic tumor growth and hence can be used in the treatment of cancer. Compounds of formula I may also be of use in the treatment and/or prevention prostaglandin-mediated proliferation disorders such as may occur in diabetic retinopathy and tumor angiogenesis. Compounds of formula I will also inhibit prostanoid-induced smooth muscle contraction by antagonizing contractile prostanoids or mimicking relaxing prostanoids and hence may be use in the treatment of dysmenorrhea, premature labor, asthma and eosinophil related disorders. It will also be of use in the treatment of Alzheimer's disease, the treatment of glaucoma, for the prevention of bone loss (treatment of osteoporosis) and for the promotion of bone formation (treatment of fractures) and other bone diseases such as Paget's disease.
By virtue of its prostanoid or prostanoid antagonist activity, a compound of formula I will prove useful as an alternative to conventional nonsteroidal anti-inflammatory drugs (NS AID'S) particularly where such non-steroidal anti-inflammatory drugs may be contraindicated such as in patients with peptic ulcers, gastritis, regional enteritis, ulcerative colitis, diverticulitis or with a recurrent history of gastrointestinal lesions; GI bleeding, coagulation disorders including anemia such as hypoprothrombinemia, haemophilia or other bleeding problems; kidney disease; thrombosis, occlusive vascular diseases; those prior to surgery or taking anti- coagulants. Compounds of formula I will also be useful as a cytoprotective agent for patients under chemotherapy.
Combinations with Other Drugs
Compounds of formula I will be useful as a partial or complete substitute for conventional antiinflammatory or analgesic compounds in preparations wherein they are presently co-administered with other agents or ingredients. Thus in further aspects, the invention encompasses pharmaceutical compositions for treating prostaglandin E2 mediated diseases as defined above comprising a non-toxic therapeutically effective amount of the compound of formula I as defined above and one or more ingredients such as another pain reliever including acetaminophen or phenacetin; a COX-2 selective inhibiting agent; a conventional NSAID; a potentiator including caffeine; an H2-antagonist, aluminum or magnesium hydroxide, simethicone, a decongestant including phenylephrine, phenylpropanolamine, pseudophedrine, oxymetazoline, ephinephrine, naphazoline, xylometazoline, propylhexedrine, or levo-desoxyephedrine; an antiitussive including codeine, hydrocodone, caramiphen, carbetapentane, or dextramethoφhan; another prostaglandin ligand including misoprostol, enprostil, rioprostil, ornoprostol or rosaprostol; a diuretic; a sedating or non-sedating antihistamine. In addition, the invention encompasses a method of treating prostaglandin E2 mediated diseases comprising: administration to a patient in need of such treatment a non-toxic therapeutically effective amount of the compound of formula I, optionally co- administered with one or more of such ingredients as listed immediately above. METHODS OF SYNTHESIS
Compounds of the present invention can be prepared according to the general synthesis schemes appearing in WO96/30358, or in accordance with U. S. Patent No. 5,391,817, incoφorated herein by reference in its entirety. Altemativley, the compounds can be synthesized using the following methods:
Method A This method make use of Z precursor containing protected acid unit
(AC(O)B). Suzuki's cross coupling between excess aryl dihalide of type 1 and an appropriate boronic acid 2 led to the biaryl 3. In most cases, Pd(PPh3)4 is used as the catalyst, Na2CO3 or CsF as the base in a refluxing mixture of DME/H2O (4/1). A second Suzuki' s coupling between 3 and the boronate 4 followed by acidic hydrolysis of the resulting ester 5 (basic hydrolysis in the cases of methyl or ethyl ester) afforded the desired acid 6. A second strategy involves the formation of the boronic acid 7 from the corresponding halide 3 and a subsequent Suzuki' s coupling with the aryl halide 8. Deprotection of the resulting ester 9 followed by coupling with an appropriate sulfonamide using EDCI/DMAP led to the acyl-sulfonamide 10.
Figure imgf000040_0001
Method B
In this method the acid group (AC(O)B) is introduce by Horner- Emmons (or Wittig) condensation. Suzuki' s coupling between the boronic acid 11 and the aryl dihalide 1 led to the aldehyde 12 under previously described conditions. The acid unit is introduce by a Homer-Emmons reaction to afford the diaryl halide 13 followed by cross coupling with the boronic acid 1 to give the desired ester 9. Alternatively, coupling between 12 and 2 followed by a Homer-Emmons reaction on the resulting aldehyde 14, can also afford the ester 9.
Figure imgf000040_0002
Method C
Upon addition of an alkylating reagent (R-Hal) to the anion of tert- butyl ester 15, generated in presence of a hindered base, monosubstituted (16) esters were obtained. The ester (16) can be further modified as described in method A to give the desired α-substituted acids 17.
Figure imgf000041_0001
Method D
For the synthesis of compounds in which the central heteroaryl Y is a 2,4-thiazole or a 2,5-thiazole (21, 24) see the procedures described in the following references; Nan'ya, S.; Ishida, H.; Kanie, K; Ito, N.; Butsugan, Y. J. Heterocyclic
Chem. 1995, 32, 1299. Zhang, M. Q.; Haemers, A.; Nanden Berghe, D.; Pattyn, S. R.; Bolaert, W. J. Heterocyclic Chem. 1991, 28, 673. Gordon, T. D.; Singh, J.; Hansen, P. E.; Morgan, P. A. Tetrahedron Lett, 1993, 34, 1901.
Figure imgf000041_0002
23 2,5-Thiazole Method E
For the synthesis of compounds in which the central heteroaryl Y is a 2,4-oxazole or a 2,5-oxazole (28, 31) see the procedures described in the following references; Huang, W.; Pei, J.; Chen, B.; Pei, W.; Ye, X. Tetrahedron, 1996, 52, 10131. Hammar, W. J.; Rustad, M. A. J. Heterocyclic Chem. 1981, 18, 885.
Figure imgf000042_0001
30 2,5-Oxazole
Method F
For the synthesis of compounds in which the central heteroaryl Y is a 4,1-imidazole (37) see the procedures described in the following references; Lopez- Alvarado, P.; Avendano, C; Menendez, J. C. /. Org. Chem. 1995, 60, 5678. Home, D. A.; Yakushijin, K.; Buchi, G. Heterocycle 1994, 39, 139. For the synthesis of aryllead triacetates see; Barton, D. H.; Finet, J. P.; Donnelly, D. M. X. J. Chem. Soc. Perkin T. 1 1992, 1365.
Figure imgf000042_0002
EXAMPLES
The species described herein can be made following the procedures set forth in the examples below, in which, unless otherwise stated:
The end products were analyzed by NMR, TLC and elementary analysis;
Intermediates were analyzed by NMR and TLC;
The compounds were purified by flash chromatography on silica gel, recrystallization and/or swish (suspension in a solvent followed by filtration of the solid); The course of reactions was followed by thin layer chromatography
(TLC) and reaction times are given for illustration only; and Some end products were converted to the sodium salt (dissolved in EtOH then addition of NaOH aq.)
The following intermediates were prepared according to literature procedures: (Phenylmethoxy)benzeneboronic acid: Johnson, C. R.; Johns,
B. A. /. Org. Chem. 1997, 62, 6046-6050.
2,4-Dibromo-5-chlorothiophene: Gronowitz, H. Acta Chem. Scand. Ser. B, 1976, 30, 439-445.
Lithium N-isopropylcyclohexylamide; lithium tert-butyl acetyl: Paquette, L. A.; Ewing, G. D. J. Org. Chem. 1975, 40, 2965-2966.
EXAMPLE 1 3-(2-(3-r5-CHLORO-2-(PHENYLMETHOXY)-3-PYRIDYLlPHENYL)PHENYL)
PROPANOIC ACID (5) Step 1 ; 2-(3-Bromophenyl)benzaldehyde:
A mixture of 2-formylbenzeneboronic acid (15.35 g, 102 rnmol), 1,3- dibromobenzene (75.25 g, 319 rnmol), 2M Na2CO3 (150 mL) and (Ph3P)4Pd (2.81 g, 24 rnmol) in DME (600 mL) was heated to 80 °C for 4 h. The mixture was cooled to r.t., quenched with IN HC1 (or aqueous NHjCl) and extracted with EtOAc. The organic layer was washed with brine, dried (MgSO ), filtered and concentrated. Purification by flash chromatography (To Hex, 0:1 to 1:1) provided the desired material as a yellow solid (21.3 g, 80%). 1H NMR (acetone-de) δ 7.4-7.8 (7H, m), 7.97 (IH, ddd), 9.95 (IH, s).
Step 2 ; Ethyl (E)-3-r2-(3-bromophenyl)phenyllprop-2-enoate:
To 2-(3-bromophenyl)benzaldehyde (6.67 g, 25.5 rnmol) and triethyl phosphonoacetate (6.89 g, 30.7 mmol) in Tol (90 mL) at 0 °C was added NaH (38.7 mmol). The reaction mixture was stirred at r.t. overnight. The reaction mixture was poured into aqueous NH CI and extracted twice with Εt2O. The combined organic extracts were washed (H2θ:brine), dried (MgSO4), filtered and concentrated.
Purification by flash chromatography (Tol) afforded the desired material as a colorless oil (8.01 g, 95%). 1H NMR (acetone-de) 5 1.25 (3H, t), 4.16 (2H, q), 6.50 (IH, d), 7.3- 7.7 (8H, m), 7.89 (IH, m). Step 3 ; (E)-3-r2-(3-bromophenyl)phenyl1prop-2-enoic acid:
The previous ester (8.01 g, 24.2 mmol) was hydrolyzed in THF:MeOH:2N LiOH (80 mL :40 mL :40 mL). After being stirred at r.t. overnight, the reaction mixture was poured into IN HCl (or H2θ:AcOH 10%) and extracted with ΕtOAc. The organic layer was washed with brine, dried (MgSO ), filtered and concentrated. Purification by a swish (Hex:ΕtOAc, 1:1) provided the title compound as a white crystalline solid (6.33 g, 86%). 1H NMR (acetone-de) δ 6.49 (IH, d), 7.3- 7.7 (8H, m), 7.90 (IH, m).
Step 4 : 3-r2-(3-Bromophenyl)phenyllpropanoic acid:
To the previous acid (7.19 g, 23.7 mmol) suspended in EtOAc (125 mL) was added PtO2 (262 mg, 1.14 mmol). After being sonicated for 2 h and stirred at r.t. for 2 days under a hydrogen atmosphere, the suspension was filtered over Celite and concentrated. Purification by a swish (Hex) afforded the title compound as a white solid (6.27 g, 86%). 1H NMR (acetone-dό) δ 2.46 (2H, t), 2.88 (2H, t), 7.18 (IH, dd), 7.28 (IH, td), 7.3-7.4 (4H, m), 7.52 (IH, t), 7.56 (IH, ddd).
Step 5 ; Methyl 3-f 2-r3-(4.4.5.5-tetramethyl-1.3.2-dioxaborolan-2-yl)phenyllphenyll propanoate: After esterification of the previous acid (1.72 g, 5.64 mmol) in ether
(20 mL) with an excess of diazomethane at 0 °C, the solution was dried (MgSO4), filtered and concentrated. To this crude ester in DMF (20 mL), diboron pinacol ester (1.57 g, 6.20 mmol), PdCl2(dppf) (138 mg, 0.169 mmol) and KOAc (1.78 g, 18.14 mmol) were added. After heating at 80 °C for 2 h, the mixture was poured into water and extracted twice with Et2O. The combined organic extracts were washed (H2O:brine), dried (MgSO4), filtered and concentrated. Purification by flash chromatography (Tol:Hex:EtOAc, 1:0:0 to 0:4:1) afforded the desired material as a yellow oil (1.42 g, 69%). 1H NMR (acetone-dό) δ 1.33 (12H, s), 2.45 (2H, t), 2.89 (2H, t), 3.54 (3H, s), 7.18 (IH, dd), 7.2-7.4 (3H, m), 7.46 (2H, m), 6.67 (IH, m), 7.75 (IH, m).
Step 6 ; 3-Bromo-5-chloro-2-(phenylmethoxy)pyridine:
A mixture of 3-bromo-5-chloropyridin-2-ol (50 g, 242 mmol), benzyl bromide (32 mL, 269 mmol) and Ag2CO3 (60 g, 218 mmol) in benzene (500mL) was refluxed for 1 h. The suspension was filtered over Celite and concentrated. The crude material was put under vacuo overnight to obtain the title compound as a yellow solid. 1H NMR (acetone-dό) δ 5.44 (2H, s), 7.2-7.4 (3H, m), 7.50 (2H, m), 8.07 (IH, d), 8.16 (IH, d).
Step 7 : Methyl 3-(2-(3-r5-chloro-2-(phenylmethoxy)-3-pyridyl1phenyl)phenyl) propanoate:
A mixture of methyl ester (404 mg, 1.10 mmol) of step 5 in this example, 3-bromo-5-chloro-2-(phenylmethoxy)pyridine (650 mg, 2.18 mmol), PdCl2(dpρf) (33 mg, 0.04 mmol) and 2M Na2CO3 (2.4 mL) in DMF (6 mL) was heated to 80 °C overnight. The mixture was quenched with IN HCl and extracted twice with Et2O. The combined organic extracts were washed (H2O :brine), dried (MgSO ), filtered and concentrated. Purification by flash chromatography (Tol) afforded the desired material as a white solid (394 mg, 78%). 1H NMR (acetone-dό) δ 2.41 (2H, t), 2.89 (2H, t), 3.50 (3H, s), 5.45 (2H, s), 7.15 (IH, dd), 7.2-7.5 (9H, m), 7.52 (IH, dd), 7.6-7.7 (2H, m), 7.88 (IH, d), 8.16 (IH, d).
Step 8 ; 3-(2-(3-r5-Chloro-2-(phenyImethoxy)-3-pyridyl1phenyl)phenyl)propanoic acid :
As described in Example 1 (step 3), the previous ester (394 mg, 0.86 mmol) was hydrolyzed in THF:MeOH:2N LiOH (4 mL:2 mL:2 mL). Purification by a swish (Hex:EtOAc) afforded the title compound as a white solid (360 mg, 94%). 1H NMR (acetone-dό) δ 2.42 (2H, t), 2.91 (2H, t), 5.45 (2H, s), 7.16 (IH, dd), 7.2-7.4 (9H, m), 7.52 (IH, td), 7.6-7.7 (2H, m), 7.89 (IH, d), 8.16 (IH, d).
EXAMPLE 2
(E)-3-(2- ( 3-r2-(PHΕNYLMΕTHOXY)PHΕNYLlPHΕNYL)PHΕNYL) PROP-2-ENOIC ACID (7) Step 1: Ethyl (E)-3-(2-|3-r2-(phenylmethoxy)phenyl1phenyljphenyl)prop-2-enoate: As described in Example 1 (step 1), a mixture of 2-(phenylmethoxy) benzeneboronic acid (545 mg, 2.39 mmol), ethyl (E)-3-[2-(3- bromophenyl)phenyl]prop-2-enoate bromide (390 mg, 1.18 mmol) of Example 1 (step 2), 2M Na2CO3 (3.5 mL) and (Ph3P)4Pd (66 mg, 0.06 mmol) in DME (7 mL) was heated to 80 °C for 2 h. Purification by flash chromatography (Tol) provided the desired material as a yellow solid (485 mg, 95%). 1H NMR (acetone-dό) δ 2.04 (3H, t), 4.12 (2H, q), 5.16 (2H, s), 6.51 (IH, d), 7.07 (IH, td), 7.20 (IH, dd), 7.2-7.4 (8H, m), 7.4-7.5 (4H, m), 7.6-7.7 (2H, m), 7.79 (IH, d) and 7.89 (IH, dd).
Step 2: (E)-3-(2-{3-r2-(Phenylmethoxy)phenyl]phenyl}phenyl)prop-2-enoic acid :
The previous ester (483 mg, 1.11 mmol) was hydrolyzed in THF:MeOH:2N LiOH (4 mL:2 mL:2 mL) at 50°C for 1 h. Purification by a swish (Hex:Εt2O) afforded the desired material as a white solid (419 mg, 93%). 1H NMR (acetone-dό) δ 5.15 (2H, s), 6.49 (IH, d), 7.05 (IH, td), 7.20 (IH, dd), 7.2-7.4 (8H, m), 7.4-7.5 (4H, m), 7.60 (2H, m), 7.80 (IH, d) and 7.89 (IH, dd).
EXAMPLE 3 (E)- 3-(2- f 5-r2-(PHENYLMETHOXY)PHENYLl-3-PYRIDYL)PHENYL)
PROP-2-ENOIC ACID (8) Step 1 ; 2-(5-Bromo-3-pyridyl)benzaldehyde: As described in Example 1 (step 1), a mixture of 2- formylbenzeneboronic acid (2.00 g, 13.3 mmol), 3,5-dibromopyridine (9.52 g, 40.2 mmol), 2M Na2CO3 (17 mL) and (Ph3P)4-Pd (762 mg, 0.66 mmol) in DME (80 mL) was heated to 80 °C for 12 h. Purification by flash chromatography (To EtOAc, 1:0 to 19 :1) provided the desired material as a yellow oil (2.85 g, 82%). 1H NMR (acetone-dό) δ 7.56 (IH, m), 7.68 (IH, t), 7.79 (IH, td), 8.03 (IH, dd), 8.11 (IH, t), 8.60 (IH, d), 8.74 (IH, d), 10.01 (IH, s).
Step 2 ; 2-{5-r2-(Phenylmethoxy)phenyl1-3-pyridyllbenzaldehyde:
As described in Example 1 (step 1), a mixture of the previous bromide (507 mg, 1.93 mmol, stepl), 2-(phenylmethoxy)benzeneboronic acid (638 mg, 2.80 mmol), 2M Na2CO3 (3.5 mL) and (Ph3P)4Pd (102 mg, 0.09 mmol) in DME (10 mL) was heated to 80 °C for 6 h. Purification by flash chromatography (To EtOAc, 9:1) provided the desired material as a yellow oil (642 mg, 91%). 1H NMR (acetone-dό) δ 5.19 (2H, s), 7.1-7.4 (9H, m), 7.51 (IH, dd), 7.63 (IH, t), 7.74 (IH, td), 8.01 (IH, dd), 8.06 (IH, t), 8.55 (IH, d), 8.83 (IH, d), 10.03 (IH, s).
Step 3 ; (E)- 3-(2-|5-r2-(Phenylmethoxy)phenyl1-3-pyridyl)phenyl)prop-2-enoic acid: As described in Example 1 (step 2), a mixture of the previous aldehyde (640 mg, 1.75 mmol, step 2), triethyl phosphonoacetate (420 uL, 2.12 mmol) and NaH (2.7 mmol) in Tol (6 mL) was stirred at it for 6 h. The crude material was not purified. According to the procedure described in step 8 of Example 1 , the crude ester was hydrolyzed in THF:MeOH:2N LiOH (6 mL:3 mL:3 mL). Purification by a swish (Hex:EtOAc) afforded the desired material as a white solid (641 mg, 90%). ]H NMR (acetone-dό) δ 5.15 (2H, s), 6.52 (IH, d), 7.09 (IH, t), 7.2-7.4 (7H, m), 7.42 (IH, m), 7.50 (4H, m), 7.94 (2H, m), 8.42 (IH, d), 8.78 (IH, d).
EXAMPLE 4 (E)-3-(2-(3-r2-(PHENYLMETHOXY)PHENYLlPHENYL}PHENYL)-N-(2- TH-(ENYl- UI-iONYL)PROP-2-ENAMIDE (55)
To (E)-3-(2-{ 3-[2-(Phenylmethoxy)phenyl]phenyl }phenyl)prop-2- enoic acid (210 mg, 0.52 mmol) of Example 2 (step 2), 2-thiophenesulfonylamide (121 mg, 0.74 mmol) and DMAP (250 mg, 2.04 mmol) in CH2C12 (3 mL) was added EDCI (142 mg, 0.74 mmol). After stirring at r.t. for 2 days, the reaction mixture was poured into IN HCl (or aqueous AcOH) and extracted with EtOAc. The organic layer was washed with brine, dried (MgSO4), filtered and concentrated. Purification by flash chromatography (Tol: EtOAc: AcOH, 1:0:0 to 20:1:0.1) and recrystallization (Hex:CH2Cl2) provided the desired material as a white solid (161 mg, 56%). 1H NMR (acetone-dό) δ 5.13 (2H, s), 6.72 (IH, d), 7.04 (IH, td), 7.1-7.2 (2H, m), 7.2-7.4 (8H, m), 7.4-7.5 (4H, m), 7.55 (IH, t), 7.62 (IH, dt), 7.7-7.8 (3H, m) and 7.92 (IH, dd).
EXAMPLE 5 . 2-METHYL-3-(2-(4-r3-(3-METHYLPHENOXY)PROPOXYlPHENYLlPHENYL)- N-(2-THffiNYLSULFONYL)PROPANAMIDE (84) Step 1 ; 3-(4-Bromophenoxy)propan-l-ol:
To 4-bromo phenol (15 lg; 0.87 mol) and 3-bromo propanol (120 mL; 1.3 mol) in 1.2 L of acetone was added portionwise K2CO3 (152 g; 1.1 mol). Final mixture was refluxed for 12h, cool to rt, filtered and concentrated. Distillation (0.2 mm Hg; 120 °C) afforded the desired material as a colorless oil (173 g; 86%). 1H NMR (acetone-dό) δ 1.95 (2H, m), 3.58 (IH, t), 3.67 (2H, m), 4.08 (2H, t), 6.90 (2H, d), 7.41 (2H, d).
Step 2 ; l-Bromo-4-r3-(3-methylphenoxy)propoxylbenzene :
To a solution of the previous alcohol (10.1 g; 44 mmol) in 110 mL of CH2C12 at -50 °C was added NEt3 (9.0 mL; 65 mmol) then dropwise MsCl (4.0 mL; 52 mmol). Final mixture was allowed to reach 0 °C then poured in saturated aqueous NaHCO3 and extracted with CH2C12 (2x). The combined organic extracts were dried over, filtered and concentrated to afford the mesylate, which was used without further purification. To a solution of m-cresol (6.6 g; 61 rnmol) in 130 mL of DMF was added portionwise NaH (1.6 g; 80%; 52 mmol). The mixture was stirred lh at rt then the mesylate was added. Final mixture was stirred 12h at rt then was poured in H2O and extracted with Et2O (3x). The combined organic extracts were washed with H2O (2x), brine, dried over MgSO4, filtered and concentrated. The excess m-cresol was removed by basic extraction (NaOH In; Hex) to afford the title compound as a white solid (13.5 g; 96%). 1H NMR (acetone-dό) δ 2.22 (2H, m), 2.27 (3H, s), 4.17 (4H, m), 6.74 (3H, m), 6.92 (2H, dd), 7.13 (IH, t), 7.41 (2H, dd).
Step 3 ; 7ert-butyl 3-(2-f4-r3-(3-methylphenoxy)propoxy1phenyl jphenyl) propanoate
To a solution of the previous aryl bromide (6.16 g; 19.1 mmol) in 70 mL of DME at -78°C was added dropwise n-BuLi (8.4 mL; 2.5 M; 21 mmol). After stirring 30 min at -78 °C, triiso-propyl boronate (4.4 mL; 19.1 mmol) was added and the solution was warmed to rt and stirred for 2h. To this solution of boronate was added Na2CO3 (28 mL; 2 M; 56 mmol), Tert-butyl 3-(2-bromophenyl)propanoate (Example 4; step 1; 4.19 g; 14.7 mmol) and Pd(PPh3)4 (0.27 g; 0.24 mmol). The final mixture was stirred at 85°C for 6h then was poured in HCl (IN) and extracted with EtOAc (2x). The combined organic extracts were washed brine, dried over Na2SO4, filtered and concentrated. Flash chromatography (Hex:EtOAc; 7:3) yielded the title compound as a yellow oil (6.69 g; 78%). 1H NMR (acetone-dό) δ 1.33 (9H, s), 2.25- 2.35 (7H, m), 2.87 (2H, t), 4.15-4.25 (4H, m), 6.76 (3H, m), 7.03 (2H, dd), 7.15 (2H, m), 7.2-7.3 (5H, m).
Step 4 ; Tert-butyl 2-methyl-3-(2-{4-r3-(3-methylphenoxy)propoxy1phenyl) phenyl )propanoate : A solution of the previous ester (2.76 g, 6.17 mmol) in THF (10 mL) was added to a solution of lithium N-isopropylcyclohexylamide (56 mL; 0.14 M; 7.8 mmol) in THF at -78 °C then 30 min later, iodomethane (1.3 mL, 21 mmol). The solution was warmed slowly (1.5 h) at 0 °C, then poured in IN HCl and extracted with EtOAc (2x). The combined organic extracts were washed with brine, dried over MgSO , filtered and concentrated. Flash chromatography (Hex:EtOAc; 7:3) afforded the title compound as a yellow oil (2.47 g; 86%). 1H NMR (acetone-dό) δ 0.88 (3H, d), 1.27 (9H, s), 2.2-2.4 (6H, m), 2.68 (IH, dd), 2.93 (IH, dd), 4.20 (2H, t), 4.24 (2H, t), 6.76 (3H, m), 7.03 (2H, dd), 7.15 (2H, m), 7.24 (5H, m).
Step 5: 2-Methyl-3-(2-{4-r3-(3-methylphenoxy)propoxy1phenyl }phenyl)propanoic acid:
To a solution of the previous ester (1.33 g; 2.88 mmol) in 9 mL of CH2CI2 at 0 °C was added TFA (4.5 mL; 58 mmol). The resulting solution was stirred at rt until all starting material was consumed as monitored by TLC. Two co- evaporation with Tol followed by flash chromatography (Tol:EtOAc:AcOH;
10:0.5:0.05) afforded the desired acid as a colorless oil (1.04 g; 89%). 1H NMR (acetone-dό) δ 0.92 (3H, d), 2.27 (5H, m), 2.52 (IH, m), 2.69 (IH, dd), 3.05 (IH, dd), 4.20 (2H, t), 4.24 (2H, t), 6.76 (3H, m), 7.02 (2H, dd), 7.1-7.3 (7H, m).
Step 6 2-Methyl-3-(2-{4-r3-(3-methylphenoxy)propoxy1phenyl }phenyl)-/V-(2-thienyl sulfonvDpropanamide :
To a solution of the previous acid (453 mg; 1.12 mmol) in 10 mL of CH2C12 at 0 °C was added oxalyl chloride (0.30 mL; 3.3 mmol) then a catalytic amount of DMF (35 μL). The resulting solution was stirred at rt was monitored by MeOH quench TLC. Two co-evaporation with Tol afforded the desired acid chloride, which was used without any further purification. The acid chloride was dissolved in 7 mL of CH2C12, cooled to 0°C, then DIPEA ( 580 mL; 3.3 mmol) and and 2- thiophenesulfonylamide (222 mg, 1.4 mmol) were added simultaneously. The resulting solution was stirred at rt for 12h, quenched with 25 mL MeOH, concentrated, poured in H2O (10% AcOH) and extracted with EtOAc (2x). The combined organic extracts were washed brine, dried over Na SO4, filtered and concentrated. Flash chromatography (CH2Cl2:MeOH 2.5%) afforded the title compound (350 mg; 56%). The carboxylic acid was converted to it' s corresponding sodium salt (EtOH : NaOH IN). 1H NMR (acetone-dό) δ 0.93 (3H, d), 2.27 (5H, m), 2.60-2.75 (2H, m), 2.91 (IH, dd), 4.22 (4H, m), 6.77 (3H, m), 7.01 (2H, d), 7.05-7.25 (8H, m), 7.74 (IH, dd), 7.94 (IH, dd). EXAMPLE 6
(E)-3- ( 2-r3-(3- ( 3-f2-(7-Cl LORO(2-OUINOLYL))NINYLlPHΕNOXY 1 PROPOXY)PHENYLlPHENYL } -N-(2-THLENYLSULFONYL)PROPANAMIDE
£98} Step 1: 3-(3-Bromophenoxy)propan-l-ol :
Prepared as described in Example 5 (step 1) from 3-bromo phenol. 1H NMR (acetone-dό) δ 1.95 (2H, m), 3.69 (3H, m), 4.12 (2H, t), 6.93 (IH, dd), 7.08 (2H, m), 7.23 (IH, t).
Step 2 ; (3-r3-(3-Bromophenoxy)propoxy]phenyl}methan-l-ol :
Prepared as described in Example 5 (step 2) from the corresponding mesylate of the previous alcohol and 3-hydroxymethyl phenol. 1H NMR (acetone-dό) δ 2.23 (2H, quint.), 4.18 (5H, m), 4.59 (2H, d), 6.81 (IH, dd), 6.90 (IH, d), 6.96 (2H, m), 7.05-7.25 (4H, m).
Step 3 ; 3-(3-r3-(Hydroxymethyl)phenoxy1propoxy)benzeneboronic acid :
To a solution of the previous aryl bromide (15.6 g; 46 mmol) in 200 mL of THF at -78°C was added dropwise n-BuLi (40 mL; 2.5 M; 100 mmol). After stirring 45 min at -78 °C, triiso-propyl boronate (27 mL; 116 mmol) was added, the solution was warmed to rt and stirred for 9h. HCl (IN) was added until the pH=l, the resulting mixture was diluted with H20 and extracted with EtOAc. The organic extract was washed with H 0 (2x), brine, dried over Na2SO4, filtered and concentrated.
Purification by swish (Hex:EtOAc:H20) yielded the title compound as a white solid
(5.3 g; 38%). 1H NMR (acetone-dό) δ
Step 4 ; 7ert-butyl 3-[2-(3-{3-r3-(hydroxymethyl)phenoxy1propoxy)phenyl) phenyl] propanoate :
The previous boronic acid (5.26 g; 17.4 mmol), tert-butyl 3-(2- bromophenyl)propanoate (5.99 g; 21 mmol), Pd(PPh3)4 (1.21 g; 1.1 mmol) and Na2CO3 (20 mL; 2 N; 40 mmol) were combined in 100 mL of DME. Nitrogen was bubbled in the suspension for 15 min then it was stirred at 80 °C for 8h. The mixture was cooled to rt, poured in saturated aqueous NH4CI then extracted with EtOAc (2x).
The combined organic extracts were dried over Na2SO , filtered and concentrated.
Flash chromatography (Tol:EtOAc; 90:10) afforded the title compound (5.18 g; 64%). 1H NMR (acetone-dό) δ 1.33 (9H, s), 2.26 (2H, m), 2.34 (2H, m), 2.88 (2H, t), 4.09 (IH, t), 4.20 (2H, t), 4.25 (2H, t), 4.58 (2H, d), 6.81 (IH, dd), 6.85-7.00 (5H, m), 7.1- 7.4 (6H, m).
Step 5 ; 7ert-butyl 3-(2-(3-r3-(3-formylphenoxy)propoxy1phenyl|phenyl) propanoate :
To a solution of the previous benzyl alcohol (5.18 g; 11.2 mmol) in 75 mL of EtOAc at 0°C was added MnO2 (30.2 g; 0.34 mol) portionwise. The suspension was stirred at rt for 4h, filtered on Celite, and concentrated to yield the title compound (4.30 g; 84%). %). Η NMR (acetone-dό) δ 1.33 (9H, s), 2.32 (4H, m), 2.87 (2H, t), 4.29 (4H, m), 6.90 (2H, m), 6.98 (IH, m), 7.17 (IH, m), 7.2-7.4 (5H, m), 7.49 (3H, m), 9.99 (IH, s).
Step 6 : Tert-butyl (E)-3-(2-r3-(3-{3-r2-(7-chloro-2-αuinolyl)vinyl1phenoxyl propoxy) phenyll phenyl) propanoate : To a suspension of (7-chloroquinolin-2-yl)methyl triphenylphosponium bromide (1.71 g; 3.3 mmol) in 15 mL THF at -78°C was added dropwise t-BuOK (3.6 mL; 1.0 M; 3.6mmol). The mixture was stirred at -78°C for 15 min, at 0 °C for 30 min then was cooled down to -78 °C. A solution of the previously described aldehyde ( 1.04 g; 2.3 mmol) in 3 mL THF was cannulated and the final mixture was stirred 5h at 0 °C. It was poured in H2O: AcOH 10% and extracted with ΕtOAc (2x). The combined organic extracts were dried over Na2SO , filtered and concentrated. Flash chromatography (Tol then To ΕtOAc; 95:5) afforded the title compound (1.39 g; 98%). 1H NMR (acetone-de) δ 1.32 (9H, s), 2.32 (4H, m), 2.88 (2H, t), 4.29 (4H, q), 6.85-7.00 (4H, m), 7.1-7.4 (8H, m), 7.44 (IH, d), 7.51 (IH, dd), 7.79 (IH, d), 7.85 (IH, d), 7.92 (IH, d), 7.99 (IH, dd), 8.30 (IH, d).
Step 7: (E)-3-(2-r3-(3-f 3-r2-(7-chloro-2-quinolyl)vinyl1phenoxy}propoxy) phenyll phenyl} propanoic acid :
To a solution of the previous ester (1.39 g; 2.24 mmol) in 60 mL of Tol was added A1C13 (0.90 g; 6.76 mmol). The suspension was stirred for 12h, poured in H2O and extracted with ΕtOAc (2x). The combined organic extracts were washed with brine, dried over Na2SO , filtered and concentrated. Flash chromatography (Tol:ΕtOAc:AcOH; 95:5:1) afforded the title compound (894 mg; 71%). 1H NMR (acetone-de) δ 2.28 (2H, m), 2.48 (2H, m), 2.94 (2H, t), 4.28 (4H, m), 6.90 (IH, m), 6.97 (3H, m), 7.1-7.4 (8H, m), 7.50 (2H, m), 7.82 (2H, m), 7.90 (IH, d), 8.02 (IH, d), 8.28 (IH, d).
Step 8: (E)-3-(2-r3-(3-{3-r2-(7-chloro(2-quinolyl))vinyl1phenoxylpropoxy) phenyllphenyl ) -N-(2-thienylsulfonyl)propanamide:
To a mixture of the previous acid (696 mg; 1.23 mmol), 2- thiophenesulfonylamide (404 mg; 2.48 mmol) and DMAP (233 mg; 1.91 mmol) in 6 mL of CH2CI2 was added ΕDCI (358 mg; 1.87 mmol). The mixture was stirred at rt for 12h, poured in saturated aqueous ΝH4CI and extracted with CH2CI2 (2x). The combined organic extracts were dried over Na2SO , filtered and concentrated. Flash chromatography (Tol:ΕtOAc:AcOH; 95:5:1) afforded the title compound (614 mg; 75%). 1H NMR (acetone-dό) δ 2.28 (2H, m), 2.55 (2H, t), 2.88 (2H, t), 4.26 (4H, q), 6.83 (IH, m), 6.88 (IH, m), 6.95 (2H, m), 7.10-7.35 (9H, m), 7.44 (IH, d), 7.49 (IH, dd), 7.75-7.90 (5H, m), 8.00 (IH, d), 8.26 (IH, d).
EXAMPLE 7
(E)-3-(2-(3-r2-(7-CHLORO(2-OUINOLYL))VINYL1PHENYL)PHENYL)-N-(2- THIEΝYLSULFOΝYL)PROP-2-EΝAMIDE (103) Step 1: (E)-2-|3-r2-(7-chloro-2-quinolyl)vinyl1phenyl|benzaldehyde: Prepared as described in Example 1 (step 1) from (E)-2-[2-(3- bromophenyl)vinyl]-7-chloroquinoline (prepared as described Example 8 (step6), from 3-bromobenzaldehyde) and 2-formyl benzeneboronic acid (1.5 eq.). Title compound was purified by swish (Hex: CH2C12). 1H NMR (CDC13) δ 7.3-7.6 (6H, m), 7.6-7.8 (6H, m), 8.0-8.1 (3H, m), 10.03 (IH, d).
Step 2: Ethyl (E)-3-(2-{3-r2-(7-chloro-2-quinolyl)vinyl1phenyl)phenyl)prop-2- enoate:
To a solution of triethylphosphono acetate (2.16 g; 9.66 mmol) in 100 mL of THF at 0 °C was added NaH (441 mg; 11.0 mmol). The reaction mixture was stirred for 30 min at 0°C, a solution of the previous aldehyde (3.40 g; 9.20 mmol) in 10 mL THF was then cannulated and the final mixture was stirred at rt for 12h. The reaction was quenched using saturated aqueous NH4CI, the THF was removed in vacuo, extracted with CH2CI2. The organic phase was washed with H2O, saturated aqueous NaHCO3, dried over Na2SO4, filtered and concentrated. Flash chromatography (To ΕtOAc; 95:5) afforded the title compound as a yellow solid
- 51 -
SUBSTITUTΕ SHEET (RULE 26) (3.54 g; 88%). 1H NMR (CDC13) δ 1.25 (3H, t), 4.20 (2H, q), 6.45 (IH, d), 7.1-7.5 (8H, m), 7.55 (3H, m), 7.67 (2H, m), 7.83 (IH, d), 7.89 (IH, d), 8.05 (IH, d).
Step 3: (E)-3-(2-f 3-r2-(7-chloro-2-quinolyl)vinyl1phenyl|phenyl)prop-2-enoic acid: To a solution of the previous ester (3.54 g; 8.05 mmol) in 50 mL of
1,4-dioxane was added 4 mL of NaOH (10 M). The mixture was stirred at 90°C for 12h, cooled down and acidified using HCl 10%. The residual precipitate ( HCl salt of the quinoline) was filtered, washed with Εt2O and dried in vacuo to yield the title compound (3.5 g; 97%) which was used without further purification. 1H NMR (dmso-dό) δ 6.49 (IH, d), 7.28 (IH, d), 7.4-8.0 (14H, m), 8.38 (IH, d).
Step 4: (E)-3-(2-(3-r2-(7-chloro(2-quinolyl))vinvnphenyl}phenyl)-N-(2-thienyl sulfonyl) prop-2-enamide:
Prepared as described in Example 4, from the previous acid, 2- thiophenesulfonamide. 1H ΝMR (dmso-dό) δ 6.63 (IH, d), 7.15 (IH, m), 7.25 (IH, d), 7.4-8.0 (16H, m), 8.40 (IH, d).
EXAMPLE 8 (E)-3-r2-(3.4-DICHLOROPHEΝYL)PHEΝYLl-N-(2-THJΕΝYI-^ULFOΝYL) PROP-2-ENAMIDE (104)
Stepl ; Methyl (E)-3-r2-(3,4-dichlorophenyl)phenyl1prop-2-enoate:
A mixture of 3,4-dichlorophenyl boronic acid ( 3.95 g; 25 mmol), ethyl 2-bromocinnamate (2.0 g; 8.3 mmol), CsF (6.30 g; 50 mmol) and Pd(PPh3)4 (1.21 g; 1.1 mmol) in 40 mL of DMΕ was stirred at 100 °C for lOh. The mixture was cooled to rt, poured in saturated aqueous NH4CI then extracted with ΕtOAc (2x). The combined organic extracts were dried over Na2SO4, filtered and concentrated. Flash chromatography (Tol) and subsequent swish (Hex) afforded the title compound (1.71 g; 47%). 1H NMR (CDC13) δ 3.75 (3H, s), 6.39 (IH, d), 7.12 (IH, dd), 7.30 (IH, m), 7.42 (3H, m), 7.50 (IH, d), 7.62 (IH, d), 7.68 (IH, m).
Step 2 ; (E)-3-r2-(3,4-dichIorophenyl)phenyl1prop-2-enoic acid:
Prepared as described in Example 2 (step 2) from the previous ester. 1H NMR (CDC13) δ 6.40 (IH, d), 7.11 (IH, dd), 7.32 (IH, dd), 7.4-7.5 (4H, m), 7.70 (2H, m). Step 3 ; (E)-3-r2-(3.4-dichlorophenyl)phenyl1-N-(2-thienylsulfonyl)prop-2-enamide: Prepared as described in Example 4 from the previous acid. lH ΝMR (CDC13) δ 6.44 (IH, d), 7.04 (IH, dd), 7.10 (IH, dd), 7.3-7.5 (5H, m), 7.66 (3H, m), 7.90 (IH, dd). HRMS (FAB+) m/z calcd for Cι9H14Cl2ΝO3S2 (M+H+): 437.9792, found 437.9791.
ELEMENTARY ANALYSIS OF REPRESENTATIVE COMPOUNDS
Figure imgf000054_0001
Figure imgf000055_0001
ASSAYS FOR DETERMI NG BIOLOGICAL ACTIVITY
The compounds of Formula I can be tested using the following assays to determine their prostanoid antagonist or agonist activity in vitro and in vivo and their selectivity. The prostaglandin receptors investigated were DP, EP1 ; EP2, EP3, EP4, FP, IP and TP.
Stable expression of prostanoid receptors in the human embryonic kidney (HEK) 293(ebna) cell line Prostanoid receptor cDNAs corresponding to full length coding sequences were subcloned into the appropriate sites of mammalian expression vectors and transfected into HEK 293(ebna) cells. HEK 293(ebna) cells expressing the individual cDNAs were grown under selection and individual colonies were isolated after 2-3 weeks of growth using the cloning ring method and subsequently expanded into clonal cell lines.
Prostanoid receptor binding assays
HEK 293(ebna) cells are maintained in culture, harvested and membranes are prepared by differential centrifugation, following lysis of the cells in the presence of protease inhibitors, for use in receptor binding assays. Prostanoid receptor binding assays are performed in 10 mM MES/KOH (pH 6.0) (EPs, FP and TP) or 10 mM HEPES/KOH (pH 7.4) (DP and IP), containing 1 mM EDTA, 10 mM divalent cation and the appropriate radioligand. The reaction is initiated by addition of membrane protein. Ligands are added in dimethylsulfoxide which is kept constant at 1 % (v/v) in all incubations. Non-specific binding is determined in the presence of 1 μM of the corresponding non-radioactive prostanoid. Incubations are conducted for 60 min at room temperature or 30 °C and terminated by rapid filtration. Specific binding is calculated by subtracting non specific binding from total binding. The residual specific binding at each ligand concentration is calculated and expressed as a function of ligand concentration in order to construct sigmoidal concentration- response curves for determination of ligand affinity.
Prostanoid receptor agonist and antagonist assays Whole cell second messenger assays measuring stimulation (EP2, EP , DP and
IP in HEK 293(ebna) cells) or inhibition (EP3 in human erythroleukemia (HEL) cells) of intracellular cAMP accumulation or mobilization of intracellular calcium (EPi, FP and TP in HEK 293(ebna) cells stably transfected with apo-aequorin) are performed to determine whether receptor ligands are agonists or antagonists. For cAMP assays, cells are harvested and resuspended in HBSS containing 25 mM HEPES, pH 7.4. Incubations contain 100 μM RO-20174 (phosphodiesterase type IN inhibitor, available from Biomol) and, in the case of the EP3 inhibition assay only, 15 μM forskolin to stimulate cAMP production. Samples are incubated at 37°C for 10 min, the reaction is terminated and cAMP levels are then measured. For calcium mobilization assays, cells are charged with the co-factors reduced glutathione and coelenterazine, harvested and resuspended in Ham's F12 medium. Calcium mobilization is measured by monitoring luminescence provoked by calcium binding to the intracellular photoprotein aequorin. Ligands are added in dimethylsulfoxide which is kept constant at 1 % (v/v) in all incubations. For agonists, second messenger responses are expressed as a function of ligand concentration and both EC50 values and the maximum response as compared to a prostanoid standard are calculated. For antagonists, the ability of a ligand to inhibit an agonist response is determined by Schild analysis and both KB and slope values are calculated.
Rat Paw Edema Assay
The method is the same as described in Chan et al (J. Pharmacol. Exp. Ther. 274: 1531-1537, 1995).
LPS-Induced Pyrexia in Conscious Rats
The method is the same as described in Chan et al (J. Pharmacol. Exp. Ther. 274: 1531-1537, 1995).
LPS-Induced Pyrexia in Conscious Squirrel Monkeys
The method is the same as described in Chan et al (Eur. J. Pharmacol. 327: 221- 225, 1997).
Acute Inflammatory Hyperalgesia Induced by Carrageenan in Rats The method is the same as described in Boyce et al (Neuropharmacology 33:
1609-1611, 1994).
Adjuvant-Induced Arthritis in Rats
Female Lewis rats (body weight -146-170 g) were weighed, ear marked, and assigned to groups (a negative control group in which arthritis was not induced, a vehicle control group, a positive control group administered indomethacin at a total daily dose of 1 mg/kg and four groups administered with a test compound at total daily doses of 0.10-3.0 mg/kg) such that the body weights were equivalent within each group. Six groups of 10 rats each were injected into a hind paw with 0.5 mg of Mycobacteήum butyricum in 0.1 mL of light mineral oil (adjuvant), and a negative control group of 10 rats was not injected with adjuvant. Body weights, contralateral paw volumes (determined by mercury displacement plethysmography) and lateral radiographs (obtained under Ketamine and Xylazine anesthesia) were determined before (day -1) and 21 days following adjuvant injection, and primary paw volumes were determined before (day -1) and on days 4 and 21 following adjuvant injection. The rats were anesthetized with an intramuscular injection of 0.03 - 0.1 mL of a combination of Ketamine (87 mg/kg) and Xylazine (13 mg/kg) for radiographs and injection of adjuvant. The radiographs were made of both hind paws on day 0 and day 21 using the Faxitron (45 kVp, 30 seconds) and Kodak X-OMAT TL film, and were developed in an automatic processor. Radiographs were evaluated for changes in the soft and hard tissues by an investigator who was blinded to experimental treatment. The following radiographic changes were graded numerically according to severity: increased soft issue volume (0-4), narrowing or widening of joint spaces (0-5) subchondral erosion (0-3), periosteal reaction (0-4), osteolysis (0-4) subluxation (0-3), and degenerative joint changes (0-3). Specific criteria were used to establish the numerical grade of severity for each radiographic change. The maximum possible score per foot was 26. A test compound at total daily doses of 0.1, 0.3, 1, and 3 mg/kg/day, indomethacin at a total daily dose of 1 mg/kg/day, or vehicle (0.5% methocel in sterile water) were administered per os b.i.d. beginning post injection of adjuvant and continuing for 21 days. The compounds were prepared weekly, refrigerated in the dark until used, and vortex mixed immediately prior to administration.

Claims

WHAT IS CLAIMED IS:
1. A method of treating or preventing a prostaglandin E mediated disease which comprises administering to a mammalian patient in need of such treatment or prevention a compound of formula I:
Figure imgf000059_0001
wherein:
R is a group Ar as defined hereinafter;
Rl is hydrogen, hydroxy, Ci-6alkyl, Cι_6alkoxy, X(CH2)pAr, or a methylenedioxy group attached to two adjacent ring carbon atoms;
R2 is -(CH2)χC(O)N(R4)S(O)yR5, -(CH2)χS(O)yN(R4)C(O)R5, -(CH2)xC(O)N(R4)C(O)R5, -(CH2)χS(O)yN(R4)S(O)yR5, -(CH2)XC02R4, or tetrazol-5-yl optionally substituted by Cι_6alkyl;
R3 is X(CH2)pAr or X(CH2)pR4 or a group of formula (a):
Figure imgf000059_0002
(a)
Ar is a group of formula (b) or (c):
Figure imgf000060_0001
or Ar is naphthyl, indolyl, pyridyl, thienyl, furyl, oxazolidinyl, oxazolyl, thiazolyl, isothiazolyl, pyrazolyl, thriazolyl, tetrazolyl, imidazolyl, imidazolidinyl, thiazolidinyl, isoxazolyl, oxadiazolyl, thiadiazolyl, moφholinyl, piperidinyl, piperazinyl, pyrrolyl, or pyrimidinyl, all of which may be unsubstituted or substituted by one or more R or R8 groups;
A is C=O or (C(R4)2)m;
each B is independently -CH2- or -O-;
R4 is hydrogen or Cι_6alkyl;
R5 is hydrogen or Ci-ioalkyl or Ar, both of which may be unsubstituted or substituted by one or two Cl, F, Br, hydroxy, XCi-5alkyl, Cι_5alkyl, NO2, tetrazol-5- yl optionally substituted by Ci_6alkyl, or R5 is N(R4)2;
R6 is hydrogen, RlO, CO2R11, Cθ2C(RlO)2O(CO)XRH, Pθ3(Rl l)2, SO2NRHRIO, NRHSO2R10, CONRllSO2R10, SO3RH, S(O)qRH, S(O)qN(Rl l)C(O)Rl0, S(O)qN(RH)S(O)qRl0, C(O)N(RH)C(O)R10, N(Rl l)C(O)Rl0, N(Rll)2, N(RH)C(0)NRH, P(O)(ORl l)Rl l, CN, - Cθ2(CH2)mC(O)N(R4)2, C(RlO)2N(Rll)2, C(0)N(R4)2, OR4 or tetrazolyl optionally substituted by Cl-6 alkyl;
R7 and R9 are independently hydrogen, RlO, OH, Ci-8 alkoxy, S(O)qRl0, N(R4)2, Br, F, I, Cl, CF3, NO2, NHCOR4, R12CO2R1 1, -X-R13-Y, -X(CR4)pOR4, S(CH2)pCO H, (CH2)pX-Rl3, -X(CH2)pCONRl lSO2R10, (CH2)pXCONRl 1SO2R10 or X(CH2)pR6 wherein each methylene group within - X(CH2)qR6 may be unsubstituted or substituted by one or two -(CH2)pAr groups; R8 is hydrogen, RlO, OH, Ci-Salkoxy, S(O)qRl0, N(R4)2, Br, F, I, Cl or NHCOR4 wherein the Ci-5 alkoxy may be unsubstituted or substituted by OH, methoxy or halogen;
RlO is hydrogen, Ar, Cl-ioalkyl, C2-10alkenyl, C2-10alkynyl, all of which may be unsubstituted or substituted by one or more OH,.CH2OH, N(R4)2 or halogen; or RlO is N(R4)2;
RU is independently hydrogen, Cl-ioalkyl, C2-10alkenyl or C2-8alkynyl, all of which may be unsubstituted or substituted by one or more OH, N(R4)2, CO2R14, halogen or XCl-5alkyl; or RU is (CH2)pAr;
Rl2 is divalent Ar, Ci-ioalkylene, Ci-ioalkylidene, C2-10alkenylene, C2-10 alkynylene, all of which may be unsubstituted or substituted by one or more of OH, CH2OH, N(R4)2 or halogen;
Rl3 is a bond, Ci-ioalkylene, Ci-ioalkenylene, Ci-ioalkylidene, Ci-ioalkynylene, all of which may be linear or branched, or phenylene, all of which may be unsubstituted or substituted by one or more OH, N(R4)2, COOH or halogen;
Rl4 is hydrogen, Ci-6 alkyl, C2-6 alkenyl or C2-7 alkynyl;
X is (CH2)p, O, NR4 or S(O)p;
Y is CH3 or X(CH2)pAr;
q is zero, one or two; p is an integer from 0 to 6; m is 1, 2 or 3; n is 1 to 4; x is 0 to 4; y is 1 or 2; the dotted line signifies the optional presence of a bond such that it represents a single or double bond.
2. A method according to Claim 1 wherein the prostaglandin mediated disease is selected from the group consisting of:
(1) pain, fever or inflammation associated with rheumatic fever, influenza or other viral infections, common cold, low back and neck pain, skeletal pain, post-partum pain, dysmenorrhea, headache, migraine, toothache, sprains and strains, myositis, neuralgia, synovitis, arthritis, including rheumatoid arthritis, degenerative joint diseases (osteoarthritis), gout and ankylosing spondylitis, bursitis, bums including radiation and corrosive chemical injuries, sunburns, pain following surgical and dental procedures as well as immune and autoimmune diseases;
(2) cellular neoplastic transformations or metastic tumor growth;
(3) diabetic retinopathy and tumor angiogenesis;
(4) prostanoid-induced smooth muscle contraction associated with dysmenorrhea, premature labor, asthma or eosinophil related disorders; (5) Alzheimer's disease;
(6) glaucoma;
(7) bone loss;
(8) osteopqrosis;
(9) promotion of bone formation; (10) Paget's disease;
(11) cytoprotection in peptic ulcers, gastritis, regional enteritis, ulcerative colitis, diverticulitis or other gastrointestinal lesions;
(12) GI bleeding and patients undergoing chemotherapy;
(13) coagulation disorders selected from hypoprothrombinemia, haemophilia and other bleeding problems;
(14) kidney disease;
(15) thrombosis;
(16) occlusive vascular disease;
(17) presurgery; and (18) anti-coagulation.
3. A method according to Claim 1 wherein the prostaglandin mediated disease is selected from the group consisting of: pain, fever or inflammation associated with rheumatic fever, influenza or other viral infections, common cold, low back and neck pain, skeletal pain, post-partum pain, dysmenorrhea, headache, migraine, toothache, sprains and strains, myositis, neuralgia, synovitis, arthritis, including rheumatoid arthritis, degenerative joint diseases (osteoarthritis), gout and ankylosing spondylitis, bursitis, bu s including radiation and corrosive chemical injuries, sunburns, pain following surgical and dental procedures as well as immune and autoimmune diseases;.
4. A method according to Claim 3 wherein the prostaglandin mediated disease is pain, fever or inflammation associated with dysmenorrhea.
5. A method according to Claim 1, wherein the compound is co- administered with other agents or ingredients.
6. A method according to Claim 5 wherein the compound is co- administered with another agent or ingredient selected from the group consisting of: (1) an analgesic selected from acetaminophen, phenacetin, aspirin, a narcotic;
(2) a cyclooxygenase-2 selective nonsteroidal anti-inflammatory drug or a conventional nonsteroidal anti-inflammatory drug;
(3) caffeine; (4) an H2-antagonist;
(5) aluminum or magnesium hydroxide;
(6) simethicone;
(7) a decongestant selected from phenylephrine, phenylpropanolamine, pseudophedrine, oxymetazoline, ephinephrine, naphazoline, xylometazoline, propylhexedrine, or levo-desoxyephedrine;
(8) an antiitussive selected from codeine, hydrocodone, caramiphen, carbetapentane and dextramethoφhan;
(9) another prostaglandin ligand selected from misoprostol, enprostil, rioprostil, omoprostol and rosaprostol; a diuretic; and (10) a sedating or non-sedating antihistamine.
7. A method according to Claim 6 wherein the compound is co- administered with a cyclooxygenase-2 selective nonsteroidal anti-inflammatory drug or a conventional nonsteroidal anti-inflammatory drug.
8. A method according to Claim 7 wherein the compound is co- administered with a conventional nonsteroidal anti-inflammatory drug selected from the group consisting of: aspirin, ibuprofen, naproxen, and ketoprofen.
9. A method according to Claim 6 wherein the compound is co- administered with a cyclooxygenase-2 selective nonsteroidal anti-inflammatory drug selected from rofecoxib, MK-663, valdecoxib, parecoxib and celecoxib.
10. A compound selected from one of the following tables:
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000069_0002
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000071_0002
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
or a pharmaceutically acceptable salt thereof, wherein
Qn represents 7-chloro-quinol-2-yl, 2-Ph-Et-SCH2 represents 2-phenylethylthiomethyl, and 3-(3-Me-Ph-oxy)Pr-oxy represents 3-(3-methylphenoxy)propyl- 1 -oxy.
11. Use of a compound of formula I, as defined in any one of claims 1 to 9, in the manufacture of a medicament for treating or preventing a prostaglandin E mediated disease.
12. Use of a compound of claim 10, in the manufacture of a medicament for treating or preventing a prostaglandin E mediated disease.
13. A compound of formula I, as defined in any one of claims 1 to 9 for use in treating or preventing a prostaglandin E mediated disease.
14. A compound of claim 10 for use in treating or preventing a prostaglandin E mediated disease.
15. A pharmaceutical composition for treating or preventing a prostaglandin E mediated disease comprising an acceptable, effective amount of a compound of formula I, as defined in any one of claims 1 to 9, in association with a pharmaceutically acceptable carrier.
16. A pharmaceutical composition for treating or preventing a prostaglandin E mediated disease comprising an acceptable, effective amount of a compound of claim 10, in association with a pharmaceutically acceptable carrier.
PCT/CA2001/000563 2000-04-24 2001-04-23 Method of treatment using phenyl and biaryl derivatives as prostaglandin e inhibitors and compounds useful therefore WO2001081312A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP01927526A EP1278734A2 (en) 2000-04-24 2001-04-23 Method of treatment using phenyl and biaryl derivatives as prostaglandin e inhibitors and compounds useful therefore
JP2001578407A JP2003531194A (en) 2000-04-24 2001-04-23 Therapeutic methods using phenyl and biaryl derivatives as prostaglandin E inhibitors and compounds useful in the methods
AU2001254555A AU2001254555A1 (en) 2000-04-24 2001-04-23 Method of treatment using phenyl and biaryl derivatives as prostaglandin E inhibitors and compounds useful therefore
CA002405170A CA2405170A1 (en) 2000-04-24 2001-04-23 Method of treatment using phenyl and biaryl derivatives as prostaglandin e inhibitors and compounds useful therefore

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US19929900P 2000-04-24 2000-04-24
US60/199,299 2000-04-24

Publications (2)

Publication Number Publication Date
WO2001081312A2 true WO2001081312A2 (en) 2001-11-01
WO2001081312A3 WO2001081312A3 (en) 2002-08-08

Family

ID=22736988

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2001/000563 WO2001081312A2 (en) 2000-04-24 2001-04-23 Method of treatment using phenyl and biaryl derivatives as prostaglandin e inhibitors and compounds useful therefore

Country Status (6)

Country Link
US (1) US6627656B2 (en)
EP (1) EP1278734A2 (en)
JP (1) JP2003531194A (en)
AU (1) AU2001254555A1 (en)
CA (1) CA2405170A1 (en)
WO (1) WO2001081312A2 (en)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004084824A3 (en) * 2003-03-24 2005-03-31 Merck & Co Inc Biaryl substituted 6-membered heterocyles as sodium channel blockers
WO2005115990A1 (en) * 2004-05-26 2005-12-08 Eisai R & D Management Co., Ltd. Cinnamide compound
WO2006037982A2 (en) * 2004-10-05 2006-04-13 Astrazeneca Ab Modulators of crth2 receptor activity for the treatment of prostaglandin d2 mediated diseases
FR2878522A1 (en) * 2004-12-01 2006-06-02 Merck Sante Soc Par Actions Si Use of new and known biphenyl derivatives as caspase-10 inhibitors in preparation of a drug to prevent and/or treat retinopathy
EP1847524A1 (en) 2006-04-21 2007-10-24 Cellzome (UK) Ltd. Terphenyl derivatives for treatment of Alzheimer's disease
WO2009064250A1 (en) * 2007-11-15 2009-05-22 Astrazeneca Ab Bis-(sulfonylamino) derivatives in therapy 065
WO2009064251A1 (en) * 2007-11-15 2009-05-22 Astrazeneca Ab Bis-(sulfonylamino) derivatives in therapy 066
US7618960B2 (en) 2005-11-24 2009-11-17 Eisai R&D Management Co., Ltd. Morpholine type cinnamide compound
US7713993B2 (en) 2006-03-09 2010-05-11 Eisai R&D Management Co., Ltd. Multi-cycle cinnamide derivatives
US7737141B2 (en) 2006-07-28 2010-06-15 Eisai R&D Management Co., Ltd. Prodrug of cinnamide compound
US7737135B2 (en) 2004-08-24 2010-06-15 Astrazeneca Ab Biphenyloxyacetic acid derivatives for the treatment of respiratory disease
US7923563B2 (en) 2004-10-26 2011-04-12 Eisai R&D Management Co., Ltd. Amorphous object of cinnamide compound
US7935815B2 (en) 2007-08-31 2011-05-03 Eisai R&D Management Co., Ltd. Imidazoyl pyridine compounds and salts thereof
US8003703B2 (en) 2003-08-21 2011-08-23 Astrazeneca Ab Phenoxiacetic acid derivatives
US8008350B2 (en) 2005-10-06 2011-08-30 Astrazeneca Ab Biphenyloxyacetic acid derivatives for the treatment of respiratory disease
US8022248B2 (en) 2004-07-08 2011-09-20 Astrazeneca Ab Substituted acids for the treatment of respiratory diseases
US8048878B2 (en) 2005-11-24 2011-11-01 Eisai R&D Management Co., Ltd. Two cyclic cinnamide compound
US8148421B2 (en) 2006-09-11 2012-04-03 Fuji Yakuhin Co., Ltd. Phenylacetic acid derivative
US8148572B2 (en) 2005-10-06 2012-04-03 Astrazeneca Ab Compounds
US8158820B2 (en) 2003-04-07 2012-04-17 Astrazeneca Ab Compounds
CN1972916B (en) * 2004-05-26 2013-03-27 卫材R&D管理有限公司 Cinnamide compound
US8507544B2 (en) 2007-07-05 2013-08-13 Astrazeneca Ab Bi-aryl amide compounds as CRTh2 receptor modulators
US8524715B2 (en) 2004-11-23 2013-09-03 Astrazeneca Ab Phenoxyacetic acid derivatives useful for treating respiratory diseases
US9145380B2 (en) 2007-12-20 2015-09-29 Astrazeneca Ab Bis-(sulfonylamino) derivatives for use in therapy
US9453000B2 (en) 2007-08-31 2016-09-27 Eisai R&D Management Co., Ltd. Polycyclic compound

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2002346620A1 (en) 2001-11-30 2003-06-17 Department Of Veteran Affairs Induction of apoptosis in cancer cells
WO2017070089A1 (en) 2015-10-19 2017-04-27 Incyte Corporation Heterocyclic compounds as immunomodulators
MD3377488T2 (en) 2015-11-19 2023-02-28 Incyte Corp Heterocyclic compounds as immunomodulators
MX2018007774A (en) 2015-12-22 2018-11-09 Incyte Corp Heterocyclic compounds as immunomodulators.
AR108396A1 (en) 2016-05-06 2018-08-15 Incyte Corp HETEROCYCLIC COMPOUNDS AS IMMUNOMODULATORS
ES2905980T3 (en) 2016-05-26 2022-04-12 Incyte Corp Heterocyclic compounds as immunomodulators
CN109890819B (en) 2016-06-20 2022-11-22 因赛特公司 Heterocyclic compounds as immunomodulators
EP3484866B1 (en) 2016-07-14 2022-09-07 Incyte Corporation Heterocyclic compounds as immunomodulators
EP3504198B1 (en) 2016-08-29 2023-01-25 Incyte Corporation Heterocyclic compounds as immunomodulators
ES2899402T3 (en) 2016-12-22 2022-03-11 Incyte Corp Pyridine derivatives as immunomodulators
LT3558990T (en) 2016-12-22 2022-12-27 Incyte Corporation Tetrahydro imidazo[4,5-c]pyridine derivatives as pd-l1 internalization inducers
PE20191532A1 (en) 2016-12-22 2019-10-23 Incyte Corp HETEROCYCLIC COMPOUNDS AS IMMUNOMODULATORS
WO2018119236A1 (en) 2016-12-22 2018-06-28 Incyte Corporation Triazolo[1,5-a]pyridine derivatives as immunomodulators
MX2020010321A (en) 2018-03-30 2021-01-08 Incyte Corp Heterocyclic compounds as immunomodulators.
MX2020012045A (en) 2018-05-11 2021-03-29 Incyte Corp Tetrahydro-imidazo[4,5-c]pyridine derivatives as pd-l1 immunomodulators.
MX2022001671A (en) 2019-08-09 2022-05-13 Incyte Corp Salts of a pd-1/pd-l1 inhibitor.
AR120109A1 (en) 2019-09-30 2022-02-02 Incyte Corp PYRIDO[3,2-D]PYRIMIDINE COMPOUNDS AS IMMUNOMODULATORS
CN114829366A (en) 2019-11-11 2022-07-29 因赛特公司 Salts and crystalline forms of PD-1/PD-L1 inhibitors
US11780836B2 (en) 2020-11-06 2023-10-10 Incyte Corporation Process of preparing a PD-1/PD-L1 inhibitor
WO2022099071A1 (en) 2020-11-06 2022-05-12 Incyte Corporation Process for making a pd-1/pd-l1 inhibitor and salts and crystalline forms thereof
US11760756B2 (en) 2020-11-06 2023-09-19 Incyte Corporation Crystalline form of a PD-1/PD-L1 inhibitor

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994002474A1 (en) * 1992-07-17 1994-02-03 Smithkline Beecham Corporation Endothelin receptor antagonists
WO1995003295A1 (en) * 1993-07-20 1995-02-02 Merck & Co., Inc. Endothelin antagonists
WO1996030358A1 (en) * 1995-03-27 1996-10-03 Smithkline Beecham Plc Phenyl derivatives useful as endothelin receptor antagonists
WO1999047497A2 (en) * 1998-03-13 1999-09-23 Merck Frosst Canada & Co. Carboxylic acids and acylsulfonamides, compositions containing such compounds and methods of treatment
WO2000020371A1 (en) * 1998-10-07 2000-04-13 Merck Frosst Canada & Co. Prostaglandin receptor ligands

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6242493B1 (en) * 1998-03-13 2001-06-05 Merck Frosst Canada & Co. Carboxylic acids and acylsulfonamides, compositions containing such compounds and methods of treatment

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994002474A1 (en) * 1992-07-17 1994-02-03 Smithkline Beecham Corporation Endothelin receptor antagonists
WO1995003295A1 (en) * 1993-07-20 1995-02-02 Merck & Co., Inc. Endothelin antagonists
WO1996030358A1 (en) * 1995-03-27 1996-10-03 Smithkline Beecham Plc Phenyl derivatives useful as endothelin receptor antagonists
WO1999047497A2 (en) * 1998-03-13 1999-09-23 Merck Frosst Canada & Co. Carboxylic acids and acylsulfonamides, compositions containing such compounds and methods of treatment
WO2000020371A1 (en) * 1998-10-07 2000-04-13 Merck Frosst Canada & Co. Prostaglandin receptor ligands

Cited By (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004084824A3 (en) * 2003-03-24 2005-03-31 Merck & Co Inc Biaryl substituted 6-membered heterocyles as sodium channel blockers
US8158820B2 (en) 2003-04-07 2012-04-17 Astrazeneca Ab Compounds
US8003703B2 (en) 2003-08-21 2011-08-23 Astrazeneca Ab Phenoxiacetic acid derivatives
US8394986B2 (en) 2003-08-21 2013-03-12 Astrazeneca Ab Phenoxiacetic acid derivatives
US7667041B2 (en) 2004-05-26 2010-02-23 Eisai R&D Management Co., Ltd. Cinnamide compound
WO2005115990A1 (en) * 2004-05-26 2005-12-08 Eisai R & D Management Co., Ltd. Cinnamide compound
US7880009B2 (en) 2004-05-26 2011-02-01 Eisai R&D Management Co., Ltd. Cinnamide compound
US7687640B2 (en) 2004-05-26 2010-03-30 Eisai R&D Management Co., Ltd. Cinnamide compound
CN1972916B (en) * 2004-05-26 2013-03-27 卫材R&D管理有限公司 Cinnamide compound
US8022248B2 (en) 2004-07-08 2011-09-20 Astrazeneca Ab Substituted acids for the treatment of respiratory diseases
US7737135B2 (en) 2004-08-24 2010-06-15 Astrazeneca Ab Biphenyloxyacetic acid derivatives for the treatment of respiratory disease
US8163727B2 (en) 2004-08-24 2012-04-24 Astrazeneca Ab Biphenyloxyacetic acid derivatives for the treatment of respiratory disease
US8722741B2 (en) 2004-08-24 2014-05-13 Astrazeneca Ab Biphenyloxyacetic acid derivatives for the treatment of respiratory disease
WO2006037982A3 (en) * 2004-10-05 2006-08-17 Astrazeneca Ab Modulators of crth2 receptor activity for the treatment of prostaglandin d2 mediated diseases
WO2006037982A2 (en) * 2004-10-05 2006-04-13 Astrazeneca Ab Modulators of crth2 receptor activity for the treatment of prostaglandin d2 mediated diseases
US7923563B2 (en) 2004-10-26 2011-04-12 Eisai R&D Management Co., Ltd. Amorphous object of cinnamide compound
US8524715B2 (en) 2004-11-23 2013-09-03 Astrazeneca Ab Phenoxyacetic acid derivatives useful for treating respiratory diseases
US7829721B2 (en) 2004-12-01 2010-11-09 Merck Patent Gmbh Specific caspase-10 inhibitors
WO2006058592A1 (en) * 2004-12-01 2006-06-08 Merck Patent Gmbh Novel specific caspase-10 inhibitors
FR2878522A1 (en) * 2004-12-01 2006-06-02 Merck Sante Soc Par Actions Si Use of new and known biphenyl derivatives as caspase-10 inhibitors in preparation of a drug to prevent and/or treat retinopathy
US8148572B2 (en) 2005-10-06 2012-04-03 Astrazeneca Ab Compounds
US8415394B2 (en) 2005-10-06 2013-04-09 Astrazeneca Ab Biphenyloxyacetic acid derivatives for the treatment of respiratory disease
US8008350B2 (en) 2005-10-06 2011-08-30 Astrazeneca Ab Biphenyloxyacetic acid derivatives for the treatment of respiratory disease
US8349897B2 (en) 2005-10-06 2013-01-08 Astrazeneca Ab Biphenyloxyacetic acid derivatives for the treatment of respiratory disease
US8048878B2 (en) 2005-11-24 2011-11-01 Eisai R&D Management Co., Ltd. Two cyclic cinnamide compound
US7618960B2 (en) 2005-11-24 2009-11-17 Eisai R&D Management Co., Ltd. Morpholine type cinnamide compound
US7713993B2 (en) 2006-03-09 2010-05-11 Eisai R&D Management Co., Ltd. Multi-cycle cinnamide derivatives
US7973033B2 (en) 2006-03-09 2011-07-05 Eisai R&D Management Co., Ltd. Multi-cyclic cinnamide derivatives
US7897632B2 (en) 2006-03-09 2011-03-01 Eisai R&D Management Co., Ltd. Multi-cyclic cinnamide derivatives
EA016908B1 (en) * 2006-04-21 2012-08-30 Орто-Макнил-Дженссен Фармасьютикэлз, Инк. Terphenyl derivatives for treatment of alzheimer's disease
US8106236B2 (en) 2006-04-21 2012-01-31 Ortho-Mcneil Pharmaceutical, Inc. Triaryl compounds and derivates thereof
EP1847524A1 (en) 2006-04-21 2007-10-24 Cellzome (UK) Ltd. Terphenyl derivatives for treatment of Alzheimer's disease
WO2007124351A1 (en) 2006-04-21 2007-11-01 Ortho-Mcneil Pharmaceutical, Inc. Terphenyl derivatives for treatment of alzheimer' s disease
US7737141B2 (en) 2006-07-28 2010-06-15 Eisai R&D Management Co., Ltd. Prodrug of cinnamide compound
US8148421B2 (en) 2006-09-11 2012-04-03 Fuji Yakuhin Co., Ltd. Phenylacetic acid derivative
US8507544B2 (en) 2007-07-05 2013-08-13 Astrazeneca Ab Bi-aryl amide compounds as CRTh2 receptor modulators
US7935815B2 (en) 2007-08-31 2011-05-03 Eisai R&D Management Co., Ltd. Imidazoyl pyridine compounds and salts thereof
US9453000B2 (en) 2007-08-31 2016-09-27 Eisai R&D Management Co., Ltd. Polycyclic compound
CN101910121A (en) * 2007-11-15 2010-12-08 阿斯利康(瑞典)有限公司 Bis-(sulfonylamino) derivatives in therapy 066
WO2009064250A1 (en) * 2007-11-15 2009-05-22 Astrazeneca Ab Bis-(sulfonylamino) derivatives in therapy 065
WO2009064251A1 (en) * 2007-11-15 2009-05-22 Astrazeneca Ab Bis-(sulfonylamino) derivatives in therapy 066
AU2008321577B2 (en) * 2007-11-15 2011-05-26 Astrazeneca Ab Bis-(sulfonylamino) derivatives in therapy 066
US9145380B2 (en) 2007-12-20 2015-09-29 Astrazeneca Ab Bis-(sulfonylamino) derivatives for use in therapy

Also Published As

Publication number Publication date
US20020082266A1 (en) 2002-06-27
AU2001254555A1 (en) 2001-11-07
CA2405170A1 (en) 2001-11-01
US6627656B2 (en) 2003-09-30
EP1278734A2 (en) 2003-01-29
WO2001081312A3 (en) 2002-08-08
JP2003531194A (en) 2003-10-21

Similar Documents

Publication Publication Date Title
US6627656B2 (en) Method of treatment using phenyl and biaryl derivatives as prostaglandin E inhibitors and compounds useful therefore
AU776831B2 (en) Carboxylic acids and acylsulfonamides, compositions containing such compounds and methods of treatment
US6211197B1 (en) Prostaglandin receptor ligands
US7348433B2 (en) Quinolinones as prostaglandin receptor ligands
EP1071648A2 (en) Carboxylic acids and acylsulfonamides, compositions containing such compounds and methods of treatment
US7446222B2 (en) Phenyl compounds
US6242493B1 (en) Carboxylic acids and acylsulfonamides, compositions containing such compounds and methods of treatment
AU772841B2 (en) Carboxylic acids and acylsulfonamides, compositions containing such compounds and methods of treatment
JP2001139575A (en) Novel pyrazolopyridine derivative
AU752820B2 (en) Biphenylene lactams as prostaglandin receptor ligands
AU756333B2 (en) Carboxylic acids and acylsulfonamides, compositions containing such compounds and methods of treatment

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 2001927526

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2001254555

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2405170

Country of ref document: CA

ENP Entry into the national phase

Ref country code: JP

Ref document number: 2001 578407

Kind code of ref document: A

Format of ref document f/p: F

WWP Wipo information: published in national office

Ref document number: 2001927526

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 2001927526

Country of ref document: EP