EP0971910A1 - Alpha-methylen gamma lactone als selektive cyclooxygenase-2 hemmer - Google Patents

Alpha-methylen gamma lactone als selektive cyclooxygenase-2 hemmer

Info

Publication number
EP0971910A1
EP0971910A1 EP98913487A EP98913487A EP0971910A1 EP 0971910 A1 EP0971910 A1 EP 0971910A1 EP 98913487 A EP98913487 A EP 98913487A EP 98913487 A EP98913487 A EP 98913487A EP 0971910 A1 EP0971910 A1 EP 0971910A1
Authority
EP
European Patent Office
Prior art keywords
phenyl
methylsulfonyl
furanone
compound
cox
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP98913487A
Other languages
English (en)
French (fr)
Inventor
Serge Leger
Erich Grimm
Petpiboon Prasit
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Merck Frosst Canada and Co
Original Assignee
Merck Frosst Canada and 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
Priority claimed from GBGB9709356.1A external-priority patent/GB9709356D0/en
Application filed by Merck Frosst Canada and Co filed Critical Merck Frosst Canada and Co
Publication of EP0971910A1 publication Critical patent/EP0971910A1/de
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/06Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C317/00Sulfones; Sulfoxides
    • C07C317/14Sulfones; Sulfoxides having sulfone or sulfoxide groups bound to carbon atoms of six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C317/00Sulfones; Sulfoxides
    • C07C317/24Sulfones; Sulfoxides having sulfone or sulfoxide groups and doubly-bound oxygen atoms bound to the same carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/56Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D307/58One oxygen atom, e.g. butenolide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/04Systems containing only non-condensed rings with a four-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/06Systems containing only non-condensed rings with a five-membered ring
    • C07C2601/08Systems containing only non-condensed rings with a five-membered ring the ring being saturated

Definitions

  • This invention relates to methods of treating cyclooxygenase mediated diseases and certain pharmaceutical compositions therefor.
  • Non-steroidal, antiinflammatory drugs exert most of their antiinflammatory, analgesic and antipyretic activity and inhibit hormone-induced uterine contractions and certain types of cancer growth through inhibition of prostaglandin G H synthase, also known as cyclooxygenase.
  • cyclooxygenase- 1 COX-1
  • constitutive enzyme as originally identified in bovine seminal vesicles.
  • COX-2 cyclooxygenase-2
  • COX-2 cyclooxygenase-2
  • This enzyme is distinct from the COX-1 which has been cloned, sequenced and characterized from various sources including the sheep, the mouse and man.
  • the second form of cyclooxygenase, COX-2 is rapidly and readily inducible by a number of agents including mitogens, endotoxin, hormones, cytokines and growth factors.
  • COX-1 constitutive enzyme
  • COX-2 inducible form
  • a selective inhibitor of COX-2 will have similar antiinflammatory, antipyretic and analgesic properties to a conventional non-steroidal antiinflammatory drug, and in addition would inhibit hormone-induced uterine contractions and have potential anti-cancer effects, but will have a diminished ability to induce some of the mechanism-based side effects.
  • such a compound should have a reduced potential for gastrointestinal toxicity, a reduced potential for renal side effects, a reduced effect on bleeding times and possibly a lessened ability to induce asthma attacks in aspirin-sensitive asthmatic subjects.
  • Such a compound will also inhibit prostanoid-induced smooth muscle contraction by preventing the synthesis of contractile prostanoids and hence may be of use in the treatment of dysmenorrhea, premature labour, asthma and eosinophil related disorders. It will also be of use in the treatment of Alzheimer's disease, for decreasing bone loss particularly in postmenopausal women (i.e. treatment of osteoporosis) and for the treatment of glaucoma.
  • the potential utilities of selective cyclooxygenase-2 inhibitors are discussed in the following articles:
  • the invention encompasses the novel compound of Formula I as well as a method of treating COX-2 mediated diseases comprising administration to a patient in need of such treatment of a non- toxic therapeutically effective amount of a compound of Formula I.
  • the invention also encompasses certain pharmaceutical compositions for treatment of COX-2 mediated diseases comprising compounds of Formula I.
  • the invention encompasses the novel compound of Formula I as well as a method of treating COX-2 mediated diseases comprising administration to a patient in need of such treatment of a non-toxic therapeutically effective amount of a compound of Formula I.
  • X is (a) O
  • Ar is (a) phenyl or
  • each of Rl, R ⁇ , R ? or R ⁇ is independently
  • each of R ⁇ or R ⁇ is independently (a) hydrogen, (b) halo,
  • R 7 is (a) NH2 or
  • a preferred embodiment of the invention is that wherein X is O.
  • Another preferred embodiment of the invention is that wherein Y is O.
  • Another preferred embodiment of the invention is that wherein X and Y are both O.
  • Another preferred embodiment of the invention is that wherein Rl and R ⁇ are both CH3.
  • R ⁇ is hydrogen or F.
  • Another preferred embodiment of the invention is that wherein R? is CH3.
  • OXONETM 2KHSO5 - KHSO4 - K 2 SO
  • alkyl means linear, branched or cyclic structures and combinations thereof, containing the indicated number of carbon atoms.
  • alkyl groups include methyl, ethyl, cyclopropyl, isopropyl, butyl, t-butyl, pentyl, hexyl, cyclohexyl, heptyl, pentadecyl, eicosyl, 3,7-diethyl-2,2-dimethyl- 4-propylnonyl, and the like.
  • Alkoxy means alkoxy groups of the indicated number of carbon atoms of a straight, branched, or cyclic configuration.
  • alkoxy groups include methoxy, ethoxy, propoxy, isopropoxy, cyclopropyloxy, cyclohexyloxy, and the like.
  • Alkylthio means alkylthio groups of the indicated number of carbon atoms of a straight, branched or cyclic configuration. Examples of alkylthio groups include methylthio, propylthio, isopropylthio, cycloheptylthio, etc. By way of illustration, the propylthio group signifies -SCH2CH2CH3.
  • Halogen includes F, Cl, Br, and I.
  • the invention encompasses pharmaceutical compositions for inhibiting COX-2 and for treating COX-2 mediated diseasesas disclosed herein comprising a pharmaceutically acceptable carrier and non-toxic therapeutically effective amount of a compound of formula I as described above.
  • the invention encompasses a method of inhibiting cyclooxygenase and treating cyclooxygenase mediated diseases, advantageously treated by an active agent that selectively inhibits COX-2 in preference to COX-1 as disclosed herein comprising: administration to a patient in need of such treatment of a non-toxic therapeutically effective amount of a compound of Formula I as disclosed herein.
  • Some of the compounds described herein contain one or more asymmetric centres 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.
  • salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids.
  • acids include acetic, adipic, aspartic, 1,5-naphthalenedisulfonic, benzenesulfonic, benzoic, camphorsulfonic, citric, 1,2-ethanedisulfonic, ethanesulfonic, ethylenediaminetetraacetic, fumaric, glucoheptonic, gluconic, glutamic, hydriodic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, 2- naphthalenesulfonic, nitric, oxalic, pamoic, pantothenic, phosphoric, pivalic, propionic, salicylic, stearic, succinic, sulfuric, tartaric, p- toluenesulfonic acid,
  • the Compound of Formula I is useful for the relief of 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, dysmenorrhea, headache, toothache, sprains and strains, myositis, neuralgia, synovitis, arthritis, including rheumatoid arthritis, degenerative joint diseases
  • Compound 1 may inhibit cellular neoplastic transformations and metastic tumour growth and hence can be used in the treatment of cancer.
  • Compound 1 may also be of use in the treatment and/or prevention of cyclooxygenase-mediated proliferative disorders such as may occur in diabetic retinopathy and tumour angiogenesis.
  • Compound I will also inhibit prostanoid-induced smooth muscle contraction by preventing the synthesis of contractile prostanoids and hence may be of use in the treatment of dysmenorrhea, premature labour, asthma and eosinophil related disorders. It will also be of use in the treatment of Alzheimer's disease, for decreasing bone loss particularly in postmenopausal women (i.e. treatment of osteoporosis) and for treatment of glaucoma.
  • compound I By virtue of its high inhibitory activity against COX-2 and/or its specificity for COX-2 over COX-1, compound I will prove useful as an alternative to conventional NSAID'S, particularly where such non- steroidal antiinflammatory drugs may be contra-indicated 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 anaemia such as hypoprothrombinemia, haemophilia or other bleeding problems; kidney disease; those prior to surgery or taking anticoagulants.
  • non-steroidal antiinflammatory drugs may be contra-indicated 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 anaemia such as hypoprothrombinemia, haemophilia or other bleeding problems; kidney disease; those prior to surgery or taking anti
  • compound 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.
  • warmblooded animals such as mice, rats, horses, cattle sheep, dogs, cats, etc.
  • pharmaceutical compositions for treating COX-2 mediated diseases as defined may optionally include one or more ingredients as listed above.
  • 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 absorption 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.
  • 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 or 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 or 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, hydroxy- propylmethycellulose, sodium alginate, polyvinyl-pyrrolidone, 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
  • 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.
  • preservatives for example ethyl, or n-propyl, p-hydroxybenzoate
  • colouring agents for example ethyl, or n-propyl, p-hydroxybenzoate
  • flavouring agents such as sucrose, saccharin or aspartame.
  • 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 emulsions.
  • 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 polyoxy-ethylene 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 purpose 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.
  • Compound I may also be administered in the form of a 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 ordinary 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 ordinary 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. (For purposes 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.
  • Dosage levels of the order of from about 0.01 mg to about 140 mg/kg of body weight per day are useful in the treatment of the above- indicated conditions, or alternatively about 0.5 mg to about 7 g per patient per day.
  • inflammation may be effectively treated by the administration of from about 0.01 to 50 mg of the compound per kilogram of body weight per day, or alternatively about 0.5 mg to about 3.5 g per patient per day.
  • 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 500 mg 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.
  • compositions for treating COX-2 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 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 dextramethorphan
  • the invention encompasses a method of treating cyclooxygenase mediated diseases comprising: administration to a patient in need of such treatment a non-toxic therapeutically effect amount of the compound of Formula I, optionally co-administered with one or more of such ingredients as listed immediately above.
  • the compounds of the present invention can be prepared according to the following methods.
  • Lactone 4 is condensed with 3 using a suitable base such as LDA to give alcohol 5.
  • Alcohol 5 is dehydrated to 6 using an acid, such as TfOH or CSA or by conversion to the mesylate; 6 is obtained as a mixture of the E and Z isomers.
  • Sulfide 6 is oxidized to the sulfone mixture 7 using MMPP or OXONETM , followed by chromatographic separation to obtain compound la.
  • the mixture of isomers 6 can be separated and the desired precursor to la oxidized.
  • the undesired isomer of 6, and compound 8, can be isomerized to the desired isomers by treatment with a protonic acid or by photochemical means.
  • the ketone 12 (Method 2) is used in place of benzophenone 4.
  • the oxidation step (6 - 7) is done with OXONETM in order to avoid formation of a pyridine-N-oxide.
  • Bromothioanisole 9 is transmetallated with an alkyl lithium, followed by reaction with aldehyde 10, to produce alcohol 11. Oxidation with a mild reagent such as Mn ⁇ 2 in ethyl acetate gives ketone 12, which is then used in place of benzophenone 3 in Method 1.
  • a Friedel-Crafts reaction between acid chloride 13 and thioanisole 2 yields the ketone 14, which upon reaction with an aryl lithium 15 yields tertiary alcohol 16.
  • Dehydration of the latter with a proton acid produces a mixture of the stereoisomers 17, which is oxidized to the mixture of sulfones 18 by MMPP or other suitable oxidant.
  • Allylic oxidation with a reagent such as chromium trioxide yields 19 as a mixture of isomers.
  • Chromatographic separation then yields the stereoisomers 20 and lb. 13 14
  • the intermediate 6 can be selectively oxidized to sulfoxide 21 by using one equivalent of an oxidizing agent such as
  • the free thiol 22 is obtained by reacting 21 with trifluoroacetic anhydride, followed by treatment with triethylamine in methanol. Oxidation of thiol 22 with CI2 in acetic acid yields a sulfonyl chloride intermediate, which reacts with ammonia to yield the isomeric mixture of sulfonamide 23. The isomers are separated by chromatography to yield 24 and the desired Ic.
  • Example 15 Example 20 Example 21 Example 23
  • the compound of Formula I can be tested using the following assays to determine their COX-2 inhibiting activity.
  • CHO transfected cell lines Chinese hamster ovary (CHO) cell lines which have been stably transfected with an eukaryotic expression vector pCDNAIII containing either the human COX-1 or COX-2 cDNA's are used for the assay. These cell lines are referred to as CHO [hCOX-1] and CHO [hCOX-2], respectively.
  • CHO[hCOX-l] cells from suspension cultures and CHO[hCOX-2] cells prepared by trypsinization of adherent cultures are harvested by centrifugation (300 x g, 10 min) and washed once in HBSS containing 15 mM HEPES, pH 7.4, and resuspended in HBSS, 15 mM HEPES, pH 7.4, at a cell concentration of 1.5 x 106 cells/ml.
  • Drugs to be tested are dissolved in DMSO to 66.7-fold the highest test drug concentration. Compounds are typically tested at 8 concentrations in duplicate using serial 3-fold serial dilutions in DMSO of the highest drug concentration.
  • Cells are then challenged in the presence or absence of drug with the AA/HBSS solution to yield a final concentration of 0.5 ⁇ M AA in the CHO[hCOX-l] assay and a final concentration of 10 ⁇ M AA in the CHO[hCOX-2] assay.
  • the reaction is terminated by the addition of 10 ⁇ l 1 N HCl followed by neutralization with 20 ⁇ l of 0.5 N NaOH.
  • the samples are centrifuged at 300 x g at 4C for 10 min, and an aliquot of the clarified supernatant is appropriately diluted for the determination of PGE2 levels using an enzyme-linked immunoassay for PGE2 (Correlate PGE2 enzyme immunoassay kit, Assay Designs, Inc.).
  • Cyclooxygenase activity in the absence of test compounds is determined as the difference in PGE2 levels of cells challenged with arachidonic acid versus the PGE2 levels in cells mock- challenged with ethanol vehicle. Inhibition of PGE2 synthesis by test compounds is calculated as a percentage of the activity in the presence of drug versus the activity in the positive control samples.
  • U 937 cells are pelleted by centrifugation at 500 x g for 5 min and washed once with phosphate-buffered saline and repelleted.
  • Cells are resuspended in homogenization buffer consisting of 0.1 M Tris-HCl, pH 7.4, 10 mM EDTA, 2 ⁇ g/ml leupeptin, 2 ⁇ g/ml soybean trypsin inhibitor, 2 ⁇ g/ml aprotinin and 1 mM phenyl methyl sulfonyl fluoride.
  • the cell suspension is sonicated 4 times for 10 sec and is centrifuged at 10,000 x g for 10 min at 4° C.
  • the supernatant is centrifuged at 100,000 x g for 1 hr at 4° C.
  • the 100,000 x g microsomal pellet is resuspended in 0.1 M Tris-HCl, pH 7.4, 10 mM EDTA to approximately 7 mg protein ml and stored at -80° C.
  • Microsomal preparations are thawed immediately prior to use, subjected to a brief sonication, and then diluted to a protein concentration of 125 ⁇ g/ml in 0.1 M Tris-HCl buffer, pH 7.4 containing 10 mM EDTA, 0.5 mM phenol, 1 mM reduced glutathione and 1 ⁇ M hematin. Assays are performed in duplicate in a final volume of 250 ⁇ l. Initially, 5 ⁇ l of DMSO vehicle or drug in DMSO are added to 20 ⁇ l of 0.1 M Tris-HCl buffer, pH 7.4 containing 10 mM EDTA in wells of a 96- deepwell polypropylene titre plate.
  • the enzyme activity is measured using a chromogenic assay based on the oxidation of N,N,N',N'-tetramethyl-p- phenylenediamine (TMPD) during the reduction of PGG2 to PGH2 by COX-2 (Copeland et al. (1994) Proc. Natl. Acad. Sci. 91, 11202-11206). Recombinant human COX-2 is purified from Sf9 cells as previously described (Percival et al (1994) Arch. Biochem. Biophys. 15, 111-118).
  • TMPD N,N,N',N'-tetramethyl-p- phenylenediamine
  • the assay mixture (180 ⁇ L) contains 100 mM sodium phosphate, pH 6.5, 2 mM genapol X-100, 1 ⁇ M hematin, 1 mg/ml gelatin , 80-100 units of purified enzyme (One unit of enzyme is defined as the amount of enzyme required to produce an O.D. change of 0.001/min at 610 nm) and 4 ⁇ L of the test compound in DMSO.
  • the mixture is preincubated at room temperature (22°C) for 15 minutes prior to initiation of the enzymatic reaction by the addition of 20 ⁇ L of a sonicated solution of 1 mM arachidonic acid (AA) and 1 mM TMPD in assay buffer (without enzyme or hematin).
  • the enzymatic activity is measured by estimation of the initial velocity of TMPD oxidation over the first 36 sec of the reaction. A non-specific rate of oxidation is observed in the absence of enzyme (0.007 - 0.010 O.D. /min) and is subtracted before the calculation of the % inhibition.
  • IC50 values are derived from 4-parameter least squares non-linear regression analysis of the log-dose vs % inhibition plot.
  • Rationale Human whole blood provides a protein and cell-rich milieu appropriate for the study of biochemical efficacy of anti-inflammatory compounds such as selective COX-2 inhibitors.
  • This assay can be used to evaluate the inhibitory effect of selective COX-2 inhibitors on PGE2 production.
  • platelets in whole blood contain a large amount of the COX-1 enzyme. Immediately following blood clotting, platelets are activated through a thrombin-mediated mechanism.
  • TxB2 thromboxane B2
  • COX-1 thromboxane B2
  • the degree of selectivity by the test compound can be determined by measuring the levels of PGE2 after LPS induction (COX-2) and TxB2 following blood clotting (COX-1) in the same assay.
  • Fresh blood is collected in heparinized tubes by venipuncture from both male and female volunteers. The subjects have no apparent inflammatory conditions and have not taken any NSAIDs for at least 7 days prior to blood collection. Plasma is immediately obtained from a 2mL blood aliquot to use as blank (basal levels of PGE2).
  • the remaining blood is incubated with LPS (100 ⁇ g/ml final concentration, Sigma Chem, #L-2630 from E. coli; diluted in 0.1% BSA (Phosphate buffered saline) for 5 minutes at room temperature.
  • LPS 100 ⁇ g/ml final concentration
  • BSA Phosphate buffered saline
  • Five hundred ⁇ L aliquots of blood are incubated with either 2 ⁇ L of vehicle (DMSO) or 2 ⁇ L of a test compound at final concentrations varying from lOnM to 30 ⁇ M for 24 hours at 37°C.
  • the blood is centrifuged at 12,000 x g for 5 minutes to obtain plasma.
  • a lOO ⁇ L aliquot of plasma is mixed with 400 ⁇ L of methanol for protein precipitation.
  • the supernatant is obtained and is assayed for PGE2 using a radioimmunoassay kit (Amersham, RPA#530) after conversion of PGE2 to its methyl
  • mice Male Sprague-Dawley rats (150-200 g) are fasted overnight and are given, po, either vehicle (1% methocel or 5% Tween 80) or a test compound. One hr later, a line is drawn using a permanent marker at the level above the ankle in one hind paw to define the area of the paw to be monitored. The paw volume (Vo) is measured using a plethysmometer (Ugo-Basile, Italy) based on the principle of water displacement. The animals are then injected subplantarly with 50 ⁇ l of 1% carrageenan solution in saline (FMC Corp, Maine) into the paw using an insulin syringe with a 25-gauge needle (i.e.
  • NSAIDs The major side effect of conventional NSAIDs is their ability to produce gastric lesions in man. This action is believed to be caused by inhibition of Cox-1 in the gastrointestinal tract. Rats are particularly sensitive to the actions of NSAIDs. In fact, rat models have been used commonly in the past to evaluate the gastrointestinal side effects of current conventional NSAIDs. In the present assay, NSAID-induced gastrointestinal damage is observed by measuring fecal 5lGr excretion after systemic injection of 51Cr-labeled red blood cells. Fecal 5lGr excretion is a well-established and sensitive technique to detect gastrointestinal integrity in animals and man.
  • Male Sprague Dawley rats (150 - 200 g) are administered orally a test compound either once (acute dosing) or b.i.d. for 5 days
  • the rats are injected via a tail vein with 0.5 mL of 51Cr-labeled red blood cells from a donor rat.
  • the animals are placed individually in metabolism cages with food and water ad lib. Feces are collected for a 48 h period and 51Cr fecal excretion is calculated as a percent of total injected dose.
  • 51Cr-labeled red blood cells are prepared using the following procedures. Ten mL of blood is collected in heparinized tubes via the vena cava from a donor rat. Plasma is removed by centrifugation and replenished with equal volume of HBSS.
  • the red blood cells are incubated with 400 ⁇ Ci of sodium bichromate for 30 min. at 37°C. At the end of the incubation, the red blood cells are washed twice with 20 mL HBSS to remove free sodium 51 c hromate. The red blood cells are finally reconstituted in 10 mL HBSS and 0.5 mL of the solution (about 20 ⁇ Ci) is injected per rat.
  • Protein-losing gastropathy (manifested as appearance of circulating cells and plasma proteins in the GI tract) is a significant and dose-limiting adverse response to standard non-steroidal anti- inflammatory drugs (NSAIDs). This can be quantitatively assessed by intravenous administration of 51CrCl3 solution. This isotopic ion can avidly bind to cell and serum globins and cell endoplasmic reticulum. Measurement of radioactivity appearing in feces collected for 24 h after administration of the isotope thus provides a sensitive and quantitative index of protein-losing gastropathy.
  • NSAIDs non-steroidal anti- inflammatory drugs
  • Groups of male squirrel monkeys (0.8 to 1.4 kg) are treated by gavage with either 1% methocell or 5% Tween 80 in H2O vehicles, (3mL/kg b.i.d.) or test compounds at doses from 1 - 100 mg/kg b.i.d. for 5 days.
  • Intravenous 51Cr (5 ⁇ Ci kg in 1 ml kg phosphate buffer saline (PBS)) is administered 1 h after the last drug/vehicle dose, and feces collected for 24 h in a metabolism cage and assessed for excreted 51Cr by gamma-counting.
  • Venous blood is sampled 1 h and 8 h after the last drug dose, and plasma concentrations of drug measured by RP-HPLC.
  • saline or LPS 2 mg/kg, Sigma Chem
  • the LPS-injected rats were given either the vehicle (1% methocel) or a test compound orally to determine whether the compound could reverse the pyrexia. Percent reversal of the pyrexia was calculated using the rectal temperature obtained at 7 h in the control (vehicle-treated) group as the reference (zero reversal) point. Complete reversal of pyrexia to the pre-LPS baseline value is taken as 100%. LPS-Induced Pyrexia in Conscious Squirrel Monkeys
  • Temperature probes were surgically implanted under the abdominal skin in a group of squirrel monkeys (Saimiri sciureus) (1.0 - 1.7 kg). This allows for the monitoring of body temperature in conscious, unrestrained monkeys by a telemetric sensing system (Data Sciences International, Minnesota). The animals were fasted and were placed in individual cages for acclimatization 13 - 14 h before use. Electronic receivers were installed on the side of the cages which pick up signals from the implanted temperature probes. At approximately 9:00 a.m. on the day of the experiment, the monkeys were restrained temporarily in training chairs and were given a bolus IN. injection of LPS, (6 ⁇ g/kg, dissolved in sterile saline).
  • the animals were returned to their cages and body temperature was recorded continuously every 5 min. Two h after injection of LPS, when the body temperature had increased by 1.5 - 2°C, the monkeys were dosed orally with either vehicle (1% methocel) or a test compound (3 mg/kg). One hundred minutes later, the difference between the body temperature and the baseline value was determined. Percent inhibition was calculated taking the value in the control group as 0% inhibition.
  • Hyperalgesia was determined by subtracting the vocalisation threshold in saline injected rats from that obtained in animals injected with carrageenan. Hyperalgesia scores for drug-treated rats were expressed as a percentage of this response. ID50 values (the dose producing 50% of the maximum observed response) were then calculated by nonlinear least squares regression analysis of mean data using GraFit (Erithacus Software).
  • mice Seventy, 6.5-7.5 week old, 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 Mycobacterium 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.
  • the animals are housed, fed and cared for according to the Guidelines of the Canadian Council on Animal Care.
  • the rats are placed in the restrainer one at a time and the box firmly secured.
  • the zero blood sample is obtained by nicking a small (1 mm or less) piece off the tip of the tail.
  • the tail is then stroked with a firm but gentle motion from the top to the bottom to milk out the blood.
  • Approximately 1 mL of blood is collected into a heparinized vacutainer tube.
  • Typical time points for determination of rat blood levels after PO dosing are:
  • the following vehicles may be used in PO rat blood level determinations :
  • PEG 200/300/400 restricted to 2 mL/kg Methocel 0.5% - 1.0%: lOmL/kg Tween 80: lOmL/kg
  • Compounds for PO blood levels can be in suspension form.
  • the solution can be placed in a sonicator for approximately 5 minutes.
  • the animals are housed, fed and cared for according to the Guidelines of the Canadian Council on Animal Care.
  • mice Male Sprague Dawley (325-375 g) rats are placed in plastic shoe box cages with a suspended floor, cage top, water bottle and food.
  • the compound is prepared as required, in a standard dosing volume of 1 mL/kg.
  • Rats are bled for the zero blood sample and dosed under CO2 sedation.
  • the rats one at a time, are placed in a primed CO2 chamber and taken out as soon as they have lost their righting reflex.
  • the rat is then placed on a restraining board, a nose cone with CO2 delivery is placed over the muzzle and the rat restrained to the board with elastics.
  • forceps and scissors With the use of forceps and scissors, the jugular vein is exposed and the zero sample taken, followed by a measured dose of compound which is injected into the jugular vein.
  • Light digital pressure is applied to the injection site, and the nose cone is removed. The time is noted. This constitutes the zero time point.
  • the 5 min bleed is taken by nicking a piece (1-2 mm) off the tip of the tail.
  • the tail is then stroked with a firm but gentle motion from the top of the tail to the bottom to milk the blood out of the tail.
  • Typical time points for determination of rat blood levels after I.V. dosing are either:
  • Vehicles The following vehicles may be used in TV rat blood level determinations:
  • DMSO dimethylsulfoxide
  • the units of CL are mL/h»kg (milliliters per hour kilogram)
  • NMR data is in the form of delta ( ⁇ ) values for major diagnostic protons, given in parts per million (ppm) relative to tetramethylsilane (TMS) as internal standard, determined at 300 MHz or 400 MHz using the indicated solvent; conventional abbreviations used for signal shape are: s. singlet; d. doublet; t. triplet; m. multiplet; br. broad; etc.: in addition "Ar" signifies an aromatic signal;
  • Examples 1, 5, 7, 9, 11, 13 and 17 were prepared according to the sequence of reactions described for Example 3 by replacing benzoyl chloride in Step 1 with the appropriate aroyl chloride and/or 4-methyl-4- hydroxyvaleric acid ⁇ -lactone in Step 2 with the appropriate ⁇ - butyrolactone.
  • Examples 2, 6, 8, 10, 12 and 14 were obtained by chromatographic separation at the Step 4 in the preparation of Examples 1, 5, 7, 9, 11 and 13 respectively.
  • Step 1 (4-(Methylthio)phenyl) phenyl ketone
  • a 3 L RBF equipped with a mechanical stirrer was charged with AICI3 (152 g) and CHCI3 (1.4 L) and cooled in an ice bath. Then benzoyl chloride (139 mL) was added over 0.5 h to the ice-cold suspension. Keeping the internal temp. ⁇ 10°C, thioanisole was added dropwise over 1 hr. After completion of addition, the resulting mixture was stirred at r.t. for 2 h. The resulting dark red-orange suspension was poured onto an ice-water mixture and stirred until decoloration.
  • Step 3 5.5-Dimethyl-f3(l-(4-(methylthio) ⁇ henyl)-l-phenyl- methylidenel tetrahydro-2-furanone. mixture of E and Z isomers.
  • Step 4 5.5-Dimethyl-3-r(E)-l-(4-(methylsulfonyl)phenyl)-l- phenylmethylideneltetrahydro-2-furanone
  • MMPP methylthio compound obtained from Step 3
  • CH2CL2 18 mL
  • MeOH 2 mL
  • MMPP 1165 mg of 80% pure
  • the resulting mixture was stirred at r.t. for 20 h.
  • the reaction was diluted with EtOAc (150 mL) and half saturated NaHCO3 (100 mL). The mixture was shaken vigorously, layers separated, the organic layer was dried over MgSO4 and concentrated.
  • Geometry of the olefin was assigned by a nOe experiments.
  • Geometry of the olefin was assigned by nOe experiments.
  • Step 2 [4-(Methylthio)phenyl1 (2-pyridyl) ketone
  • EtOAc 100 mL
  • manganese (IV) oxide 2.50 + 2.72 g
  • the resulting suspension was stirred at r.t. for a total of 2 days.
  • the reaction was filtered through a pad of celite, washed with EtOAc and concentrated.
  • the crude product was purified by crystallization from 5% EtOAc in hexane (100 mL) to give the title compound.
  • Step 4 5.5-Dimethyl-3-r(Z)-l-r4-(methylthio)phenyl1-l-(2- pyridyl)methylidene1tetrahydro-2-furanone
  • Step 5 5.5-Dimethyl-3-r(Z)-l-(4-(methylsulfonyl)phenyl)-l-(2- pyridyl)methylidene1tetrahydro-2-furanone
  • Step 2 CyclopentyH4-(methylsulfonyl)phenvnphenylmethanol A solution of the ketone obtained from Step 1 (3.52 g) in Et2 ⁇
  • Step 3 4- rCyclopentyliden(phenyl)methyl] phenyl methyl sulfide
  • Step 4 4-rCyclopentyliden(phenyl)methyl1phenyl methyl sulfone
  • Chromium trioxide (4.04 g) was suspended in CH2CI2 (35 mL) at -20°C and 3,5-dimethylpyrazole was added in one portion. After stirring for 0.25 h at -20°C, a solution of the compound described in Example 18 (615 mg) in CH2CI2 (4 mL) was added and the reaction mixture was stirred for 1 h while maintaining the temperature at -20°C. Sodium hydroxide (15 mL, 5N) was added and the mixture was stirred for 1 h at 0°C. The reaction was diluted with water and extracted with CH2CI2. The organic layer was washed once with IN HCl, once with water, once with brine, dried over MgSO4 and concentrated.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
EP98913487A 1997-04-02 1998-03-31 Alpha-methylen gamma lactone als selektive cyclooxygenase-2 hemmer Withdrawn EP0971910A1 (de)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US4253597P 1997-04-02 1997-04-02
US42535P 1997-04-02
GBGB9709356.1A GB9709356D0 (en) 1997-05-07 1997-05-07 Alpha-methylene gamma lactones as selective cyclooxygenase-2 inhibitors
GB9709356 1997-05-07
PCT/CA1998/000302 WO1998043966A1 (en) 1997-04-02 1998-03-31 Alpha-methylene gamma lactones as selective cyclooxygenase-2 inhibitors

Publications (1)

Publication Number Publication Date
EP0971910A1 true EP0971910A1 (de) 2000-01-19

Family

ID=26311502

Family Applications (1)

Application Number Title Priority Date Filing Date
EP98913487A Withdrawn EP0971910A1 (de) 1997-04-02 1998-03-31 Alpha-methylen gamma lactone als selektive cyclooxygenase-2 hemmer

Country Status (5)

Country Link
EP (1) EP0971910A1 (de)
JP (1) JP2001518087A (de)
AU (1) AU6818298A (de)
CA (1) CA2285567A1 (de)
WO (1) WO1998043966A1 (de)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6180651B1 (en) 1996-04-04 2001-01-30 Bristol-Myers Squibb Diarylmethylidenefuran derivatives, processes for their preparation and their uses in therapeutics
DK1303265T3 (da) 2000-07-20 2007-11-12 Lauras As Anvendelse af COX-2-inhibitorer, som immunostimulerende midler ved behandling af HIV eller AIDS
DK1406895T3 (da) 2001-06-13 2008-12-01 Magnachem Int Lab Inc Lactonformuleringer og fremgangsmåde til anvendelse deraf
AU2003287546A1 (en) 2002-11-05 2004-06-07 Magnachem International Laboratories, Inc. Synthetic lactone formulations and method of use
PL1534305T3 (pl) 2003-05-07 2007-03-30 Osteologix As Leczenie zaburzeń chrząstek i kości solami strontu rozpuszczalnymi w wodzie
WO2005102315A1 (en) 2004-04-23 2005-11-03 Magnachem International Laboratories, Inc. Synthetic lactone formulations for pain control
EP2350645A2 (de) 2008-10-24 2011-08-03 Magnachem International Laboratories, Inc. Verfahren zur suche nach mit rad9 selektiv wechselwirkenden verbindungen
IL305573A (en) 2021-03-15 2023-10-01 Saul Yedgar Hyaluronic acid conjugated with dipalmitoyl phosphatidyl ethanolamine in combination with non-steroidal anti-inflammatory drugs (NSAIDs) for the treatment or suppression of inflammatory diseases
WO2023150719A1 (en) * 2022-02-03 2023-08-10 Purdue Research Foundation Alpha-methylene and aminomethyl lactones and lactams for treatment of clostridioides difficile infection (cdi)

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2747123B1 (fr) * 1996-04-04 1998-06-26 Union Pharma Scient Appl Nouveaux derives diarylmethylidene tetrahydrofurane, leurs procedes de preparation, et leurs utilisations en therapeutique
FR2751964B1 (fr) * 1996-08-01 1998-10-30 Union Pharma Scient Appl Nouveaux derives diarylmethylene carbocycliques, leurs procedes de preparation, et leurs utilisations en therapeutique

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9843966A1 *

Also Published As

Publication number Publication date
WO1998043966A1 (en) 1998-10-08
AU6818298A (en) 1998-10-22
CA2285567A1 (en) 1998-10-08
JP2001518087A (ja) 2001-10-09

Similar Documents

Publication Publication Date Title
EP0912518B1 (de) Substituierte pyridine als selektive cyclooxygenase inhibitoren
US5922742A (en) Pyridinyl-2-cyclopenten-1-ones as selective cyclooxygenase-2 inhibitors
CA2234642C (en) 3,4-diaryl-2-hydroxy-2,5-dihydrofurans as prodrugs to cox-2 inhibitors
EP1012142B1 (de) 2,3,5-trisubstituierte pyridine als inhibitoren von cyclooxygenase-2
US6057319A (en) 3,4-Diaryl-2-hydroxy-2,5-dihydrofurans as prodrugs to cox-2 inhibitors
EP0975604B1 (de) Pyridazinone als inhibitoren von cyclooxygenase-2
US6046217A (en) 2,3,5-trisubstituted pyridines as inhibitors of cyclooxygenase-2
US6133292A (en) Diaryl-5-alkyl-5-methyl-2-(5H)-furanones as selective cyclooxygenase-2-inhibitors
EP0863134A1 (de) 2-(3,5-difluorophenyl)-3-(4-methylsulfonylphenyl)-2-cyclopenten-1-on zur Verwendung als Inhibitor der Cyclooxygenase-2
US6465509B2 (en) Pyrones as inhibitors of cyclooxygenase-2
WO1998043966A1 (en) Alpha-methylene gamma lactones as selective cyclooxygenase-2 inhibitors
AU2001268881A1 (en) Pyrones as inhibitors of cyclooxygenase-2
AU707199B2 (en) Diphenyl stilbenes as prodrugs to COX-2 inhibitors
CA2283100A1 (en) (methylsulfonyl)phenyl-2-(5h)-furanones with oxygen link as cox-2 inhibitors
AU741790B2 (en) Diaryl-5-alkyl-5-methyl-2(5H)-furanones as selective cyclooxygenase-2 inhibitors
US6004960A (en) Pyridazinones as inhibitors of cyclooxygenase-2
US6004950A (en) 2-aminopyridines as inhibitors of cyclooxygenase-2
EP1015431B1 (de) 2-aminopyridine als inhibitoren von cyclooxygenase-2
CA2252401C (en) Pyridinyl-2-cyclopenten-1-ones as selective cyclooxygenase-2 inhibitors
CA2202345A1 (en) 2-(3,5-difluorophenyl)-3-(4-(methylsulfonyl)phenyl)-2-cyclopenten-1-one useful as an inhibitor of cox-2

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19991102

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LI LU NL PT SE

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20001004