IE903874A1 - Substituted benzoylbenzene-, biphenyl- and 2-oxazole-¹alkanoic acid derivatives as inhibitors of pla2 and¹lipoxygenase - Google Patents

Substituted benzoylbenzene-, biphenyl- and 2-oxazole-¹alkanoic acid derivatives as inhibitors of pla2 and¹lipoxygenase

Info

Publication number
IE903874A1
IE903874A1 IE387490A IE387490A IE903874A1 IE 903874 A1 IE903874 A1 IE 903874A1 IE 387490 A IE387490 A IE 387490A IE 387490 A IE387490 A IE 387490A IE 903874 A1 IE903874 A1 IE 903874A1
Authority
IE
Ireland
Prior art keywords
arh
compound
ethyl acetate
acid
lower alkyl
Prior art date
Application number
IE387490A
Original Assignee
American Home Prod
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 American Home Prod filed Critical American Home Prod
Publication of IE903874A1 publication Critical patent/IE903874A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • 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
    • C07C205/00Compounds containing nitro groups bound to a carbon skeleton
    • C07C205/45Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by at least one doubly—bound oxygen atom, not being part of a —CHO group
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C255/00Carboxylic acid nitriles
    • C07C255/01Carboxylic acid nitriles having cyano groups bound to acyclic carbon atoms
    • C07C255/32Carboxylic acid nitriles having cyano groups bound to acyclic carbon atoms having cyano groups bound to acyclic carbon atoms of a carbon skeleton containing at least one six-membered aromatic ring
    • C07C255/40Carboxylic acid nitriles having cyano groups bound to acyclic carbon atoms having cyano groups bound to acyclic carbon atoms of a carbon skeleton containing at least one six-membered aromatic ring the carbon skeleton being further substituted by doubly-bound oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C45/00Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds
    • C07C45/004Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reaction with organometalhalides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C45/00Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds
    • C07C45/61Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups
    • C07C45/63Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups by introduction of halogen; by substitution of halogen atoms by other halogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C45/00Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds
    • C07C45/61Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups
    • C07C45/67Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups by isomerisation; by change of size of the carbon skeleton
    • C07C45/68Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups by isomerisation; by change of size of the carbon skeleton by increase in the number of carbon atoms
    • C07C45/72Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups by isomerisation; by change of size of the carbon skeleton by increase in the number of carbon atoms by reaction of compounds containing >C = O groups with the same or other compounds containing >C = O groups
    • 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
    • C07C59/76Unsaturated compounds containing keto groups
    • C07C59/90Unsaturated compounds containing keto groups containing singly bound oxygen-containing groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/66Esters of carboxylic acids having esterified carboxylic groups bound to acyclic carbon atoms and having any of the groups OH, O—metal, —CHO, keto, ether, acyloxy, groups, groups, or in the acid moiety
    • C07C69/73Esters of carboxylic acids having esterified carboxylic groups bound to acyclic carbon atoms and having any of the groups OH, O—metal, —CHO, keto, ether, acyloxy, groups, groups, or in the acid moiety of unsaturated acids
    • C07C69/738Esters of keto-carboxylic acids or aldehydo-carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/12Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D215/14Radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/02Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings
    • C07D263/30Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D263/32Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Pulmonology (AREA)
  • Immunology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Nitrogen And Oxygen As The Only Ring Hetero Atoms (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Abstract

There are disclosed compounds of the formula: A(CH2)nO-B, wherein A is phenoxyethyl, phenoxyphenyl or a group having formula (a), wherein X is -N- or (b); Z is (c), (d), (e), (f), -S- or -O-; R<1> is hydrogen, lower alkyl or phenyl; R<2> is hydrogen or lower alkyl; or R<1> and R<2> taken together form a benzene ring; R<3> is hydrogen or lower alkyl; n is 1 -2; B is (g), (h), (i) or (j), wherein R<4> and R<5> are each, independently, hydrogen or lower alkyl; R<6> is halo or nitro; R<7> is (k) or (l); R<8> is lower alkyl; m is 0-3; and the pharmacologically acceptable salts thereof, and their use in the treatment of inflammatory conditions, such as rheumatoid arthritis, ulcerative colitis, psoriasis and other immediate hypersensitivity reactions; in the treatment of leukotriene-mediated naso-bronchial obstructive air-passageway conditions, such as allergic rhinitis, allergic bronchial asthma and the like; and as gastric cytoprotective agents.

Description

SUBSTITUTED BENZOYLBENZENE-, BIPHENYLAND 2-OXAZOLE- ALKANOIC ACID DERIVATIVES AS INHIBITORS OF PLA2 AND LIPOXYGENASE This invention relates to novel substituted benzoylbenzene-, biphenyl- and 2oxazole- alkanoic acid derivatives possessing lipoxygenase inhibitory', phospholipase A2 inhibitory and leukotriene antagonist activity, which are useful as anti-inflammatory, antiallergic and cytoprotective agents.
It is now well-established that arachidonic acid (AA) is metabolized in mammals by two distinct pathways. The metabolism of arachidonic acid by cyclooxygenase enzymes results in the production of prostaglandins and thromboxanes. The physiological activity of the prostaglandins has already been amply elucidated in recent years. It is now known that prostaglandins arise from the endoperoxides PGG2 and PGH2 by the cyclooxygenase pathway of arachidonic acid metabolism. These endoperoxides are also the precursors of the thromboxanes (Tx) A2 and B2. TxA2 is a vasoconstrictor which stimulates platelet aggregation. In the normal situation, the vasoconstrictive and platelet aggregating properties of the thromboxanes are balanced by another product arising from the endoperoxides in the cyclooxygenase pathway, prostacyclin (PGI2), which is a vasodilator with platelet aggregation inhibitory activity. In the event prostacyclin synthesis is impaired and/or platelet activation is enhanced, then thrombosis and vasoconstriction is favored. The role of prostanoids in haemostasis and thrombosis are reviewed by R.J. Gryglewski, CRC Crit, Rev. Biochem., 2. 291 (1980) and J.
B. Smith, Am. J. Pathol,. 99, 743 (1980). Cyclooxygenase metabolites are known to participate directly in the inflammatory response [see Higgs et al., Annals of Clinical Research, J_6, 287-299 (1984)]. This is through their vasodepressor activities, participation in pain and fever augmentation of peptide mediator vascular permeability and edema forming properties. Finally, various aspects of cell mediated immunity are influenced by cyclooxygenase products.
The other pathway of AA metabolism involves lipoxygenase enzymes and results in the production of a number of oxidative products called leukotrienes. The latter are designated by the LT nomenclature system, and the most significant products of the lipoxygenase metabolic pathway are the leukotrienes B4, C4 and D4. The substance denominated slow-reacting substance of anaphylaxis (SRS-A) has been shown to consist of a mixture of leukotrienes, with LTC4 and LTD4 as the primary products and having varying amounts of other leukotriene metabolites [see Bach et al., J. Immun., 215, 115-118 (1980); Biochem. Biophvs. Res. Commun.. 93. 1121-1126 (1980)].
The significance of these leukotrienes is that a great deal of evidence has been accumulated showing that leukotrienes participate in inflammatory reactions, exhibit chemoIE 903874 October 1989 3ED2:OCO:GT:jj AHP-9498 PATENT -2- tactic activities, stimulate lysosomal enzyme release and act as important factors in the immediate hypersensitivity reaction. It has been shown that LTC4 and LTD4 are potent bronchoconstrictors of the human bronchi [see Dahlen et al., Nature, 288. 484-486 (1980) and Piper, Int, Arch. Appl. Immunol., 76. suppl. 1, 43 (1985)] which stimulate the release of mucus from airways in vitro [Marom et al., Am. Rev. Resp. Dis.. 126. 449 (1982)], are potent vasodilators in skin [see Bisgaard et al.. Prostaglandins. 23. 797 (1982)], and produce a wheal and flare response [Camp et al., Br, J, Pharmacol.. 80. 497 (1983)]. The nonpeptide leukotriene, LTB4, is a powerful chemotactic factor for leukocytes [see A. W. Ford-Hutchinson, J. Roy, Soc. Med., 74. 831-833 (1981), which stimulates cell accumulation and affects vascular smooth muscle [see Bray, Br. Med, Bull., 39, 249 (1983)]. The activity of leukotrienes as mediators of inflammation and hypersensitivity is extensively reviewed in Bailey and Casey, Ann. Reports Med. Chem., 19. 87 (1986).
Phospholipase A2 (PLA2) is the critical rate limiting enzyme in the arachidonic acid (AA) cascade since it is responsible for the hydrolysis of esterified AA from the C-2 position of membrane phospholipids. This reaction generates two products (1) free AA which is then available for subsequent metabolism by either the cyclooxygenase or lipoxygenase enzymes and (2) lysophospholipid. When alkyl-arachidonoyl-glycerophosphatidylcholine is acted upon by the PLA2 the generation of platelet activating factor (PAF) is initiated; PAF is pro-inflammatory in its own right [see Wedmore et al., Br. J. Pharmacol.. 74. 916-917 (1981)]. In this regard it may be noted that the anti-inflammatory steroids are thought to inhibit eicosanoid synthesis by inducing the synthesis of a PLA2 inhibitory protein denominated macrocortin or lipomodulin [see Flower et al., Nature. London, 278.456 (1979) and Hirata et al., Proc. Natn. Acad. Sci. U.S.A., 77, 2533 (1980)].
As the initial step leading to subsequent conversion of AA to the various eicosanoids by the cyclooxygenase and lipoxygenase pathways, the PLA2-mediated release of AA from membrane phospholipids is a critical event in attempting to deal with the various physiological manifestations which are based on the activity of the eicosanoids and/or PAF. Thus, while PLA2 has been shown to be required for platelet aggregation [Pickett et al., Biochem. J„ 160. 405 (1976)], cardiac contraction and excitation [Geisler et al., Pharm. Res. Commun.. 9,117 (1977)], as well as prostaglandin synthesis [Vogt, Adv, Prostagl. Thromb. Res.. 3, 89 (1978)], the inhibition of PLA2 is indicated in the therapeutic treatment of both PAF induced or cyclooxygenase and/or lipoxygenase pathway product-mediated physiological conditions.
There is also evidence that products of the cyclooxygenase/lipoxygenase pathways play key roles in both the pathogenesis of gastric mucosal damage due to extracellular (gastric and intestinal contents, microorganisms, and the like) or intracellular (ischemia, October 1989 3ED3:OCO:GT:jj AHP-9498 PATENT -3- viruses, etc.) agents, as well as in cytoprotection against such damage. Thus, on the one hand prostaglandins exert a cytoprotective effect on the gastric mucosa [see Robert, Gastroenterology. 77. 761-767 (1979)] and this action of the prostaglandins, especially of the E series, is considered to be of importance in the treatment of gastro-intestinal ulceration [see Isselbacher, Drugs, 33 (suppl.), 38-46 (1987)]. On the other hand, ex vivo experiments have shown that gastric mucosal tissue from ethanol-pretreated rats is capable of LTC4 generation and that this LTC4 production is quantitatively related to the severity of the ethanol damage [see Lange et al., Naunvn-Schmiedeberg's Arch, Pharmacol. Suppl.. 330. R27, (1985)]. It has also been demonstrated that LTC4 can induce vasoconstriction in both venous and arteriolar vessels in the rat submucosa [see Whittle, IUPHAR Ninth Int. Cong, of Pharm.. S30-2, London, England (1984)]. This is significant since ethanol-induced lesion formation in gastric mucosa may be multifactorial with, for example, stasis of gastric blood flow contributing significantly to the development of the hemorrhagic necrotic aspects of the tissue injury [see Guth et al., Gastroenterology. 87. 1083-90 (1984)]. Moreover, in the anesthetized cat, exogenous LTD4 evokes both increased pepsin secretion and decreased transgastric potential [Pendleton et al., Eur. J. Pharmacol.. 125. 297-99 (1986)]. A particularly significant recent finding in this regard is that 5-lipoxygenase inhibitors and some leukotriene antagonists protect the gastric mucosa against lesions induced by the oral or parenteral administration of most nonsteroidal anti-inflammatory drugs [see Rainsford, Agents and Actions, 21. 316-319 (1987)]. Platelet activating factor (PAF) is also implicated as a mediator of gastrointestinal damage, and it has been recently shown that 5-lipoxygenase inhibitors inhibit PAF-induced gastric mucosal damage (Gastroenterology. 96, A55, A434, 1989). Accordingly, a significant body of evidence implicates the involvement of lipoxygenase products in the development of pathological features associated with gastric mucosal lesions, such as for example, those induced by ethanol exposure and administration of non-steroidal anti-inflammatory drugs. Thus, compounds which inhibit the biological effects of leukotrienes and PAF and/or which control the biosynthesis of these substances, as by inhibiting 5-lipoxygenase, are considered to be of value as cytoprotective agents.
Accordingly, the biological activity of the leukotrienes and SRS's, and of lipoxygenase as the enzyme leading to the metabolism of AA to leukotrienes, indicates that a rational approach to drug therapy to prevent, remove or ameliorate the symptoms of allergies, anaphylaxis, asthma and inflammation and for gastric cytoprotection must focus on either blocking the release of mediators of these conditions or antagonizing their effects. Thus, compounds which inhibit the biological effects of the leukotrienes and SRS's and/or which control the biosynthesis of these substances, as by inhibiting the PLA2-mediated release of arachidonic acid from membrane phospholipids, or by inhibiting lipoxygenase, are considered October 1989 3ED4:OCO:GT:jj AHP-9498 PATENT -4- to be of value in treating such conditions as allergic bronchial asthma, allergic rhinitis, as well as in other immediate hypersensitivity reactions and in providing gastric cytoprotection.
It has now been found that certain novel substituted benzoylbenzene-, biphenvland 2-oxazole- alkanoic acid derivatives inhibit PLA2 and lipoxygenase, and antagonize products of the lipoxygenase pathway, and so are useful as anti-inflammatory, anti-allergic and cytoprotective agents. The present invention provides novel compounds having the following formula: A(CH2)nO-B wherein A is phenoxyethyl, phenoxyphenyl or a group having the formula Rl .X, R2' I •Z wherein R3 I X is -N- or -C-; R3 R3 R3 R3 R3 III II Z is -C=C- , -C=N- , -N=C- , -N-, -S- or -O-; R1 is hydrogen, lower alkyl or phenyl; R2 is hydrogen or lower alkyl; or R1 and R2 taken together form a benzene ring; R3 is hydrogen or lower alkyl; n is 1 - 2; B is Ο R6 Ο October 1989 3ED5:OCO:GT:jj AHP-9498 PATENT -5- wherein R4 and R5 are each, independently, hydrogen or lower alkyl; R6 is halo or nitro; O R411 8 1 5 R7 is -C-R8 or -CHCOOR5 ; R8 is lower alkyl; m is 0 - 3 ; and the pharmacologically acceptable salts thereof.
The term lower alkyl refers to moieties having 1 - 6 carbon atoms in the carbon chain. The term halo refers to fluoro, chloro or bromo.
The grouping A embraces, inter alia. 5- or 6- membered unsaturated nitrogen, sulfur or oxygen containing mono- or benzofused-heterocycles, optionally substituted with lower alkyl or phenyl. The foregoing definition embraces the following heterocyclic moieties: furyl, pyrrolyl, thienyl, oxazolyl, thiazolyl, imidazolyl, pyridyl, pyrazinyl, pyrimidinyl, benzofuranyl, benzothienyl, benzothiazolyl, indolyl, benzoxazolyl, quinolinyl, quinazolinyl, benzimidazolyl, quinoxalinyl, quinazolinyl and the like. Especially preferred are quinolinyl, benzothiazolyl, and benzimidazolyl.
The compounds of the invention can form pharmacologically acceptable salts from pharmacologically acceptable organic and inorganic acids such as hydrochloric, hydrobromic, sulfonic, sulfuric, phosphoric, nitric, maleic, fumaric, benzoic, ascorbic, pamoic, succinic, methanesulfonic, acetic, propionic, tartaric, citric? lactic, malic, mandelic, cinnamic, palmitic, itaconic and benzenesulfonic. The compounds which are carboxylic acids are capable of forming alkali metal and alkaline earth carboxylates and carboxylates of pharmacologically acceptable cations derived from ammonia or a basic amine. Examples of the latter include but are not limited to cations such as ammonium, mono-, di-, and trimethylammonium, mono-, diand triethylammonium, mono-, di- and tripropylammonium (iso and normal), ethyldimethylammonium, benzyldimethylammonium, cyclohexylammonium, benzylammonium, dibenzylammonium, piperidinium, morpholinium, pyrrolidinium, piperazinium, 1-methylpiperidinium, 4-ethylmorpholinium, 1-isopropylpyrrolidinium, 1,4-dimethylpiperazinium, 1-n-butyl-piperidinium, 2-methylpiperidinium, l-ethyl-2-methylpiperidinium, mono-, di- and triethanolammonium, ethyl diethanolammonium, n-butylmonoethanolammonium, tris(hydroxymethyl)methylammonium, phenylmonoethanolammonium, and the like.
The compounds of the invention can be prepared by the following reaction schemes. When it is desired to prepare compounds having the formula October 1989 3ED6:OCO:GT:jj AHP-9498 PATENT O 4-methoxybenzonitrile, for example, is reacted with 3-bromotoluene, followed by reaction with bromine in ethylene bromide to yield the intermediate 3-bromomethyl-[4'-methoxy] benzophenone.
II O to yield the hydroxy carboxylic acid intermediate: The bromo intermediate is reacted with sodium cyanide to yield the cyano intermediate, which is hydrolyzed in the presence of base to yield the carboxylic acid, which in turn is demethylated NaCN dioxane / H2O CH3°y^ CH3°x^\ -CHoCN KOH Δ II O October 1989 3ED7:OCO:GT:jj AHP-9498 PATENT The hydroxy carboxylic acid intermediate is converted to the methyl ester with methanol in the presence of p-toluenesulfonic acid followed by reaction with an appropriate haloalkyl-A compound, where A is as defined hereinbefore and hal is halo, to yield the desired final product as the methyl ester.
HO. ch2cooh ch3oh p-toluenesulfonic acid The ester can be hydrolyzed by conventional methods to yield the desired final product in free carboxylic acid form.
October 1989 3ED8:OCO:GT:jj AHP-9498 PATENT -8- Compounds of the invention having the formula can be prepared by several routes. Compounds in which R6 is nitro and R7 is the -C-R8 moiety can be prepared as follows; for example: 4-bromo-3-nitroacetophenone is reacted with 4-iodoanisole in the presence of copper bronze, to yield the intermediate methoxy-containing biphenyl, which is demethylated with aluminum bromide to yield the hydroxy intermediate The latter is then reacted with an appropriate haloalkyl-A compound, where A is as defined hereinbefore and hal is fcalo, to yield the desired final product.
Compounds in which R6 is halo and R7 is the -CHCOOR5 moiety can be prepared by a process which utilizes the 4-methoxy-biphenyl intermediate of the preceding scheme. Thus, the 4-acetyl-4-methoxy-2-nitrobiphenyl intermediate of the previous scheme is subjected to reduction with stannous chloride to yield the intermediate amino derivative, which is then subjected to replacement of the amino group with a halo group. For example, the amino group can be replaced with fluorine via a diazonium fluoroborate transitory intermediate prepared from the amino intermediate using sodium nitrite and tetrafluoroboric acid. The resulting October 1989 3ED9:OCO:GT:jj AHP-9498 PATENT -9- acetyl-fluoro-methoxy biphenyl intermediate is converted to the corresponding carboxylic acid followed by demethylation with hydrogen bromide to yield the 2-fluoro-4'-hydroxy-[l,l'biphenyl]-4-acetic acid intermediate: HO2C-CH2 OH October 1989 3ED10:0CO:GT:jj AHP-9498 PATENT - 10- The latter carboxylic acid intermediate is esterified with methanol in the presence of p-toluenesulfonic acid and the latter is reacted with an appropriate haloalkyl-A compound, where A is as defined hereinbefore and hal is halo, to yield the desired final product as the methyl ester.
The ester can be hydrolyzed by conventional methods to yield the desired final product in its free carboxvlic acid form.
-¾ Compounds of the invention having the formula R4 I CH(CH2)mCOOR5 or R4 CH(CH2)mCOOR5 can be prepared as follows. Benzaldehyde and 4-methoxybenzaldehyde are reacted to yield 4methoxvbenzoin, which is converted to the hemisuccinate by reaction with succinic anhydride. The latter is reacted with urea and acetic acid to yield the intermediate 4-(4-methoxyphenyl)-5phenyl-2-oxazole-propionic acid.
CH3O, CHO acH° EtOH/Δ CH3O, O toluene / Δ October 1989 3EDll.OCO:GT:jj AHP-9498 PATENT -11-o=c—CH2CH2COOH CH’V^ i I -C-CHo II nh2cnh2 HOAc/Δ CH3O, XL, -CH2CH2COOH 0X“ The latter intermediate is demethylated with hydrogen bromide and esterified with methanol to yield the corresponding hydroxy methyl ester intermediate, which is then reacted with an appropriate haloalky-A compound, where A is as defined hereinbefore and hal is halo, to yield the desired final product as the methyl ester.
CH3Ok N HO OX0' -CH2CH2COOH CH3OH, 'p-toluenesulfonic acid / Δ XL· QX CH2CH2COOH -CH2CH2COOCH3 CX A(CH2)nhal K2CO3 / 18-crown-( October 1989 3ED12:OCO:GT:jj AHP-9498 PATENT - 12 - - CH2CH2COOCH3 The ester can be hydrolyzed by conventional methods to yield the desired final product in its free carboxylic acid form.
The conventional starting materials used in the reaction sequences outlined 5 above are available commercially or can be prepared by methods known in the art. Thus, for example, the intermediate compound 2-bromomethylquinoline can be prepared by the following reaction sequence: The benzo-fused heterocyclic compounds used in the above reaction sequences are also either commercially available or can be prepared by methods conventional in the art. Thus, for example, such intermediates as l-methyl-2-chloromethylbenzimidazole, 2-chloromethylbenzthiazole and 2-chloromethylbenzoxazole can be prepared by the following reaction scheme wherein X is O, S or NCH3. The reaction is preferably carried out at a controlled low temperature in an organic solvent, such as methylene chloride.
The compounds of the invention, by virtue of their ability to inhibit the activity of PLA2 enzyme, as well as that of lipoxygenase enzyme and to antagonize mediators arising from the enzymatic pathway, are useful in the treatment of conditions mediated by products of October 1989 3ED13:OCO:GT:jj AHP-9498 PATENT -13-the oxidation of arachidonic acid. Accordingly, the compounds are indicated in the treatment of such diseases as rheumatoid arthritis, inflammatory bowel disease, osteoarthritis, tendinitis, bursitis, psoriasis (and related skin inflammation) and similar conditions involving inflammation. Moreover, by virtue of their ability to antagonize the effect of LTC4, LTD4 and LTE4, which are the constituents of SRS-A, they are useful for the inhibition of symptoms induced by these leukotrienes. Accordingly, the compounds are indicated in the prevention and treatment of those disease states in which LTC4, LTD4 and LTE4 are causative factors, for example allergic rhinitis, allergic bronchial asthma and other leukotriene mediated naso-bronchial obstructive air-passageway conditions, as well as in other immediate hypersensitivity reactions, such as allergic conjunctivitis. The compounds are especially valuable in the prevention and treatment of allergic bronchial asthma.
The cornpounds of the invention are cytoprotective agents and are considered especially useful when administered with conventional non-steroidal anti-inflammatory drugs, whose major side effect is gastrointestinal irritation. The cytoprotective effect of the compounds of the invention significantly reduces the gastroirritant impact of conventional antiinflammatory drugs. This effect is based not only on the ability of the compounds of the invention to inhibit the biological effects of leukotrienes and/or control the biosynthesis of these substances, as by inhibiting lipoxygenase, but also by a shunting effect, whereby the control of the lipoxygenase pathway shunts the oxidation of arachidonic acid into the cyclooxygenase pathway, giving rise to an increase in the formation of cytoprotective prostaglandins. These biological effects make the compounds of the invention* especially useful in treating such conditions as erosive esophagitis, inflammatory bowel disease and induced hemorrhagic lesions such as those induced by alcohol or non-steroidal anti-inflammatory drugs (NSAID's), hepatic ischemia, noxious agent induced damage or necrosis of hepatic, pancreatic, renal or myocardial tissue; liver parenchymal damage caused by hepatotoxic agents such as carbon tetrachloride and D-galactosamine; ischemic renal failure; disease-induced hepatic damage; bile salt-induced pancreatic or gastric damage; trauma or stress-induced cell damage; and glycerolinduced renal failure.
When the compounds of the invention are employed in the treatment of allergic airway disorders, as anti-inflammatory agents and/or as cytoprotective agents, they can be formulated into oral dosage forms such as tablets, capsules and the like. The compounds can be administered alone or by combining them with conventional carriers, such as magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, low melting wax, cocoa butter and the like. Diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, tabletdisintegrating agents and the like may be employed. The compounds may be encapsulated with October 1989 3ED14:OCO.GT:jj AHP-9498 PATENT - 14-or without other carriers. In all cases, the proportion of active ingredients in said compositions both solid and liquid will be at least to impart the desired activity thereto on oral administration. The compounds may also be injected parenterally, in which case they are used in the form of a sterile solution containing other solutes, for example, enough saline or glucose to make the solution isotonic. For administration by inhalation or insufflation, the compounds may be formulated into an aqueous or partially aqueous solution, which can then be utilized in the form of an aerosol.
The dosage requirements vary with the particular compositions employed, the route of administration, the severity of the symptoms presented and the particular subject being treated. Treatment will generally be initiated with small dosages less than the optimum dose of the compound. Thereafter the dosage is increased until the optimum effect under the circumstances is reached. In general, the compounds of the invention are most desirably administered at a concentration that will generally afford effective results without causing any harmful or deleterious side effects, and can be administered either as a single unit dose, or if desired, the dosage may be divided into convenient subunits administered at suitable times throughout the day.
The PLA2 and lipoxygenase inhibitory and leukotriene antagonist effects, as well as the anti-inflammatory and potential gastroirritant effects of the compounds of the invention, may be demonstrated by standard pharmacological procedures which are described more full in the examples given hereinafter.
These procedures, inter alia, determine; the specificity of action of the compounds of the invention as PLA2 inhibitors as measured by their ability to inhibit the synthesis of LTB4 and PGE2 by rat glycogen-elicited polymorphonuclear leukocytes, as well as measure their ability to inhibit arachidonic acid release mediated by human source PLA2. The pharmacological testing additionally demonstrates the ability of the compounds of the invention to inhibit, in vivo, the lipoxygenase and cyclooxygenase pathways of arachidonic acid metabolism.
The following examples show the preparation and pharmacological testing of compounds within the invention.
October 1989 3ED15:OCO:GT:jj AHP-9498 PATENT - 15-Example 1 l-r2-Nitro-4,.(2-auinolinvlmethoxv)-fkl,-biDhenvU4-vUethanone A. 4-Acetyl-4'-methoxv-2-nitro biphenyl A stirred mixture of 4-iodoanisole (43.65 g, 0.187 mole), 4-bromo-3-nitro acetophenone (40.6 g, 0.166 mole) and copper powder (copper bronze, 36 g, 0.567 mole) kept under nitrogen is placed in an oil bath heated at 80°C. The temperature is slowly raised to 110°C and the mixture is kept at this temperature for 5 days (TLC, 8:2 hexane-ethyl acetate). Upon cooling the mixture is dissolved in dichloromethane and filtered through a Celite pad. The filtrate and washings are evaporated and the residual thick, dark brown oil (58.4 g) is flash chromatographed (on silica Merck 60, preabsorbed in dichloromethane, eluted with 9:1 hexane-ethyl acetate to remove the impurities and 8:2 hexane-ethyl acetate to recover the main product) to provide 16.2 g (32%) of the title compound (yellow solid, m.p. 124-126°C).
NMR (CDC13, 400 MHz): δ 2.67 (s, 3H, COCH3), 3.85 (s, 3H, OCH3), 6.97 (d, 2H, J 8.74 Hz, ArH), 7.27 (d, 2H, J 8.74 Hz, ArH), 7.56 (d, 1H, J 8 Hz, ArH), 8.15 ( d, 1 H, J 8 Hz, ArH), 8.34 (s, 1H, ArH) MS (El, m/z): 271 (M)+ B. 4-Acetvl-4'-hydroxy-2-nitro biphenyl To a stirred solution of AlBr3 (12.6 g, 47.4 mmole) in benzene (45 mL) is added dropwise under nitrogen a solution of the methylether (5 g, 18.45 mmole) of Step A in benzene (12 mL) over 30 minutes. The resulting solution is stirred at room temperature for 3.5 hours. (TLC, 8:2 hexane-ethyl acetate). The mixture is cooled in an ice bath and the complex is decomposed by the dropwise addition of 6N-HC1 (ca. 37 mL). The organic layer is separated and the aqueous phase is reextracted with ether (3x). The combined extracts are concentrated to a small volume and extracted again with 2.5N-NaOH (2x50 mL + 1x10 mL). The basic extracts are cooled and acidified with concentrated HCl (to pH 2). The solid is collected and dried (4.27 g, 90%). It is used in the next step without further purification.
NMR (CDC13, 400 MHz): δ 2.67 (s, 3H, COCH3), 5.03 (broad, 1H, OH), 6.91 (d, 2H, J 8.56 Hz, ArH), 7.23 (d, 2H, J 8.57 Hz, ArH), 7.55 (d, 1H, J 7.9 Hz, ArH), 8.15 (d, 1H, J 8.1 Hz, ArH), 8.34 (s, 1H, ArH).
MS (El, m/z): 257 (b.p., M)+ October 1989 3ED16:OCO:GT:jj AHP-9498 PATENT -16-C. l-r2-Nitro-4'-f2-quinolinyl')il.l’-biphenvl-4-vnethanone A mixture of the phenol (4.4 g, 17.12 mmole) of Step B, powdered anhydrous potassium carbonate (2.37 g, 17.12 mmole), 18-crown-6 (0.453 g, 1.71 mmole) and acetonitrile (38 mL) is stirred at room temperature under nitrogen for 15 minutes. 2-Chloro5 methylquinoline (3.34 g, 18.83 mmole, free base freshly prepared from the hydrochloride salt) is added and the mixture is refluxed for 10 hours. (TLC, 7:3 hexane-ethyl acetate). A 10% excess of potassium carbonate, 18-crown-6 and the chloromethylquinoline is added and reflux continued for another 4 hours. The solvent is removed and the residue is diluted with water and extracted with ethyl acetate (3x). The extracts are washed and dried (MgSO4). The residue is flash chromatographed (on silica Merck 60, preabsorbed in dichloromethane and eluted in order of increasing polarity with 7:3, 1:1 and 1:3 hexane-ethyl acetate followed by pure ethyl acetate) to provide the pure title compound (2.59 g). Recrystailization from toluene yields a yellow solid, m.p. 160-162°C (2.05 g, 30%).
NMR (CDC13,400 MHz): δ 2.66 (s, 3H, COCH3), 5.43 (s, 2H, OCH2Ar), 7.10 (d, 2H, J 8.7 Hz, ArH), 7.27 (d, 2H, J 8.7 Hz, ArH), 7.56 (m, 2H, ArH), 7.68 (d, 1H, J 8.49 Hz, ArH), 7.75 (dt, 1H, ArH), 7.84 (d, 1H, J 8.1 Hz, ArH), 8.09 (d, 1H, J 8.5 Hz, ArH), 8.14 (dd, 1H, ArH), 8.22 (d, 1H, J 8.49 Hz, ArH), 8.34 (s, 1H, ArH) MS (El, m/z): 398 (M)+, 256, 158, 142 (b.p.) Analysis for: Calculated: Found: C24H18N2O4 C, 72.35; Η, 4.55; N, 7.03 C, 71.96; Η, 4.75; N, 6.80.
*E 903874 October 1989 3ED17:OCO:GT:jj AHP-9498 PATENT -17-Eaampk 2 2-Fluoro-4'-(2-quinolinvlmethoxv)-fl.r-biDhenYll-4^acgtk_ ..acid A. 4-Acetyl-4'-methoxy-2-amino biphenyl To a stirred, warm solution of tin (Π) chloride (49.4 g, 218.9 mmole) in a mixture of concentrated HCl (72 mL) and ethanol (99 mL) is added over a period of 45 minutes the nitro derivative (10.7 g, 39.5 mmole) of Example IA. The resulting yellow solution is refluxed for 3.5 hours (TLC, 1:1 hexane-ethyl acetate). The ethanol is removed and the residue is poured into a mixture of 50% NaOH (360 mL) and ice. The resulting solid is extracted (dichloromethane, 3x), the extracts are washed with water and dried (Na2SO4). Removal of the solvent provides a yellow solid (9.31 g, 97.8%), m.p. 152154°C.
NMR (CDC13, 400 MHz): δ 2.59 (s, 3H, COCH3), 3.80 (s, 3H, OCH3), 6.97 (d, 2H, J 8.7 Hz, ArH), 7.23 (d, 1H, J 7.4 Hz, ArH), 7.40 (d, 2H, J 8.7 Hz, ArH), 7.48 (d, 1H, J 7.3 Hz, ArH), 7.49 (s, 1H, ArH).
MS (El, m/z): 241 (b.p., M)+, 226 (M-CH3)+, 198 (M-COCH3)+, 83.
B. 4-Acetyl-4,-methoxv-2-fluoro biphenyl To a stirred, ice cold mixture of the aniline (9.2 g, 38.2 mmole) in tetrahydrofuran (26 mL), water (9.8 mL) and HBF4 (48%, 35.1 mL) is slowly added a solution of sodium nitrite (2.82 g, 40.85 mmole) in water (5 mL). The internal temperature is kept below 5°C during the addition. The mixture is then stirred for an additional 20 minutes at 0-5°C. The diazonium fluoroborate is filtered off and washed with 10% HBF4 and 10% methanol in ether and dried in vacuo. The salt is decomposed by heating at 70°C in xylene (95 mL). When the decomposition subsides, the mixture is refluxed for another 2.5 hours (TLC, 1:1 hexane-ethyl acetate, UV). The xylene is removed and the residue is extracted with ethyl acetate (3x) and ether. The combined extracts are washed with 10% sodium carbonate and brine and dried (MgSO4). Removal of the solvent provides an amber oil (6.03 g) which is purified by flash chromatography (on silica Merck 60, preabsorbed in dichloromethane and eluted with 95:5 hexane-ethyl acetate). The title compound is obtained as a yellow solid 3.12-4.75 g, (33-51% depending on the run); m.p. 100-101°C.
NMR (CDC13, 400 MHz): δ 2.62 (s, 3H, COCH3), 3.86 (s, 3H, OCH3), 7.00 (d, 2H, J 8.9 Hz, ArH), 7.50-7.80 (m, 5H, ArH).
MS (El, m/z): 244 (M)+, 229 (b.p., M-CH3)+ October 1989 3ED18:OCO:GT:jj AHP-9498 PATENT -18-C. 2-Fluoro-4'-methoxv-fl.r-biphenvll-4-acetic acid A mixture of sulfur (0.468 g, 14.6 mmole), morpholine (2.57 mL) and the ketone (3.95 g, 16.2 mmole) of Step A is refluxed for 17 hours (TLC, acid treated silica plate, 8:2 hexane-ethyl acetate). Upon cooling, glacial acetic acid (9.9 mL), sulfuric acid (1.6 mL) and water (4 mL) are added and the reflux resumed for 30 hours. Water is then added and the mixture is extracted with ether (3x). The combined extracts are concentrated to a smaller volume and extracted with 10% sodium carbonate. The basic extracts are carefully acidified in the cold with concentrated HCl (to pH 2). The title acid is extracted with ether (3x) and the extracts are washed and dried (MgSO4). Removal of the solvent provides a tan to brown solid (2.37 g, 56.3%) melting at 140-142°C.
NMR (CDC13, 400 MHz): δ 3.68 (s, 2H, CH2COO), 3.85 (s, 3H, OCH3), 6.96-7.50 (m, 7H, ArH).
MS (EI, m/z): 260 (M)+, 215 (b.p., M-COOHf.
D. 2-Fluoro-4'-hvdroxv-iLr-biphenyll-4-acetic acid To a solution of the methylether (1.31 g, 5.04 mmole) of Step C in glacial acetic acid (17 mL) is added dropwise 48% HBr in acetic acid (25 mL) and the mixture is refluxed for 4.5 hours (TLC, 7:3 hexane-ethyl acetate). A little water is added and the mixture is extracted with ether (3x). The extracts are washed and dried (MgSO4). Removal of the solvent provides the title compound as a tan solid (1.13 g, 92%), m.p. 208-210°C.
NMR (DMSO-d6, 400 MHz): δ 3.61 (s, 2H, CH2COO), 6.83 (d, 2H, J 8.64 Hz, ArH), 7.1-7.42 (m, 5H, ArH), 9.61 (s, IH, COOH).
MS ( C I, m/z): 246 (M)+, 201 (b.p., M-COOH)+· E. 2-Fluoro-4'-hydroxy-fT.r-biphenyll-4-acetic acid methvlester A solution of the acid (1.1 g, 4.47 mmole) of Step D in methanol (10 mL) containing p-toluenesulfonic acid ' H2O (0.159 g) is refluxed for 1.5 hours (TLC, acid treated silica plate, hexane-ethyl acetate 7:3). The solvent is removed, the residue is dissolved in ethyl acetate, washed with brine and dried (MgSO4). The tan solid (1.16 g, m.p. 115-118°C, quantitative yield) is used as such in the next step.
NMR (CDC13, 400 MHz): δ 3.65 (s, 2H, CH2COO), 3.73 (s, 3H, COOCH3), 6.88 (d, 2H, J 8.8 Hz, ArH), 7.10 (m, 2H, ArH), 7.32-7.44 (m, 3H, ArH).
MS (EI, m/z): 260 (M)+, 201 (b.p., M-COOCH3)+ .
October 1989 3ED19.OCO.GT.jj AHP-9498 PATENT - 19-F. 2-Fluoro-4'-(2-quinolinvlmethoxy)-[l.r-biphenvn-4-acetic acid methvlester A stirred mixture of the phenol (1.16 g, 4.46 mmole) of Step E, powdered anhydrous potassium carbonate (0.616 g, 4.46 mmole), 18-crown-6 (0.118 g, 0.445 mmole) and acetonitrile (10 mL) is stirred at room temperature under nitrogen for 15 minutes. 2-ChloromethylquinoIine (0.871 g, 4.9 mmole, free base freshly prepared from the hydrochloride salt) is then added and the mixture is placed in an oil bath heated at 65°C for 5 hours. A 10% excess of potassium carbonate, 18-crown-6 and the chloromethylquinoline is added and the heating continued for another 6 hours (TLC, 19:1 dichloromethane-methanol or 7:3 hexane-ethyl acetate). The solvent is removed and the residue is diluted with water and extracted with ethyl acetate (3x). The extracts are washed and dried (MgSO4). Removal of the solvent provides a tan solid which is purified by flash chromatography (on silica Merck 60, preabsorbed with dichloromethane, eluted with 7:3 hexane-ethyl acetate). The title compound thus obtained (1.55 g, 87%) is recrystallized from methanol. The off-white solid melts at 99-101 °C.
NMR (CDC13, 400 MHz): δ 3.64 (s, 2H, CH2COO), 3.72 (s, 3H, COOCH3), 5.43 (s, 2H, OCH2Ar), 7.1 (m, 4H, ArH), 7.35 (t, 1H, ArH), 7.47 (d, 2H, ArH), 7.55 (t, 1H, ArH), 7.69 (d, 1H, ArH), 7.74 (t, 1H, ArH), 7.84 (d, 1H, ArH), 8.09 (d, 1H, ArH), 8.20 (d, 1H, ArH).
MS (El, m/z): 401 (M)+, 142, 114 (b.p.) Analysis for: C25H20FNO3 Calculated: C, 74.80; H, 5.02; N, 3.49.
Found: C, 74.68; H, 4.65; N, 3.49.
!E 903874 October 1989 3ED20.0CO:GT:jj AHP-9498 PATENT -20-G. 2-Fluoro-4'-(2-quinolinylmethoxy)-il.r-biphenyll-4-acetic acid A solution of the ester (1.69 g, 4.21 mmole) of Step F, in dry tetrahydrofuran (20 mL) is treated dropwise under nitrogen with ΙΝ-LiOH (12.6 mL) and the mixture is stirred for 3 hours at room temperature (TLC, 19:1 dichloromethane-methanol or 1:1 hexane5 ethyl acetate). The solvent is removed, the residue is treated with water and neutralized (to pH 6.5) with 10% acetic acid. The acid is extracted with ethyl acetate (large volume needed) and the extracts are dried (MgSO4) and evaporated to dryness to yield an off-white solid (1.65 g, quantitative yield, m.p. 190-193°C, dec.). Recrystallization from ethyl acetate provides a white solid (1.32 g, 80%, m.p. 195-196°C dec.). The analytical sample is dried in vacuo at 40°C.
NMR (DMSO-d6, 400 MHz): δ 3.62 (s, 2H, CH2COO), 5.41 (s, 211, CH2OAr), 7.15 (m, 4H, ArH), 7.41 (t, 1H, J 8 Hz, ArH), 7.48 (d, 2H, ArH), 7.61 (t, 1H, ArH), 7.69 (d, 1H, ArH), 7.78 (dt, 1H, ArH), 8.01 (rn, 2H, ArH), 8.42 (d, 1H, ArH), 12.42 (s, COOH).
MS (+ FAB, m/z): 388 (M)+Analysis for: Calculated: Found: C24H18FNO3 C, 74.41; H, 4.68; N, 3.62. C, 74.28; H, 4.48; H, 3.69.
October 1989 3ED21:OCO:GT:jj AHP-9498 PATENT -21-Example 3 3-i4-(2-Ouinolinvlmethoxv)henzovllbenzene acetic acid A. 3-Methvl-f4'-methoxv1-benzophenone A 3-neck flask equipped with a condenser, mechanical stirrer and dropping funnel is charged under nitrogen with 1.925 g (79.19 g.a.) of magnesium turnings and enough ether to cover the turnings. A few drops of a solution of 3-bromotoluene (15.79 g, 92.28 mmole) in ether (40 mL) is then added along with a crystal of iodine to initiate the reaction. The remainder of the solution is then added dropwise and the mixture is refluxed until most of the magnesium has disappeared. After cooling, a solution of 4-methoxybenzo1 o nitrile (10 g, 75.1 mmole, dried in vacuo over P2O5) is added in one portion. The mixture is refluxed for 2 hours (TLC, no starting material present), cooled (ice bath) and slowly treated with cold water (130 mL) followed by dilute H2SO4 (1:1, v/v, 25 mL). The decomposition of the complex is completed by refluxing the mixture for 4 hours (followed by TLC, 8:2 ether-ethyl acetate). Following stirring overnight at room temperature, the layers are separated and extracted with ether (3x). The extracts are washed with 5% NaHCO3, dried (MgSO4) and evaporated to dryness. The crude material (amber oil, 13.93 g) is purified by flash chromatography (on silica Merck-60, eluted with 8:2 petrolether-ethyl acetate) to provide the title compound as a light yellow oil (12.5 g, 73.5%).
NMR (CDC13, 400 MHz): δ 2.4 (s, 3H, CH3), 3.9 (s, 3H, OCH3), 6.96 (d, J 8.8 Hz, 2H, ArH), 7.38 (m, 2H, ArH), 7.53 (d, J 6.9 Ηζ,ΙΗ, ArH), 7.57 (s, IH, ArH), 7.82 (d, J 8.7 Hz,2H, ArH).
MS (EI, m/z): 226 (M)+, 135 (b.p.), 91.
October 1989 3ED22:OCO:GT:ij AHP-9498 PATENT -22-B. 3-Bromomethyl-f4'-methoxvl-benzophenone A solution of the benzophenone (17.5 g, 77.4 mmole) of Step A in ethylene bromide (26.5 mL) (containing a small amount of benzoyl peroxide) is heated at reflux. A solution of bromine (12.7 g, 79.6 mmole) in ethylene bromide (15 mL) is added dropwise over 30 minutes while the mixture is irradiated with a Photolamp (300 W). Reflux is continued for 17 hours (TLC, 9:1 petrolether-ethylacetate, traces of starting material still present). The solvent is removed in vacuo and the residue (brown oil, 39.22 g) is purified by flash chromatography (on silica Merck-60, preabsorbed in dichloromethane, eluted with 9:1 perolether-ethyl acetate) to give unreacted starting material (2.59 g, ca. 15%) along with the desired product (14.36 g, 61% or 71.5% based on recovered unreacted starting material) and some mixed fractions (ca. 5.20 g). The light yellow solid melts at 58-61 °C and it is used as such in the next step.
NMR (CDC13, 400 MHz): δ 3.88 (s, 3Η, OCH3), 4.52 (s, 2H, CH2Br), 6.96 (d, J 8.8 Hz, 2H, ArH) 7.44 (t, J 7.6 Hz, 1H, ArH), 7.58 (d, J 7.8 Hz, 1H, ArH), 7.66 (d, J 7.6 Hz, 1H, ArH), 7.76 (s, 1H, ArH), 7.81 (d, J 8.8 Hz, 1H, ArH).
MS (El, m/z): 306/304 (1 bromine, M)+, 225, 135 (b.p.). Trace of dibromo at 386/384/382 (possibly CH3O Br o C. 3-Cvanomethyl-r4'-methoxyl-benzophenone The bromo compound (14 g, 45.9 mmole) of Step B, is dissolved in dioxane (30 mL) and a solution of NaCN (7 g) in water (28.5 mL) is added. The mixture is refluxed for 6 hours (TLC, petrolether-ethyl acetate 8:2), charcoalized if needed and extracted with ether (3x). The extracts are dried (MgSO4) and evaporated to dryness to yield a brown oil (13.44 g). The crude product is purified by flash chromatography (on silica Merck-60, preabsorbed in dichloromethane, eluted with 6:4 hexane-ethyl acetate) to provide the pure product (10.69 g, 92%) as a light yellow oil that sets up upon standing. The nearly colorless solid melts at 70-71 °C.
NMR (CDC13, 400 MHz): δ 3.80 (s, 2H, CH2CN), 3.87 (s, 3H, OCH3), 6.95 (d, J 8.6 Hz, 2H, ArH), 7.48 (t, J 7.7 Hz, 1H, ArH), 7.54 (d, J 7.6 Hz, 1H, ArH), 7.68 ( s+d, J 7.6 Hz, 2H, ArH), 7.79 (d, J 8.6 Hz, 2H, ArH).
MS (El, m/z): 251 (M)+, 135 (b.p.).
October 1989 3ED23:OCO:GT:jj AHP-9498 PATENT -23-D. ?-[4-Methoxvbenzovll-phenvlaceric acid The nitrile (4 g, 15.9 mmole) of Step C, is dissolved in 40% NaOH (40 mL) and the solution is heated at reflux under nitrogen for 7 hours (TLC, toluene-methanol 9:1). Water is added while cooling in an ice bath. The solution is washed with ethyl acetate and then acidified in the cold with concentrated HCl (to pH 2). The acid is extracted with ethyl acetate (3x) and the extracts are dried (MgSO4) and evaporated to dryness to yield the crude product (yellow solid, 3.56 g, 82%), m.p. 138-140°C.
NMR (CDC13, 400 MHz): δ 3.72 (s, 2H, CH2COO), 3.88 (s, 3H, OCH3), 6.95 (d, J 8.8 Hz, 2H, ArH), 7.43 (t, 1H, ArH), 7.48 (d, 1H, ArH), 7.65 (d. 1H, ArH), 7.68 (s, 1H, ArH), 7.81 (d, J 8.6 Hz, 2H, ArH).
MS (El, m/z): 270 (M)+, 211 (M-CH2COOH)+, 135 (b.p.), 107.
E. 3-f4-Hydroxybenzovll-phenylacetic acid An intimate mixture of the acid (8.1 g, 0.030 mole) of Step D, and pyridine hydrochloride (13.87 g, 0.120 mole) is stirred under nitrogen in an oil bath heated at 20015 210°C for 7 hours (TLC, toluene-methanol 9:1, dichloromethane-methanol 9:1). After cooling, the mixture is dissolved in dichloromethane. The solution is extracted with 1NNaOH, the extract acidified in the cold with concentrated HCl and extracted with ethyl acetate (3x). After drying (MgSO4) the solvent is removed to provide the crude title compound as a tan solid (7.61 g, quantitative yield), m. p. 147-149°C.
NMR (DMSO-d6, 400 MHz): δ 3.67 (s, 2H, CH2COO), 6.88 (d, J 8.84 Hz, 2H, ArH), ca. 7.5 (m, 4H, ArH), 7.65 (d, J 8.8 Hz, 2H, ArH), 10.4 (s, 1H, OH), ca. 12.3 (s, 1H, COOH).
MS (m/z): 257 (M+H)+, 217, 131, 91 (b.p.).
October 1989 3ED24:OCO:GT:jj AHP-9498 PATENT -24-F. 3-F4-Hvdroxvbenzoyll-phenvlacetic acid methvlester A mixture of the acid (8.56 g, 33.4 mmole) of Step E and p-toluenesulfonic acid monohydrate (1.05 g, 5.6 mmole) in methanol (70 mL) is refluxed for 2.5 hours (TLC, methanol-toluene 1:9). The methanol is evaporated and the residue is dissolved in ethyl acetate and washed with brine. After drying (MgSO4) the solvent is removed to yield a tan solid (8.68 g, 96.2%, m.p. 111-113°C). The crude product is used as such in the next step.
NMR (CDC13, 400 MHz): δ 3.69 (s, 2H, CH2COO), 3.69 (s, 3H, COOCH3), 6.86 (d, J 8.4 Hz, 2H, ArH), 7.41 (t, 1H, 7.58 Hz, 1H, ArH), 7.47 (d, J 7.56 Hz, 1H, ArH), 7.62 (d, J 7.4 Hz, 1H, ArH), 7.64 (s, 1H, ArH), 7.74 (d, 2H, J 8.4 Hz, ArH).
MS (m/z): 271(M+H)t, 217, 131,91 (b.p.).
G. 3-f4-(2-Ouino1inylmethoxy)benzoyllbenzene acetic acid methvlester A mixture of the phenol (4 g,14.8 mmole) of Step F, powdered anhydrous K2CO3 (2.05 g, 14.8 mmole) and 18-crown-6 (0.4 g, 1.48 mmole) in acetonitrile (35 mL) is stirred at room temperature under nitrogen for 15 minutes. 2-Chloromethylquinoline (2.9 g, 16.28 mmole, freshly prepared from the hydrochloride salt) is added in one portion and the mixture is heated in an oil bath kept at 65-70°C for 8 hours (TLC, toluene-methanol 9:1). A 10 % excess of K2CO3 , crown ether and chloromethylquinoline is added and the heating is continued for another 8 hours. The acetonitrile isxvaporated and the residue is partitioned between water and ethyl acetate. The organic layer is dried (MgSO4) and evaporated to yield a tan solid (6.57 g). The crude product is purified by flash chromatography (on silica Merck-60, preabsorbed in dichloromethane, eluted with petrolether-ethyl acetate 7:3) to give the title compound as a light yellow solid (5.03 g, 82.7%) m.p. 93-95°C.
NMR (CDC13, 400 MHz): δ 3.67 (s,5H, CH2COO + OCH3), 5.45 (s, 2H, ArCH2O), 7.08 (d, J 8.8 Hz, 2H, ArH), 7.40 (t, J 7.8 Hz, 1H, ArH), 7.46 (d, J 7.7 Hz, 1H, ArH), 7.55 (t, J 7.3 Hz, 1H, ArH), 7.6-7.66 (m, 3H, ArH), 7.7-7.82 (m, 4H, ArH), 8.07 (d, 1H, ArH), 8.20 (d, J 8.4 Hz, 1H, ArH).
MS (El, m/z): 411 (M)+, 142, 121 (b.p.).
October 1989 3ED25:OCO:GT:jj AHP-9498 PATENT Έ903874 -25-H. 3-f4-(2-Ouinolinvlmethoxv)benzoyllbenzene acetic acid To a solution of the ester (5 g, 12.16 mmole) of Step G, in dry tetrahydrofuran (66 mL) is added ΙΝ-LiOH (37 mL, 37 mmole) and the mixture is stirred under nitrogen at room temperature for 2.5 hours (TLC, toluene-MeOH 9:1). The tetrahvdrofuran is evaporated and the residue is diluted with water, acidified (to pH 6.5) with 10% acetic acid and extracted with ethyl acetate (3x). The extracts are washed with brine, dried (MgSO4) and evaporated to dryness. The crude product (4.94 g, pale yellow solid) is recrystallized from ethyl acetate to provide 3.65 g (75%) of the pure title compound (white solid, m.p. 146-147°C).
NMR (DMSO-d6, 400 MHz): δ 3.68 (s, 2H, CH2COO), 5.48 (s, 2H, ArCH2O), 7.22 (d, 2H, J 8.8 Hz, ArH), 7.47 (m, IH, ArH), 7.53 (m, 2H, ArH), 7.62 (m, 2H, ArH), 7.69 (d, J 8.4 Hz, IH, ArH) 7.74-7.82 (m, 3H, ArH), 8.2 (m, 2H, ArH), 8.43 (d, J 8.5 Hz, IH, ArH), 12.39 ( IH, COOH). MS (EI, m/z): 397 (b.p., M)+, 380 (M-0H)+, 142 . Analysis for: C25H19NO4 Calculated: Found: C, 75.57; H, 4.78; N, 3.53. C, 75.22; H, 4.76; N, 3.39.
Example 4 3-r4-(2-Naphthalenvlmethoxy)benzovHbenzene acetic acid A. 3-f4-(2-Naphthalenylmethoxv’)benzovllbenzene acetic acid methvlester A mixture of the phenol (1 g, 3.7 mmole) of Example 3F, powdered anhhydrous K2CO3 (0.48 g, 3.7 mmole), 18-crown-6 (0.098 g, 0.37 mmole) and acetonitrile (10 mL) is stirred under nitrogen for 15 minutes. 2-Bromomethylnaphthalene (0.496 g, 4.07 mmole) is added and the mixture is placed in an oil bath heated at 65-70°C for 10 hours (TLC, dichloromethane-ethyl acetate 8:2). A 10% excess of K2CO3, crown ether and bromomethvlnaphthalene is added and the heating is continued for another 4 hours. The acetonitrile is evaporated and the residue dissolved in water and extracted with ethyl acetate (3x). The extracts are washed with ΙΝ-NaOH and brine, dried (MgSO4) and evaporated to dryness.
The crude product (1.49 g, waxy solid) is used as such in the next step.
NMR (CDC13, 400 MHz): δ 3.7 (s, 5H, OCH3+ CH2COO), 5.32 (s, 2H, ArCH2O), 7.08 (d, J 8.7 Hz, 2H, ArH), 7.4-7.56 (m, 5H, ArH), 7.63-7.68 (m, 2H, ArH), 7.82-7.92 (m, 6H, ArH).
MS (m/z): 410 (M)+, 141 (b.p.).
October 1989 3ED26:OCO:GT:jj AHP-9498 PATENT -26-B. 3-i4-(2-Naphthalenvlmethoxy)benzovllhenzene acetic acid A solution of the ester (1.29 g, 3.15 mmole) of Step A, is treated dropwise with ΙΝ-LiOH and the mixture is stirred under nitrogen overnight. The solvent is evaporated and the residue is dissolved in water, acidified in the cold with 10% acetic acid (to pH 3) and extracted with ethyl acetate (3x). The extracts are dried (MgSO4) and evaporated to dryness. The residue (1.24 g, quantitative yield) is recrystallized by dissolving it in a relatively large volume of warm ethyl acetate-dichloromethane followed by concentrating to half volume.
The precipitate is collected and dried at 45°C in vacuo (0.610 g, 48.8%), m.p. 150-152°C.
NMR (DMSO-d6, 400 MHz): 5 3.70 (s, 2H, CH2COO), 5.40 (s, 2H, ArCH2), 7.20 (d, 2H, ArH), 7.45-7.60 (m, 7H, ArH), 7.75 (d, 2H, ArH), 7.95 (m, 3H, ArH), 8.02 (s, IH, ArH), 12.47 (broad s, IH, COOH).
MS (+FAB, m/z): 397 (M+H)+, 217, 141. Analysis for: C26H20O4 Calculated: C, 78.78; H, 5.09. Found: . 78.12; H, 5.13.
Examplf.-5 -Phenvl-4-14-(2-0uinolinvlmefhoxv)-phenvl1-2-oxazole propanoic acid -¼ A. 4-M ethox y ben zoi n To a solution of KCN (5 g) in water (35 mL) is added 4-methoxybenzaldehyde (27.2 g, 0.2 mole), benzaldehyde (21.2 g, 0.2 mole) and 95% ethanol (70 mL). The mixture is refluxed under nitrogen for 4.5 hours and the ethanol removed in vacuo. Water (200 mL) is added to the residue and then distilled off at reduced pressure (to remove remaining unreacted bezaldehyde). The procedure is repeated twice and the residual water azeotroped with ethanol. The crude product (56.3 g, orange semi-solid) is purified by flashchromatography (on silica Merck-60, preabsorbed in dichloromethane-ethyl acetate and eluted with hexane-ethyl acetate 8:2) to yield a light yellow solid (20.1 g, 41.5%), m.p. 99-101°C.
NMR (CDC13, 400 MHz): δ 3.82 (s, 3H, OCH3), 4.62 (broad s, IH, OH), 5.88 (s, IH, CHOH), 6.86 (d, 2H, J 8.94Hz, ArH), 7.22-7.38 (m, 5H, ArH), 7.91 (d, 2H, J 8.94Hz, ArH).
MS ( C I, m/z): 243 (b.p., M+H)+, 225, 197, 137 (M-PhCOf October 1989 3ED27:OCO:GT:jj AHP-9498 PATENT -27 B. 4-Methoxvbenzoin hemisuccinate A mixture of 4-methoxybenzoin (20 g. 0.083 mole) and succinic anhydride (9.1 g, 0.091 mole) in toluene (6 mL) is heated for 7 hours under nitrogen at 135°C (internal temp.). The solution is poured into 0.5N-NaHCO3, the organic layer was separated and re5 extracted with 0.5N-NaHCO3. The combined extracts are washed with ether and then acidified in the cold with concentrated HCl. The liberated oil is extracted with ethyl acetate (3x), the extracts washed with water and dried (MgSO4). Removal of the solvent yields a yellow solid (20.89 g, 73.8%), m.p. 104-108°C. It is used in the next step without further purification.
NMR (CDC13, 400 MHz): δ 2.72-2.82 (mm, 4H, CH2CH2COO), 3.82 (s, 3H, OCH3), 6.86 (d, 2H, 19.1Hz, ArH), 7.34-7.46 (m, 5H, ArH), 7.92 (d, 2H, J 9.1Hz, ArH).
MS (EL m/z): 342 (M)+, 135 (b.p.).
C. 4-(4-Methoxyphenyl)-5-phenyl-2-oxazole-propanoic acid A mixture of the crude 4-methoxybenzoin hemisuccinate (20.8 g, 0.061 mole) of Step B, urea (8.7 g, 0.146 mole) and acetic acid (60 mL) is heated at reflux under nitrogen for 5.5 hours. The mixture is cooled and poured into ice water. The liberated oil is extracted with ethyl acetate (3x). The extracts are washed with water until neutral and then extracted with saturated sodium carbonate. The combined^aqueous extracts are carefully acidified in the cold with concentrated HCl and extracted with ethyl acetate. The organic extract is dried (MgSO4) and evaporated to dryness to provide a waxy yellow oil (19.6 g). Purification of the residue by flash chromatography (on silica Merck-60, eluant: dichloromethane-ethyl acetate 8:2) yields a pale yellow solid (14.3 g, 72.7%), m.p. 100-101°C.
NMR (CDC13, 400 MHz): δ 2.96 (t, 2H, CH2C), 3.20 (t, 2H, CH2COO), 3.83 (s, 3H, OCH3), 6.90 (d, 2H, ArH), 7.28-7.38 (m, 3H, ArH), 7.54-7.62 (m, 4H, ArH).
MS (El, m/z): 323 (M)+, 278 (b.p., M-COOH)+, 152, 77.
October 1989 3ED28:OCO:GT:jj AHP-9498 PATENT -28D. 4-(4-Hvdroxvphenvl)-5-phenyl-2-oxazole-propanoic acid To a solution of the methoxyacid (5.6 g, 17.3 mmole) of Step C, in acetic acid (55 mL) is added 48% HBr (84 mL) and the mixture is heated at reflux under nitrogen for 8 hours (TLC, 1:1 hexane-ethyl acetate). After cooling, water is added and the solution extracted with ethyl acetate (3x). The extract is dried (MgSO4) and evaporated to dryness. The residue (brown waxy oil, 5.25 g, 99%) is used in the next step without further purification. For analytical characterization a small sample is flash-chromatographed (on silica Merck-60, eluant: dichloromethane -methanol 98:2 and 95:5).
NMR (DMSO-d6, 400 MHz): δ 2.75 (t, 2H, J 7.14Hz, CH2C), 3.02 (t, 2H, J 7.1Hz, CH2COO), 6.77 (d, 2H, J 8.7Hz, ArH), 7.35 (d, 2H, J 8.55Hz, ArH), 7.41 (t, 3H, J 7.l2HzAArH), 7.50 (d, 2H, J 7Hz, ArH), 9.64 (broad s, exchangeable).
MS (El, m/z): 309 (M)+, 264 (M-C00H)+, 121,105, 77.
E. 5-Phenvl-4-(4-hydroxyphenvl)-2-oxazole propanoic acid methylester A solution of the crude acid (5 g, 16.18 mmole) of Step D, in methanol (40 15 mL), containing a small amount of p-toluenesulfonic acid ' H2O (0.58 g) is refluxed for 2.5 hours. The methanol is evaporated and the residue is partitioned between ethyl acetate and 20% NaCl. The extracts are washed, dried (MgSO4) and evaporated to yield a thick oil (ca. 4.8 g). The residue is flash-chromatographed (on silica Merck-60, preabsorbed in dichloromethane, eluted with a dichloromethane-ethyl acetate gradient from 90:10 to 75:25) to yield a white solid (3.56 g, 68%), m.p. 115-116°C.
NMR (CDC13, 400 MHz): δ 2.92 (t, 2H, J 7.4 Hz, CH2C), 3.20 (t, 2H, J 7.4Hz, CH2COO), 3.71 (s, 3H, OCH3), 6.74 (d, 2H, J 8.59 Hz, ArH), 7.26-7.36 (m, 3H, ArH), 7.40 (d, 2H, J 8.7Hz, ArH), 7.54 (d, 2H, J 7.56Hz, ArH).
MS (El, m/z): 323 (M)+, 264 (M-COOCH3)+, 105, 77 (b.p.).
October 1989 3ED29:0CO:GT:jj AHP-9498 PATENT -29F. 5-Phenvl-4-f4-(2-Quinolinvlmethoxv)phenyll-2-oxazole propanoic acid methvlester A mixture of the ester (2.46 g, 7.61 mmole) of Step E, powdered anhydrous K2CO3 (1.05 g, 7.60 mmole), l8-crown-6 (0.223 g, 0.843 mmole) and acetonitrile (33 mL, ex-sieves) is stirred at room temperature under nitrogen for 15 minutes. 2-Chloromethyl5 quinoline (free base, freshly prepared from the hydrochloride salt, 1.35 g, 7.60 mmole) is added and the mixture is placed in an oil bath heated at 65°C for 10 hours (N.B. A 10% excess of the chloromethylquinoline, 18-crown-6 and K2CO3 is added after 6 hours). The solvent is removed and the residue is partitioned between ethyl acetate and water. The extracts are washed (brine), dried (MgSO4) and evaporated to yield a yellow solid. The crude product is flash chromatographed (on silica Merck-60, eluant: toluene and then toluenemethanol 97.5:2.5) to provide the title compound (3.5 g, quantitative yield).
NMR (CDC13, 400 MHz): δ 2.90 (t, 2H, J ca. 7.2Hz, CH2C), 3.16 (t, 2H, J 7.2 Hz, CH2COO), 3.72 (s, 3H, OCH3), 5.41 (s, 2H, ArCH2O), 7.02 (d, 2H, J 8.8Hz, ArH), 7.29-7.36 (m, ca. 4H, ArH), 7.54-7.84 (m, ca. 6H, ArH), 7.83 (d, 1H, J 8.1Hz, ArH), 8.08 (t, 1H, J 8.5Hz, ArH), 8.19 (d, 1H, J 8.5Hz, ArH).
MS (+FAB, m/z): 487 (M+Na)+, 465 (M+H)+ October 1989 3ED30:OCO:GT:jj AHP-9498 PATENT 903874 - 30-G. 5-Phenvl-4-f4-(2-auinolinvlmethoxy)phenvl1-2-oxazole propanoic acid A solution of the ester (3.4 g, 7.32 mmole) of Step F, in dry tetrahydrofuran (37 mL) is treated dropwise under nitrogen with ΙΝ-LiOH (21.98 mL, 3 equiv.) and stirred at room temperature for 3 hours (TLC, dichloromethane-methanol 97:3 or toluene-methanol 95:5). The solvent is evaporated, the residue is dissolved in water, neutralized in the cold with 10% acetic acid (to pH 5.5-6) and extracted with ethyl acetate. The extracts are washed with brine, dried (MgSO4) and evaporated to yield a pale yellow solid (3.18 g, quantitative yield). The crude product is recrystallized from warm ethyl acetate (containing enough dichloromethane to obtain a clear solution) to yield a first crop of crystals (2.63 g, m.p. dec. 192-194°C). A second crop is obtained by concentrating the mother liquors (0.327 g, m.p. dec. 192-193°C). The combined yield is 85.8%.
IR (KBr, cm'1): 1720 (CO).
NMR (DMSO-d6, 400 MHz): δ 2.76 (t, 2H, J 7Hz, CH2C), 3.03 (t, 2H, J 7Hz, CH2COO), 5.38 (s, 2H, ArCH2O), 7.11 (d, 2H, J 8.8Hz, ArH), 7.36-7.56 (m, 7H, ArH), 7.61 (t, 1H, ArH), 7.69 (d, 1H, J 8.5Hz, ArH), 7.78 (t, 1H, ArH), 8.00 (t, J 7.9Hz, 2H, ArH), 8.42 (d, 1H, J 8.5Hz, ArH).
MS (El or C I, m/z): 451 (M+H)\ 310 (b.p.).
Analysis for Calculated: 20 Found: ^28^22^2^4 C, 74.65; H, 4.92; N, 6.22. C, 74.20; H, 4.86; N, 6.00.
October 1989 3ED31:OCO:GT.lj AHP-9498 PATENT -31Examptf-6 4-i4-i2-Naphthalen vlmethoxvlphen vll-5-phenvl-2-oxazole propanoic ax id A. 4-i4-[2-Naphthalenvlmethoxvlphenvll-5-phenvl-2-oxazole propanoic acid methvlester A mixture of the hydroxyester (1.5 g, 4.6 mmole) of Example 5E, powdered anhydrous K2CO3 (0.636 g,4.6 mmole), 18-crown-6 (0.123 g, 0.46 mmole) and acetonitrile (18 mL) is stirred at room temperature under nitrogen for 15 minutes. 2-Bromomethylnaphthalene (1.13 g, 5.1 mmole) is added and the mixture is placed in an oil bath heated at 70°C for 8-9 hours (TLC, hexane-ethyl acetate 9:1 or dichloromethane-methanol 9:1). The solvent is evaporated and the residue dissolved in water and extracted with ethyl acetate. The extracts are washed and dried (MgSO4). Removal of the solvent yields a tan solid (2.17 g, quantitative yield). A sample is recrystallized from methanol (containing enough dichloromethane to obtain a clear solution) by concentrating to small volume and cooling in an ice bath. The white solid is collected and dried overnight in vacuo, m.p. 13415 135°C.
IR (KBr, cm’1): 1740 (CO).
NMR (CDCI3-400 MHz): δ 2.89 (t, 2H, J 7.5Hz, CH2C), 3.16 (t, 2H, J 7.5Hz, CH2COO), 3.71 (s, 3H, OCH3), 5.2 (s, 2H, ArCH2O), 7.00 (d, 2H, J 8.6Hz, ArH), 7.25-7.35 (m, 3H, ArH), 7.46-7.58 (m, 7H, ArH), 7.8-7.9 (m, 4H, ArH).
MS (CI, m/z): 464 (M+H)+, 324.
Analysis for: Calculated: Found: C3oH25N04 C, 77.73; H, 5.44; N, 3.02. C, 77.44; H, 5.36; N, 3.03.
October 1989 3ED32:OCO:GT:jj AHP-9498 PATENT -32B. 4-i4-(2-Naphtha)envlmethoxy)phenyH-5-phenvl-2-oxazole propanoic acid A solution of the ester (1.49 g, 3.21 mmole) of Step A, in dry tetrahydrofuran (18mL) containing ΙΝ-LiOH (9.6 mL) is stirred under nitrogen overnight at room temperature (TLC, 75:25 hexane-ethyl acetate). The solvent is evaporated, the residue dissolved in water and acidified (to pH 5) with dilute HCl. The mixture is extracted with ethyl acetate, the extracts are dried (MgSO4) and evaporated to yield the crude product (1.39 g, m.p. 145150°C). For purification, it is dissolved in hot ethyl acetate (containing enough dichloromethane to obtain a clear solution), concentrated to half volume and precipitated with ether. The white solid melts at 151-152°C (1.07 g, 58%).
IR (KBr, cm'1): 1720 (CO).
NMR (DMSO-d6, 400 MHz): δ 2.78 (t, 2H, CH2C), 3.03 (t, 2H, J 7Hz, CH2COO), 5.29 (s, 2H, ArCH2O), 7.10 (d, 2H, J 8.9Hz, ArH), 7.34-7.60 (m, 10H, ArH), 7.90-8.00 (m, 4H, ArH), 12.28 (s, 1H, COOH).
MS (El, m/z): 450 (M+H)+, 310.
Analysis for: Calculated: Found: C29H23NO4 C, 77.48; H, 5.15; N, 3.11. C, 76.40; H, 5.16; N, 3.04.
,E 903874 October 1989 3ED33:OCO:GT:jj AHP-9498 PATENT -33’ Example 7 4-Γ4-ΚΊ-Methyl-1 H-benzimidazol-2-vl)methoxvlphenyn-5-phenvl-2-oxazole propanoic acid A. 4-f4-i(l-Methyl-1 H-benzimidazol-2-vl)methoxvlphenvn-5-phenvl-2-oxazole 5 propanoic acid methvlester A mixture of the ester (0.5 g, 1.55 mmole) of Example 5E, powdered anhydrous Κ2ΟΟ3 (0.214 g, 1.55 mmole), 18-crown-6 (0.0416 g, 0.155 mmole) and acetonitrile (6 mL) is stirred under nitrogen at room temperature for 15 minutes. 2-Chloromethyl-lmethylbenzimidazole (0.307 g, 1.7 mmole) is added and the mixture is placed in an oil bath heated at 65-70°C for 4 hours (TLC, dichloromethane-ethyl acetate 9:1, iodine visualization). A 10% excess of K2CO3,2-chloromethyl-1 -methyl-benzimidazole and 18-crown-6 is added at this point and the heating continued for another 10 hours. The solvent is evaporated, the residue dissolved in water and extracted with ethyl acetate. The extracts are washed, dried (MgSO4) and evaporated to dryness. The residue (1.64 g) is purified by flash-chromato15 graphy (on silica Merck-60, preabsorbed in dichloromethane containing a small amount of methanol, eluted with dichloromethane-ethyl acetate 8:2) to yield 1.03 g (71.2%) of a light yellow solid, m.p. 142-144°C (dec).
NMR (CDC13, 400 MHz): δ 2.87 (t, 2H, J 7.1Hz, CH2C), 3.16 (t, 2H, J 7.8Hz, CH2COO), 3.72 (s, 3H, COOCH3), 3.90 (s, 3H, NCH3), 5.41 (s, 2H, ArCH2O), 7.07 (d, 2H, J 8.8Hz, ArH), 7.25-7.40 (m, 7H, ArH), 7.5-7.6 (m, 3H, ArH), 7.78 (d, 1H, ArH).
MS (+C I, m/z): 468 (M+H)+, 324, 293, 147.
October 1989 3ED34:OCO:GT:jj AHP-9498 PATENT - 34-B. 4-14-1(1-Methvl-1 H-benzimidazol-2-vl)methoxv]-5-phenvl-2-oxazole propanoic acid A solution of the ester (1 g, 2.14 mmole) of Step A, in tetrahydrofuran (13 mL) containing ΙΝ-LiOH (6.42 mL) is stirred under nitrogen at room temperature for 1 hour (TLC, dichloromethane-ethanol 9:1). The solvent is evaporated, water added and the pH adjusted to 6.5 with 10% acetic acid. The light yellow precipitate is collected, washed with water and dried in vacuo. It is redissolved in hot ethyl acetate (containing enough methanol to obtain a clear solution), concentrated to a smaller volume and cooled in an ice bath. The crystals are collected and dried (0.642 g, 66.2%, m.p. 222-224°C).
NMR (DMSO-d6, 400 MHz): δ 2.76 (t, 2H, J 7Hz, CH2C), 3.03 (t, 2H, J 7Hz, CH2COO), 3.86 (s, 3H, NCH3), 5.43 (s, 2H, ArCH2O), 7.14-7.66 (m, 13H, ArH).
MS (CI, m/z): 454 (M+H)+, 147 (b.p.).
Analysis for: Calculated: 15 Found: C27H23N3O4 C, 71.51; H, 5.11; N, 9.27. C, 71.62; H, 5.17; N, 9.40.
Example 8 The compounds 5- and 12-hydroxyeicosatetraenoic acid (5-HETE and 12HETE) and LTB4 are early arachidonic acid oxidation prdducts in the lipoxygenase cascade, which have been shown to mediate several aspects of inflammatory and allergic response. This is especially true with respect to 5,12-diHETE, which is also denoted as LTB4 [see FordHitchinson, J. Rov. Soc. Med.. 74. 831 (1981)]. Compounds which inhibit the PLA2mediated release of arachidonic acid thereby effectively prevent the oxidation of arachidonic acid to the various leukotriene products via the lipoxygenase cascade. Accordingly, the specificity of action of PLA2 inhibitors can be determined by the activity of test compounds in this assay, which measures the ability of compounds to inhibit the synthesis of LTB4 by rat glycogen-elicited polymorphonuclear leukocytes (PMN) in the presence of exogenous substrate.
The assay is carried out as follows: Rat polymorphonuclear leukocytes (PMNs) are obtained from female Wistar rats (15030 200 g) which receive an injection of 6% glycogen (10 ml i.p.). Rats are sacrificed 18-24 hours post injection by CO2 asphyxiation and the elicited cells are harvested by peritoneal lavage using physiological saline (0.9% NaCI). The exudate is centrifuged at 400 xg for 10 October 1989 3ED35:OCO:GT:jj AHP-9498 PATENT -35-10 minutes. The supernatant fluid is discarded and the cell pellet is resuspended to a concentration of 2.0 χ 107 cells/mL in HBSS containing Ca++ and Mg++ and 10 μΜ L-cysteine.
To 1 mL aliquots of cell suspension, test drugs or vehicle are added, then preincubated at 37*C for 10 minutes. A23187 (1 μΜ), [3H]-AA (3.0 pCi/mL) and unlabeled AA (1 μΜ) are then added and the samples are further incubated for 10 minutes. The reaction is terminated by centrifugation and pelleting cells. Supernatants are then analyzed by HPLC analysis on a 15 cm x 4.6 mm ID supelcosil LC-18 (Supelco)(3M) column, using a two solvent system at a flow rate of 1.4 mL total flow as follows: Solvent A: 70:30 17.4 mM H3PO4:CH3CN Solvent B. CH3CN Gradient: (system is equilibrated with Solvent A) Time Percent A Percent B 0 100 0 15.0 100 0 20.0 65 35 40.0 65 35 42.0 10 90 50.0 10 90 20 50.1 100 0 Percent solvent changes are accomplished in a linear fashion.
Injections: 140 pL of each supernatant is injected directly onto column and 3H arachidonic acid metabolites are monitored using an on-line radioactivity detector (Ramona, IN/US, Fairfield, NJ).
Standards: 104 - 2.0 χ 104 dpm of eicosanoids of interest are injected in 90 pL EtOH cocktail.
Co-chromatography with standard [3H] leukotriene B4 (LTB4) in medium of stimulated 30 PMN exposed to drug is compared to that found in medium of stimulated cells exposed to no drug, generating percent inhibition.
Results are expressed as percent inhibition at a given compound dose or as an IC50 value.
October 1989 3ED36:OCO:GT:jj AHP-9498 PATENT -36-Testing compounds of the invention in this assay gave the following results: Table I Compound of Example No. % Inhibition ketoprofen -50* (at ΙΟμΜ) 1 95 (at 0.5μΜ) 2 91 (αιΟ.5μΜ) 3 87 (at ΙΟμΜ) 4 38 (at 0.5μΜ) 8 (at ΙΟμΜ) 5 96 (at ΙΟμΜ) 6 95 (at ΙΟμΜ) 6A 81 (3ΐΟ.5μΜ) 94 (at ΙΟμΜ) 7 63 (at 0.5μΜ) 85 (at ΙΟμΜ) * a negative value denotes potentiation of cyclooxygenase (PGE2 synthesis) Example 9 The procedure of Example 8 is also employed for the determination of the extent to which compounds of the invention inhibit the synthesis of the arachidonic acid cyclooxygenase oxidation product PGE2.
In this assay, the procedure of Example 8 is carried out as described. However, in order to determine cyclooxygenase activity, the samples are co-chromatographed with authentic reference pH]-PGE2.
The results are calculated as in Example 8 and presented below: Table Π Compound of Example No. ketoprofen % Inhibition 87 (at ΙΟμΜ) -13* (at 0.5μΜ) -22* (at 0.5μΜ) (at ΙΟμΜ) October 1989 3ED37:OCO:GT:jj AHP-9498 PATENT -37-Table II (cont.) Compound of Example No. 6A % Inhibition -8* (at 0.5μΜ) -31* (at ΙΟμΜ) -275* (at ΙΟμΜ) -191* (at ΙΟμΜ) -12* (at 0.5μΜ) -79* (at ΙΟμΜ) -29* (at 0.5μΜ) -268* (at ΙΟμΜ) * Negative values denote a potentiation of cyclooxygenase (PGE2 synthesis).
Example 10 The compounds of the invention are tested in an in vitro isolated phospholipase A2 assay to determine the ability of the test compounds to inhibit the release of arachidonic acid from an arachidonic acid-containing substrate by the action of phospholipase A2 enzyme from human and non-human sources.
This assay is carried out as follows: Into a 15 mL polypropylene tube are added the following: Agent Volume. uL Final Cone.3H-AA E. coli substrate 1 25 5 nmoles PL CaCl2(0.1M) 2 5 5 mM Tris-HCl (0.5M) pH 7.5 3 20 100 mM Water 4 25 Drug/vehicle 5 1 50 μΜ PLA2 25 Volume yielding 12% hydrolysis in 10 min. 100 * pre-incubate at room temperature 30 min prior to substrate addition.
Prepared by adding 2 mL deionized and distilled water to 2 mL 3H-arachidonate labeled E. coli (lower count), to which is added 1 mL of 3H-arachidonate labeled E. coli (higher count) to yield a total of 5 m substrate (containing 1000 nmoles phospholipid). 2 Stock 0.1 m CaCl2, required for enzyme activity.
October 1989 3ED38:OCO:GT:jj AHP-9498 PATENT -38-3 Stock 0.5 m Trisma-Base.
Stock 0.5 M Trisma-HCl. Adjust pH to 7.5 (optimum for enzyme). 4 Deionized and distilled water.
Stock 10 mM prepared in dimethyl sulfoxide. Make 1:2 dilution with dimethyl sulfoxide and add 1 pL to 100 pL assay tube. 6 Two human PLA2 enzymes are used: a) Semi-purified human platelet acid extract PLA2 (in 10 mM sodium acetate buffer, pH 4.5). Remove protein precipitate by centrifugation at about 2200 rpm for 10 minutes. b) Purified human synovial fluid.
Incubate the lOOpL reaction mixture for 10 minutes at 37*C in a shaking water bath. The reaction is terminated by the addition of 2 mL tetrahydrofuran, followed by vortexing. NH2 columns (100 pg/mL - Analytichem International) are conditioned with 0.5 mL tetrahydrofuran followed by 0.5 mL tetrahydrofuran/water (2 mL.O.l mL, v/v).
The sample is loaded onto the columns and slowly drawn through them. The hydrolyzed arachidonic acid retained in the columns is eluted therefrom with 1 mL tetrahydrofuran/glacial acetic acid (2%). The arachidonic acid is transferred to scintillation vials and quantitated by β-counting analysis. A total counts sample is prepared by pipetting 25 pL 3H-arachidonate E. coli directly into a scintillation vial to which is added 1 mL tetrahydrofuran. 10 mL aquasol (scintillation cocktail) is added to all samples.
Calculations: [3H]AA dpm (sample) - [3H]AA dpm (nonspecific hydrolysis) % hydrolysis = - x 100 total counts dpm vehicle dpm - drug dpm % change = - x 100 vehicle dpm Activity of Standard Drugs: _LC-5 0 (U.MJ_ Human Platelet Human Synovial PLA2. PL A 2. 3.2 0.14 Drug Arachidonic Acid Monoalide 8.6 .2 October 1989 3ED39:OCO:GT:jj AHP-9498 PATENT -39-When tested in this assay, the compounds of the invention gave the following results: Table ΙΠ % Inhibition at 10 U.M Compound of Example No. HSF* ketoprofen -16.9** .5 .1 * human synovial fluid ** negative values denote a potentiation of HSF Example 11 The compounds of the invention are evaluated for their ability to inhibit the lipoxygenase and/or cyclooxygenase pathways of arachidonic acid metabolism in the in vivo murine zymosan peritonitis assay.
This assay is carried out as follows: Male CD-I mice (8 weeks old) are placed in plastic boxes in groups of six. Animals are injected with 1 mL i.p. of either 1% zymosan in pyrogen free 0.9% saline or saline (unstimulated control). Compounds are dosed orally 1 hour prior to zymosan injection. Twenty minutes after zymosan injection, the mice are asphyxiated by CO2 inhalation and the peritoneal cavity is lavaged with 2 mL ice cold Hanks Balanced Salt Solution (HBSS) without CaCl2, MgSCL · 7H2O and MgCl2 · 6H2O. Peritoneal lavage fluid from each mouse is removed by syringe and placed in 5 mL plastic test tubes put on ice and volume is noted. Preparation of samples for evaluation by ELISA is as follows: Samples are centrifuged at 800 xg for 15 minutes; 1 mL of the supernatant is added to 8 mL ice cold methanol and kept at 70’C overnight to precipitate protein; and samples are then centrifuged at 800 xg for 15 minutes, followed by a drying procedure in a Savant speed vac concentrator. The samples are reconstituted with 1 mL ice cold ELISA buffer and stored at -70*C until assayed. The assay for eicosanoids (LTC4 and 6-keto-PGFia) is performed according to conventional ELISA procedures.
Compounds to be tested orally are suspended in 0.5% Tween 80. Compounds to be tested intraperitoneally are suspended in 0.5% methylcellulose in 0.9% saline.
The total metabolite level in lavage fluid/mouse is calculated and the significance is determined by a one-way analysis of variance with LSD comparisons to control (p < 0.05). Drug effects are expressed as a percent change from control values.
October 1989 3ED40:0CO:GT:jj AHP-9498 PATENT -40The activity of standard drugs in this assay is as follows: Compound _EDsn mg/kg__ LT£4 6-keto-PGFin/TxB? BW755C <10 22.0 Phenidone 24.0 <30.0 Indomethacin Not Active 0.126 Ibuprofen Not Active 7.0 When tested in this assay a compound of the invention and the antiinflammatoiy compound etodolac gave the following results: Tabtejy Compound of Example No. Dose mg/kg % Inhibition LTCa 6-keto-PGF 5 10 (i.p.) * 86 -27 ** intraperitoneally administered negative values denote potentiation The results show that the compound of the invention exerts a potent inhibitory effect on the lipoxygenase pathway but not on the cyclooxygenase pathway.

Claims (12)

1. A compound having the formula A(CH 2 ) n O-B 5 wherein A is phenoxyethyl, phenoxyphenyl or a group having the formula wherein R 3 I X is -N- or -C-; R 3 R 3 R 1 R 3 R 3 III II Z is -C=C-, -C=N-, -N=C-, -N-, -S- or -O-; R 1 is hydrogen, lower alkyl or phenyl; R 2 is hydrogen or lower alkyl; or R 1 and R 2 taken together form a benzene ring; R 3 is hydrogen or lower alkyl; n is 1 -
2. ; B is wherein R 4 and R 5 are each, independently, hydrogen or lower alkyl; R 6 is halo or nitro; 12 October 1989 3. ED42:OCO:GT:jj AHP-9498 PATENT -42-R 4 R 7 is -C-R 8 or -CHCOOR 5 ; R 8 is lower alkyl; m is 0 - 3 ; and the pharmacologically acceptable salts thereof. 5 2. The compound of Claim 1, having the name l-[2-nitro-4'-(2-quino linylmethoxy)-[ 1,1 ’-biphenyl]4-yl]ethanone.
3. The compound of Claim 1, having the name 2-fluoro-4'-(2-quinolinyl methoxy- [ 1,1 '-biphenyl]-4-acedc acid.
4. The compound of Claim 1, having the name 3-[4-(2-quinolinyl 10 methoxy)benzoyl]benzene acetic acid.
5. The compound of Claim 1, having the name 3-[4-(2-naphthalenyl methoxy)benzoyl]benzene acetic acid.
6. The compound of Claim 1, having the name 5-phenyl-4-[4-(2-quino linylmethoxy)-phenyl]-2-oxazole propanoic acid. 15
7. The compound of Claim 1, having the name 4-[4-[2-naphthalenyl methoxy]phenyl]-5-phenyl-2-oxazole propanoic acid.
8. The compound of Claim 1, having the name 4-[4-[2-naphthalenyl methoxy]phenyl]-5-phenyl-2-oxazole propanoic acid methylester.
9. The compound of Claim 1, having the name 4-[4-[(l-methyl-1 H-benz 20 imidazol-2-yl)methoxy]phenyl]-5-phenyl-2-oxazole propanoic acid.
10. A compound having the formula given and defined in Claim 1, or a pharmacologically acceptable salt thereof, substantially as hereinbefore described and exemplified.
11. A process for preparing a compound having the formula given and defined in Claim 1, or a pharmacologically acceptable salt thereof, substantially as hereinbefore described and exemplified. -43
12. A compound having the formula given and defined in Claim 1, or a pharmacologically acceptable salt thereof, whenever prepared by a process claimed in Claim 11.
IE387490A 1989-10-27 1990-10-26 Substituted benzoylbenzene-, biphenyl- and 2-oxazole-¹alkanoic acid derivatives as inhibitors of pla2 and¹lipoxygenase IE903874A1 (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US42767789A 1989-10-27 1989-10-27

Publications (1)

Publication Number Publication Date
IE903874A1 true IE903874A1 (en) 1991-05-08

Family

ID=23695804

Family Applications (1)

Application Number Title Priority Date Filing Date
IE387490A IE903874A1 (en) 1989-10-27 1990-10-26 Substituted benzoylbenzene-, biphenyl- and 2-oxazole-¹alkanoic acid derivatives as inhibitors of pla2 and¹lipoxygenase

Country Status (11)

Country Link
EP (1) EP0497888A1 (en)
JP (1) JPH05502021A (en)
KR (1) KR927003552A (en)
AU (1) AU647425B2 (en)
BR (1) BR9007783A (en)
CA (1) CA2067138A1 (en)
FI (1) FI921864A0 (en)
HU (1) HUT64750A (en)
IE (1) IE903874A1 (en)
PT (1) PT95690A (en)
WO (1) WO1991006538A1 (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5229516A (en) * 1989-10-27 1993-07-20 American Home Products Corporation Substituted indole-, indene-, pyranoindole- and tetrahydrocarbazole-alkanoic acid derivatives as inhibitors of PLA2 and lipoxygenase
US5021576A (en) * 1989-10-27 1991-06-04 American Home Products Corporation 2-Anilino phenylacetic acid derivatives
US5190968A (en) * 1991-09-30 1993-03-02 Merck Frosst Canada, Inc. (Polycyclic-arylmethoxy) indoles as inhibitors of leukotriene biosynthesis
US5290798A (en) * 1991-09-30 1994-03-01 Merck Frosst Canada, Inc. (hetero-arylmethoxy)indoles as inhibitors of leukotriene biosynthesis
US5308850A (en) * 1991-09-30 1994-05-03 Merck Frosst Canada, Inc. (Bicyclic-hetero-arylmethoxy)indoles as inhibitors of leukotriene biosynthesis
US5374635A (en) * 1993-03-29 1994-12-20 Merck Frosst Canada, Inc. Furo[3,2-b]pyridines and thieno[3,2-b]pyridines as inhibitors of leukotriene biosynthesis
AU684316B2 (en) * 1993-10-15 1997-12-11 Shionogi & Co., Ltd. Oxazolinone derivative having intracellular phospholipase A2 inhibitor activity
EP1113801A4 (en) * 1998-09-17 2002-10-02 Bristol Myers Squibb Co METHOD FOR TREATING ATHEROSCLEROSIS EMPLOYING AN aP2 INHIBITOR AND COMBINATION
AU2004224825A1 (en) * 2003-03-26 2004-10-07 Crc For Asthma Limited Therapeutic and prophylactic compositions and uses therefor

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1710984A (en) * 1923-03-14 1929-04-30 Ingram Harry Closure for tumblers, jars, and other containers
US3578671A (en) * 1967-11-06 1971-05-11 Wyeth John & Brother Ltd Oxazoles
US4659728A (en) * 1985-02-25 1987-04-21 American Home Products Corporation Hydroxy substituted 4,5-diphenyl-2-oxazole propanoic acid
US4681940A (en) * 1985-11-19 1987-07-21 American Home Products Corporation 5-[3-[[2-quinolyl]methoxy]phenyl]-1,3-oxazoles
US4920133A (en) * 1987-11-03 1990-04-24 Rorer Pharmaceutical Corp. Quinoline derivatives and use thereof as antagonists of leukotriene D4
US4920132A (en) * 1987-11-03 1990-04-24 Rorer Pharmaceutical Corp. Quinoline derivatives and use thereof as antagonists of leukotriene D4

Also Published As

Publication number Publication date
AU647425B2 (en) 1994-03-24
HUT64750A (en) 1994-02-28
JPH05502021A (en) 1993-04-15
WO1991006538A1 (en) 1991-05-16
FI921864A (en) 1992-04-24
KR927003552A (en) 1992-12-18
HU9201395D0 (en) 1992-07-28
CA2067138A1 (en) 1991-04-28
PT95690A (en) 1991-09-13
FI921864A0 (en) 1992-04-24
EP0497888A1 (en) 1992-08-12
AU6722090A (en) 1991-05-31
BR9007783A (en) 1992-08-11

Similar Documents

Publication Publication Date Title
AU643996B2 (en) Substituted indole-, indene-, pyranoindole- and tetrahydrocarbazole- alkanoic acid derivatives as inhibitors of PLA2 and lipoxygenase
US5420289A (en) Substituted indole-, indene-, pyranoindole- and tetrahydrocarbazole-alkanoic acid derivatives as inhibitors of PLA2 and lipoxygenase
US5229516A (en) Substituted indole-, indene-, pyranoindole- and tetrahydrocarbazole-alkanoic acid derivatives as inhibitors of PLA2 and lipoxygenase
US4772703A (en) 2-(phenoxymethyl)-quinazolines as antiallergic and antiinflammatory agents
AU654292B2 (en) Naphthalenepropionic acid derivatives as anti-inflammatory/antiallergic agents
US5218124A (en) Substituted benzoylbenzene-, biphenyl- and 2-oxazole-alkanoic acid derivatives as inhibitors of pla2 and lipoxygenase
GB2181135A (en) Substituted benzene derivatives
AU647425B2 (en) Substituted benzoylbenzene-, biphenyl- and 2-oxazole- alkanoic acid derivatives as inhibitors of PLA2 and lipoxygenase
US5021576A (en) 2-Anilino phenylacetic acid derivatives
DE4127842A1 (en) New 5-(omega-arylalkyl)-2-thienyl alkanoic acid derivs. as LTB inhibitors - for treating eczema, psoriasis, acne, arthritis, asthma, rhinitis and inflammatory skin disorders
US5071988A (en) Substituted benzoylbenzene-, biphenyl- and 2-oxazole-alkanoic acid derivatives
EP0301813B1 (en) Naphthalenepropionic acid derivatives
PT775133E (en) NAFTALENE AMIDES WITH LEUCOTRIENE ANTAGONIST ACTION
US5364944A (en) Substituted benzoylbenzene-, biphenyl- and 2-oxazole- alkanoic acid compounds
US5159085A (en) 2-anilino phenylacetic acid derivatives as inhibitors of PLA2 and lipoxygenase
RU2037481C1 (en) Derivatives of substituted benzoylbenzene-, biphenyl or 2-oxazolealkanoic acid or their pharmacologically acceptable salts, and a method of their synthesis
Musser et al. Substituted indole-, indene-, pyranoindole-, and tetrahydrocarbazole-alkanoic acid derivatives as inhibitors of PLA 2 and lipoxygenase
US5428171A (en) 2-substituted quinoline dioic acids
CA1331000C (en) Naphthalenepropionic acid derivatives as anti-inflammatory/antiallergic agents