WO1991006537A2 - Substituted indole-, indene-, pyranoindole- and tetrahydrocarbazole- alkanoic acid derivatives as inhibitors of pla2 and lipoxygenase - Google Patents

Substituted indole-, indene-, pyranoindole- and tetrahydrocarbazole- alkanoic acid derivatives as inhibitors of pla2 and lipoxygenase Download PDF

Info

Publication number
WO1991006537A2
WO1991006537A2 PCT/US1990/006251 US9006251W WO9106537A2 WO 1991006537 A2 WO1991006537 A2 WO 1991006537A2 US 9006251 W US9006251 W US 9006251W WO 9106537 A2 WO9106537 A2 WO 9106537A2
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
compound
acetic acid
indole
quinolinylmethoxy
Prior art date
Application number
PCT/US1990/006251
Other languages
French (fr)
Other versions
WO1991006537A3 (en
Inventor
John Henry Musser
Anthony Frank Kreft, Iii
Amedeo Arturo Failli
Christopher Alexander Demerson
Uresh Shantilal Shah
James Albert Nelson
Original Assignee
American Home Products Corporation
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 Products Corporation filed Critical American Home Products Corporation
Priority to KR1019920700971A priority Critical patent/KR920702345A/en
Priority to BR909007790A priority patent/BR9007790A/en
Publication of WO1991006537A2 publication Critical patent/WO1991006537A2/en
Publication of WO1991006537A3 publication Critical patent/WO1991006537A3/en
Priority to FI921865A priority patent/FI921865A0/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom 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
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/50Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton
    • C07C323/62Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atom of at least one of the thio groups bound to a carbon atom of a six-membered aromatic ring of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/18Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D209/22Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with an aralkyl radical attached to the ring nitrogen atom
    • 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
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems

Definitions

  • This invention relates to novel substituted indene-, indole-, pyrano- indole- and tetrahydrocarbazole alkanoic acid derivatives possessing lipoxygenase inhibitory, phospholipase A 2 inhibitory and leukotriene antagonist activity, which are useful as anti-inflammatory, antiallergic and cytoprotective agents.
  • arachidonic acid AA
  • arachidonic acid AA
  • the metabolism of arachidonic acid by cyclooxy ⁇ genase enzymes results in the production of prostaglandins and thromboxanes.
  • the physiological activity of the prostaglandins has already been amply elucidated in recent years.
  • TxA 2 is a vasoconstrictor which stimulates platelet aggregation.
  • PKI 2 prostacyclin
  • peptide mediator vascular permeability and edema forming properties 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, C 4 and D4.
  • SRS-A slow-reacting substance of anaphylaxis
  • LT and LTD 4 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. Pis.. 126.
  • Phospholipase A2 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.
  • AA arachidonic acid
  • PAF platelet activating factor
  • 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)].
  • PLA2 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.
  • 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.
  • prostaglandins exert a cytoprotective effect on the gastric mucosa [see Robert, Gastroenterologv.2 , 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)].
  • 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.
  • exogenous LTD 4 evokes both increased pepsin secretion and decreased transgastric potential [Pendleton et al., Eur. J. Pharmacol.. 125. 297-99 (1986)].
  • 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 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 (Gastroenterologv.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-inflamma- tory 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.
  • 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 cyto- protection must focus on either blocking the release of mediators of these conditions or antagonizing their effects.
  • compounds which inhibit the biological effects of the leukotrienes and SRS's and/or which control the biosynthesis of these substances are considered 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.
  • novel substituted indene-, indole-, pyranoindole- and tetrahydrocarbazole 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 is C 4 -C8 alkyl, phenoxyethyl, phenoxyphenyl or a group having the formula
  • R 3 I X is -N- or -C- ;
  • R 1 is hydrogen, lower alkyl, phenyl or phenyl substituted with trifluoromethyl
  • R 2 is hydrogen or lower alkyl
  • R 1 and R 2 taken together form a benzene ring;
  • R 3 is hydrogen or lower alkyl;
  • n is 1 - 2;
  • B is
  • R 4 is -CO 2 R 2 , -N(OH)CNH 2 , -N(OH)CR 7 , -NHCNHOH ,
  • R 5 is ; or phenyl or phenyl substituted by halo, lower alkylthio, lower alkylsulfinyl or lower alkylsulfonyl; R 6 is A(CH2) n O- or halo; with the proviso that when R 6 is halo, R 5 is
  • R 7 is lower alkyl; Y is -CH2- or -O- ; R8 is lower alkyl or -(CH2) m CO2R 3 ; O
  • R 9 is -CR 10 or -CH 2 R 10 ;
  • R 10 is lower alkyl, phenyl, phenyl substituted with carboxy, halo, loweralkylthio or loweralkylsulfinyl, pyridyl, furanyl or quinolinyl;
  • R 11 is lower alkyl or phenyl; and the pharmacologically acceptable salts thereof.
  • the terms “lower alkyl” and “lower alkoxy” refer 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, benzothia- zolyl, indolyl, benzoxazolyl, quinolinyl, quinazolinyl, benzimidazolyl, quinoxalinyl, quinazolinyl and the like. Especially preferred are quinolinyl, benzothiazolyl, benzimidazolyl and 2-phenylthiazole.
  • 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-, di- and triethylammonium, mono-, di- and tripropylammonium (iso and normal), ethyldimethylammonium, benzyldimethylammonium, cyclohexylammonium, benzy- lammonium, dibenzylammonium, piperidinium, morpholinium, pyrrolidinium, piperazinium, 1-methylpiperidinium, 4-ethylmo holinium, 1-isopropylpyrrolidinium, 1,4-dimethylpiperazinium, 1-n-butyl-piperidinium, 2-methylpiperidinium, l-ethyl-2- methylpiperidinium, mono-, di- and triethanolammonium, ethyl diethanolammonium, n-butylmonoethanolammonium, tris
  • the compounds of the invention can be prepared by the following reaction schemes. When it is desired to prepare compounds having the formula
  • indanone intermediate is then subjected to demethylation and the resulting hydroxy- indanone is reacted with an appropriate haloalkyl-A compound where A is as defined hereinbefore and hal is halo
  • R 5 group is lower alkylthiophenyl
  • R 5 is a lower- alkylsulfinylphenyl grouping
  • R 5 is a lower- alkylsulfinylphenyl grouping
  • the intermediate hydroxyindanone can be reacted with cyanoacetic acid to give the corresponding hydroxyindene-3-acetic acid, which is then esterified before introduction of the R 5 -methylene grouping in order to facilitate the purification of desired intermediates. Since introduction of the R5-methylene grouping is accompanied by ester hydrolysis, the intermediate free acid is re-esterified to facilitate purification followed by reaction with an appropriate halo- alkyl- A group to yield the desired final product
  • the final product free acids can be esterified via conventional methods and in like fashion, final product esters can be hydrolyzed by known procedures to yield the corresponding free acids.
  • X is -CH2-
  • X is -CH2-
  • 4-methoxyphenylhydrazine with, for example, 2-carbomethoxymethyl-2-alkylcyclohexanone, followed by ring closure to yield an intermediate tetrahydro-methoxy-lH-carbazole-1 -acetic acid
  • the latter intermediate is demethylated with hydrobromic acid to yield the corresponding hydroxy compound, which is then reacted with an appropriate halo- loweralkyl-A compound by one of several routes.
  • A(CH2) n hal two equivalents of the starting material, A(CH2) n hal, are reacted with a metal derivative of the hydroxycarbazole-1 -acetic acid to form an intermediate ester ether which is hydrolyzed to yield the desired final products.
  • the metal derivative of the hydroxycarbazole-1 -acetic acid may be prepared by treating the acid with an alkali metal alkoxide, such as sodium methoxide.
  • A, n, m and R 7 are as defined hereinbefore and hal denotes chloro, bromo or iodo.
  • the desired final products can be prepared by the alkylation of alkyl esters of the hydroxycarbazole-1-acetic acid:
  • X is -O- can be prepared by the above-outlined reaction sequences which dif ⁇ fer only in the preparation of the starting tetrahydro-6-hydroxypyrano[3,4-b]indole-l- lower alkanoic acid esters.
  • the latter can be prepared by the reaction of, for example, a 5-benzyloxytryptophol with a loweralkyl-(R 7 -carbonyl)lower alkanoate in boron trifluoride etherate to yield an intermediate l-R 7 -tetrahydro-6-(phenylmethoxy)pyrano- [3,4-b]indole-l-alkanoic acid ester, which is then subjected to reduction to yield the corresponding 6-hydroxy ester
  • Another scheme by which the starting tetrahydro-6-hydroxypyrano[3,4- bjindole 1-alkanoic acid ester can be prepared involves reducing 7-loweralkyltryptophol to 7-loweralkyl-2,3-dihydrotryptophol, followed by reacting the latter with potassium nitrosodisulfonate to yield a 7-loweralkyl-5-hydroxytryptophol, which is then reacted with a suitable lower alkyl-3-methoxy-lower alkanoate to yield the desired intermediate tetrahydro-6-hydroxypyrano[3,4-b]indole-l-loweralkanoic acid ester
  • carbazole final product free acids can be esterified via conventional methods and in like fashion, final product esters can be hydrolyzed by known procedures to yield the corresponding free acids.
  • A(CH2)nhal compound can be prepared by reacting 5-hydroxy-2-methyl-lH-indole-3-loweralkanoic acid with an appropriate A(CH2)nhal compound in the presence of sodium methoxide/methanol to yield the intermediate A(CH2) n O-containing indole alkanoic acid derivative, which is then esterified and reacted with a suitable substituted benzyl or benzoyl halide to yield the desired final product in ester form, which can be converted to the free acid form by conventional methods.
  • the ⁇ -methyl tetrahydrocarbazole acetic acid ester intermediate is reacted under reflux with chloranil in xylene under a nitrogen atmosphere to yield the corresponding ⁇ -methylcarbazole acetic acid, which is reduced by hydrogenation to remove the benzyloxy group, yielding the 6-hydroxy- ⁇ -methyl carbazole acetic acid ester intermediate.
  • the intermediate compound 2-bromomethylquinoline can be prepared by the following reaction sequence:
  • 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.
  • such intermediates as l-methyl-2-chloromethylbenzimidazole, 2-chloromethylbenzthiazole and 2-chloromethylbenzoxazole can be prepared by the following reaction scheme
  • reaction is preferably carried out at a controlled low temperature in an organic solvent, such as methylene chloride.
  • E and Z stereoisomers and the individual isomers can be differentiated by the prefixes E and Z, as assigned by the accepted sequence rules procedures. Accordingly, the present invention embraces the E, Z and mixed isomer forms of those final product compounds exhibiting this form of stereoisomerism.
  • 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 the oxidation of arachidonic acid.
  • the com ⁇ pounds 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.
  • diseases as rheumatoid arthritis, inflammatory bowel disease, osteoarthritis, tendinitis, bursitis, psoriasis (and related skin inflammation) and similar conditions involving inflammation.
  • LTC4, LTD4 and LTE4 which are the constituents of SRS-A, they are useful for the inhibition of symptoms induced by these leukotrienes.
  • 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 hyper- sensitivity reactions, such as allergic conjunctivitis.
  • the compounds are especially valuable in the prevention and treatment of allergic bronchial asthma.
  • the compounds 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 cyto ⁇ protective effect of the compounds of the invention significantly reduces the gastro- irritant impact of conventional anti-inflammatory 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.
  • 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-inflamma ⁇ tory drugs (NS AID'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 glycerol-induced renal failure.
  • NS AID's non-steroidal anti-inflamma ⁇ tory drugs
  • the compounds of the invention 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 carboxymethyl- cellulose, low melting wax, cocoa butter and the like.
  • Diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, tablet-disintegrating agents and the like may be employed.
  • the compounds may be encapsulated with or without other carriers.
  • the proportion of active ingredients in said compositions both solid and liquid will be at least to impart the desired activity thereto on oral administra ⁇ tion.
  • 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.
  • 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.
  • 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 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 and non-human source PLA2.
  • the procedures further measure the ability of the compounds of the invention to inhibit, in vivo, the activity of exogenously administered PLA2.
  • the pharmacological testing additionally demonstrates the ability of the compounds of the invention to inhibit, in vivo, the lipoxygenase and cyclooxy ⁇ genase pathways of arachidonic acid metabolism; the in vitro, leukotriene antagonist activity of the compounds of the invention; and also measures the in vivo activity of the compounds as anti-inflammatory agents in the rat carrageenan paw edema assay. Finally, the potential of the compounds to induce acute gastroirritation in rats is measured in a test procedure.
  • Example 1 3-[(4-ChlorophenyI)methylene]-[2-methyl-6-(2-quinoIinyl- mgthQ ⁇ y)1-3H-i ⁇ nJene-l -acetic n-i
  • Step B 0.227 mole of Step B, is added to warm (50"C) polyphosphoric acid (420 g). The mixture is then placed in an oil bath heated at 95"C for 90 minutes (TLC, 9:1 dichloromethane-methanol, UV). The warm mixture is poured into water (3 L) and stirred overnight at room temperature. It is then extracted with ether and ethyl acetate and the combined extracts are washed with saturated NaHCO3 to neutral. The organic phase is washed with brine and dried (MgSO4). Removal of the solvent provides the title compound as an amber oil.
  • the basic extracts are neutralized (ice bath) with concentrated HCl and the oil that separates is extracted with ether (3 times).
  • the combined extracts are washed with brine, dried (MgSO4) and evaporated to yield an oil that readily solidifies.
  • the crude material is flash chromatographed (on silica Merck- 60, gradient CH2G2, CH2 ⁇ 2-ethyl acetate 85:15) to provide the pure title compound (13.60 g, nearly colorless solid, 82%).
  • Triethylphosphonoacetate (22.41 g, 100 mmole) is added dropwise under nitrogen to a stirred and cooled (0°C) slurry of NaH (57% in oil, 4.27 g, 100 mmole) in dry toluene (250 mL). The cooling bath is removed and stirring continued at room temperature for 60 minutes whereby an almost homogeneous solution is obtained. A solution of the indanone (15.15 g, 50 mmole) of Step E, in toluene (50 mL) is then added dropwise.
  • the flask is placed in an oil bath heated at 95 ⁇ C for 19 hours (TLC, traces of starting material present, dichloromethane-ethyl acetate 9:1, UV, Vaughn's).
  • TLC traces of starting material present, dichloromethane-ethyl acetate 9:1, UV, Vaughn's.
  • the reddish solution is cooled, diluted with water and extracted with ethyl acetate (3 times).
  • the combined extracts are washed with brine, dried (anhydrous K2CO3) and evaporated to dryness.
  • the dark orange solution is cooled, the methanol evaporated, more water added and the gelatinous precipitate is collected, washed with water and dried to provide the sodium salt of the title compound as a bright yellow solid.
  • the latter is slurried in water and neutralized (to pH 6-6.5) with 10% acetic acid.
  • the mixture is extracted with ethyl acetate (large volume), the extract is washed with brine and evaporated to dryness.
  • the residue is azeotroped with benzene to provide an orange solid [7.52 g, mixture of Z (major) and E (minor) isomers]. This material is slurried in ether, stirred for 30 minutes, filtered and dried (4.32 g).
  • the slurry (bright yellow precipitate) is treated dropwise with water (65 mL) and a little methanol and refluxing is continued for another 5.5 hours (reaction followed by TLC).
  • the brown solution is cooled, the methanol evaporated, more water added and the gelatinous precipitate is collected, washed with water and dried to provide the sodium salt of the title compound as a yellow solid.
  • the latter is slurried in water and neutralized (to pH 6-6.5) with 10% acetic acid.
  • the mixture is extracted with ethyl acetate flarge volume), the extract is washed with brine and evaporated to dryness.
  • the residue is azeotroped with benzene to provide a yellow solid (4.47 g, 52%).
  • the precipitate is filtered, slurried in water, collected, washed with water and dried in vacuo (over P2O5) to yield the crude acid as a yellow solid.
  • the solid is redissolved in a large volume of warm (30-35 * C) ethyl acetate (containing some methanol and dichloromethane) and filtered.
  • the filtrate is concentrated in vacuo until precipitation starts and diluted with ether.
  • the precipitate is collected, washed with ether and dried (fluffy solid, m.p. 136-137"C, sintering and foaming).
  • the crude acid is recrystallized from a hot mixture of methanol and ethanol (large volume needed to dissolve, concentrate to smaller volume after filtration).
  • the analytical sample is dried overnight in vacuo at 35"C.
  • the yellow solid m.p. 218-
  • the crude sodium salt is slurried in water and acidified (to pH 6.5) with 10% acetic acid.
  • the acid is collected, washed and dried in vacuo. It is again washed with ether and dried (1.02 g, 32%, m.p. 209-21 l'C, dec).
  • the NMR shows it to be a mixture of Z and E isomers in approximately 8:1 ratio.
  • the crude product is flash chromatographed (on silica Merck-60, toluene-hexane-isopropanol 50:50:1 as eluent) to yield the pure title compound (0.630 g, yellow foam that solidifies upon trituration with ethanol, 69.6% over 2 steps).
  • Mixture of Z and E isomers in approxi ⁇ mately 4:1 ratio (based upon the position of the 2-CH3 signal in the NMR spectrum).
  • a solution of potassium nitrosodisulfonate (17.0 g, 0.063 mol) in pH 7 buffer (760 mL) is added over a 20 minute period to a stirring solution of 7- ethyl-2,3-dihydrotryptophol (5.0 g, 0.026 mol) in 350 mL of acetone.
  • the reaction mixture is extracted with ethyl acetate (4 x 300 mL).
  • the combined ethyl acetate layers are washed with distilled water (2 x 200 mL), once with brine (200 mL), dried (MgSO 4 ) and concentrated to afford 5.6 g of crude product.
  • the crude product is loaded onto a flash chromatography column. The next day, it is flashed using 70% ethyl acetate/hexane to afford 2.5 g of title compound.
  • a solution of sodium hydroxide (3.9 g, in 20 mL H2O) is added and stirred at room temperature followed by addition of 2-chloromethyl quinoline hydrochloride (8.4 g, 0.0392 mol).
  • the reaction mixture is heated at 70-80'C for 3.5 hours, cooled and diluted with water (100 mL).
  • the mixture is made acidic with IN HCl to pH 4, and extracted with ethyl acetate (3 x 400 mL).
  • the combined organic layers are washed with water (3 x 200 mL), once with brine (200 mL), dried over MgS04, filtered and concen ⁇ trated to give 10.0 g of crude product.
  • Step B (800 mL), tetrahydrofuran (100 mL) and the acid (21.0 g, 0.056 mol) of Step B, is hydrogenated at 30 psi for 18 hours.
  • the reaction mixture is filtered and concentrated to give 21.0 g of crude product, which is used in the next reaction without further purification.
  • the reaction mixture is cooled, poured into water (1500 mL) and made acidic with IN HCl to pH 4.
  • reaction mixture is cooled to room temperature and washed with 5% sodium bicarbon ⁇ ate, (1 x 200 mL), water (200 mL) and brine (100 mL), dried (MgSO4), filtered and concentrated to give 8.0 g of crude product as a thick oil, which is flash chro ⁇ matographed on silica gel using 20% ethyl acetate/hexane as an eluent to afford 5.5 g oil which solidifies upon standing, m.p. 96-98"C.
  • Step A and 10% Pd/C catalyst (600 mg) in ethanol (150 mL) is hydrogenated for 6 hours in a Parr shaker. Filtration of the catalyst and evaporation of the solvent affords the title compound (3.75 g, 89%) as a cream-colored foam homogeneous by TLC.
  • Trimethylsilyldiazomethane (40 mL, 10% by weight methylene chloride) is added dropwise to a mixture consisting of 1 -ethyl- l,3,4,9-tetrahydro-(2- quinolinylmethoxy)pyrano[3,4-b]indole-l-acetic acid (1.0 g, 0.002 mol) as prepared in Example 9, methylene chloride (150 mL) and methanol (15 mL). The reaction mixture is stirred at room temperature for 1/2 hour, and is concentrated to give 1.2 g of oily residue, which is very difficult to crystallize. The ester is converted to the hydrochloride salt by adding ether/HCl.
  • the reaction mixture is cooled to room temperature and washed with 5% sodium bicarbonate (1 x 200 mL), water (200 mL) and brine (200 mL). It is dried (MgSO 4 ), filtered and concentrated to give 5.0 g product as thick oil.
  • the crude product is flash chromatographed with 15% ethyl acetate/hexane as an eluent to afford 2.5 g product, m.p. 128-129°C.
  • Step A A mixture consisting of 10% Pd/c on charcoal (0.7 g), ethanol (150 mL) and l,3,4,9-tetrahydro-l,l-dimethyl-6-(phenylmethoxy)pyrano[3,4- bjindole (4.2 g, 0.014 mol) of Step A, is hydrogenated at 30 psi for 18 hours. The reaction mixture is filtered and concentrated to give 3.5 g product which is used in the next reaction without further purification.
  • a mixture consisting of 5-benzyloxytryptophol (7.0 g, 0.0262 mol), diethyl ketone (10 mL), benzene (400 mL) and p-toluene sulfonic acid is refluxed for 3.5 hours using a Dean-Stark tap to remove the water.
  • the reaction mixture is cooled to room temperature, washed with 5% sodium bicarbonate, water (200 mL), brine (200 mL), dried (MgSO 4 ), filtered and concentrated to give 9.9 g of crude product.
  • the crude product is flash chromatographed on silica gel using 15% ethyl acetate/hexane as an eluent to afford 6.5 g of pure title compound.
  • Step A (6.5 g, 0.0193 mol), of Step A, is hydrogenated at 30 psi for 18 hours.
  • the reaction mixture is filtered and concentrated to give 5.2 g of product which is used in the next reaction without further purification.
  • a mixture consisting of the 6-hydroxy compound (5.2 g, 0.0212 mol) of Step B, dimethyl sulfoxide (150 mL), sodium hydroxide (6.0 g, 0.15 mol in 20 mL water), and 2-chloromethylquinoline hydrochloride (6.0 g, 0.028 mol) is heated to 70-80'C for 6 hours.
  • the reaction mixture is cooled to room temperature, diluted with water (800 mL), and extracted with ethyl acetate (3 x 150 mL).
  • the combined organic layers are washed once with water (2 x 150 mL), once with brine (100 mL), dried (MgSO4), filtered and concentrated to give 9.9 crude product.
  • the crude product is flash chromatographed on silica gel using 25% ethyl acetate/hexane as an eluent to afford 4.5 g pure product as a thick oil.
  • the oil is dissolved in ether
  • a solution of sodium nitrite (3.8 g, 0.055 mol in 20 mL of H2O), is added dropwise to an ice cold stirring suspension of 4-benzyloxyaniline (13.0 g, 0.055 mol in 150 mL of concentrated HCl).
  • the reaction mixture is stirred for 90 minutes at -8° to lO'C.
  • a solution of SnC • 2H2O (32.0 g, 0142 mol in 50 mL concentrated HCl) is added and stirred for 1 hour at O'C.
  • the reaction mixture is removed from the ice bath and is stirred at room temperature for 20 hours. The mixture is then filtered and washed with water to give 13.0 g (95% yield) of product, m.p. 182-185'C.
  • the title compound is prepared according to the method of Example 16 using 4-(2-quinolinylmethoxy)benzyl chloride. A crystalline solid is obtained, m.p. 182-185°C.
  • Example 36 l-[(4-Chlorophenyl)methyl]-2-methyI-5-(2- nvridinvlmethoxv lH-indole-3-acetic acid
  • the title compound is prepared according to the method of Example 37 using 4-chloromethyl-[(4-trifluoromethyl)-2-phenyl]thiazole. White crystals are obtained, m.p. 196-197°C.
  • the compounds 5- and 12-hydroxyeicosatetraenoic acid (5-HETE and 12-HETE) and LTB4 are early arachidonic acid oxidation products in the lipoxygenase cascade, which have been shown to mediate several aspects of inflammatory and aller ⁇ gic response. This is especially true with respect to 5,12-diHETE, which is also denoted as LTB 4 [see Ford-Hitchinson, J. Rov. Soc Med.. J_ , 831 (1981)].
  • Com ⁇ pounds which inhibit the PLA2-mediated release of arachidonic acid thereby effectively prevent the oxidation of arachidonic acid to the various leukotriene products via the lipoxygenase cascade.
  • PLA 2 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 polymorpho- nuclear leukocytes (PMN) in the presence of exogenous substrate.
  • PMN polymorpho- nuclear leukocytes
  • the assay is carried out as follows:
  • Rat polymorphonuclear leukocytes are obtained from female Wistar rats (150-200 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% NaCl). The exudate is centrifuged at 400 xg for 10 minutes. The supernatant fluid is discarded and the cell jjellet is resuspended to a concentration of 2.0 x 10 7 cells/mL in HBSS containing Ca ++ and Mg++ and 10 ⁇ M L-cysteine.
  • test drugs or vehicle are added, then preincubated at 37'C for 10 minutes.
  • A23187 (1 ⁇ M), [ H]-AA (3.0 ⁇ Ci/mL) and unlabeled AA (1 ⁇ M) are then added and the samples are further incubated for 10 minutes.
  • the reaction is terminated by centrifugation and pelleting cells.
  • Supematants 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:
  • Percent solvent changes are accomplished in a linear fashion.
  • Results are expressed as percent inhibition at a given compound dose or as an IC50 value.
  • Example 43 The procedure of Example 43 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.
  • the procedure of Example 43 is carried out as described.
  • the samples are co- chromatographed with authentic reference [3H]-PGE2-
  • 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:
  • 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.
  • 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 ⁇ -counting analysis.
  • a "total counts" sample is prepared by pipetting 25 ⁇ L 3 H-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.
  • the ability of the compounds of the invention to inhibit paw edema induced by the exogenous administration of PLA 2 is measured in the in vivo PLA 2 murine paw edema assay.
  • the assay is carried out as follows:
  • Non-fasted, male CD-I mice (8 weeks old; 31-36 grams) are placed in plastic boxes in groups of six. The right hind paw volume is measured using mercury plethysmography (zero time). Compounds are dosed orally (0.5 mL of 0.5% Tween- 80) 1 or 3 hours prior to PLA2 injection or intravenously (0.2 mL in 0.3% dimethylsulfoxide/saline) 3 minutes prior to PLA ⁇ injection.
  • a solution of purified PLA2, from the diamond back cotton mouth snake (A. piscivorus piscivorus) is prepared in saline at a concentration of 6 ⁇ g/mL.
  • the paw edema is calculated by subtracting the zero time volume from the volume recorded at each time period. Mean paw edema for each treatment group is then calculated and expressed as ( ⁇ L + S.E.). Drug effects are expressed as a percent change from control (vehicle) values. Statistical significance is determined by a one ⁇ way analysis of variance with LSD comparison to control (p ⁇ 0.05). EDso's are determined using repression analysis.
  • Example 47 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.
  • mice 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 die peritoneal cavity is lavaged with 2 mL ice cold Hanks Balanced Salt Solution (HBSS) without CaCt ⁇ , MgSO 4 • 7H2O and MgCl • 6H2O.
  • HBSS Hanks Balanced Salt Solution
  • 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-PGF ⁇ ⁇ ) 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.
  • the activity of standard drugs in this assay is as follows:
  • the LTD4 antagonist activity of the compounds of the invention is assessed in the in vitro isolated guinea pig trachea assay. This assay is carried out as follows:
  • guinea pigs 350-400 g are euthanized by a blow to the head, the neck is opened and die trachea removed.
  • the trachea is maintained in aerated physiological salt solution, cleared of connective tissue and fat and cut into rings approximately 2 mm in width (usually containing two cartilaginous segments per ring).
  • Two pieces of silk suture are then passed through the lumen of the tracheal ring and are tied around the cartilage, one on each side of the trachealis muscle.
  • the tracheal ring is suspended between a glass hook and a force displacement transducer in a 10 mL organ bath for measurement of isometric tension.
  • Tissues are maintained at 37° C in aerated (95% CO2/5% CO2) physiological salt solution of the following composition: NaCl (100 mM), KH2PO4 (1.18 mM), KCl (4.74 mM), CaCl (2.5 mM), MgSO4 • 7 H 2 O (1.19 mM), NaHCL 3 (25 mM), dextrose (11.1 mM) and indomethacin (1 ⁇ M).
  • the tracheal rings are maintained at 2 g resting tension and equilibrated for 45 minutes (with frequent washing and readjustment of resting tension).
  • the tracheal rings are first contracted by the addition of carbachol (3xlO" 6 M), to determine tissue responsiveness and establish a reference contraction. On attainment of a stable level of contraction (approximately 30 minutes), the tissues are washed several times until baseline tension has been restored and dien re-equilibrated for 30 minutes. The tissues are then incubated for 45 minutes with a test antagonist (either lxlO ⁇ M or lxlO ⁇ M) or 10 ⁇ L of an appropriate solvent control (control, non-treated). One tissue in each group serves as the control.
  • a test antagonist either lxlO ⁇ M or lxlO ⁇ M
  • 10 ⁇ L of an appropriate solvent control control, non-treated
  • L-cysteine (lxl0" 2 M final bath concentration) is added to inhibit bioconversion of LTD4 to LTE 4 . Only one LTD 4 concentration-response curve is constructed in each tissue.
  • LTD4 antagonist activity is determined by comparison of the concentration response curves of LTD4 in the presence and absence of antagonist. Assessment of the relative rightward shift of the antagonist treated curve relative to the solvent (control) treated tissue is calculated as a concentration ratio (Eq. A) and used in subsequent calculations to derive an antagonist pK ⁇ value (Eqs B and C). In the event that the maximum response to LTD4 is depressed, the EC50 for that particular curve is determined, an "apparent" pK ⁇ reported, and the compound reported as "non-competitive.”
  • the compounds of the invention are further tested in the rat carrageenan paw edema assay to determine their ability to inhibit the acute inflammatory response.
  • This assay is carried out as follows: 140-180 g Male Sprague-Dawley rats, in groups of 6 animals are injected subcutaneously in the right paw with 0.1 mL of 1% carrageenan at zero time. Mercury plethysmographic readings (mL) of the paw are made at zero time and 3 hours later. Test compounds are suspended or dissolved in 0.5% methylcellulose and given perorally 1 hour prior to carrageenan administration. The increase in paw volume (edema in mL) produced by the carrageenan is measured. Paw edema is calculated (3 hour volume minus zero time volume), and percent inhibition of edema is determined. Unpaired Student's t-test is used to determine statistical significance.
  • Phenylbutazone 26.2 ( 2.3, 291.0) When tested in this assay, a compound of the invention and the anti-inflammatory drug etodolac gave the following results:
  • Example 50 The assay of tiiis Example measures the ability of the compounds tested to inhibit 5-lipoxygenase in human whole blood. This assay is carried out as follows: Blood is obtained in 50-100 ml quantities from male donors. White blood cell counts and differentials are made. Two ml of blood are placed in a 15 ml polypropylene test tube. Compounds are solubilized in dimethylsulfoxide and diluted 1:10 in 10% bovine serum albumin in phosphate buffered saline, pH 7.4 resulting in a final dimethylsulfoxide concentration of 0.1% in the blood.
  • Plasma is removed from sample tubes, placed in 15 ml polypropylene test tubes containing 8 ml methanol, and then vortexed to precipitate protein. Samples are stored at -70"C overnight. The next day, samples are centrifuged at 200 x g for 15 minutes at 4°C to pellet the precipitate. Samples are dried in a Savant speed vac concentrator, reconstituted to original volume with ice cold RIA or ELISA buffer, and stored at -70°C until assayed.
  • the assay for eicosanoids (LTB4, TxB2, and PGE2) is performed as described by the manufacturer of the [ 3 H]-RIA kit or ELISA kit (LTB 4 -Amersham, TxB2 and PGE2 - Caymen Chemical).
  • the total eicosanoid level in 2 ml of blood is calculated and reported as ng/10 6 neutrophils. Significance is determined by a one-way analysis of variance with least significant difference (LSD) comparisons to control (p ⁇ 0.05) and ICso's ( ⁇ M) are determined by regression analysis (Finney, 1978). Drug effects are expressed as percent change from control values.
  • Compounds tested in vitro are solubilized in dimethylsulfoxide and diluted 1:10 in 10% bovine serum albumin in phosphate buffer saline resulting in a final dimethylsulfoxide concentration of 0.1% in the blood.

Abstract

There are disclosed compounds of the formula: A(CH2)nO-B, wherein A is C4-C8 alkyl, phenoxyethyl, phenoxyphenyl or a group having formula (a) or (b), wherein X is -N- or (c); Z is (d), (e), (f), (g), -S- or -O-; R1 is hydrogen, lower alkyl, phenyl or phenyl substituted with trifluoromethyl; R2 is hydrogen or lower alkyl; or R?1 and R2¿ taken together form a benzen ring; R3 is hydrogen or lower alkyl; n is 1-2; B is (h), (i), (j) or (k); wherein R4 is -CO¿2R?2, (l), (m), (n), (o) or (p); m is 0-3; R5 is (q); or phenyl or phenyl substituted by halo, lower alkylthio, lower alkylsulfinyl or lower alkylsulfonyl; R6 is A(CH¿2?)nO- or halo; with the proviso that when R?6¿ is halo, R5 is (r); R7 is lower alkyl; Y is -CH¿2?- or -O-; R?8¿ is lower alkyl or -(CH¿2?)mCO2R?3; R9¿ is (s) or -CH¿2?R?10; R10¿ is lower alkyl, phenyl, phenyl substituted with carboxy, halo, loweralkylthio or loweralkylsulfinyl, pyridyl, furanyl or quinolinyl; R11 is lower alkyl or phenyl; 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 INDOLE-, INDENE-, PYRANOINDOLE- AND TETRAHYDROCARBAZOLE- ALKANOIC ACID DERIVATIVES AS INHIBITORS OF P A, AND LTP XYGENASE
This invention relates to novel substituted indene-, indole-, pyrano- indole- and tetrahydrocarbazole 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 cyclooxy¬ genase 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 haemo- stasis and thrombosis are reviewed by RJ. Gryglewski, CRC Crit. Rev. Biochem.. __, 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, 16, 287-299 (1984)]. This is through their vasode- pressor 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 chemotactic activities, stimulate lysosomal enzyme release and act as important factors in the immediate hypersensitivity reaction. It has been shown that LT 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. Pis.. 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. Rov. Soc. Med..74, 831-833 (1981), which stimulates cell accumulation and affects vascular smooth muscle [see Bray, Br. Med. Bull..22, 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.. 2, 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 intra- cellular (ischemia, 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, Gastroenterologv.2 , 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 LTQ can induce vasoconstriction in both venous and arteriolar vessels in the rat submucosa [see Whittle, IUPHAR Ninth Int. Cong, of Pharm.. S30- !__ 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., Gastroenterologv. 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 (Gastroenterologv.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-inflamma- tory 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 cyto- protection 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 phospho¬ lipids, or by inhibiting lipoxygenase, are considered 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 indene-, indole-, pyranoindole- and tetrahydrocarbazole 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 C4-C8 alkyl, phenoxyethyl, phenoxyphenyl or a group having the formula
Figure imgf000006_0001
wherein
R3 I X is -N- or -C- ;
R3 R3 R3 R3 R3
I I I I I Z is -C= C- , -C=N- , -N=C- , -N- , -S- or -O- ;
R1 is hydrogen, lower alkyl, phenyl or phenyl substituted with trifluoromethyl;
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
Figure imgf000007_0001
wherein
O O O
R4 is -CO2R2 , -N(OH)CNH2 , -N(OH)CR7 , -NHCNHOH ,
O o
II
-CN(OH)R7 or -CNHSO2Rn ; m is 0
R5 is ; or phenyl or phenyl substituted by
Figure imgf000007_0002
halo, lower alkylthio, lower alkylsulfinyl or lower alkylsulfonyl; R6 is A(CH2)nO- or halo; with the proviso that when R6 is halo, R5 is
Figure imgf000007_0003
R7 is lower alkyl; Y is -CH2- or -O- ; R8 is lower alkyl or -(CH2)mCO2R3 ; O
R9 is -CR10 or -CH2R10 ;
R10 is lower alkyl, phenyl, phenyl substituted with carboxy, halo, loweralkylthio or loweralkylsulfinyl, pyridyl, furanyl or quinolinyl;
R11 is lower alkyl or phenyl; and the pharmacologically acceptable salts thereof. The terms "lower alkyl" and "lower alkoxy" refer 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, benzothia- zolyl, indolyl, benzoxazolyl, quinolinyl, quinazolinyl, benzimidazolyl, quinoxalinyl, quinazolinyl and the like. Especially preferred are quinolinyl, benzothiazolyl, benzimidazolyl and 2-phenylthiazole.
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-, di- and triethylammonium, mono-, di- and tripropylammonium (iso and normal), ethyldimethylammonium, benzyldimethylammonium, cyclohexylammonium, benzy- lammonium, dibenzylammonium, piperidinium, morpholinium, pyrrolidinium, piperazinium, 1-methylpiperidinium, 4-ethylmo holinium, 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, phenyl- monoethanolammonium, 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
Figure imgf000008_0001
β-methoxybenzaldehyde, for example, is reacted with propionic anhydride, followed by reduction and ring closure to yield the intermediate 6-methoxy-2-methyl-l-indanone
Figure imgf000009_0001
CH3
The indanone intermediate is then subjected to demethylation and the resulting hydroxy- indanone is reacted with an appropriate haloalkyl-A compound where A is as defined hereinbefore and hal is halo
Figure imgf000009_0002
The latter intermediate, where n=l, is reacted with tiiethylphosphonoacetate in sodium hydride to yield an intermediate indene-3-acetic acid ethyl ester, which is then hydrolyzed and concomitantly reacted with an appropriate reactant to introduce the desired R5-methylene group onto the indene-3-acetic acid moiety, exemplified below by the introduction of a p-chlorophenylmethylene grouping
Figure imgf000009_0003
Figure imgf000010_0001
Compounds in which the R5 group is lower alkylthiophenyl can be prepared by using an appropriate loweralkylthiobenzaldehyde. The compounds in which R5 is a lower- alkylsulfinylphenyl grouping can be prepared from the loweralkylthiophenyl-containing compounds by reaction with 30% H2Q2 in chloroform/acetic acid.
Figure imgf000010_0002
In an alternative preparation scheme, the intermediate hydroxyindanone can be reacted with cyanoacetic acid to give the corresponding hydroxyindene-3-acetic acid, which is then esterified before introduction of the R5-methylene grouping in order to facilitate the purification of desired intermediates. Since introduction of the R5-methylene grouping is accompanied by ester hydrolysis, the intermediate free acid is re-esterified to facilitate purification followed by reaction with an appropriate halo- alkyl- A group to yield the desired final product
Figure imgf000011_0001
Figure imgf000011_0002
Compounds having the formula halo
Figure imgf000012_0001
can be prepared by reacting a halo-indene-3-acetic acid with a A(CH2)n-containing benzaldehyde prepared from the corresponding individual components
NaOC2H5 /EtOH
A(CH2)nhal A(CH2)n-O- V- CHO CHO
OH
Figure imgf000012_0002
In all of the above-oudined reaction schemes, the final product free acids can be esterified via conventional methods and in like fashion, final product esters can be hydrolyzed by known procedures to yield the corresponding free acids.
Compounds of the invention having the formula
Figure imgf000012_0003
in which X is -CH2- can be prepared by initially reacting 4-methoxyphenylhydrazine with, for example, 2-carbomethoxymethyl-2-alkylcyclohexanone, followed by ring closure to yield an intermediate tetrahydro-methoxy-lH-carbazole-1 -acetic acid
1) toluene /Δ
2) glacial acetic acid /Δ
Figure imgf000013_0001
Figure imgf000013_0002
The latter intermediate is demethylated with hydrobromic acid to yield the corresponding hydroxy compound, which is then reacted with an appropriate halo- loweralkyl-A compound by one of several routes. In one such preparative sequence, two equivalents of the starting material, A(CH2)nhal, are reacted with a metal derivative of the hydroxycarbazole-1 -acetic acid to form an intermediate ester ether which is hydrolyzed to yield the desired final products. The metal derivative of the hydroxycarbazole-1 -acetic acid may be prepared by treating the acid with an alkali metal alkoxide, such as sodium methoxide.
A(CH2)nhal + NaOCH3
Figure imgf000013_0003
Figure imgf000013_0004
Figure imgf000014_0001
In the above sequence, A, n, m and R7 are as defined hereinbefore and hal denotes chloro, bromo or iodo.
In an alternative sequence, it is possible to use only one equivalent of starting material A(CH2)nhal with the metal derivative to obtain the desired final product directly, without proceeding through the ethyl ester intermediate.
In yet another reaction sequence, the desired final products can be prepared by the alkylation of alkyl esters of the hydroxycarbazole-1-acetic acid:
Figure imgf000014_0002
hydrolysis
Figure imgf000014_0003
Figure imgf000014_0004
where A, n, m, R3, R7, R8 and hal are as defined hereinbefore. Hydrolysis is carried out using a dilute hydroxide, such as for example sodium hydroxide. Compounds of the invention having the formula
Figure imgf000014_0005
in which X is -O- can be prepared by the above-outlined reaction sequences which dif¬ fer only in the preparation of the starting tetrahydro-6-hydroxypyrano[3,4-b]indole-l- lower alkanoic acid esters. The latter can be prepared by the reaction of, for example, a 5-benzyloxytryptophol with a loweralkyl-(R7-carbonyl)lower alkanoate in boron trifluoride etherate to yield an intermediate l-R7-tetrahydro-6-(phenylmethoxy)pyrano- [3,4-b]indole-l-alkanoic acid ester, which is then subjected to reduction to yield the corresponding 6-hydroxy ester
Figure imgf000015_0001
The latter intermediate can then be reacted as outlined earlier with an appropriate A(CH2)nhal compound to yield the desired final compounds.
Another scheme by which the starting tetrahydro-6-hydroxypyrano[3,4- bjindole 1-alkanoic acid ester can be prepared involves reducing 7-loweralkyltryptophol to 7-loweralkyl-2,3-dihydrotryptophol, followed by reacting the latter with potassium nitrosodisulfonate to yield a 7-loweralkyl-5-hydroxytryptophol, which is then reacted with a suitable lower alkyl-3-methoxy-lower alkanoate to yield the desired intermediate tetrahydro-6-hydroxypyrano[3,4-b]indole-l-loweralkanoic acid ester
Figure imgf000016_0001
As with the previously described indene-1 -acetic acid derivatives, the carbazole final product free acids can be esterified via conventional methods and in like fashion, final product esters can be hydrolyzed by known procedures to yield the corresponding free acids.
Compounds of the invention having, for example, the formula
Figure imgf000016_0002
can be prepared by reacting 5-hydroxy-2-methyl-lH-indole-3-loweralkanoic acid with an appropriate A(CH2)nhal compound in the presence of sodium methoxide/methanol to yield the intermediate A(CH2)nO-containing indole alkanoic acid derivative, which is then esterified and reacted with a suitable substituted benzyl or benzoyl halide to yield the desired final product in ester form, which can be converted to the free acid form by conventional methods.
Figure imgf000017_0001
H
Figure imgf000017_0002
H
Figure imgf000017_0003
Compounds of the invention having the formula
Figure imgf000017_0004
can be prepared via the following reaction sequence: 4-benzyloxyaniline is reacted with sodium nitrite in the presence of stannous chloride to yield 4-benzyloxyhydrazine, which is reacted with α-methyl-3-oxocyclohexanone acetic acid to yield an intermediate α-methyl-tetrahydrocarbazole acetic acid, which is esterified with ethanol to yield the intermediate ethyl ester.
Figure imgf000018_0001
The α-methyl tetrahydrocarbazole acetic acid ester intermediate is reacted under reflux with chloranil in xylene under a nitrogen atmosphere to yield the corresponding α-methylcarbazole acetic acid, which is reduced by hydrogenation to remove the benzyloxy group, yielding the 6-hydroxy-α-methyl carbazole acetic acid ester intermediate.
Figure imgf000018_0002
xylene / N2
Figure imgf000018_0003
Figure imgf000018_0004
Figure imgf000019_0001
The latter intermediate is then hydrolyzed to give the free carboxylic acid intermediate, which is then reacted with an appropriate haloalkyl-A compound, where A is as hereinbefore defined and hal is halo, to yield the desired final product.
Figure imgf000019_0002
Figure imgf000019_0003
The conventional starting materials used in the reaction sequences outlined 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:
Figure imgf000019_0004
N^CH,
Figure imgf000020_0001
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
Figure imgf000020_0002
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. Certain compounds within the scope of the invention exist in the form of
E and Z stereoisomers and the individual isomers can be differentiated by the prefixes E and Z, as assigned by the accepted sequence rules procedures. Accordingly, the present invention embraces the E, Z and mixed isomer forms of those final product compounds exhibiting this form of stereoisomerism. 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 the oxidation of arachidonic acid. Accordingly, the com¬ pounds 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 hyper- sensitivity reactions, such as allergic conjunctivitis. The compounds are especially valuable in the prevention and treatment of allergic bronchial asthma.
The compounds 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 cyto¬ protective effect of the compounds of the invention significantly reduces the gastro- irritant impact of conventional anti-inflammatory 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-inflamma¬ tory drugs (NS AID'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 glycerol-induced 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 carboxymethyl- cellulose, low melting wax, cocoa butter and the like. Diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, tablet-disintegrating agents and the like may be employed. The compounds may be encapsulated with 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 administra¬ tion. 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 and non-human source PLA2. The procedures further measure the ability of the compounds of the invention to inhibit, in vivo, the activity of exogenously administered PLA2. The pharmacological testing additionally demonstrates the ability of the compounds of the invention to inhibit, in vivo, the lipoxygenase and cyclooxy¬ genase pathways of arachidonic acid metabolism; the in vitro, leukotriene antagonist activity of the compounds of the invention; and also measures the in vivo activity of the compounds as anti-inflammatory agents in the rat carrageenan paw edema assay. Finally, the potential of the compounds to induce acute gastroirritation in rats is measured in a test procedure.
The following examples show the preparation and pharmacological testing of compounds within the invention. Example 1 3-[(4-ChlorophenyI)methylene]-[2-methyl-6-(2-quinoIinyl- mgthQχy)1-3H-iιnJene-l -acetic n-i
A. p-Methoxy-α-methyl cynnamic acid A mechanically stirred mixture of 4-methoxybenzaldehyde (47.6 g,
0.35 mole), propionic anhydride (78.3 g, 0.602 mole) and sodium propionate
(33.5 g, 0.35 mole) is placed under nitrogen in an oil bath heated at 150"C for 15 hours. Upon addition of water (130 mL) under vigorous stirring a yellow solid is obtained. It is redissolved in 2N-KOH (pH 11) and the solution is extracted with ether. The aqueous phase is acidified (ice bath) with concentrated HCl. The white solid is collected, washed with water and dried to provide the title compound (47.69 g, 71%, m.p. 153-154βC).
NMR (DMSO-d6, 400 MHz): δ 2.01 (d, J 1.34 Hz, 3H, 2-CH ), 3.77 (s, 3H, OCH3), 6.98 (d, J 8.81 Hz, 2H, ArH), 7.43 (d, J 8.7 Hz, ArH), 7.53 (s, IH, ArCH=C).
MS (El, m/z): 192 (b.p., M)+.
Analysis for: C11H12O3.
Calculated: C, 68.74; H, 6.29.
Found: C, 68.80; H, 6.43.
B. p-Methoxy-α-methyl hydrocynnamic acid
A mixture of the acid (44 g, 0.229 mole) of Step A, and 10% Pd/C (4.4 g) in dry methanol (500 mL) is hydrogenated overnight at 50 psi. The mixture is filtered (Solka-Hoc) and the filtrate evaporated to dryness to yield a pale yellow oil, which is used without further purification (44.5 g). NMR (CDCI3, 400 MHz): δ 1.16 (d, J 6.9 Hz, 3H, 2-CH3), 2.62 (dd, IH, ArCH) and 2.70 (m, IH, ArCH), 3.00 (dd, IH, CH), 3.78 (s, 3H, OCH3), 6.82 (d, 2H, ArH), 7.09 (d, 2H, ArH). MS (El, m/z): 194 (M)+, 121 (b.p.).
C. 6-Methoxy-2-methyl- 1 -indanone A mechanically stiπed mixture of the crude propionic acid (44 g,
0.227 mole) of Step B, is added to warm (50"C) polyphosphoric acid (420 g). The mixture is then placed in an oil bath heated at 95"C for 90 minutes (TLC, 9:1 dichloromethane-methanol, UV). The warm mixture is poured into water (3 L) and stirred overnight at room temperature. It is then extracted with ether and ethyl acetate and the combined extracts are washed with saturated NaHCO3 to neutral. The organic phase is washed with brine and dried (MgSO4). Removal of the solvent provides the title compound as an amber oil. It is purified by flash chromatography (on silica Merck-60, dichlσromethane as eluent) to yield 25.58 g (64%) of a pale yellow oil. NMR (CDC13, 400 MHz): δ 1.31 (d, J 7.4 Hz, 3H, 2-CH3), 2.65 (dd, IH, ArCH) and 2.74 (m, IH, ArCH), 3.32 (dd, IH, CH), 3.83 (s, 3H, OCH3), 7.20 (m,
2H, ArH), 7.34 (d, IH, ArH). MS (El, m/z): 176 (M)+, 161 (b.p., M-CH3)+.
D. 6-Hvdroxy-2-methyl- 1 -indanone To a stirred solution of anhydrous aluminum bromide (69.82 g,
0.261 mole) in anhydrous benzene (250 mL) kept under nitrogen is added drop wise over 30 minutes a solution of the methoxy indanone (18 g, 0.102 mole) of Step C, in benzene (60 mL). The mixture is gently refluxed for 3 hours (TLC, dichloromethane- ethyl acetate 8:2, UV), cooled in an ice bath and treated dropwise with 6N-HC1 (ca. 200 mL) to decompose the aluminum complex. The aqueous phase is extracted with ether (3 times), the extracts are concentrated at reduced pressure and extracted with 2.5N-NaOH (2x75 mL and 1x50 mL). The basic extracts are neutralized (ice bath) with concentrated HCl and the oil that separates is extracted with ether (3 times). The combined extracts are washed with brine, dried (MgSO4) and evaporated to yield an oil that readily solidifies. The crude material is flash chromatographed (on silica Merck- 60, gradient CH2G2, CH2θ2-ethyl acetate 85:15) to provide the pure title compound (13.60 g, nearly colorless solid, 82%).
NMR (CDCI3, 400 MHz): δ 1.31 (d, J 7.44 Hz, 3H, 2-CH3), 2.65 (dd, IH, ArCH) and 2.76 (m, IH, ArCH), 3.32 (dd, IH, CH), 6.68 (s, IH, OH), 7.18 (dd, IH, ArH), 7.30 (m, 2H, ArH).
MS (El, m/z): 162 (M)+, 147 (b.p., M-CH3)+, 133 (M-C2H5)+.
E. 6-(2-Ouinolinylmethoxy)-2-meth yl- 1 -indanone
A mixture of the phenol (15.16 g, 93.58 mmole) of Step D, powdered anhydrous potassium carbonate (12.93 g, 93.6 mmole), 18-crown-6 (2.47 g, 9.36 mmole) and dry acetonitrile (200 mL) is stirred at room temperature under a nitrogen for 15 minutes. 2-Chloromethylquinoline (free base, freshly prepared from 18.29 g or 102.96 mmole of the hydrochloride salt) is added in one portion and the mixture is placed in an oil bath heated at 65QC for 11 hours (TLC, dichloromethane- methanol 9:1, UV). The solvent is removed in vacuo and the residue is partitioned between ethyl acetate and water. The organic phase is washed with brine, dried (MgSO4) and evaporated to yield an oil which solidifies upon trituration with hexane (ca. 31 g). The crude product is purified by flash chromatography (on silica Merck- 60, absorbed in methylene chloride, eluted with CH2θ2-ethyl acetate 90:10 and 85:15) to provide 5.95 g of slightly impure material together with 24.55 g of the pure title compound (pale yellow solid, 84.8%).
NMR (CDC13, 400 MHz): δ 1.29 (d, J 7.36 Hz, 3H, 2-CH3), 2.65 (dd, IH, ArCH) and 2.73 (m, IH, ArCH), 3.32 (dd, IH, CH), 5.41 (s, 2H, ArCH2O), 7.3-
7.38 (m, 3H, ArH), 7.55 (t, J ca. 7 Hz, IH, ArH), 7.64 (d, J 8.5 Hz, IH,
ArH), 7.74 (t, J ca 7 Hz, IH, ArH), 7.83 (d, J 8 Hz, IH, ArH), 8.11 (d, J 8.4 Hz, IH, ArH), 8.20 (d, J 8.4 Hz, IH, ArH).
MS (El, m/z): 303 (M)+, 142 (b.p., CιoH8N)+, 115.
F. 2-Methyl-5-(2-quinolinylmethoxy)-indene-3-acetic acid ethylester
Triethylphosphonoacetate (22.41 g, 100 mmole) is added dropwise under nitrogen to a stirred and cooled (0°C) slurry of NaH (57% in oil, 4.27 g, 100 mmole) in dry toluene (250 mL). The cooling bath is removed and stirring continued at room temperature for 60 minutes whereby an almost homogeneous solution is obtained. A solution of the indanone (15.15 g, 50 mmole) of Step E, in toluene (50 mL) is then added dropwise. The flask is placed in an oil bath heated at 95βC for 19 hours (TLC, traces of starting material present, dichloromethane-ethyl acetate 9:1, UV, Vaughn's). The reddish solution is cooled, diluted with water and extracted with ethyl acetate (3 times). The combined extracts are washed with brine, dried (anhydrous K2CO3) and evaporated to dryness. The residue (heavy brown oil) is flash chromatographed (on silica Merck-60, eluted using a gradient dichloromethane — > dichloromethanerethyl acetate 84:16) to provide the title compound as a mixture of endo and exo isomers (16.3 g, 87.4%, oil that solidifies upon standing) together with unreacted, more polar starting material (1.84 g, 12% recovery). MS (El, m/z): 373 (M)+, 344 (M-C2H5)+, 300 (M-COOC2H5)+, 143 (b.p.), 115.
G. 3-r(4-Chlorophenyl)methylene1-r2-methyl-6-(2-quinolinylmethoxy)]-3H- indene-1 -acetic acid To a vigorously stirred mixture of the ester (mixture of isomers, 6.9 g,
18.5 mmole) prepared as described in Step F, and p-chlorobenzaldehyde (2.86 g, 20.3 mmole, 1.1 equiv.) in dry methanol (45 mL) is added dropwise under nitrogen 25% methanolic sodium methoxide (8 mL, 2 equiv.). The mixture is warmed until it becomes homogeneous (ca. 65*C, bath temp.) and then refluxed for a total of 13 hours. After overnight at room temperature, the slurry (bright yellow precipitate) is treated dropwise with water (35 mL) and a little methanol and refluxing is continued for another 4 hours (reaction followed by TLC). The dark orange solution is cooled, the methanol evaporated, more water added and the gelatinous precipitate is collected, washed with water and dried to provide the sodium salt of the title compound as a bright yellow solid. The latter is slurried in water and neutralized (to pH 6-6.5) with 10% acetic acid. The mixture is extracted with ethyl acetate (large volume), the extract is washed with brine and evaporated to dryness. The residue is azeotroped with benzene to provide an orange solid [7.52 g, mixture of Z (major) and E (minor) isomers]. This material is slurried in ether, stirred for 30 minutes, filtered and dried (4.32 g). The solid is extracted portionwise with hot ethyl acetate (containing some methanol and dichloromethane) until almost completely dissolved. The filtrate is concentrated in vacuo until precipitation occurs, diluted with ether and the yellow solid collected and dried (3.1 g, m.p. 218-220"C). Concentration of the mother liquors provides additional product (1.1 g, m.p. 200-203*C). Combined yield is 4.4 g (51%). The NMR spectrum is consistent [see: Shuman et al., J. Org. Chem.. 42, 1914, (1977)] with the Z-isomer. NMR (DMSO-dβ, 400 MHz): δ 2.11 (s, 3H, CCH3), 3.54 (s, 2H, CH2COO), 5.326
(s, 2H, OCH2Ar), 6.60 (dd, IH ArH), 6.95 (d, J 2.35 Hz, IH), 7.14 (m, 2H,
ArH), 7.52 (m, 4H, ArH), 7.60 (t, J 7.03 Hz, IH, ArH), 7.66 (d, J 8.39 Hz, IH, ArH), 7.77 (t, IH, ArH), 7.99 (m, 2H, ArH), 8.4 (d, J=8.9 Hz, IH,
ArH). UV (λ max, MeOH, nm): 232 (ε 39,594), 285.2 (14,830), 287.8 (14,921), 288
(14,921), 338.2 (10,839). MS (+FAB, m/z): 468 (1C1, M+H)+, 237,131,91 (b.p.). Analysis for: C29H22CINO3
Calculated: C, 74.43; H, 4.74; N, 2.99. Found: C, 74.54; H, 4.67; N, 3.13.
Example 2 2-Methyl-3-[[4-(methylthio)ρhenyl]methvlene]-6-(2- αuinoIinvlmethoxv)-3H-indene-l -acetic acid
To a vigorously stirred mixture of the ester (mixture of isomers, 6.7 g,
17.96 mmole), prepared as described in Example 1, Step F, and (4- methylthio)benzaldehyde (3.05 g, 19.06 mmole, 1.1 equiv.) in dry methanol (75 mL) is added dropwise under nitrogen 25% methanolic sodium methoxide (7.7 mL, 2 equiv.). The mixture is warmed until it becomes homogeneous (ca. 65"C, bath temp.) and then refluxed for a total of 13 hours (TLC, CH2θ2-ethyl acetate 9:1). After overnight at room temperature, the slurry (bright yellow precipitate) is treated dropwise with water (65 mL) and a little methanol and refluxing is continued for another 5.5 hours (reaction followed by TLC). The brown solution is cooled, the methanol evaporated, more water added and the gelatinous precipitate is collected, washed with water and dried to provide the sodium salt of the title compound as a yellow solid. The latter is slurried in water and neutralized (to pH 6-6.5) with 10% acetic acid. The mixture is extracted with ethyl acetate flarge volume), the extract is washed with brine and evaporated to dryness. The residue is azeotroped with benzene to provide a yellow solid (4.47 g, 52%). This material is slurried in ether, stirred for 30 minutes, filtered and dried (4.13 g, m.p. 205-207 βC, dec). The solid is extracted portionwise with hot ethyl acetate (containing some methanol and dichloromethane) until almost completely dissolved. The filtrate is concentrated in vacuo until precipitation occurs and diluted with ether. The yellow solid is collected and dried (3.5 g, 35.4%, m.p. 207-209'C, dec). The NMR spectrum is consistent [see: Shuman et al., J. Org. Chem.. 42, 1914, (1977)] with the Z-isomer.
NMR (DMSO-dβ, 400 MHz): δ 2.11 (s, 3H, CCH3), 2.51 (s, 3H, SCH3), 3.53 (s, 2H, CH2COO), 5.32 (s, 2H, OCH2Ar), 6.60 (dd, IH, ArH), 6.95 (d, IH, J 2.35 Hz), 7.13 (s, IH, ArH), 7.30 (m, 3H, ArH), 7.47 (d, 2H, J 8.4 Hz, ArH), 7.60 (t, J 7.0 Hz, IH, ArH), 7.66 (d, IH, J 8.49 Hz, ArH), 7.77 (t,
IH, J ca. 7 Hz, ArH), 7.99 (m, 2H, ArH), 8.4 (d, IH, J 8.49 Hz, ArH). UV (λ max, MeOH, nm): 231.5 (ε 37,382), 303 (9,739), 307.5 (9,342), 315
(10,525), 352.5 (13,407). MS (+FAB), m/z): 480 (M+H)+. Analysis for: C30H25NO3S
Calculated: C, 75.13; H, 5.25; N, 2.92. Found: C, 75.30; H, 5.25; N, 2.95.
Example 3 2-Methyl-3-[[(4-methylsulfinyI)phenyI]methylene]-6-(2- αuino1vlmethoxv -3H-indene-1 -acetic acid monohvdrate
A slurry of the thioether (1.67 g, 3.48 mmole) of Example 2 in a cold mixture of chloroform and glacial acetic acid (65:35, 40 mL) is treated dropwise (via syringe) with 30% H2O2 (0.403 mL). The mixture is stirred for 30 minutes in the cold and then at room temperature for 24 hours. A clear solution is obtained after 2 hours and a small amount of H2O2 is added after 4 and 7 hours, respectively, to drive the reaction to completion (reaction followed by TLC, aliquot treated with water and extracted with ethyl acetate). The chloroform is evaporated, the residue is diluted with water and neutralized in the cold with the calculated amount of NH4OH. The precipitate is filtered, slurried in water, collected, washed with water and dried in vacuo (over P2O5) to yield the crude acid as a yellow solid. The solid is redissolved in a large volume of warm (30-35*C) ethyl acetate (containing some methanol and dichloromethane) and filtered. The filtrate is concentrated in vacuo until precipitation starts and diluted with ether. The precipitate is collected, washed with ether and dried (fluffy solid, m.p. 136-137"C, sintering and foaming). NMR (DMSO-d6, 400 MHz): δ 2.12 (s, 3H, 2-CH3), 2.80 (s, 3H, SOCH3), 3.55 (s, 2H, CH2COO), 5.32 (s, 2H, ArCH2O), 6.58 (dd, IH, ArH), 6.96 (d, J 2.3 Hz, IH), 7.14 (d, J 8.36 Hz, IH, ArH), 7.21 (s, IH, ArH), 7.58-7.80 (m 7H, ArH), 7.98 (m, 2H, ArH), 8.39 (d, J 8.5 Hz, IH, ArH), 12.36 (s, IH, COOH). MS (+FAB, m/z): 518 (M+Na)+, 496 (M+H)+.
Analysis for: C30H25NO4S • H2O Calculated: C, 70.19; H, 5.30; H, 2.73. Found: C, 69.15; H, 4.93; N, 2.65.
Example 4 5-Fluoro-2-methyl-l-[[4-(2-quinoIinylmethoxy)- nhenvll ethvlene1.1H-indene-3-acetic acid
A. 4-IY2-quinolinylVmethoxy]-benzaldehvde
To a solution of sodium metal (0.9 g, 39.13 g.a.) in absolute ethanol (50 mL) is added dropwise under nitrogen a solution of 4-hydroxybenzaldehyde (5 g, 40.94 mmole) in absolute ethanol (50 mL). The mixture is gently refluxed for 1 hour and then treated dropwise with a solution of 2-chloromethylquinoline (free base, 7.24 g, 40.76 mmole), freshly prepared from the HCl salt) in ethanol (50 mL). The mixture is refluxed for 24 hours, the solvent is evaporated and the residue is partitioned between water and ethyl acetate. The organic layer is washed with 5% NaOH (pH 8), water, brine to neutral and dried (MgSO4). Removal of the solvent yields the title com¬ pound as a yellow solid (9.81 g). The crude material is further purified by flash chromatography (on silica Merck-60, hexane-ethyl acetate 9:1 to remove less polar impurities and 8:2 to elute the product). Yield: 7.37 g (71.5%), m.p. 82-83βC. NMR (CDCI3, 200 MHz): δ 5.5 (s, 2H, ArCH2O), 7.2-8.6 (m, 9H, ArH), 9.80 (s, IH, CHO). B. 5-Huoro-2-methyl-l-[r4-(2-quinolinylmethoxy)phenyl]methylene1-lH-indene- 3-acetic acid
To a mixture of 5-fluoro-2-methyl-indene-3-acetic acid (prepared according to the procedure disclosed in U.S. 3,654,349) (2 g, 9.7 mmole) and the aldehyde (3.2 g, 12.19 mmole) of Step A, in dry methanol (40 mL) is added dropwise under nitrogen 25% methanolic sodium methoxide (6.48 mL). The mixture is warmed until homogeneous and then refluxed for a total of 16 hours. The solvent is evaporated and the residue is slurried in water. The gelatinous precipitate is collected, washed with water and dried. It is then slurried in etiier, filtered and dried to provide the sodium salt of the title compound as a yellow solid (2.03 g, 46.3%). The salt is slurried in water and neutralized (to pH 6-6.5) with 10% acetic acid. The acid is extracted with ethyl acetate (large volume), dried (MgSO4) and evaporated to dryness.
The crude acid is recrystallized from a hot mixture of methanol and ethanol (large volume needed to dissolve, concentrate to smaller volume after filtration). The analytical sample is dried overnight in vacuo at 35"C. The yellow solid (m.p. 218-
221 βC, dec) is a mixture of Z and E isomers in approximately 6:1 ratio (NMR).
NMR (DMSO-d6, 400 MHz): δ 1.84 (s, 2-CH3, minor E isomer), 2.12 (s, 2-CH , major Z isomer), 3.55 (s, 2H, CH2COO), 5.41 (s, ArCH2θ, possibly E isomer), 5.44 (s, ArCH2θ, possibly Z isomer), 6.70 (dt, IH, ArH), 6.85-7.3 (m, IH, ArH), 7.18 (d, 2H, J 8.7 Hz, ArH), 7.28 (s, IH, ArH), 7.32-7.40
(m, IH, ArH), 7.51 (d, 2H, J 8.6 Hz, ArH), 7.62 (t, IH, J ca. 7 Hz, ArH),
7.68-7.76 (m, IH, ArH), 7.79 (t, J, 7 Hz, IH, ArH), 8.01 (nm, 2H, ArH),
8.44 (d, IH, 8.5 Hz, ArH), 12.4 (broad s, IH, COOH).
MS (Cl, m/z): 452 (M+H)+, 408 (M-COOH)+. Analysis for: C29H22FNO3
Calculated: C, 77.15; H. 4.91; N, 3.10. Found: C, 77.08; H, 4.93; N, 3.12.
Example 5 2-MethyI-3-[[4-(methylthio)phenyl]methyIene]-6- IY2-naphthvnmethoxv1-3H-indene-1 -acetic acid
A. 6-[(2-Naphthyl)methoχy]-2-methyl-indanone
A mixture of the indanone (14 g, 86.4 mmole) prepared as described in Example 1, Step D, anhydrous K2CO3 (11.24 g, 81.4 mmole), 18-crown-6 (2.29 g, 6.4 mmole) and 2-bromomethylnaphthalene (11.6 g, 95 mmole) in acetonitrile (275 mL) is heated under nitrogen for 24 hours (in an oil bath set at 70°C). The acetonitrile is evaporated and the residue is dissolved in water, extracted with ethyl acetate, dried (MgSO4) and evaporated to dryness. The crude product (tan solid) is flash chromatographed (on silica Merck-60, preabsorbed in CH2CI2, eluted with 8:2 hexane-ethyl acetate) to provide the title compound as a white solid (13.49 g, 52%). NMR (CDCI3, 400 MHz): δ 1.30 (d, J 3.2 Hz, 3H, 2-CH3), 2.70 (m, 2H, ArCH2C),
3.35 (m, IH, CHCO), 5.25 (s, 2H, ArCH2O), 7.32 (m, 3H, ArH), 7.50 (m,
3H, ArH), 7.85 (m, 4H, ArH). MS (El, m z): 302 (M)+, 141.
B. I) 6-[(2-Naphthyl)methoxy]-2-methyl-indan-l-ylidene-acetic acid ethylester (Z isomer) II) 6-[(2-Naphthyl)methoxy]-2-methyl-indan-l-ylidene-acetic acid ethylester
(E isomer) HI) 5-[(2-Naphthyl)methoxy1-2-methyl-indene-3-acetic acid ethyl ester Tiiethylphosphonoacetate (19.8 g, 88.3 mmole) is added dropwise under nitrogen to a stirred and cooled suspension of NaH (60% in oil, 3.53 g, 88.2 mmole) in toluene (225 mL). The cooling bath is removed. A clear solution is obtained after 10 minutes at room temperature. A solution of the indanone (13.3 g, 44 mmole) of Step A, in toluene (40 mL) is added dropwise and the mixture is placed in an oil bath heated at 100"C for 54 hours (follow by TLC , hexane-ethyl acetate 7:3). Water is added, the layers are separated and the aqueous phase is reextracted with ethyl acetate (3 times). The extracts are dried (K2CO3) and evaporated to dryness to yield the crude product as an amber oil (21.5 g), mixture of endo and exo isomers. Flash chromatography of this crude material (on silica Merck-60, preabsorbed in CH2CI2, hexane-ethyl acetate 9:1) provides small amounts of each of the pure isomers (i.e. I, II and HI) together with mixed fractions and unreacted starting material (ca. 5.24 g). The structures of the isomers are confirmed by MS and NMR and assigned by NOE. The mixture of endo and exo esters is routinely used in the next step (combined yield 8.94 g or 54.7%; 90% based upon recovered starting material). Isomer I
NMR (CDCI3, 400 MHz): δ 1.26 (d, J 6.8 Hz, 3H, 2-CH3), 1.32 (t, J 7 Hz, 3H, ester CH3), 2.54 (d, J 15.1 Hz, IH, ArCHC), 3.1 (m, 2H, ArCHC + CCH), 4.22 (q, J 7.2 Hz, 2H, ester CH2), 5.28 (s, 2H, ArCH2O), 5.85 (s, IH, C=CH-COO), 7.04 (dd, IH, J 8.4 Hz and 2.5 Hz, ArH), 7.17 (d, J 8.4 Hz, 1H, ArH), 7.49 (m, 2H, ArH), 7.59 (d, J 8.4 Hz, IH, ArH), 7.82 (m, 3H, ArH), 7.95 (s, IH, ArH), 8.61 (s, IH, ArH). MS (El, m/z): 372 (M)+, 231, 141 (b.p.).
Isomer II NMR (CDC13, 400 MHz): δ 1.21 (d, 3H, J 7 Hz, 2-CH3), 1.33 (t, 3H, J 7.1 Hz, ester CH3), 2.56 (d, J 16.3 Hz, IH, ArCHC), 3.18 (dd, J 16.3 and 7.1 Hz, IH, ArCHC), 3.88 (m, IH, CCH), 4.22 (q, J ca. 7 Hz, 2H, ester CH2), 5.24 (s, 2H, ArCH2O), 6.19 (s, IH, C=C-COOEt), 7.06 (dd, J 8.4 and 2.4 Hz, IH, ArH), 7.17 (d, J 2.3 Hz, IH, ArH), 7.23 (d, J 8.4 Hz, IH, ArH), 7.49 (m, 2H, ArH), 7.54 (dd, J 8.4 and 1.6 Hz, IH, ArH), 7.88 (m, 4H, ArH).
MS (Cl, m/z): 373 (M+H)+, 261 (b.p.), 233
Isomer III
NMR (CDCI3, 400 MHz): δ 1.20 (t, J 7 Hz, 3H, ester CH3), 2.11 (s, 3H, 2-CH3),
3.28 (s, 2H, ArCH2), 3.48 (s, 2H, CH2COO), 4.09 (q, J 7.1 Hz, 2H ester CH2), 5.25 (s, 2H, ArCH2O), 6.79 (dd, J 8.1 and 2.3 Hz, IH, ArH), 6.98 (s,
IH, ArH), 7.24 (d, 8.8 Hz, IH, ArH), 7.48 (m, 2H, ArH), 7.56 (dd, J 8.4 and 1.7 Hz, IH, ArH), 7.87 (m, 4H, ArH). MS (Cl, m/z): 373 (M+H)+, 285
C. 2-Methyl-3-rr4-(methylthio)Dhenyllmethylene1-6-r(2-naphthvnmethoxy1-3H- indene-1-acetic acid
To a mixture of the ester (mixture of isomers prepared as described above (2.5 g, 6.71 mmole) and 4-(methylthio)-benzaldehyde (1.124 g, 7.38 mmole) in anhydrous methanol (60 mL) is added dropwise under nitrogen 25% methanolic sodium methoxide (2.9 mL). The mixture is refluxed for 16 hours, the solvent removed and the residue slurried in water and stirred overnight (the initial yellow oil turns into a solid). The water is evaporated in vacuo and the residue is triturated with ether (to remove unreacted aldehyde), filtered, washed with ether and dried(yellow solid, 2.65 g). The crude sodium salt is slurried in water and acidified (to pH 6.5) with 10% acetic acid. The acid is collected, washed and dried in vacuo. It is again washed with ether and dried (1.02 g, 32%, m.p. 209-21 l'C, dec). The NMR shows it to be a mixture of Z and E isomers in approximately 8:1 ratio.
NMR (DMSO-d6, 400 MHz): δ 1.81 (s, 2-CH3, minor isomer E), 2.11 (s, 2-CH , major isomer Z), 2.51 (s, 3H, SCH3), 3.54 (s, 2H, CH2COO), 5.23 (s,
ArCH2θ, Z isomer) and 5.28 (s, ArCH2θ, E isomer), 6.60 (dd, IH, ArH), 6.90 (d, J 2.2 Hz, IH, ArH), 7.12 (s, IH, ArH), 7.27-7.36 (m, 3H, ArH), 7.46-7.62 (m, 5H, ArH), 7.88-8.00 (m, 4H, ArH), 12.33 (broad s, IH, COOH).
MS (Cl, m/z): 479 (M+H)+, 435 (M+H-COO)+, 141 (CnH9, naphthyl)+. Analysis for: C31H26O3S
Calculated: C, 77.80; H, 5.48.
Found: C, 77.44; H, 5.47.
Example 6 3-[(4-ChIorophenyI)methyIene]-[2-methyl-6-(2- αuinolinvlmethoxv)1-3H-indene-l-acetic acid
A. 5-Hvdκ.xy-2-methyl indene-3-acetic acid
A mixture of the indanone (1.81 g, 11.17 mmole) of Example 1, Step D, cyanoacetic acid (1.05 g, 12.3 mmol) ammonium acetate (0.17 g), acetic acid (0.66 g) and toluene (5 mL) is heated at reflux with water removal (Dean-Stark) for 24 hours (TLC, CH2θ2-ethyl acetate 8:2). The toluene is evaporated and the residual yellow solid is redissolved in ethanol (6 mL) containing 2.2 N-KOH (1.4 mL). A solution of KOH (2.2 g, 85) in water (15 mL) is added and the solution is refluxed under nitrogen for 18 hours. The ethanol is evaporated, the residue is diluted with water and extracted with ether (2 times). The aqueous layer is acidified in the cold with 6N-HC1 (to pH 3) and extracted with ethyl acetate. The extracts are washed with brine, dried (MgSO4) and evaporated to provide the crude title compound together with traces of unreacted indanone. It is used as such in the next step.
NMR (DMSO-d6, 200 MHz): δ 2.02 (s, 3H, 2-CH3), 3.2 (s, 2H, ArCH2), 3.4 (s, 2H, CH2COO), 6.48 (d, IH, ArH), 6.6 (s, IH, ArH), 7.1 (d, IH. ArH), 9.08 (broad, IH, OH).
B. 5-Hvdroxy-2-methyl-indene-3-acetic acid methylester
To a solution of the crude acid as prepared in Step A, above, in dry methanol (120 mL) is added p-toluenesulfonic acid monohydrate (1.9 g) and the mixture is gendy refluxed for 1.5 hours. The methanol is evaporated, the residue dissolved in ethyl acetate, washed with brine and dried (MgSO4). Removal of the solvent provides a brown oil that readily solidifies upon drying in vacuo. The residue is flash chromatographed (silica Merck-60, absorbed in dichloromethane, eluted with dichloromethane-ethylacetate 98:2 and 94:6) to yield the pure title compound as an off- white solid (9.4 g, 80%). NMR (CDCI3, 400 MHz): δ 2.1 (s, 3H, 2-CH3), 3.26 (s, 2H, ArCH2), 3.49 (s, 2H, CH2COO), 3.67 (s, 3H, COOCH3), 4.89 (broad s, OH), 6.59 (dd, IH, ArH), 6.75 (d, IH, J 2.4 Hz, ArH), 7.18 (d, IH, 7.9 Hz, ArH). MS (El, m/z): 218 (M)+, 158 (b.p.).
C. 3-r(4-Chlorophenyl)methylene1-[2-methyl-6-hvdroχy]-3H-indene-l-acetic acid
To a solution of sodium metal (0.170 g, 7.39 g.a.) in anhydrous methanol (2.5 mL) under nitrogen is added dropwise with cooling a solution of the ester (0.537 g, 2.46 mmole) of step B, in methanol (5 mL) followed by p-chloro- benzaldehyde (0.381 g, 2.71 mmole, 1.1 equiv.). The mixture is stirred in an oil bath heated at 65-70°C for 6 hours followed by standing overnight at room temperature (TLC, CH2Cl2-ethyl acetate 93:7). Water (10 mL) and methanol (2 mL) are added and the mixture is again refluxed under nitrogen for 3 hours (TLC, CH2θ2-ethyl acetate 90:10). The methanol is evaporated, the residue is dissolved in water, and washed with ether (2 times). The aqueous phase is acidified in the cold with 6N-HC1 (to pH 3) and extracted with ethyl acetate. The extracts are washed (brine) and evapo¬ rated to yield a greenish-yellowish solid (0.8 g). The NMR spectrum shows that it is a mixture of the title compound and 5-hydroxy-2-methyl-indene-3-acetic acid. They are best separated as their methylesters (see below).
D. 3-r(4-Chlorophenyl)methylene1-r2-methyl-6-hvdroxyr-3H-indene-l-acetic acid methylester
A solution of the crude acids (2.46 mmole, obtained as described in Step C, above) in methanol (10 mL) containing p-toluenesulfonic acid monohydrate (60 mg) is refluxed for 5 hours. The methanol is evaporated and the residue dissolved in ethyl acetate, washed with brine and dried (MgSO4). The solvent is evaporated and the residue is flash chromatographed (on silica Merck-60, absorbed in 1:1 dichloromethane-hexane, eluted with toluene-isopropanol 95:5) to provide the title compound (less polar spot, 0.103 g, yellow solid) together with some impure material (0.390 g, oil that solidifies upon standing). The Z configuration is confirmed by NOE. The more polar spot is identical (NMR, TLC) with methyl-5-hydroxy-2- methylindene-3-acetate.
NMR (CDCI3, 400 MHz): δ 2.17 (s, 3H, 2-CH3), 3.55 (s, 2H, CH2COO), 3.69 (s, 3H, COOCH3), 6.33 (dd, IH, ArH), 6.67 (d, J 2.3 Hz, IH, ArH), 7.01 (s, IH), 7.14 (d, J 8.3 Hz, IH, ArH), 7.40 (dd, 4H, ArH). MS (El, m/z): 342/340 (1 Cl, M)+, 231 (b.p.) E. 3-r(4-Chlorophenyl)methylenel-[2-methyl-6-(2-quinolinylmethoxy 1-3H- indene-1 -acetic acid methylester
A mixture of the ester (0.410 g, 1.2 mmole) of Step D, anhydrous potassium carbonate (0.17 g), and 18-crown-6 (0.050 g) in acetonitrile (7 mL) is stirred at room temperature for 15 minutes. 2-Chloromethylquinoline is added (free base, 0.149 g, 1.42 mmole, freshly prepared from the hydrochloride salt) and die deep red mixture is stirred under nitrogen for 15 hours in an oil bath heated at 65βC. A 10% excess of potassium carbonate, crown ether and the 2-chloromethylquinoline is then added and the reflux is continued for another 2 hours (TLC, hexane-isopropanol 93:7). The solvent is evaporated and the residue is partitioned between water and ethyl acetate. The organic phase is washed (brine), dried (MgSO4) and evaporated to dry¬ ness. The crude product is identical (NMR, TLC) with the material described in Example 7, below.
F. 3-r(4-Chlorophenyl)methylene1-r2-methyl-6-(2-quinolinylmethoxyΗ-3H- indene-1-acetic acid
1N-KOH (3.72 mL) is added dropwise to a warm solution of the ester (0.600 g, 1.24 mmole) of Step E, in methanol (7 mL). The mixture is refluxed under nitrogen for 3 hours. After stirring overnight at room temperature (TLC, hexane- isopropanol 93:7), the methanol is evaporated and the residue washed with ether. The solid is slurried in water, neutralized in the cold with 10% acetic acid (to pH 6-6.5) and extracted with ethyl acetate (vigorous stirring is necessary to achieve solution). The extracts are washed with brine, dried (MgSO4) and evaporated to yield the crude acid (0.250 g, 43%). It is recrystallized from hot ethyl acetate/methylene chloride (dissolve in a large volume and concentrate the filtrate until precipitation occurs). The pure title compound (m.p. 216-218βC) is identical (TLC, NMR) with the material described in Example 1, Step G. (Z isomer).
Example 7 3-[(4-ChIorophenyl)methyIene]-[2-methyI-6-(2-quinoIinyl- τnethoxv)1-3H-indene-1. acetic acid methvlester
A suspension of the acid (1.88 mmole, crude material before purifica¬ tion, obtained as described in Example 1, Step G), in methanolic HCl (15 mL) is refluxed until the reaction is complete by TLC (dichloromethane-methanol 9:1, UV). The methanol is evaporated and the residue is partitioned between ethyl acetate and dilute NH4OH. The extracts are washed with brine, dried (K2CO3) and evaporated to dryness to provide a yellow oil that foams in vacuo (0.900 g). The crude product is flash chromatographed (on silica Merck-60, toluene-hexane-isopropanol 50:50:1 as eluent) to yield the pure title compound (0.630 g, yellow foam that solidifies upon trituration with ethanol, 69.6% over 2 steps). Mixture of Z and E isomers in approxi¬ mately 4:1 ratio (based upon the position of the 2-CH3 signal in the NMR spectrum). NMR (CDCI3, 400 MHz): δ 1.81 (s, 2-CH3, minor isomer E), 2.17 (s, 2-CH3, major isomer Z), 3.56 (s, 2H, CH2COO), 3.62 (s, 3H, OCH3) 5.42 (s, 2H, ArOCH2), 6.52 (dd, IH, ArH), 6.89 (d, J 2.4 Hz, IH), 7-8.3 (m, 12H, ArH). MS (+FAB, m/z): 482 (1C1, M+H)+, 360, 217, 143 (b.p.)+, 91.
Example 8 l,8-Diethyl-l,3,4,9-tetrahydro-6-(2-quinolinyl- methoxv .Dvranor3.4-b1indole-l-acetic acid
A. 7-Ethyl-2.3-dihydrotryptophol A mixture consisting of 7-ethyltryptophol (28.0 g, 0.148 mol) and
250 mL of trifluoroacetic acid is stirred at room temperature. Sodium borohydride pellets (5.4 g, 0.145 mol) are added over a 4 hour period. After addition is complete, the reaction is stirred for 1 hour. The reaction mixture is poured onto ice and neutral¬ ized with 50% NaOH to pH 10. The aqueous layer is extracted with ether (3 x 200 mL). The ether layers are combined and extracted with 5% HCl solution (3 x 200 mL). The combined acidic solution is then made alkaline with 50% NaOH and extracted with ether (3 x 200 mL). The combined etiier layers are washed with water (2 x 200 mL), once with brine (200 mL), dried (MgSU4), filtered and concen¬ trated to afford 17.3 g of oil. Flash chromatography using 60% ethyl acetate/hexane, and then 80% ethyl acetate/ hexane affords 13.5 g of solid title compound, m.p. 73- 75*C.
NMR (CDCI3): δ 6.98 (d, J=8.3 Hz, IH, Ar), 6.93 (d, J=5.3 Hz, IH, Ar), 6.74 (t, J=7.5 Hz, IH, Ar), 3.7 (m, 3H, OCH OH). 3.68 (m, 2H, NH-CH2), 3.56 (m, IH), 3.33 (m, IH, NH-CH2), 2.49 (q, J-7.6 Hz, 2H, CH2CH3), 2.11 (m, IH, CH2CH2), 1.80 (m, IH, CH2CH2), 1.21 (t, J=7.6 Hz, CH2CH3).
MS: m/e 191 (M+), 130, 118.
B. 7-Ethyl-5-hvdroxytrvptophol
A solution of potassium nitrosodisulfonate (17.0 g, 0.063 mol) in pH 7 buffer (760 mL) is added over a 20 minute period to a stirring solution of 7- ethyl-2,3-dihydrotryptophol (5.0 g, 0.026 mol) in 350 mL of acetone. Ten minutes after the addition is complete, the reaction mixture is extracted with ethyl acetate (4 x 300 mL). The combined ethyl acetate layers are washed with distilled water (2 x 200 mL), once with brine (200 mL), dried (MgSO4) and concentrated to afford 5.6 g of crude product. The crude product is loaded onto a flash chromatography column. The next day, it is flashed using 70% ethyl acetate/hexane to afford 2.5 g of title compound.
NMR (DMSO-dό): δ 8.43 (s, IH, 6.97 (d, J=2.3 Hz, 2H, arom), 6.60 (d, J=2.1 Hz, IH, arom), 6.40 (d, J-2,1 Hz, IH, arom), 4.55 (t, J-5.3 Hz, IH, CH2OH), 3.57 (m, 2H), 3.32 (s, IH, Ar-OH), 2.7 (m, 4H), 1.20 (t, J=7.5 Hz, 3H).
MS: 205 (M)+, 174, 159. Analysis for: C12H15NO2 Calculated: C, 70.22; H, 7.37; N, 6.82. Found: C, 70.15; H, 7.52; N, 6.60.
C. 1.8-Diethyl-1.3.4.9-tetrahvdro-6-hydroxypyranor3.4-b1indole-l-acetic acid methyl ester
A mixture consisting of 7-ethyl-5-hydroxytryptophol (15.5 g, 0.076 mol), 20% tetrahydrofuran/methylene chloπde (1200 mL), methyl 3-methoxy- 2-pentenoate (17.5 g, 0.0121 mol) and boron trifluoride etherate (23.0 mL, 0.187 mol) is stirred at room temperature for 28 hours. The reaction mixture is diluted with 100 mL of methylene chloride and washed with 5% NaHCO3 (3 x 500 mL), once with water, dried (MgSO4) and concentrated to give 30.3 g of crude. Flash chromatography using 25% ethyl acetate/hexane as an eluent afforded 16.0 g of title compound, m.p. 153-154"C. NMR (CDCI3): δ 8.90 (bs, IH), 6.74 ( d, J=23 Hz, H, arom), 6.49 (d, J=2.4 Hz, IH, arom), 4.50 (s, IH, Ar-OH) , 4.00 (m, 2H), 3.7 (s, 3H), 3.0-2.70 (m, 6H), 2.20 (m, 2H), 1.33 (t, J=7.6 Hz, 3H), 0.82 (t, J=7.6 Hz, 3H). MS: 317 (M)+, 288, 244. Analysis for: C18H23NO4 -Calculated: C, 68.12; H, 7.30; N, 4.41. Found: C, 67.73; H, 7.23; N, 4.24.
D. 1.8-Diethyl- 1.3.4.9-tetrahvdro-6-hydroxypyranor3.4-b "lindole- 1 -acetic acid
A mixture consisting of the methyl ester (43.1 g, 0.136 mol) of Step
C, potassium hydroxide (2.4 g, 0.407 mol), 1600 mL of methanol and 160 mL water is refluxed for 5 hours under a nitrogen atmosphere. The excess methanol is evaporated and water (1000 mL) is added to the residue. The mixture is extracted with ether (3 x 500 mL). The aqueous layer is made acidic with 5% HCl (pH 2), then extracted with chloroform (4 x 300 mL). The combined chloroform layers are washed with water (2 x 500 mL), brine (500 mL), dried (MgSO4), and concentrated under vacuo to yield 43.0 g of foam. Flash chromatography using 15% acetone/toluene as eluent, affords 40.0 g of foam. This is crystallized from aceto- nitrile/toluene to afford 35.4 g of pure title compound, m.p. 172-173βC. IH NMR (DMSO-d6): δ 8.44 (s, IH, -NH), 6.51 (d, J=2.1 Hz, IH, Ar), 6.39 (d,
J=1.96 Hz, IH, Ar), 3.89 (m, 2H), 2.89 (q, 2H), 2.87 (d, J=13.5 Hz, IH, CH2COOH), 2.75 (d, J=15.8 Hz, IH, CH2COOH), 2.71-2.57 (m, 2H), 1.99
(q, J=7.2 Hz, 2H), 1.20 (t, J=7.6 Hz, 3H), 0.60 (t, J=8.5 Hz, 3H). IR (KBr, cm-l): 3590 (OH), 1720 (CO). UV (MeOH, nm): 276 (9940), 295 (6400). MS: m/e 303 (M+), 244. Analysis for: C17H21NO4
Calculated: C, 67.31; H, 6.98; N, 4.62. Found: C, 66.99; H, 6.80; N, 4.96.
E. 1.8-Diethyl-1.3.4.9-tetrahvdro-6-(2-quinolinylmethoxy)pyranor3.4-b1indole-l- acetic acid A mixture consisting of l,8-diethyl-l,3,4,9-tetrahydro-6-hydroxy- pyrano[3,4-b]indole acetic acid methyl ester (5.0 g, 0.0158 mol), dimethylsulfoxide (250 mL), and potassium iodide (150 mg) is stirred at room temperature. A solution of sodium hydroxide (3.9 g, in 20 mL H2O) is added and stirred at room temperature followed by addition of 2-chloromethyl quinoline hydrochloride (8.4 g, 0.0392 mol). The reaction mixture is heated at 70-80'C for 3.5 hours, cooled and diluted with water (100 mL). The mixture is made acidic with IN HCl to pH 4, and extracted with ethyl acetate (3 x 400 mL). The combined organic layers are washed with water (3 x 200 mL), once with brine (200 mL), dried over MgS04, filtered and concen¬ trated to give 10.0 g of crude product. The crude product is chromatographed on silica gel using 7% methanol/methylene chloride as eluent to afford 2.9 g pure product. The latter is dissolved in 200 cc ethyl acetate and precipitated by dropwise addition to petroleum ether (700 mL) to afford 2.7 g of the title compound, m.p. 179-18 l'C.
-B. NMR (400 MHz, DMSO-dβ): δ 8.38 (d, J=8.5 IH, 4Q), 8.0 (d, J=8.6 IH, 8-Q),
7.97 (d, J-7.2, 1H-5Q), 7.76 (dt, Jι=6.8, J2=1.5 IH, 7Q), 7.70 (d, J=8.5 IH 3Q), 7.58 (dt, Jι=6.9, J2=l.l IH 6Q), 6.86 (d, J=2.2 IH ar.), 6.67 (d, J-2.2 1H ar.), 5.32 (s, 2H-O£H2Q), 3.6 (m, 2H), 2.79 (m, 2H), 2.74 (D, J=15.2 IH), 2.64 (d, J=14.2, IH), 2.55 (m, 2H, 2.0 (q, J=7.2, 2H, CH2CH3), 1.22 (5, J=7.5 3H CH2CH3), 0.66 (t, J=7.2 3H, 8 CH2CH3).
IR: (KBr): 3400 (NH, OH), 1740 (C=O) cm-1. Mass: (M+Η), 445, 239, 217.
Analysis for: C27H28N2O4
Calculated: C, 72.95; H, 6.35; N, 6.30.
Found: C, 71.04; H, 6.19; N, 6.21.
Example 9 l-EthyI-l,3,4,9-tetrahydro-6-(quinolinylmethoxy)- nvranor3.4-h1in dole- l -acetic aci d
A. l-Ethyl-1.3.4.9-tetrahydro-6-(phenylmethoxy pyranor3.4-b1indole-l-acetic acid methyl ester
A mixture consisting of 5-benzyloxytryptophol (24.0 g, 0.090 mol), methylene chloride (600 mL), methyl 3-methoxy-2-pentenoate (16.8 g, 0.177 mol) and borontrifluoride etherate (2 mL) is stirred at room temperature for 3 hours. The reaction mixture is washed with 5% NaHCO3 (2 x 200 mL), water (2 x 200 mL) and once with brine (200 mL), dried (MgSQφ), filtered and concentrated to give 38.0 g crude product. The crude product is flash chromatographed on silica gel using 20% ethyl acetate/hexane as an eluent to afford 29.0 g pure title compound, m.p. 98- 100'C.
IH NMR, CDCI3): δ 8.87 (BS, IH NH), 7.47 (d, J=7.3, 2H Bz), 7.36 (t, J=7.6, 2H Bz), 7.29 (m, IH), 7.24 (d, J=7.5, IH), 7.04 (d, J=2.4, IH), 6.91 (dd, Jl=8.7, J2=2.4, IH), 5.10 (s, 2H O-CH2-BZ) 4.06-3.90 (m, 2H), 3.71 (s, 3H CH2COOCH3), 2.99 (d, J=16.6, IH), 2.89 (d, J=16.6 IH), 2.75 (m, 2H),
2.0 (m, 2H), 0.81 (t, J=7.4 3H CH2CH3). Mass: (M+) 379, 350, 288. Analysis for: C23H25NO4 Calculated: C, 72.80; H, 6.64; N, 3.69. Found: C, 72.61; H, 6.63; N, 3.69.
B. 1 -Eth yl- 1.3.4.9-tetrah vdro-6-(phen ylmethoxy)p yranoβ .4-bl indole- 1 -acetic acid
A mixture consisting of the ester (29.0 g, 0.0765 mol) of Step A, methanol (800 mL), water (50 mL) and potassium hydroxide (16.0 g, 0.29 mol) is refluxed for 4 hours. The reaction mixture is cooled to room temperature, concentrated and diluted with water (500 mL). The aqueous layer is washed with ether
(2 x 200 mL) and made acidic with a 1:1 mixture of concentrated HCl and water to pH 2.71 is extracted with chloroform (4 x 300 mL) and the combined chloroform layers are washed once with water (2 x 300 mL), once with brine (300 mL), dried
(MgSO4), filtered and concentrated to give 26.0 g of title compound, m.p. 163-165*C.
-H NMR (400 MHz, CDC13): δ 8.39 (BS, NH), 7.47 (d, J=7.4, 2H), 7.38 (dt,
Jl=7.1, J2=1.8, 2H), 7.30 (m, IH), 7.22 (d, J=8.7, IH), 7.04 (d, J=2.4 IH),
6.90 (dd, Jι=8.8, J2=2.4 IH), 5.10 (S-OCH2 Bz) 4.08 (m, 2H), 3.02 (d, J=16.4 IH), 2.95 (d, J=16.3, IH), 2.80 (q, J=5.0 2H), 2.06 (m, 2H), 0.86 (t,
J=7.3 3H CH2CH3).
IR (KBr): 3380 (NH), 1710 (CO) cm-1-
Mass: (M+) 365, ions 336, 306.
Analysis for: C22H23NO4 Calculated: C, 72.31; H, 6.34; N, 3.83.
Found: C, 72.17; H, 6.29; N, 3.65.
C. 1 -Ethyl-1.3.4.9-tetrahydro-6-hydroxy-pyranor3.4-b1indole- 1 -acetic acid
A mixture consisting of 10% Pd/c on charcoal (3.5 g), ethanol
(800 mL), tetrahydrofuran (100 mL) and the acid (21.0 g, 0.056 mol) of Step B, is hydrogenated at 30 psi for 18 hours. The reaction mixture is filtered and concentrated to give 21.0 g of crude product, which is used in the next reaction without further purification.
*H NMR (DMSO-d6): δ 10.36 (s, NH), 7.16 (d, J=7.4, IH Ar), 6.68 (d, J=2.2 IH Ar), 6.53 (dd, Jι=8.5, J2=2.2 IH Ar), 3.89 (m, 2H), 2.82 (d, J=13.6, IH), 2.62 (d, J=13.6, IH), 2.58 (m, 2H), 2.0-1.90 (m, 2H), 0.61 (t, J=7.3 3H
CH2CH3). IR (KBr): Broad peak. 3700-3300 cm-1 (NH, OH), 1710 cπr* (C=O). Mass: M ion: 275, ion 246, 216, 202. Analysis for: C15H17NO4 Calculated: C, 65.44; H, 6.22; N, 5.09. Found: C, 68.82; H, 6.72; N, 4.81.
D. 1 -Ethyl- 1 ,3.4.9-tetrahvdro-6-(quinolinylmethoxy)pyrano[3.4-blindole- 1 -acetic acid
A mixture consisting of the 6-hydroxy pyrano acid (21.0 g, 0.076 mol) of Step C, dimethyl sulfoxide (900 mL), sodium hydroxide (180.0 g in 100 mL H2O) and 2-chloromethylquinoline hydrochloride (23.0 g, 0.0107 mol) is heated to 70-80βC for 1 hour. The reaction mixture is cooled, poured into water (1500 mL) and made acidic with IN HCl to pH 4. It is extracted with ethyl acetate (4 x 500 mL) and the combined organic layers are washed once with water (2 x 300 mL), once with brine (300 mL), dried (MgSO4), filtered and concentrated to give 33.0 crude product, which is chromatographed on silica gel using 7% methanol/methylene chloride as eluent to afford 13.9 g pure product. The pure product is dissolved in 100 mL ethyl acetate, and precipitated by adding dropwise to petroleum ether (3000 mL), and filtered to give 13.8 g product, which is crystallized from benzene to give 12.0 g of pure product, m.p. 105'C (dec).
IH NMR DMSO-d6: δ 10.59 (s, IH, NH), 8.39 (d, J=8.5, IH, 4Q), 8.01 (d, J=8.5, IH, 8Q), 7.97 (d, J=8.2, IH, 5Q), 7.78 (dt, Jι=6.9, J2=1.4, IH 7Q), 7.70 (d, J=8.5, IH, 3Q), 7.60 (dt, Jι=6.9, J =1.0, IH, 6Q), 7.21 (d, J=8.7, IH ar.), 7.06 (d, J=22.3, IH Ar), 6.82 (dd, Jι=8.7, J2=2.5, IH Ar.), 5.34 (s, 2H, O-CH -O). 3.92-3.86 (m, 2H), 2.82 (d, J=13.6, IH), 2.62 (d, J=18.6, IH),
2.60 (m, 2H), 2.01-1.91 (m, 2H), 0.61 (t, J=7.3, CH2CH3). IR (KBr): 3400 cm-l (NH,OH) 1705 cm-l (C=O). Mass: FAB: M+H 417 ion M+ 357. Analysis for: C25H25N2O4 Calculated: C, 72.10; H, 5.81; N, 6.73. Found: C, 72.55; H, 5.91, N, 6.48.
Example 10 l-MethyI-l,3,4,9-tetrahydro-6-(2-quinoIinyl-
Figure imgf000040_0001
A. l-Methyl-1.3.4.9-tetrahvdro-6-(phenylmethoxy)pyranor3.4-b1indole-l-acetic acid, ethyl ester
A mixture consisting of 5-benzyloxytryptophol (4.5 g, 0.017 mol), benzene (200 mL), ethyl acetoacetate (3.4 g, 0.026 mol) and p-toluenesulfonic acid (0.5 g) is refluxed for 2.5 hours using a Dean-stock trap to remove the water. The reaction mixture is cooled to room temperature and washed with 5% sodium bicarbon¬ ate, (1 x 200 mL), water (200 mL) and brine (100 mL), dried (MgSO4), filtered and concentrated to give 8.0 g of crude product as a thick oil, which is flash chro¬ matographed on silica gel using 20% ethyl acetate/hexane as an eluent to afford 5.5 g oil which solidifies upon standing, m.p. 96-98"C. H NMR 400 MHz CDC13: δ 8.96 (bs, IH, NH), 7.47 (d, J=7.0, 2H ar., O-Bz), 7.36 (dt, Jι=7.1, J2=1.8, 2H Ar., O-CH2Bz), 7.3 (m, IH, OCH2-Bz), 7.23 (d, J=8.7 IH Ar.), 7.04 (d, J=2.5, IH Ar.), 6.90 (dd, Jι=8.7, J2=2.3 IH Ar), 5.10 (s, 2H, O-CH2-0), 4.22-4.14 (m, 2H), 4.01 (m, 2H), 2.98 (d, J=16.5, IH), 2.85 (d, J=16.4, IH), 2.79 (t, J= 2H), 1.7 (s, 3H, CH2), 1.3 (t, J= 3H
COOCH2CH3).
Mass: El: M/e: 379, ions 292, 158.
B. l-Methyl-1.3.4.9-tetrahvdro-6-(phenylmethoxy)pyrano[3.4-b1indole-l-acetic acid A mixture consisting of the ester (5.5 g, 0.013 mol) of Step A., methanol (300 mL), water (20 mL) and potassium hydroxide (4.0 g, 0.071 mol) is refluxed for 4 hours. The reaction mixture is cooled to room temperature, concen¬ trated, diluted with water (200 mL) and made acidic witii a 1:1 solution of concentrated HCl:water to pH of 1. The mixture is extracted with chloroform (3 x 150 mL), the combined chloroform extracts are washed once with water (2 x 100 mL), once with brine (100 mL), dried (MgSO4), filtered and concentrated to give 4.6 g of product, which is used in the next reaction, m.p. 157-158"C (dec).
IH NMR (400 MHz, DMSO-dβ): δ 11.98 (bs, IH, COOH), 10.62 (s, IH, NH),
7.45 (d, J=7.1, 2H-Bz), 7.38 (dt, Jι=7.1, J2=1.8, 2H, Bz), 7.30 (m, IH, Bz), 7.18 (d, J=8.7, IH Ar.), 6.99 (d, J=2.3, IH Ar.), 6.76 (dd, Jι=8.7,
J2=2.5, IH Ar.), 5.07 (s, 2H, OCH20), 3.95-3.84 (m, 2H), 2.78 (d, J=13.8,
IH), 2.68 (d, J=13.8, IH), 2.60 (t, J=5.4, 2H), 1.56 (s, 3H).
Mass: El: M/e: 351, ions 292, 260, 130.
C. 1 -Methyl-1.3.4.9-tetrahvdro-6-hvdroxy-pyranor3.4-b1indole- 1 -acetic acid A mixture consisting of the acid (4.5 g, 0.013 mol) of Step B, ethanol
(150 mL), tetrahydrofuran (20 mL) and 10% Pd/c on charcoal (800 mg) is hydro¬ genated at 30 psi for 18 hours. The mixture is filtered and concentrated to give 3.1 g of product, which is used into next reaction without further purification. IH NMR (400 MHz, DMSO-dό): δ 7.19 (d, J=8.5, IH Ar.), 6.70 (d, J=2.5, IH Ar.), 6.55 (dd, Jι=8.5, J2=2.5, IH Ar.), 3.90 (m, 2H), 2.77 (d, Jι=16.0,
IH), 2.66 (d, J=16.0, IH), 2.55 (m, 2H), 1.55 (s, 3H). Mass: El: M/e: 261, ions: 216, 160, 101. D. l-MethvI-1.3.4.9-tetrahvdro-6-(2-quinolinylmethoxy)pyranor3.4-b]indole-l- acetic acid
A mixture consisting of 1 -methyl- l,3,4,9-tetrahydro-6-hydroxy- pyrano[3,4-b]indole-l-acetic acid (3.0 g, 0.011 mol) of Step C, dimethyl sulfoxide (300 mL), potassium iodide (100 mg) and sodium hydroxide (2.5 g in 20 mL water) is stirred at room temperature. 2-Chloromethylquinoline hydrochloride (5.8 g,
0.022 mol) is added and the reaction mixture is heated for 3.5 hours, cooled and diluted with water (700 mL). The mixture is made acidic to pH 4 with IN HCl, extracted with ethyl acetate (4 x 300 mL) and the combined organic layers are washed once with water (2 x 200 mL), once with brine (200 mL), dried (MgSO4), filtered and concentrated to yield crude product, which is flash chromatographed on silica gel using 10% methanol/methylene chloride as an eluent to give 1.0 g product. The latter is dissolved in ethyl acetate (20 mL) and precipitated by adding dropwise to petroleum ether (500 mL) to give 1.0 g pure compound, m.p. 175-177βC (dec). IH NMR (400 MHz DMSO-dβ): δ 11.2 (bs, IH, NH), 8.37 (d, J=8.5, IH 4Q), 8.0
(d, J=8.7 IH, 8Q), 7.97 (d, J=7.6, IH, 5Q), 7.77 (dt, Jι=6.9, J2=1.4, IH,
7Q), 7.69 (d, J=8.5, IH, 3Q), 7.58 (dt, Jι=7.0, J2=1.0, IH, 6Q), 7.18 (d,
J=8.7, IH Ar.), 7.03 (d, J=2.3 IH Ar.), 6.80 (dd, Jι=8.7, J2=2.3 IH Ar),
5.34 (s, 2H, OCH2Q), 3.88-3.84 (m, 2H), 2.62 (bs, 2H), 2.48 (bs, 2H), 1.56 (s, 3H-CJ^).
IR (KBr): 3400 cm -l (broad NH, OH, ), 1700 cm-l (C=θ). Mass: El: M/e: M+ 402, ions 202, 157. Analysis for: C24H22N2O4 Calculated: C, 71.63; H, 5.57; N, 6.96. Found: C, 69.20; H, 5.38; N, 6.60.
Example 11 l-Ethyl-2,3,4,9-tetrahydro-6-(2-quinoIinyl- metlrøxyHH-carfrazQle-l-acetic acid
A. l-Ethyl-2.3.4.9-tetrahydro-6-methoxy-lH-carbazole-l-acetic acid methyl ester A mixture consisting of 4-methoxyphenylhydrazine (25.0 g,
0.162 mol), 2-carbomethoxymethyl-2-ethylcyclohexanone (42.0 g, 0.212 mol), and toluene (1000 mL) is refluxed for 24 hours under a nitrogen atmosphere and the water is removed using a Dean-Stark trap. The reaction mixture is cooled and concentrated to give crude hydrazone. (= 56 g). The crude hydrazone (56 g) is dissolved in glacial acetic acid (500 mL) and refluxed for 25 minutes. The reaction mixture is cooled to room temperature, poured into water (2000 mL) and extracted with ethyl acetate (4 x 500 mL). The organic layers are combined, washed with IN NaOH (2 x 500 mL), water (2 x 500 mL), once with brine (500 mL), dried (MgSO4), filtered and concentrated to give 47.0 g of crude product. The crude product is flash chromatographed using 10% ethyl acetate/hexane as an eluent to afford 18.0 g product; m.p.72-74*^. IH NMR (400 MHz, CDCI3): δ 8.94 (bs, IH, NH), 7.19 (d, J=8.0, IH Ar.), 6.90 (d, J=2.3 IH Ar.), 6.76 (dd, Jι=8.6, J2=2.3, IH Ar.), 3.81 (s, 3H, COOCH3), 3.64 (s, 3H, OCJH3), 2.68-2.62 (m, 4H), 1.95-1.61 (m, 6H), 0.8
(t, Jι=7.6, 3H, CH2CH3). Mass: El: M/e: 301, ions 272, 228 (base peak).
B. l-Ethyl-2.3.4.9-tetrahvdro-6-hvdroxy-lH-carbazole-l-acetic acid
A mixture consisting of the methoxy compound (18.0 g, 0.060 mol) of Step A, and 48% hydrobromic acid (450 mL) is heated at 125*C for 6 hours. The reaction mixture is cooled, diluted with water (1500 mL) and extracted with ethyl acetate (3 x 500 mL). The organic layers are combined and washed once with water (2 x 500 mL), once with brine (500 mL), dried <MgSO4), filtered and concentrated to give 15.8 g product as a foam. H NMR (400 MHz, DMSO-dβ): δ 10.21 (s, IH, COOH), 8.45 (bs, IH, NH), 7.03 (d, J=8.5, IH Ar.), 6.63 (d, J=1.9, IH Ar.), 6.48 (dd, Jι=7.3, J2=1.3, IH Ar.), 2.54-2.46 (m, 4H), 1.98-1.66 (m, 6H), 0.70 (t, J=7.3, CH2CH3). Mass: El: M/e: 273 (M+) ions 244, 214, 198.
C. 1 -Ethyl-2.3,4.9-tetrahvdro-6-(2-quinolinylmethoxy 1 H-carbazole- 1 -acetic acid A mixture consisting of the hydroxy acid (8.0 g, 0.028 mol) of Step
B, dimethyl sulfoxide (500 mL), potassium iodide (100 mg), and sodium hydroxide (7.0 g in 300 mL H2O 0.175 mol) is stirred at room temperature, for 15 minutes. 2-Chloromethylquinoline hydrochloride (11.8 g, 0.055 mol) is added and the reaction mixture is heated to 70-80βC for 3 hours. The mixture is cooled to room temperature and poured into water (1500 mL), made acidic to pH 4 with IN HCl, and extracted with ethyl acetate (3 x 500 mL). The organic layers are combined, washed once with water (2 x 300 mL), once with brine (300 mL). The organic layers are combined, washed once with water (2 x 300 mL), once with brine (300 mL), dried (MgSO4), filtered and concentrated to give 9.0 g of crude product which is flash chro- matographed using 10% methanol/methylene chloride as an eluent to give 3.0 g pure product, which is dissolved in ethyl acetate (20 mL) and added dropwise to petroleum ether (500 mL). The precipitate is filtered off to give 2.5 g product, m.p. 120-125°C. IH NMR (400 MHz, DMSO-dό): δ 10.80 (bs, IH, NH), 8.37 (d, J=8.5, IH, 4Q),
8.0 (d, J=8.5, IH, 8Q), 7.98 (d, J=7.2, IH 5Q), 7.77 (dt, Jι=6.9, J =1.5, IH, 7Q), 7.69 (d, J=8.5, IH, 3Q), 7.61 (dt, Jι=6.9, J2=l.l, IH, 6Q), 7.14
(d, J=8.7, IH Ar.), 6.96 (d, J=2.4, IH Ar.), 6.76 (dd, Jι=8.6, J2=2.4, IH
Ar.), 5.32 (s, 2H -CH2Q), 2.52 (bs, 2H), 1.82-1.72 (m, 8H), 0.71 (t, J=7.3,
3H, CH2CH3). IR (KBr): 3410 cm"1 (NH, OH, broad), 1710 cm"1 (C=O). Mass: M+ 415
Analysis for: C26H26N2O3
Calculated: C, 75.34; H, 6.32; N, 6.76.
Found: C, 73.49; H, 6.27; N, 6.48.
Example 12 l,3,4,9-Tetrahydro-l-propyl-6-(2-quinolinyl- methoxv)pvranor3.4-b1indole-l-acetic acid
A. Ethyl-l-propyl-l.S^.g-tetrahvdro-ό-fphenylmethoxy^pyranorS^-blindole-l- acetate
A solution of 5-benzyloxytryptophol (4.72 g, 17 mmol) and ethyl butyrylacetate (3.16 g, 20 mmol) in methylene chloride (75 ml) is treated dropwise with boron trifluoride etherate (3 mL) and stirred under nitrogen at room temperature overnight. The reaction mixture is diluted with methylene chloride (250 mL) and sequentially washed with 5% aqueous sodium bicarbonate (2 x 100 mL) and water
(2 x 50 mL). The organic phase is dried (MgSO4), filtered, and evaporated to afford 7.0 g of a crude dark oil. Purification by flash column chromatography (ethyl acetate:hexane, 1:9 eluent) on silica gel (50:1 ratio, 230-400 mesh) affords the title compound (5.5 g, 79%) as a slightly amber oil homogeneous by TLC. lR NMR (200 MHz, CDCI3): δ 8.93 (bs, IH, NH), 7.25-7.80 (m, 5H, C6H5), 7.22
(d, J=9.3 Hz, IH, 8- ArH), 7.04 (d, J=2.8 Hz, IH, 5-ArH), 6.91 (dd, J=9.3, 2.8 Hz, IH, 7- ArH), 5.12 (s, 2H, OCH Ar . 3.83-4.40 (m, 4H, CH2 ester,
3-H's), 3.03, 2.86 (d, J=17.2 Hz, 2H, CH2CO2C2H5), 2.60-2.87 (m, 2H,
4-H's), 1.82-2.20 (m, 2H, l-CH2(α)), 1.25 (t, J=7.0 Hz, 3H, CH3 ester),
1.00-1.50 (m, 2H, l-CH2(β)), 0.86 (t, J=7.0 Hz, 3H, I-CH3). B. Ethyl- 1 -propyl- 1.3.4.9-tetrahydro-6-hydroxypyranor3.4-b]indole- 1 -acetate
A suspension of the 6-benzyloxypyranoindole (5.4 g, 13.25 mmol) of
Step A, and 10% Pd/C catalyst (600 mg) in ethanol (150 mL) is hydrogenated for 6 hours in a Parr shaker. Filtration of the catalyst and evaporation of the solvent affords the title compound (3.75 g, 89%) as a cream-colored foam homogeneous by TLC.
-H NMR (200 MHz, CDC13): δ 8.90 (bs, IH, NH), 7.20 (d, J=9.3 Hz, IH,
8-ArH), 6.92 (d, J=2.8 Hz, IH, 5-ArH), 6.75 (dd, J=9.3, 2.8 Hz, IH,
7-ArH), 4.90 (bs, IH, OH), 3.85-4.40 (m, 4H, CH2 ester, 3-H's), 3.02, 2.88
(d, J=17.2 Hz, 2H, £H2CO2C2H5), 2.57-2.88 (m, 2H, 4-H's), 1.82-2.20 (m, 2H, l-CH2(α)), 1.25 (t, J=7.0 Hz, 3H, CH3 ester), 1.00-1.60 (m, 2H, l-CH2(β)), 0.87 (t, J=7.0 Hz, 3H, I-CH3).
C. 1 ,3.4.9-Tetrahvdro- 1 -propyl-6-(2-quinolinylmethoxy)pyranor3.4-blindole- 1 - acetic acid
A solution of the 6-hydroxypyranoindole (3.5 g, 11 mmol) of Step B, in dimethylsulfoxide (80 mL) is treated with 2.5N NaOH (20 mL, 50 mmol) and stirred at 40eC under nitrogen for 5 minutes. 2-Chloromethylquinoline hydrochloride (4.71 g, 22 mmol) is added as a solid in one aliquot and the reaction mixture heated to 80*C for 30 minutes. After cooling, the pH of the reaction is adjusted to 12.0 with 2.5N NaOH. After extraction with ether (3 x 250 mL), the aqueous layer is carefully acidified to pH 3.0 with 2N HCl and 0.1N HCl (near end point) followed by rapid extraction with ether (4 x 300 mL). The combined ether extracts are washed witii water (2 x 200 mL), dried (MgSO4), and filtered. Evaporation of the solvent affords a crude yellow foam (4.16 g, 88%). Purification by flash column chromatography (methanohmethylene chloride, 5:95 eluent) on silica gel (100:1 ratio, 230-400 mesh) affords an amber foam (2.08 g, 61%) homogeneous by TLC. A concentrated methylene chloride solution of the foam is added dropwise to cooled, rapidly stirred, petroleum ether (200 mL). Filtration of the resulting precipitate yields the title com¬ pound as a cream colored amorphous solid (3.2 g, 61%) homogeneous by TLC. After high vacuum drying (36 hours) the compound still retains solvent of crystallization as indicated by NMR, m.p. 100-110°C (dec). IR (KBr): 3400 (NH, OH), 1710 (C=O) cm-1. -H NMR (400 MHz, CDC13): δ 8.93 (s, IH, NH), 8.26 (d, J=8.5 Hz, IH, 4-Q), 8.19 (d, J=8.6 Hz, IH, 8-Q), 7.85 (d, J=8.2 Hz, IH, 5-Q), 7.81 (d, J=8.5 Hz, IH, 3-Q), 7.76 (dt, J=7.0, 1.2 Hz, IH, 7-Q), 7.58 (t, J=7.0 Hz, IH, 6-Q), 7.07 (d, J=2.4 Hz, IH, 5-ArH), 6.97 (d, J=8.8 Hz, IH, 8-ArH), 6.84 (d, J=8.8, 2.4 Hz, IH, 7-ArH), 5.49 (s, 2H, OCH^O). 4.03 (ddd,
J=11.4, 4.7, 4.3 Hz, IH, 3-Heq.), 3.92 (ddd, J=11.4, 7.2, 4.8 Hz, IH, 3— Hax), 3.05 (d, J=16.8 Hz, IH, 4-Hax), 2.66 (ddd, J=15.4, 4.7, 4.7 Hz, IH, 4-Heq.), 1.91-2.14 (m, 2H, l-CH (α)), 1.08-1.48 (m, 2H, l-CH2(β)), 0.88 (t, J=7.0 Hz, 3H, I-CH3). MS (El); m/e 430 (1.4, M+), 160 (50, QCH2OH2 Θ), 143 (100, QCH3®).
Analysis for; C26H26N2O4
Calculated: C, 72.54; H, 6.09; N, 6.51.
Found: C, 72.64; H, 7.52; N, 5.41.
Example 13 l-Ethyl-l,3,4,9-tetrahydro-6-(2-quinoIinylmethoxy)- pγranor3ι4-fr1ind9lc-l-acetic acid, methyl ester
Trimethylsilyldiazomethane (40 mL, 10% by weight methylene chloride) is added dropwise to a mixture consisting of 1 -ethyl- l,3,4,9-tetrahydro-(2- quinolinylmethoxy)pyrano[3,4-b]indole-l-acetic acid (1.0 g, 0.002 mol) as prepared in Example 9, methylene chloride (150 mL) and methanol (15 mL). The reaction mixture is stirred at room temperature for 1/2 hour, and is concentrated to give 1.2 g of oily residue, which is very difficult to crystallize. The ester is converted to the hydrochloride salt by adding ether/HCl. The hydrochloride salt is recrystallized from methanol/ether to afford 0.75 g pure compound, m.p. 185-187*C. -R NMR (400 MHz, DMSO-d6): δ 8.71 (d, J=8.2 Hz, IH, 4Q), 8.21 (d,
J=8.2 Hz, IH, 8Q), 8.13 (d, J=8.2 Hz, IH, 5Q), 7.93-7.89 (m, 2H, 3Q,
7Q), 7.76 (t, J=7.8 Hz, 6Q), 7.23 (d, J=8.6 Hz, IH Ar), 7.10 (d, J=2.6 Hz,
IH Ar), 6.86 (dd, Jι=8.6, J2=2.3, IH Ar.), 5.5 (s, 2H, OCH -q). 3.91 (m,
2H), 3.51 (s, 311, OC ~ 2.91 (d, J=13.7 Hz, IH), 2.74 (d, J=13.5 Hz, IH), 2.56 (m, 2h„ 1.93 (q, J=7.4 Hz, 2H), 0.60 (t, J=7.2 Hz, 3H,
CH2£H3). IR (KBr) 3400 cm-1, 3200 cπr1 (NH, OH), 1740 cm-1 (C=O). Mass: El (m/e) M+ 430, ions 401, 357. Analvsis for: C26H26N2O4-HCI Calculated: C, 66.88; H, 5.83; N, 6.00. Found: C, 66.58; H, 5.75; N, 5.96.
Example 14 l,3,4,9-Tetrahydro-l,l-dimethyl-6-(2-quinoIiπyI- methoxv)pvran or3.4- h1i n d ol e
A. 1 ,4.9-Tetrahydro- 1.1 -dimethyl-6-(phenylmethoxy)pyranor3.4-b]indole
A mixture consisting of 5-benzyloxytryptophol (3.2 g, 0.012 mol), acetone (7 mL), benzene (300 mL) and p.-toluene sulfonic acid (500 mg) is refluxed for 3.5 hours using a Dean-Stark trap to remove the water. The reaction mixture is cooled to room temperature and washed with 5% sodium bicarbonate (1 x 200 mL), water (200 mL) and brine (200 mL). It is dried (MgSO4), filtered and concentrated to give 5.0 g product as thick oil. The crude product is flash chromatographed with 15% ethyl acetate/hexane as an eluent to afford 2.5 g product, m.p. 128-129°C.
B. 1 ,3.4,9-Tetrahydro- 1.1 -dimethyl-6-hvdroxy-pyranor3.4-blindole
A mixture consisting of 10% Pd/c on charcoal (0.7 g), ethanol (150 mL) and l,3,4,9-tetrahydro-l,l-dimethyl-6-(phenylmethoxy)pyrano[3,4- bjindole (4.2 g, 0.014 mol) of Step A, is hydrogenated at 30 psi for 18 hours. The reaction mixture is filtered and concentrated to give 3.5 g product which is used in the next reaction without further purification.
C. 13.4.9-Tetrahydro-l.l-dimethyl-6-(2-quinolinylmethoxy)pyrano["3.4-b1indole A mixture consisting of the 6-hydroxy compound (3.5 g, 0.016 mol) of Step B, dimethylformamide (100 mL), potassium carbonate, anhydrous (6.0 g, 0.043 mol), potassium iodide (100 mg) and 2-chloromethylquinoline (2.9 g, 0.016 mol) is heated at 60°C for 24 hours. The reaction mixture is cooled to room temperature, diluted with water (600 mL), and extracted with ethyl acetate (3 x 150 mL). The combined organic layers are washed with 2.5N NaOH, water (2 x 100 mL), once with brine (100 mL), dried (MgSO4), filtered and concentrated to give 3.8 g of crude product. The crude product is flash chromatographed on silica gel using 40% ethyl acetate/hexane as an eluent to afford 2.3 g of product which is recrystallized from methanol to afford 1.69 g pure title compound, m.p. 164-165βC. JH NMR (400 MHz, DMSO-d6): δ 10.68 (s, IH, NH), 8.38 (d, J=8.6 Hz, IH, 4Q), 8.0 (d, J=8.4 Hz, IH, 8Q), 7.97 (d, J=8.1 Hz, IH, 5Q), 7.77 (t, J=7.1 Hz, IH, 7Q), 7.69 (d, J=8.6 Hz, IH, 3Q), 7.59 (t, J=7.1 Hz, IH, 6Q), 7.18 (d, J*=8.7 Hz, IH Ar), 7.04 (d, J=2.4 Hz, IH Ar), 6.82 (dd, Jl=8.6, J2=2.4 Hz, IH Ar), 5.34 (s, 2H, OCH2Q), 3.85 (t, J=5.3 Hz, 2H), 2.56 (t, J=5.3 Hz, 2H), 1.44 (s, 6H, (CH-^-A IR (KBr): 3410 cm- (broad NH). Mass: El: m/e: 358 (M+), 343 143. Analysis for: C23H22N2O2 Calculated: C, 77.07; H, 6.19; N, 7.81. Found: C, 76.76; H, 6.25; N, 7.70.
Example 15 l,3,4,9-Tetrahydro-l,l-diethyI-6-(2-quinoIinyl- meth o v) p vra n o r3.4 -h1 in d ol e
A. 1.3.4.9-Tetrahydro-l.l-diethyl-6-(phenylmethoxy')pyranor3.4-blindole
A mixture consisting of 5-benzyloxytryptophol (7.0 g, 0.0262 mol), diethyl ketone (10 mL), benzene (400 mL) and p-toluene sulfonic acid is refluxed for 3.5 hours using a Dean-Stark tap to remove the water. The reaction mixture is cooled to room temperature, washed with 5% sodium bicarbonate, water (200 mL), brine (200 mL), dried (MgSO4), filtered and concentrated to give 9.9 g of crude product. The crude product is flash chromatographed on silica gel using 15% ethyl acetate/hexane as an eluent to afford 6.5 g of pure title compound.
B. 1.3.4.9-Tetrahydro- 1.1 -diethyl-6-(hvdroxy')pyranor3 ,4-blindole
A mixture consisting of 10% Pd/c on charcoal (1.0 g), ethanol
(125 mL), and l,3,4,9-tetrahydro-l,l-diethyl-6-(phenylmethoxy)pyrano[3,4-b]indole
(6.5 g, 0.0193 mol), of Step A, is hydrogenated at 30 psi for 18 hours. The reaction mixture is filtered and concentrated to give 5.2 g of product which is used in the next reaction without further purification.
C. 1.3.4.9-Tetrahydro- 1.1 -diethyl-6-(2-quinolinylmethoxy)pyrano[3 ,4-blindole
A mixture consisting of the 6-hydroxy compound (5.2 g, 0.0212 mol) of Step B, dimethyl sulfoxide (150 mL), sodium hydroxide (6.0 g, 0.15 mol in 20 mL water), and 2-chloromethylquinoline hydrochloride (6.0 g, 0.028 mol) is heated to 70-80'C for 6 hours. The reaction mixture is cooled to room temperature, diluted with water (800 mL), and extracted with ethyl acetate (3 x 150 mL). The combined organic layers are washed once with water (2 x 150 mL), once with brine (100 mL), dried (MgSO4), filtered and concentrated to give 9.9 crude product. The crude product is flash chromatographed on silica gel using 25% ethyl acetate/hexane as an eluent to afford 4.5 g pure product as a thick oil. The oil is dissolved in ether
(150 mL) and ether/HCl is added dropwise. The precipitate is filtered off and the product crystallized from methanol/ether to give 2.6 g product as a yellow crystalline solid, m.p. 164-166'C.
-H NMR (400 MHz, DMSO-dό): δ 10.64 (s, IH, NH), 8.70 (d, J=8.6 Hz, IH, 4Q), 8.21 (d, J=8.5 Hz, IH, 8Q), 8.13 (d, J=8.1 Hz, IH, 5Q), 7.95-7.88 (m, 2H, 3 and 7 Q), 7.74 (t, J=7.3 Hz, IH, 6Q), 7.21 (d, J=8.6 Hz, IH Ar), 7.08 (d, J=2.4 Hz, IH Ar), 6.84 (dd, Jι=8.7, J2=2.4 Hz, IH Ar.), 5.50 (s,
2H, OCH2Q), 3.85 (t, J=5.3 Hz, 2H), 2.57 (t, J=5.4 Hz, 2H), 1.80 (m, 4H, CH2CH3 1,1-diethyl), 0.71 (t, J=7.3 Hz, 6H, CH~CH~. 1,1-diethyl).
IR: (KBr) 3410 cm"1 (NH).
Mass: El: M/e: 386 (M+) ions 357, 216, 142, 128. Analysis for: C25H26N2O2ΗCI
Calculated: C, 70.99; H, 6.43; N, 6.62
Found: C, 71.20; H, 6.43; N, 6.66.
Example 16 l-[(4-Chlorophenyl)methyI]-2-methyl-5-(2-quinoIinylmethoxy)-lH- indole-3-acetic acid hydrochloride quarter hvdrate
A. 2-Methyl-5-(2-quinolinylmethoxyVlH-indole-3-acetic acid, one tenth hydrate
To a previously degassed mixture of dimethylformamide (70 mL) and sodium methoxide/methanol solution (25 wt %, 2 equivalents, 8.8 mL) is added 5-hydroxy-2-methyl-lH-indole-3-acetic acid (4.0 g, 19.5 mmol) at room temperature. After 15 minutes 2-chloromethylquinoline (3.46 g, 19.5 mmol) is added and the mixture stirred overnight. The dimethylformamide is evaporated and the mixture parti¬ tioned between pH 4 buffer solution and ethyl acetate. The organic phase is dried (MgSO4), filtered and partially concentrated. A solid is filtered off and crystallized using ethanol - water. This gives the title compound as a solid (2.96 g), 44% yield, m.p. 208-210βC.
Analysis for: C21H18N2O3 • 0.1 H2O Calculated: C, 72.44; H, 5.26; N, 8.04. Found: C, 72.29; H, 5.38; N, 7.93. B. 2-Methyl-5-(2-quinohnylmethoxy)-lH-indole-3-acetic acid methyl ester.
To a stirring mixture of the indole- acetic acid substrate (1.2 g, 3.4 mmol) in tetrahydrofuran (25 mL) is added slowly, a freshly prepared diazomethane/diethyl ether solution at ambient temperature. Upon consumption of starting material, a few drops of acetic acid are added. The solvent is evaporated and methylene chloride is added. This organic phase is washed with water and dried (MgSO4). Solvent removal gives the product as an oil (1.2 g). Yield is quantitative.
C. l-r(4-Chlorophenyl)methyl]-2-methyl-5-(2-quinolinylmethoxy)-lH-indole-3- acetic acid methyl ester To a stirring mixture of the indole-ester substrate (0.8 g, 2.2 mmol) in dimethylformamide (15 mL) is added sodium hydride (0.058 g, 2.4 mmol). The reaction is stirred for 30 minutes at ambient temperature. 4-Chloro-benzylchloride (0.35 g, 2.2 mmol) is then added. After 2 hours the dimethylformamide is evaporated and the residue partitioned using pH 4 buffer solution and ethyl acetate. The organic phase is separated, dried (MgSO4) and filtered. Solvent removal produces a solid that is flash chromatographed using chloroform as eluent, then crystallized with acetonitrile to yield the product as a crystalline solid (0.162 g 16% yield) m.p. 132-134*C. Analysis for: C29H25N2O3CI Calculated: C, 71.82; H, 5.20; N, 5.78. Found: C, 71.48; H, 5.17; N, 5.88.
*
D. l-[(4-Chlorophenyl methvn-2-methyl-5-(2-quinolinylmethoxy -lH-indole-3- acetic acid hydrochloride quarter hydrate
The above ester (2.2 g, 4.5 mmol) is combined with tetrahydrofuran (50 mL), IN NaOH solution (50 mL) and heated to reflux (3.5 hours). The tetra- hydrofuran is removed in vacuo and the aqueous phase acidified with IN HCl. A crude solid is filtered and recrystallized using ethanol to produce 1.16 g of product as the hydrochloride salt (50% yield), m.p.249-251βC. Analysis for: C28H23CIN2O3 • HCL • 1/4 H2O Calculated: C, 65.69; H, 4.82; N, 5.47. Found: C, 65.64; H, 4.92; N, 5.41. Example 17 l-(4-Chlorobenzoyl)-2-methyl-5-(2-quinoIinyImethoxy)- IH-indole-3-acetic acid three quarters hvdrate
A. l-(4-Chlorobenzoyl)-2-methyl-5-(2-quinolinylmethoxy)-lH-indole-3-acetic acid methyl ester one tenth hvdrate
To a solution of 2-methyl-5-(2-quinolinylmethoxy)-lH-indole-3-acetic acid methylester (0.95 g, 2.6 mmol) (see Example 16B) in dimethylformamide (10 mL) at 0βC is added sodium hydride (0.12 g, 50% dispersion) and the mixture stirred 20 minutes. 4-Chlorobenzoyl chloride (0.45 g, 2.6 mmol) is then added and the mixture stirred overnight; 100 mL of 5% acetic acid is used to quench the reaction. After extraction with a 1:1 mixture of etheπethyl acetate, the organic phase is washed with water, saturated NaHCO3 solution, water and brine. It is then separated and dried (MgSO4). Solvent removal gives a crude solid which is flash chromatographed using methylene chloride-methanol (99-1) as eluent The product is isolated and crystallized from acetonitrile (0.20 g, m.p. 117-118"C), yield is 15%. Analysis for: C29H23CIN2O4 • 0.1 H2O Calculated: C, 69.55; H, 4.66; N, 5.59. Found: C, 69.35; H, 4.48; N, 5.56.
B. l-(4-Chlorobenzoyl)-2-methyl-5-(2-quinolinylmethoxy)-lH-indole-3-acetic acid three quarters hvdrate
To a mixture of the above ester (0.5 g, 1.0 mmol) in acetonitrile (20 mL) is added iodotrimethyl silane (0.46 g, 2.2 mmol). After refluxing for 24 hours the mixture is diluted with ethyl acetate then washed with 10% Na2S2θ3 solu¬ tion, water and 15% NaHCO3 solution. A precipitate forms which is filtered off, washed with IN HCl and dried in vacuo to give the title compound as a solid (0.05 g, 10% yield), m.p. 191-193βC. Analysis for: C28H21CIN2O4 . 0.75 H2O Calculated: C, 67.47; H, 4.55; N, 5.62. Found: C, 67.68; H, 4.22; N, 5.94. Example 18 2-Methyl-5-(2-quinolinylmethoxy)-l,2-(quinoIinyl- methvn- l H-in dole-3-acetic aj d
To a stirred suspension of sodium hydride (1.4 g, 29.2 mmol) in dimethylformamide (40 mL) at O'C is added 5-hydroxy-2-methyl-lH-indole-3-acetic acid (2.0 g, 9.76 mmol). After 1 hour 2-chloromethylquinoline (3.44 g, 19.48 mmol) is added and the reaction mixture is allowed to warm to room tempera¬ ture. After stirring overnight water is added and the pH was adjusted to 5 witii 0.5N HCl. The reaction mixture is extracted with methylene chloride, the organic extract is dried over MgSO4 and evaporated to a solid which upon sequential trituration with ethanol and hot ethanol affords 2.5 g (52% ) of a beige solid, m.ρ. 198-200°C. Analysis for: C31H25N3O3 Calculated: C, 76.37; H, 5.17; N, 8.62. Found: C, 76.72; H, 5.24; N, 8.74.
Example 19 l-[(4-Chlorophenyl)methyl]-5-(hexyIoxy)- 2-methvl-1H-indole-3-acetic a£i_d
A. 5-(Hexyloxy)-2-methyl- lH-indole-3-acetic acid
To a solution of 5-hydroxy-2-methyl-lH-indole-3-acetic acid (7.17 g, 35.0 mmol) in methanol (40 mL) is added sodium methoxide (70 mmol). The solu¬ tion is evaporated, dimethylformamide (40 mL) is added followed by hexyl iodide (35.0 mmol). After 2 days the solvent is removed and the residue is partitioned between ethyl acetate and pH=4 buffer. The organic layer is separated, dried over MgSO4 and evaporated to 8.6 g of a brown solid. Recrystallization from ethanol/ ater affords 6.23 g (62%) of white crystals, m.p. 65-67'C. Analysis for: C17H23NO3 Calculated: C, 70.56; H, 8.01; N, 4.84. Found: C, 70.52; H, 7.98; N, 4.83.
B. 1 -[(4-Chlorophenyl)meth vn-5-(hexyloxy)-2-methyl- lH-indole-3-acetic acid The acid from part A is converted to the methyl ester (m.p. 148- 150°C) via diazomethane (see Example 16B). To a solution of die ester (4.5 g, 14.83 mmol) in dimethylformamide (40 mL) is added sodium hydride (1.42 g, 29.6 mmol). After 30 minutes 4-chlorobenzylchloride (2.38 g, 14.83 mmol) is added. After stirring overnight, the solvent is removed and the residue is partitioned between ethyl acetate and pH=4 buffer. The organic layer is separated, dried over MgSO4 and evaporated to a red solid. Sequential recrystallization from ether and isopropanol gives 2.2 g of crystals (36%), m.p. 148-150'C. Analysis for: C24H28CINO3
Calculated: C, 69.64; H, 6.82; N, 3.38. Found: C, 70.00; H, 6.71; N, 3.46.
Example 20 2-MethyI-5-(2-quinoIinylmethoxy)-l-(2-quinolinyl- methyl)-lH-indQle-3-carfrpχylic acid ethyl ester
Under an atmosphere of nitrogen, a mixture of ethyl-5-hydroxy-2- methyl-3-indolyl carboxylate (3.5 g, 16 mmol), finely powdered anhydrous potassium carbonate (2.2 g, 16 mmol) and 18-crown-6 (0.48 g, 1.8 mmol) in dry acetonitrile (50 mL) is stπxed at room temperature for 0.5 hours. In one portion 2-chloromethylquinoline (free base, 2.85 g, 16 mmol) is added and the mixture is stirred at 55°C overnight. The reaction (found to be complete by TLC analysis) is filtered hot.
The filtrate affords the desired product (0.75 g, 9%) as a tan solid, m.p. 162-163°C. Recrystallization from ethyl acetate gives pure product (0.42 g, m.p. 162-163βC).
Analysis for: C32H27N3O3
Calculated: C, 76.63; H, 5.43; N, 8.3.
Found: C, 76.20; H, 5.59; N, 8.11.
Example 21 α-Methyl-6-(2-quinolinylmethoxy)-9-(2-quinolinyl- methvO-9H-carbazole-2-acetic acid
A. 4-Benzyloxyphenylhvdrazine hydrochloride
A solution of sodium nitrite (3.8 g, 0.055 mol in 20 mL of H2O), is added dropwise to an ice cold stirring suspension of 4-benzyloxyaniline (13.0 g, 0.055 mol in 150 mL of concentrated HCl). The reaction mixture is stirred for 90 minutes at -8° to lO'C. A solution of SnC • 2H2O (32.0 g, 0142 mol in 50 mL concentrated HCl) is added and stirred for 1 hour at O'C. The reaction mixture is removed from the ice bath and is stirred at room temperature for 20 hours. The mixture is then filtered and washed with water to give 13.0 g (95% yield) of product, m.p. 182-185'C.
B. 6-Benzyloxy-α-methyl-1.2.3.4-tetrahydrocarbazole-2-acetic acid ethyl ester
A mixture containing 4-benzyloxyphenylhydrazine hydrochloride (37.0 g, 0.148 mol), α-methyl-3-oxocyclohexane acetic acid (40.0 g, 0.235 mol), glacial acetic acid (650 mL) and H2O (100 mL) is stirred at room temperature under a nitrogen atmosphere for 2.5 hours. The mixture is then refluxed for 20 minutes, cooled and poured into 200 mL of ice/H2θ. The precipitate is filtered to give 80 g of red colored crude product The crude product is dissolved in ether (600 mL) concentrated H2SO4
(5 mL) is added and the mixture refluxed for 24 hours. The reaction mixture is cooled, concentrated and diluted with ether (100 mL), then washed with IN NaOH (2 x 200 mL), water (2 x 200 mL) and finally once with brine (200 mL), dried over anhydrous MgSO4, filtered and concentrated to give 40.0 g crude product as a thick oil.
C. 6-Benzyloxy-α-methyl carbazole-2-acetic acid ethyl ester
A mixture consisting of 6-benzyloxy-α-methyl- 1,2,3 ,4-tetrahydro- carbazole-2-acetic acid ethyl ester (15.0 g, 0.0398 mol) xylene (500 mL), and chloranil (10.0 g, 0.0408 mol) is refluxed in the dark under a nitrogen atmosphere for 24 hours. The reaction mixture is concentrated to 200 mL and the precipitate is removed by filtration. The precipitate is triturated with benzene (3 x 100 mL), the solvent fractions are combined and washed with IN NaOH (3 x 150 mL), washed with water until neutral to pH paper, dried over MgSO4, filtered and concentrated to give 15.8 crude product. The crude product is flash chromatographed on silica gel using 15% ethyl acetate hexane as an eluent to give 7.5 g product as a thick oil.
D. 6-Hydroxy-α-methyl carbazoIe-2-acetic acid ethyl ester
A mixture consisting of 6-benzyloxy-α-methyl carbazole-2-acetic acid ethyl ester (7.3 g, 0.0195 mol) and ethanol (130 mL) is subjected to hydrogenation over 10% Pd/c (0.9 g) for 18 hours at 35 psi. The reaction mixture is filtered and concentrated to give 5.0 g product as a foam.
E. 6-Hydroxy-α-methyl carbazole-2-acetic acid
A mixture consisting of 6-hydroxy-α-methyl carbazole-2-acetic acid ethyl ester (5.0 g, 0.021 mol), potassium hydroxide (5.0 g, 0.089 mol), methanol - 54 -
Analvsis for: C29H26CIN3O3 • 3/4 H2O Calculated: C, 67.83; H, 5.39; N, 8.18. Found: C, 67.87; H, 5.10; N, 8.39.
Example 23 l-[(4-Chlorophenyl)methyI]-2-methyl-N-(phenylsulfonyl)-
5-f2-auinolinvlmethoxv)- lH-indole-3-acetamide
To a solution of the acid of Example 16 (l.Og, 2.12mmol), benzene- sulfonamide (0.34g, 2.12mmol) and benzotriazole-l-yloxytris(dimethylamino)- phosphonium hexafluorophosphate (0.94g, 2.12mmol) in methylene chloride(25 mL) is added tiiethylamine (0.6mL, 2 equiv). The reaction mixture is stirred overnight and then is quenched by addition of brine. After sequential washes with water, 0.5N HCl and water, the organic layer is dried over MgSO4 and evaporated to afford a crude solid, which upon toluene recrystallization affords a crystalline solid , m.p. 194-
Analysis for: C34H28CIN3O4S
Calculated: C, 66.93; H, 4.63; N, 6.89. Found: C, 67.01; H, 4.66; N, 7.33.
Example 24 l-[(4-Fluorophenyl)methyI]-2-methyI-5-(2-quinolinyl- methQχy)-lH-indftle-3-acetic acid hemihydrate
The title compound is prepared according to the method of Example 16 using 4-fluorobenzyl bromide. A crystalline solid is obtained, m.p. 199-202°C. Analysis for: C28H23FN2O3 • 1/2 H2O Calculated: C, 72.55; H, 5.21; N, 6.04. Found: C, 72.84; H, 5.31; N, 5.68.
Example 25 l-[(4-Bromophenyl)methyI]-2-methyI-5-(2-quinoIinyl- methoxy -lH-indole-3-acetic acid hemihvdrate
The title compound is prepared according to the method of Example 16 using 4-bromobenzyl bromide. A crystalline solid is obtained, m.p. 215-217°C. - 53 -
(300 mL), and water (20 mL) is refluxed for 15 hours. The reaction mixture is cooled concentrated and acidified with a 1:1 solution of concentrated HCl and water to pH 1. The aqueous layer is extracted with ether (4 x 300 mL), washed with water (200 mL), brine (200 mL), dried over MgSO4, filtered and concentrated to give 2.6 g of product as a foam.
F. α-Methyl-6-(2-quinolinylmethoxy -9-(2-quinolinylmethyl)-9H-carbazole-2- acetic acid
A mixture consisting of 6-hydroxy-α-methyl carbazole-2-acetic acid
(2.6 g, 0.010 mol), dimethyl sulfoxide (200 mL), sodium hydroxide (2.0 g in 20 mL H O, 0.05 mol) and 2-chloromethylquinoline-HCl (4.0 g, 0.018 mol) is heated to 80°C for 3 hours. The reaction mixture is cooled, poured into water
(1000 mL), and the pH is adjusted to 4 with IN HCl. The aqueous solution is extracted with ethyl acetate (4 x 300 mL) and the organic layers are combined, washed with water (2 x 300 mL), once with brine (300 mL), dried over MgSO4, filtered and concentrated to give 3.0 g of crude product. The latter is chromatographed on silica gel using 10% methanol/methylene chloride as an eluent to give 8 g of product, m.p.
197-199'C.
Analysis for: C35H27N3O3
Calculated: C, 78.19; H, 5.06; N, 7.82. Found: C, 76.01; H, 5.02; N, 7.36.
Example 22 l-[(4-Chlorophenyl)methyl]-2-methyI-N-(methyl)-N-(hydroxy)-5-(2- αuinolinylmethoxy -lH-indole-3-acetamide three quarters hvdrate
To a solution of the acid of Example 16 (0.2 g, 0.42mmol), N-methyl- hydroxylamine hydrochloride (0.035 g, 0.42mmol) and benzotriazole-l-yloxytris(di- methylamino)phosphonium hexafluorophosphate (0.188g, 0.42mmol) in methylene chloride (5 mL) is added tiiethylamine (0.12mL, 2 equiv). The reaction mixture is stirred overnight and then is quenched by addition of brine. After sequential washes with water, 0.5N HCl and water, the organic layer is dried over MgSO4 and evaporated to afford a crude solid, which upon chloroform recrystallization(twice) affords a crystalline solid , m.p. 192-194°C. This material is washed sequentially with 0.5N NaOH and water and dried to afford white crystals, m.p. 191-193°C. 56 -
Example 29 2-MethyI-l-pentyl-5-(2-quinolinylmethoxy)- lH-indole-3-acetic acid hemihvdrate
The title compound is prepared according to the method of Example 16 using pentyl iodide. A crystalline solid is obtained, m.p. 90-93°C. Analysis for: C26H28N2O3 • 1/2 H2O Calculated: C, 73.38; H, 6.86; N, 6.58. Found: C, 73.37; H, 7.11; N, 6.41.
Example 30 l-HexyI-2-methyl-5-(2-quinolinylmethoxy)-lH- indole-3-acetic acid three quarters hvdrate
The title compound is prepared according to the method of Example 16 using hexyl iodide. A crystalline solid is obtained, m.p. 122-124°C. Analysis for: C27H30N2O3 • 3/4 H2O Calculated: C, 73.03; H, 7.15; N, 6.30. Found: C, 73.25; H, 7.10; N, 6.30.
Example 31 l-HeptyI-2-methyI-5-(2-quinolinyI- methoxv)-lH-indole-3-acetic acid
The title compound is prepared according to the method of Example 16 using heptyl iodide. A crystalline solid is obtained, m.p. 127-131°C. Analysis for: C28H32N2O3 Calculated: C, 75.65; H, 7.26; N, 6.30. Found: C, 75.80; H, 7.19; N, 6.26.
Example 32 l-MethyI-2-methyI-5-(2-quinoIinyI- methoxv -lH-indole-3-acetic acid
The title compound is prepared according to the method of Example 16 using methyl iodide. A crystalline solid is obtained, m.p. 186-188°C. - 55 -
Analysis for: C28H23BrN2θ3 1/2 H2O Calculated: C, 64.12; H, 4.61; N, 5.34. Found: C, 64.40; H, 4.76; N, 5.01.
Example 26 l-[(Phenyl)methyl-2-methyl]-5-(2-qiiinoIinyl- methoxy)-lH-indole-3-acetic acid tenth hvdrate
The title compound is prepared according to the method of Example 16 using benzyl bromide. A crystalline solid is obtained, m.p. 199-202°C. Analysis for: C28H24N2O3 • 0.1 H2O Calculated: C, 76.72; H, 5.56; N, 6.39. Found: C, 76.30; H, 5.37; N, 6.34.
Example 27 l-[(4-Carboxyphenyl)methyI]-2-methyl-5-(2- quinolinylmethoxy -lH-indole-3-acetic acid
The title compound is prepared according to the method of Example 16 using 4-(chloromethyl)benzoic acid. A crystalline solid is obtained, m.p. 232-234°C. Analysis for: C29H24N2O5 Calculated: C, 72.49; H, 5.03; N, 5.83. Found: C, 72.44; H, 5.18; N, 5.98.
Example 28
2-Methyl-5-(2-quinolinylmethoxy)-l-[[4-(2-quinolinyImethoxy)- nhenvnmethvn-lH-indole-3-acetic acid hemihvdrate
The title compound is prepared according to the method of Example 16 using 4-(2-quinolinylmethoxy)benzyl chloride. A crystalline solid is obtained, m.p. 182-185°C.
Analysis for: C38H31N3O4 • 1/2 H2O Calculated: C, 75.73; H, 5.35; N, 6.97. Found: C, 75.41; H, 5.33; N, 6.78. Analvsis for: C22H20N2O3
Calculated: C, 73.32; H, 5.59; N, 7.77.
Found: C, 73.03; H, 5.69; N, 7.57.
Example 33 l-[(4-ChIorophenyI)methyI]-5-(2-quinoIinyI- methoxv)-lH-indole-3-acetic acid
The title compound is prepared according to the method of Example 16 using 5-hydroxy-lH-indole-3-acetic acid. The desired product is obtained as white crystals, m.p. 158-159°C. Analysis for: C27H21CIN2O3 Calculated: C, 70.97; H, 4.63; N, 6.13. Found: C, 71.13; H, 4.67; N, 6.03.
Example 34 l-[(4-ChIorophenyI)methyI]-2-methyl-5-(2-naphthalenyl- methoxv). l H-in dol e-3-acetic aci d
The title compound is prepared according to the method of Example 16 using 2-(bromomethyl)naphthalene. A crystalline solid is obtained, m.p. 168-170°C. Analysis for: C29H24CINO3 Calculated: C, 74.12; H, 5.15; N, 2.98. Found: C, 74.43; H, 5.31; N, 2.74.
Example 35 l-[(4-Chlorophenyl)methyI]-2-methyI-5-
(Dhenvlmethoxv)-lH-indole-3.acetic acid
The title compound is prepared according to the method of Example 16 using benzyl chloride. A crystalline solid is obtained, m.p. 140-142°C. Analysis for: C25H22CINO3 Calculated: C, 71.51; H, 5.28; N, 3.34. Found: C, 71.54; H, 5.29; N, 3.47. Example 36 l-[(4-Chlorophenyl)methyl]-2-methyI-5-(2- nvridinvlmethoxv lH-indole-3-acetic acid
The title compound is prepared according to the method of Example 16 using 2-(chloromethyl)pyridine. A crystalline solid is obtained, m.p. 231-233°C. Analysis for: C24H21CIN2O3 Calculated: C, 68.49; H, 5.03; N, 6.66. Found: C, 68.61; H, 5.12; N, 6.39.
Example 37 l-[(4-Chlorophenyl)methyI]-2-methyI-5-(2-benzo- thia7.olvlmethoxv>-l H-indole-3-acetic acid
A. 2-Methyl-5-(2-benzothiazolylmethoxy -lH-indole-3-acetic acid ethyl ester
To a mixture of 5-hydroxy-2-methyl-lH-indole-3-acetic acid ethyl ester (5.6 g, 17.15 mmol) and cesium carbonate (15.0 g, 46.04 mmol) in dimethylsulfoxide (70 mL) that has been previously stirred for 30 minutes is added 2-(chloromethyl)- benzothiazole (4.6 g, 25.08 mmol). After 1 hour the reaction is poured into ice water and extracted with ethyl acetate. The organic layer is washed sequentially with 0.1N NaOH, water and brine, dried over MgSO4 and evaporated to a black oil which is purified by flash chromatography (eluant: ethyl acetate-hexane) to afford 5.4g (59%) of the alkylated ester.
B. l-r(4-Chlorophenyl methyll-2-methyl-5-(2-benzothiazolylmethoxy)-lH-indole- 3-acetic acid
The title compound is prepared according to the method of Example 16 using 2-methyl-5-(2-benzothiazolylmethoxy)-lH-indole-3-acetic acid ethyl ester and 4- chlorobenzyl chloride; however, the hydrolysis is carried out in aqueous methanol. Recrystallization from acetonitrile affords white crystals, m.p. 175-176°C. Analysis for: C26H21CIN2O3S Calculated: C, 65.47; H, 4.44; N, 5.87. Found: C, 65.17; H, 4.61; N, 5.81. Example 38 l-[(4-Chlorophenyl)methyl]-2-methyl-5-[(2-phenyl-
4-thiazolvnmethoxv1-lH-indole-3-acetic acid
The title compound is prepared according to the method of Example 37 using 4-(chloromethyl)-2-phenylthiazole. White crystals are obtained, m.p. 150- 1510Q
Analysis for: C28H23CIN2O3S Calculated: C, 66.86; H, 4.61; N, 5.57. Found: C, 66.46; H, 4.59; N, 5.59.
Example 39 l-[(4-ChIorophenyl)methyl]-2-methyl-5-[[2-(4-trifluoromethyl- Dhenvl>-4-thia7θlvnmethoxv1-l H-indole-3-acetic a_ti_
The title compound is prepared according to the method of Example 37 using 4-chloromethyl-[(4-trifluoromethyl)-2-phenyl]thiazole. White crystals are obtained, m.p. 196-197°C.
Analysis for: C29H22CIF3N2O3S Calculated: C, 61.00; H, 3.88; N, 4.91. Found: C, 60.95; H, 3.97; N, 4.89.
Example 40 l-[(Phenyl)methyI]-2-methyI-5-[(2-phenyl-4- thiazolvnmethoxvl-lH-indole-3-acetic acid
The title compound is prepared according to the method of Example 37 using 4-(chloromethyl)-2-phenylthiazole and benzyl chloride. White crystals are obtained from toluene, m.p. 150-151°C. Analysis for: C28H24N2O3S
Calculated: ' C, 71.77; H, 5.16; N, 5.98. Found: C, 71.53; H, 5.11; N, 5.90. Example 41 2-Methyl-5-(2-quinoIinylmethoxy)-l-[[4-(2-quinolinyl- methoxv phenvnmethvlene1-lH-indene-3-acetic acid
A. 2-Methyl-5-(2-quinolinylmethoxy -lH-indene-3-acetic acid methyl ester A mixture of 2-methyl-5-hydroxy-lH-indene-3-acetic acid methyl ester
(1.8 g, 8.25 mmol), powdered anhydrous potassium carbonate (1.13 g, 8.25 mmol) and 18-crown-6 (0.219 g) in acetonitrile (21 mL) is stirred at room temperature for 15 minutes. 2-(Chloromethyl)quinoline (1.6 g, 9.08 mmol) is then added and the mixture is stirred for 15 hours at 65°C. After the solvent is removed, the residue is partitioned between water and ethyl acetate. The organic phase is dried over MgSO4 and evaporated. The crude product is purified by flash chromatography (eluant: methylene chloride-ethyl acetate) to afford 2.35 g (79%) of the desired product.
B. 2-Methyl-5-('2-quinolinylmethoxyVl-rr4-f2-quinolinylmethoxy)phenyn methylenel-lH-indene-3-acetic acid To a solution of the above ester (1.77 g, 4.93 mmol) in anhydrous methanol (20 mL) is added 4-[(2-quinolinyl)methoxy]benzaldehyde (1.63 g., 6.19 mmol) followed by dropwise addition of 25% methanolic sodium methoxide(3.46 mL). After the reaction mixture is refluxed for 13 hours, the solvent is removed and water is added. The pH is adjusted to 6.5 and the resulting precipitate is extracted sequentially with methylene chloride and ethyl acetate. The combined extracts are dried over MgSO4 and evaporated to a crude solid which is triturated with diethyl ether and then redissolved in hot ethyl acetate. After the insolubles are filtered off, the solution is concentrated until precipitation occurs. The precipitate is filtered, washed diethyl ether and hexane and dried to afford 1.31 g(45%) of the desired product as a Z E mixture (7.6/1), m.p. 191-193°C(dec). Analysis for: C39H30N2O4 Calculated: C, 79.30; H, 5.12; N, 4.74. Found: C, 79.11; H, 5.05; N, 4.59. Example 42
N-[[l-[(4-ChIorophenyI)methyl]-2-methyl-5-(2-quinolinyl- methoxv)-1 H-indol-3-vnmethvn-N,-hvdroxvurea
To a solution of the acid of Example 16 (0.72 g, 1.52 mmol), in benzene (25 mL) is added triethylamine (0.23 mL, 1.1 equiv) followed by diphenyl- phosphoryl azide (0.46 g, 1.1 equiv). After the reaction mixture is heated at 90°C for 1 hour, a solution of hydroxylamine hydrochloride (0.23 g, 2.2 equiv.) in a mixture of triethylamine (0.46 mL) and water (0.25 mL) is added to it, and the reaction mixture is heated at 90°C overnight The reaction is quenched by addition of ammonium chloride solution, and the resulting precipitate is filtered off, washed with water and ethyl acetate and dried to afford 0.45 g (59%) of a white solid, m.p. 184- 186°C. Analysis for: C28H25CIN4O3 Calculated: C, 67.13; H, 5.03; N, 11.18. Found: C, 66.85; H, 5.06; N, 10.85.
Example 43
The compounds 5- and 12-hydroxyeicosatetraenoic acid (5-HETE and 12-HETE) and LTB4 are early arachidonic acid oxidation products in the lipoxygenase cascade, which have been shown to mediate several aspects of inflammatory and aller¬ gic response. This is especially true with respect to 5,12-diHETE, which is also denoted as LTB4 [see Ford-Hitchinson, J. Rov. Soc Med.. J_ , 831 (1981)]. Com¬ pounds which inhibit the PLA2-mediated 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 polymorpho- nuclear 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 (150-200 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% NaCl). The exudate is centrifuged at 400 xg for 10 minutes. The supernatant fluid is discarded and the cell jjellet is resuspended to a concentration of 2.0 x 107 cells/mL in HBSS containing Ca++ and Mg++ and 10 μM 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 μM), [ H]-AA (3.0 μCi/mL) and unlabeled AA (1 μM) are then added and the samples are further incubated for 10 minutes. The reaction is terminated by centrifugation and pelleting cells. Supematants 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:
Figure imgf000064_0001
Percent solvent changes are accomplished in a linear fashion.
Injections: 140 μL of each supernatant is injected direcdy onto column and 3H arachidonic acid metabolites are monitored using an on-line radioactivity detector (Ramona, IN/US, Fairfield, NJ). Standards: 104 - 2.0 x 104 dpm of eicosanoids of interest are injected in 90 μL EtOH cocktail.
Co-chromatography with standard [3H] leukotriene B4 (LTB4) in medium of stimulated 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.
Testing compounds of the invention in this assay gave the following results: Table l
Figure imgf000065_0001
negative number denotes potentiation
Example 44
The procedure of Example 43 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 43 is carried out as described. However, in order to determine cyclooxygenase activity, the samples are co- chromatographed with authentic reference [3H]-PGE2-
The results are calculated as in Example 43 and presented below:
Table π
Figure imgf000066_0001
* Denotes a potentiation of cyclooxygenase (PGE2 synthesis). Exa nle 45
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:
Figure imgf000067_0001
100 * pre-incubate at room temperature 30 min prior to substrate addition.
1 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.
3 Stock 0.5 m Trisma-Base.
Stock 0.5 M Trisma-HCl. Adjust pH to 7.5 (optimum for enzyme). Deionized and distilled water.
5 Stock 10 mM prepared in dimethyl sulfoxide. Make 1:2 dilution with dimethyl sulfoxide and add 1 uL to 100 μL 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 lOOμL 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 μg/mL - Analytichem International) are conditioned with 0.5 mL tetrahydrofuran followed by 0.5 mL tetrahydrofuran/water (2 mL:0.1 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 μL 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 Λ nn
% change = fe v x 100 vehicle dpm
Activity of Standard Drugs: I C S Q (μ M )
Human Platelet Human Synovia!
Drug PL A2_ PL A2_
Arachidonic Acid 8.6 3.2
Monoalide 25.2 0.14
When tested in this assay, the compounds of the invention gave the following results:
Compound of Example No. sulindac indomethacin
1
2 ' 4
16 17 18
Figure imgf000068_0001
Table III (cont'd.)
% Inhibition at 10 μM ICsn (μM)
Compound of Example No. HP* HSF** HP HSF 19 71 45
23 64
24 56
25 11
26 92 2.6 27 45
28 41
29 53 15.5
30 46 16.4
31 32 32 36
33 38
34 33.7
35 28 52.9
36 46 37 39
38 31
39 46
40 42
41 90 3.8 * humanplatelet
** humansynovialfluid
Example 6
The ability of the compounds of the invention to inhibit paw edema induced by the exogenous administration of PLA2 is measured in the in vivo PLA2 murine paw edema assay.
The assay is carried out as follows:
Non-fasted, male CD-I mice (8 weeks old; 31-36 grams) are placed in plastic boxes in groups of six. The right hind paw volume is measured using mercury plethysmography (zero time). Compounds are dosed orally (0.5 mL of 0.5% Tween- 80) 1 or 3 hours prior to PLA2 injection or intravenously (0.2 mL in 0.3% dimethylsulfoxide/saline) 3 minutes prior to PLA^ injection. A solution of purified PLA2, from the diamond back cotton mouth snake (A. piscivorus piscivorus) is prepared in saline at a concentration of 6 μg/mL. Fifty (50) μL (0.3 μg) of this PLA2 solution is injected subcutaneously into the right hind paw with a plastic 1 mL plastic syringe (27 gauge, 1" needle). Paw volume of the injected paw is measured again at 10 minutes, 30 minutes and 60 minutes after PLA2 injection. Animals are euthanized with CO2 at the completion of the study.
The paw edema is calculated by subtracting the zero time volume from the volume recorded at each time period. Mean paw edema for each treatment group is then calculated and expressed as (μL + S.E.). Drug effects are expressed as a percent change from control (vehicle) values. Statistical significance is determined by a one¬ way analysis of variance with LSD comparison to control (p < 0.05). EDso's are determined using repression analysis.
The activity of standard drugs in this assay is as follows:
Figure imgf000070_0001
Figure imgf000070_0002
19 30 (i.p.) -30
60 (i.p.) -34
* intravenous ** peroral *** intraperitoneal
The results show that the compounds of the invention are effective in vivo in inhibiting edema induced by die exogenous administration of snake venom PLA2.
Example 47 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 die peritoneal cavity is lavaged with 2 mL ice cold Hanks Balanced Salt Solution (HBSS) without CaCtø, MgSO4 • 7H2O and MgCl • 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-PGFια) 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. The activity of standard drugs in this assay is as follows:
Compound ED -JQ mg/kg p.o.
LT& 6-keto-PGFι,v/TxBo
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 anti- inflammatoiy compound etodolac gave the following results:
Table V
Figure imgf000072_0001
* perorally administered
** negative values denote potentiation
The results show that the compounds of the invention exert an inhibitory effect on both the lipoxygenase pathway and the cyclooxygenase pathway.
Example 48
The LTD4 antagonist activity of the compounds of the invention is assessed in the in vitro isolated guinea pig trachea assay. This assay is carried out as follows:
Male Hartley guinea pigs (350-400 g) are euthanized by a blow to the head, the neck is opened and die trachea removed. The trachea is maintained in aerated physiological salt solution, cleared of connective tissue and fat and cut into rings approximately 2 mm in width (usually containing two cartilaginous segments per ring). Two pieces of silk suture are then passed through the lumen of the tracheal ring and are tied around the cartilage, one on each side of the trachealis muscle. The tracheal ring is suspended between a glass hook and a force displacement transducer in a 10 mL organ bath for measurement of isometric tension. Tissues are maintained at 37° C in aerated (95% CO2/5% CO2) physiological salt solution of the following composition: NaCl (100 mM), KH2PO4 (1.18 mM), KCl (4.74 mM), CaCl (2.5 mM), MgSO4 • 7 H2O (1.19 mM), NaHCL3(25 mM), dextrose (11.1 mM) and indomethacin (1 μM). The tracheal rings are maintained at 2 g resting tension and equilibrated for 45 minutes (with frequent washing and readjustment of resting tension).
The tracheal rings are first contracted by the addition of carbachol (3xlO"6M), to determine tissue responsiveness and establish a reference contraction. On attainment of a stable level of contraction (approximately 30 minutes), the tissues are washed several times until baseline tension has been restored and dien re-equilibrated for 30 minutes. The tissues are then incubated for 45 minutes with a test antagonist (either lxlO^M or lxlO^M) or 10 μL of an appropriate solvent control (control, non-treated). One tissue in each group serves as the control. Twenty minutes prior to the construction of the LTD4 cumulative concentration-response curve, L-cysteine (lxl0"2M final bath concentration) is added to inhibit bioconversion of LTD4 to LTE4. Only one LTD4 concentration-response curve is constructed in each tissue.
All responses to LTD4 in an individual tissue are measured as a percentage of the reference contraction of that tissue to carbachol. LTD4 antagonist activity is determined by comparison of the concentration response curves of LTD4 in the presence and absence of antagonist. Assessment of the relative rightward shift of the antagonist treated curve relative to the solvent (control) treated tissue is calculated as a concentration ratio (Eq. A) and used in subsequent calculations to derive an antagonist pKβ value (Eqs B and C). In the event that the maximum response to LTD4 is depressed, the EC50 for that particular curve is determined, an "apparent" pKβ reported, and the compound reported as "non-competitive."
EC50 treated tissue
A) Concentration Ratio (CR) =
EC50 control
[Test Compound]
B) KB =
CR-1
C) -log KB = pKB
If a compound is found to be active and/or depress the maximal response to LTD4, then a range of concentrations of the test compound should be used generating multiple concentration ratios which would then be used to perform a Schild analysis, and determination of a pA2 value where appropriate. The activity of reference leukotriene antagonists in this assay is as follows:
Figure imgf000074_0001
The above results demonstrate that the compounds tested have significant leukotriene antagonist activity as measured in the in vitro isolated guinea pig trachea assay.
Example 49
The compounds of the invention are further tested in the rat carrageenan paw edema assay to determine their ability to inhibit the acute inflammatory response. This assay is carried out as follows: 140-180 g Male Sprague-Dawley rats, in groups of 6 animals are injected subcutaneously in the right paw with 0.1 mL of 1% carrageenan at zero time. Mercury plethysmographic readings (mL) of the paw are made at zero time and 3 hours later. Test compounds are suspended or dissolved in 0.5% methylcellulose and given perorally 1 hour prior to carrageenan administration. The increase in paw volume (edema in mL) produced by the carrageenan is measured. Paw edema is calculated (3 hour volume minus zero time volume), and percent inhibition of edema is determined. Unpaired Student's t-test is used to determine statistical significance.
The activity of standard drugs in this assay is as follows:
Drug Oral EDsn (95% C.L mg ke
Indomethacin 3.7 ( 0.6, 23.8)
Aspirin 145.4 (33.1, 645.6)
Phenylbutazone 26.2 ( 2.3, 291.0) When tested in this assay, a compound of the invention and the anti-inflammatory drug etodolac gave the following results:
Table Vπ
Figure imgf000075_0001
* administered perorally
The results show that the compounds tested have activity in the rat carrageenan paw edema assay, evidencing an effect on the acute inflammatory response.
Example 50 The assay of tiiis Example measures the ability of the compounds tested to inhibit 5-lipoxygenase in human whole blood. This assay is carried out as follows: Blood is obtained in 50-100 ml quantities from male donors. White blood cell counts and differentials are made. Two ml of blood are placed in a 15 ml polypropylene test tube. Compounds are solubilized in dimethylsulfoxide and diluted 1:10 in 10% bovine serum albumin in phosphate buffered saline, pH 7.4 resulting in a final dimethylsulfoxide concentration of 0.1% in the blood. Then, compounds are added to the blood in a shaking water bath at 37*C for 10 minutes prior to the addition of 30 μM calcium ionophore (A23187; Sigma). After ionophore administration, whole blood samples are mixed and incubated for 20 minutes at 37* C in a shaking water bath. Incubation is terminated by placing samples in an ice bath and immediately adding ethylene glycol-bis-(β-aminoethyl ether)-N,N,N',N'-tetraacetic acid (10 mM). Samples are mixed and centrifuged at 1200 x g for 15 minutes at 4*C. Preparation of samples for evaluation by RIA or ELISA is carried out by the following protocol. Plasma is removed from sample tubes, placed in 15 ml polypropylene test tubes containing 8 ml methanol, and then vortexed to precipitate protein. Samples are stored at -70"C overnight. The next day, samples are centrifuged at 200 x g for 15 minutes at 4°C to pellet the precipitate. Samples are dried in a Savant speed vac concentrator, reconstituted to original volume with ice cold RIA or ELISA buffer, and stored at -70°C until assayed. The assay for eicosanoids (LTB4, TxB2, and PGE2) is performed as described by the manufacturer of the [3H]-RIA kit or ELISA kit (LTB4-Amersham, TxB2 and PGE2 - Caymen Chemical). The total eicosanoid level in 2 ml of blood is calculated and reported as ng/106 neutrophils. Significance is determined by a one-way analysis of variance with least significant difference (LSD) comparisons to control (p < 0.05) and ICso's (μM) are determined by regression analysis (Finney, 1978). Drug effects are expressed as percent change from control values. Compounds tested in vitro are solubilized in dimethylsulfoxide and diluted 1:10 in 10% bovine serum albumin in phosphate buffer saline resulting in a final dimethylsulfoxide concentration of 0.1% in the blood.
The results for compounds of the invention tested in this assay are presented in Table DC.
Figure imgf000076_0001

Claims

WHAT IS CLAIMED TS:
A compound having the formula
A(CH2)nO-B wherein
A is C4-C8 alkyl, phenoxyethyl, phenoxyphenyl or a group having the formula
Figure imgf000077_0001
wherein
R3
I
X is -N- or -C-
R3 R3 R3 R3 R3 I I I I
Z is -C= C- , -C=N- , -N=C- , -N- , -S- or -O- ;
R1 is hydrogen, lower alkyl, phenyl or phenyl substituted with trifluoromethyl; 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
Figure imgf000077_0002
wherein
O O O
A - 11 11 7 11
R4 is -CO2R2 , -N(OH)CNH2 , -N(OH)CR7 , -NHCNHOH , O O
II 7 II π
-CN(OH)R7 or -CNHSO2Rπ ; m is 0
R >55 ;i„s ; or phenyl or phenyl substituted by
Figure imgf000078_0001
halo, lower alkylthio, lower alkylsulfinyl or lower alkylsulfonyl; R6 is A(CH2)nO- or halo; with the proviso that when R6 is halo, R5 is
Figure imgf000078_0002
R7 is lower alkyl; Y is -CH2- or-O- ; R8 is lower alkyl or -(CH2)mCO2R3 ; O
R9 is -CR10 or -CH2R10 ;
R1^ is lower alkyl, phenyl, phenyl substituted with carboxy, halo, loweralkylthio or loweralkylsulfinyl, pyridyl, furanyl or quinolinyl;
R11 is lower alkyl or phenyl; and the pharmacologically acceptable salts thereof.
2. The compound of Claim 1, 3-[(4-chlorophenyl)methylene]-[2-methyl-6- (2-quinoUnylmethoxy)]-3H-indene- 1 -acetic acid.
3. The compound of Claim 1, 2-methyl-3-[[4-(methylthio)phenyl]- methylene]-6-(2-quinolinylmethoxy)-3H-indene-l-acetic acid.
4. The compound of Claim 1, 2-methyl-3-[[(4-methylsulfinyl)phenyl]- methylene]-6-(2-quinolylmethoxy)-3H-indene-l-acetic acid.
^. The compound of Claim 1,
5-fluoro-2-methyl-l-[[4-(2-quinolinyl- meth φhenyl] methylene]- lH-indene-3-acetic acid.
6. The compound of Claim 1, 2-methyl-3-[[4-methylthio)phenyl]- methylene]-6-[(2-naphthyl)methoxy]-3H-indene-l-acetic acid.
7. The compound of Claim 1, 3-[(4-chlorophenyl)methylene]-[2-methyl-6- (2-quinolinylmethoxy)]-3H-indene- 1 -acetic acid
8. The compound of Claim 1 , 3-[(4-chlorophenyl)methylene]-[2-methyl-6- (2-quinolinylmethoxy)]-3H-indene-l-acetic acid methylester.
9. The compound of Claim 1, l,8-diethyl-l,3,4,9-tetrahydro-6-(2- quinolinylmethoxy)pyrano[3,4-b]indole-l-acetic acid.
10. The compound of Claim 1, 1 -ethyl- 1,3,4,9- tetrahydro-6-(quinolinyl- methoxy)pyrano[3,4-b]indole-l-acetic acid
11. The compound of Claim 1, 1 -methyl- l,3,4,9-tetrahydro-6-(2- quinolinylmethoxy)pyrano[3,4-b]indole-l-acetic acid.
12. The compound of Claim 1, l-ethyl-2,3,4,9-tetrahydro-6-(2-quinolinyl- methoxy)-lH-carbazole-l -acetic acid.
13. The compound of Claim 1, l,3,4,9-tetrahydro-l-propyl-6-(2- quinolinylmethoxy)pyrano[3,4-b]indole-l-acetic acid.
14. The compound of Claim 1, l-ethyl-l,3,4,9-tetrahydro-6-(2-quinolinyl- methoxy)pyrano[3,4-b]indole-l-acetic acid, methyl ester.
15. The compound of Claim 1, l,3,4,9-tetrahydro-l,l-dimethyl-6-(2- quinolinylmethoxy)pyrano[3,4-b]indole.
16. The compound of Claim 1, l,3,4,9-tetrahydro-l,l-diethyl-6-(2- quinolinylmethoxy)pyrano[3,4-b]indole.
17. The compound of Claim 1, l-[(4-chlorophenyl)methyl]-2-methyl-5-(2- quinolinylmethoxy)- lH-indole-3-acetic acid.
18. The compound of Claim 1, l-[(4-chlorophenyl)methyl]-2-methyl-5-(2- quinolinylmethoxy)-lH-indole-3-acetic acid methyl ester.
19. The compound of Claim 1, l-(4-chlorobenzoyl)-2-methyl-5-(2- quinolinylmethoxy)- lH-indole-3-acetic acid.
20. The compound of Claim 1, l-(4-chlorobenzoyl)-2-methyl-5-(2- quinolinylmethoxy)-lH-indole-3-acetic acid methyl ester.
21. The compound of Claim 1, 2-methyl-5-(2-quinolinylmethoxy)-l,2- (quinolinylmethyl)- lH-indole-3-acetic acid.
22. The compound of Claim 1, l-[(4-chlorophenyl)methyl]-5-(hexyloxy)-2- methyl-lH-indole-3-acetic acid.
23. The compound of Claim 1, 2-methyl-5-(2-quinolinylmethoxy)-l-(2- quinolinylmethyl)-lH-indole-3-carboxylic acid ethyl ester.
24. The compound of Claim 1, α-methyl-6-(2-quinolinylmethoxy)-9-(2- quinolinylmethyl)-9H-carbazole-2-acetic acid.
25. The compound of Claim 1, l-[(4-chlorophenyl)methyl]-2-methyl-N- (methyl)-N-(hydroxy)-5-(2-quinolinylmethoxy)-lH-indole-3-acetamide.
26. The compound of Claim 1, l-[(4-chlorophenyl)methyl]-2-methyl-N- (phenylsulfonyl)-5-(2-quinolinylmethoxy)-lH-indole-3-acetamide.
27. The compound of Claim 1, l-[(4-fluorophenyl)methyl]-2-methyl-5-(2- quinolinylmethoxy)- lH-indole-3-acetic acid.
28. The compound of Claim 1, l-[(4-bromophenyl)methyl]-2-methyl-5-(2- quinolinylmethoxy)- lH-indole-3-acetic acid.
29. The compound of Claim 1, l-[(phenyl)methyl-2-methyl]-5-(2-quinolin- ylmethoxy)-lH-indole-3-acetic acid.
30. The compound of Claim 1, l-[(4-carboxyphenyl)methyl]-2-methyl-5- (2-quinolinylmethoxy )- lH-indole-3-acetic acid.
31. The compound of Claim 1, 2-methyl-5-(2-quinolinylmethoxy)-l-[[4-(2- quinolinylmethoxy)phenyl]methyl]- lH-indole-3-acetic acid.
32. The compound of Claim 1, 2-methyl-l-pentyl-5-(2-quinolinylmethoxy)- lH-indole-3-acetic acid.
33. The compound of Claim 1 , 1 -hexyl-2-methyl-5-(2-quinolinylmethoxy)- lH-indole-3-acetic acid.
34. The compound of Claim 1, l-heptyl-2-methyl-5-(2-quinolinylmethoxy)- lH-indole-3-acetic acid.
35. The compound of Claim 1, l-methyl-2-methyl-5-(2-quinolinyl- methoxy)-lH-indole-3-acetic acid.
36. The compound of Claim 1, l-[(4-chlorophenyl)methyl]-5-(2-quinolinyl- methoxy)-lH-indole-3-acetic acid.
37. The compound of Claim 1, l-[(4-chlorophenyl)methyl]-2-methyl-5-(2- naphthalenylmethoxy)-lH-indole-3-acetic acid.
38. The compound of Claim 1, l-[(4-chlorophenyl)methyl]-2-methyl-5- (phenylmethoxy)- lH-indole-3-acetic acid.
39. The compound of Claim 1, l-[(4-chlorophenyl)methyl]-2-methyl-5-(2- pyridinylmethoxy)-lH-indole-3-acetic acid . '
40. The compound of Claim 1, l-[(4-chlorophenyl)methyl]-2-methyl-5-(2- benzothiazolylmethoxy)- lH-indole-3-acetic acid.
41. The compound of Claim 1 , l-[(4-chlorophenyl)methyl]-2-methyl-5-[(2- phenyl-4-thiazolyl)methoxy]-lH-indole-3-acetic acid.
42. The compound of Claim 1, l-[(4-chlorophenyl)methyl]-2-methyl-5-[[2- (4-trifluoromethylphenyl)-4-thiazolyl]methoxy]-lH-indole-3-acetic acid.
43. The compound of Claim 1, l-[(phenyl)methyl]-2-methyl-5-[(2-phenyl- 4-thiazolyl)methoxy]- lH-indole-3-acetic acid.
44. The compound of Claim 1, 2-methyl-5-(2-quinolinylmethoxy)-l-[[4-(2- quinolinylmethoxy)phenyl]methylene]-lH-indene-3-acetic acid.
45. The compound of Claim 1, N-[[l-[(4-chlorophenyl)methyl]-2-methyl-5-
(2-quinolinylmethoxy)-lH-indol-3-yl]methyl]-N'-hydroxyurea.
46. A process for preparing a compound of Claim 1 comprising one of the following:
a) where the final product has the formula
Figure imgf000082_0001
wherein R6 is A(CH2)nO-
i) etherifying a compound of formula
Figure imgf000082_0002
wherein R3 is as defined above, with a compound of formula
A(CH2)n-X where A and n are as defined above and X is a leaving group, followed by reaction with tiiethylphosphonoacetate, hydrolysis of the resulting acetic acid ester intermediate and then reaction with an appropriately substituted benzaldehyde to introduce the desired R5CH= moiety into the intermediate to yield die desired final product; or
ϋ) reacting an intermediate having the formula
Figure imgf000082_0003
wherein R3 in the -COOR3 moiety is lower alkyl, with an appropriately substituted benzaldehyde to introduce the desired R5CH= moiety into said intermediate and then etherifying said intermediate with a compound of formula
A(CH2)n-X where A and n are as defined above, and X is a leaving group and, if necessary, hydrolyzing the intermediate to yield a final product in which R3 is hydrogen; or b) where the final product has the formula
Figure imgf000083_0001
wherein R is halo, R represents A(CH2)nO-r- --— and R and n are as defined above, reacting a compound of formula halo
Figure imgf000083_0002
with a compound having the formula
Figure imgf000083_0003
where A and n are as defined above; and, if desired, esterifying the final product free acid; c) where the final products have the formula
Figure imgf000083_0004
wherein Y, R2, R3, R4, R7, R8, R9, A and n are as defined above,
i) etherifying a compound of formula
Figure imgf000083_0005
Figure imgf000084_0001
with a compound of formula
A(CH2)n-X where A and n are as defined above and X is a leaving group, and, if necessary, wherein the intermediate bears the ester function -CO2R3 or -CO2R2, hydrolyzing the intermediate, to yield a compound in which R2 or R3 in the moiety -CO2R2 or -CO2R3 is hydrogen; and further
ii) reacting die intermediate compound
Figure imgf000084_0002
with a compound having the formula R9hal wherein R9 is as defined above and hal represents a halo atom, to yield die final product
Figure imgf000084_0003
d) hydrolyzing a compound of formula I, II, III or IV, wherein the moiety -CO2R2 or -CO2R3 is an ester function, to yield a compound in which R2 or R3 in said moiety is hydrogen or a pharmacologically acceptable salt thereof; or e) converting a compound of formula I, π, HI or IV to a pharmacologically acceptable salt
47. A process as claimed in Claim 46 substantially or as hereinbefore described and illustrated in any one of Examples IE, IF, 1G, 2, 3, 4, 5B, 5C, 6C, 6E, 6F, 7, 8E, 9D, 10D, 11C, 12C, 13, 14C, 15C, 16A, 16C, 16D, 17, 18, 19, 20, 21F and 22 through 42.
PCT/US1990/006251 1989-10-27 1990-10-27 Substituted indole-, indene-, pyranoindole- and tetrahydrocarbazole- alkanoic acid derivatives as inhibitors of pla2 and lipoxygenase WO1991006537A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
KR1019920700971A KR920702345A (en) 1989-10-27 1990-10-27 Substituted indole-, indene-, pyranoindole- and tetrahydrocarbazole-alkanoic acid derivatives as inhibitors of PLA_2 and lipoxygenase
BR909007790A BR9007790A (en) 1989-10-27 1990-10-27 DERIVATIVES OF INDOL-, INDENO-, PYRANINDOL-AND TETRAHYDROCARBAZOL-ALKANOIC ACIDS REPLACED AS PLA2 AND LIPOXYGENASE INHIBITORS
FI921865A FI921865A0 (en) 1989-10-27 1992-04-24 SUBSTITUERADE INDOL-, INDEN-, PYRANOINDOL- OCH TETRAHYDROCARBAZOLE-ALKANSYRADERIVAT SOM INHIBITORER AV PLA2 OCH LIPOXIGENAS.

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US596,134 1984-04-02
US42826089A 1989-10-27 1989-10-27
US428,260 1989-10-27
US59613490A 1990-10-11 1990-10-11

Publications (2)

Publication Number Publication Date
WO1991006537A2 true WO1991006537A2 (en) 1991-05-16
WO1991006537A3 WO1991006537A3 (en) 1991-10-17

Family

ID=27027687

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1990/006251 WO1991006537A2 (en) 1989-10-27 1990-10-27 Substituted indole-, indene-, pyranoindole- and tetrahydrocarbazole- alkanoic acid derivatives as inhibitors of pla2 and lipoxygenase

Country Status (11)

Country Link
EP (1) EP0502106A1 (en)
JP (1) JPH05502222A (en)
KR (1) KR920702345A (en)
AU (1) AU643996B2 (en)
BR (1) BR9007790A (en)
CA (1) CA2070422A1 (en)
FI (1) FI921865A0 (en)
HU (1) HUT63407A (en)
IE (1) IE903872A1 (en)
PT (1) PT95692A (en)
WO (1) WO1991006537A2 (en)

Cited By (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0468789A2 (en) * 1990-07-26 1992-01-29 Merck Frosst Canada Inc. (Quinolin-2-ylmethoxy)heterotetrahydrocarbazoles as inhibitors of the biosynthesis of leukotrienes
EP0468785A2 (en) * 1990-07-26 1992-01-29 Merck Frosst Canada Inc. (Quinolin-2-ylmethoxy)tetrahydrocarbazoles as inhibitors of the biosynthesis of leukotrienes
US5190968A (en) * 1991-09-30 1993-03-02 Merck Frosst Canada, Inc. (Polycyclic-arylmethoxy) indoles as inhibitors of leukotriene biosynthesis
EP0535923A1 (en) * 1991-09-30 1993-04-07 Merck Frosst Canada Inc. (Azaarylmethoxy)indoles as inhibitors of leukotriene biosynthesis
EP0535924A1 (en) * 1991-09-30 1993-04-07 Merck Frosst Canada Inc. (Bicyclic-azaarylmethoxy)indoles as inhibitors of leukotriene biosynthesis
EP0535926A1 (en) * 1991-09-30 1993-04-07 Merck Frosst Canada Inc. (Hetero-arylmethoxy) indoles as inhibitors of leukotriene biosynthesis
WO1993016069A1 (en) * 1992-02-13 1993-08-19 Merck Frosst Canada Inc. (azaaromaticalkoxy)indoles as inhibitors of leukotriene biosynthesis
EP0574774A1 (en) * 1992-06-17 1993-12-22 Bayer Ag Substituted benzothiazolyl- and quinoxalyl methoxy-phenyl acetic acid derivatives
WO1994001407A2 (en) * 1992-07-10 1994-01-20 American Home Products Corporation Indole and indene derivatives as pla2 and lipoxygenase inhibitors
US5308850A (en) * 1991-09-30 1994-05-03 Merck Frosst Canada, Inc. (Bicyclic-hetero-arylmethoxy)indoles as inhibitors of leukotriene biosynthesis
WO1994022869A1 (en) * 1993-03-29 1994-10-13 Merck Frosst Canada Inc. FURO[3,2-b]PYRIDINES AND THIENO[3,2-b]PYRIDINES AS INHIBITORS OF LEUKOTRIENE BIOSYNTHESIS
US5410061A (en) * 1991-10-24 1995-04-25 Lilly Industries Limited Pharmaceutical compounds
DE4338770A1 (en) * 1993-11-12 1995-05-18 Matthias Dr Lehr Indole-2-alkanoic acids and their derivatives as inhibitors of phospholipase A¶2¶
EP0675110A1 (en) * 1994-04-01 1995-10-04 Eli Lilly And Company 1H-Indole-3-glyoxylamide sPLA2 inhibitors
FR2721610A1 (en) * 1994-06-28 1995-12-29 Adir Novel (thia) cycloalkyl [b] indole derivatives, process for their preparation and pharmaceutical compositions containing them
US5496957A (en) * 1993-09-01 1996-03-05 Virginia Commonwealth University Tryptamine analogs with 5-HT1D selectivity
US5504101A (en) * 1994-05-06 1996-04-02 Allelix Biopharmaceuticals, Inc. 5-HT-1D receptor ligands
WO1998005637A1 (en) * 1996-08-01 1998-02-12 Merckle Gmbh Acylpyrroldicarboxylic acids and acylindoldicarboxylic acids and their derivatives and inhibitors of the cytosolic phospholipase a¿2?
US5948779A (en) * 1997-12-12 1999-09-07 Cell Pathways, Inc. Substituted condensation products of n-benzyl-3-indenyl acetamides with heterocyclic aldehydes
US5965619A (en) * 1996-06-13 1999-10-12 Cell Pathways Inc. Method for treating patients having precancerous lesions with substituted indene derivatives
US5998477A (en) * 1996-06-13 1999-12-07 Cell Pathways Inc. Substituted methoxy benzylidene indenyl-acetic and propionic acids for treating patients with precancerous lesions
US6028116A (en) * 1998-04-03 2000-02-22 Cell Pathways, Inc. Substituted condensation products of 1H-indenyl-hydroxyalkanes with aldehydes for neoplasia
US6063818A (en) * 1996-06-13 2000-05-16 Cell Pathways Inc. Substituted benzylidene indenyl formamides, acetamides and propionamides
US6121321A (en) * 1996-06-13 2000-09-19 Cell Pathways, Inc. Substituted methoxy benzylidene indenyl acetic and propionic acids for treating patients with precancerous lesions
WO2001005761A1 (en) * 1999-07-19 2001-01-25 Eli Lilly And Company sPLA2 INHIBITORS
WO2001066110A2 (en) * 2000-03-09 2001-09-13 Eli Lilly And Company METHOD FOR THE TREATMENT OF RENAL DYSFUNCTION WITH sPLA2 INHIBITORS
WO2002020478A1 (en) * 2000-08-24 2002-03-14 Dublin City University N-benzylindole-3-acetic acid derivatives for use in the treatment of drug resistan cancer
WO2002079154A1 (en) * 2001-03-28 2002-10-10 Eli Lilly And Company Substituted carbazoles as inhibitors of spla2
US6500853B1 (en) 1998-02-28 2002-12-31 Genetics Institute, Llc Inhibitors of phospholipase enzymes
US6635771B2 (en) 2001-12-03 2003-10-21 Wyeth N-benzhydryl indole compounds
US6635670B2 (en) 1999-07-19 2003-10-21 Eli Lilly And Company sPLA2 inhibitors
US6797708B2 (en) 2001-12-03 2004-09-28 Wyeth Inhibitors of cytosolic phospholipase A2
GB2407318A (en) * 2003-10-23 2005-04-27 Oxagen Ltd Substituted Indol-3-yl acetic acid derivatives
US6984735B2 (en) 2001-12-03 2006-01-10 Wyeth Process for making an aldehyde
US7101875B2 (en) 2001-12-03 2006-09-05 Wyeth Methods for treating arthritic disorders
US7557135B2 (en) 2005-05-27 2009-07-07 Wyeth Inhibitors of cytosolic phospholipase A2
US7579002B2 (en) * 2003-12-05 2009-08-25 Wisconsin Alumni Research Foundation Method for improving body weight uniformity and increasing carcass yield in animals
US7605156B2 (en) 2001-12-03 2009-10-20 Wyeth Methods for the use of inhibitors of cytosolic phospholipase A2
WO2010011316A1 (en) * 2008-07-23 2010-01-28 Arena Pharmaceuticals, Inc. SUBSTITUTED 1,2,3,4- TETRAHYDROCYCLOPENTA[b]INDOL-3-YL) ACETIC ACID DERIVATIVES USEFUL IN THE TREATMENT OF AUTOIMMUNE AND INFLAMMATORY DISORDERS
GB2431927B (en) * 2005-11-04 2010-03-17 Amira Pharmaceuticals Inc 5-Lipoxygenase-activating protein (FLAP) inhibitors
US7713964B2 (en) 2001-12-03 2010-05-11 Wyeth Llc Methods for treating asthmatic conditions
US7977359B2 (en) 2005-11-04 2011-07-12 Amira Pharmaceuticals, Inc. 5-lipdxygenase-activating protein (FLAP) inhibitors
US8415484B2 (en) 2008-08-27 2013-04-09 Arena Pharmaceuticals, Inc. Substituted tricyclic acid derivatives as S1P1 receptor agonists useful in the treatment of autoimmune and inflammatory disorders
US8853419B2 (en) 2010-01-27 2014-10-07 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and salts thereof
US9085581B2 (en) 2010-03-03 2015-07-21 Arena Pharmaceuticals, Inc. Processes for the preparation of S1P1 receptor modulators and crystalline forms thereof
US10301262B2 (en) 2015-06-22 2019-05-28 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compund1) for use in SIPI receptor-associated disorders
US11007175B2 (en) 2015-01-06 2021-05-18 Arena Pharmaceuticals, Inc. Methods of treating conditions related to the S1P1 receptor
US11426412B2 (en) 2017-10-18 2022-08-30 Jubilant Epipad LLC Imidazo-pyridine compounds as PAD inhibitors
US11459338B2 (en) 2017-11-24 2022-10-04 Jubilant Episcribe Llc Heterocyclic compounds as PRMT5 inhibitors
US11478448B2 (en) 2017-02-16 2022-10-25 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of inflammatory bowel disease with extra-intestinal manifestations
US11529341B2 (en) 2018-03-13 2022-12-20 Jubilant Prodel LLC Bicyclic compounds as inhibitors of PD1/PD-L1 interaction/activation
US11534424B2 (en) 2017-02-16 2022-12-27 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of primary biliary cholangitis
US11629135B2 (en) 2017-11-06 2023-04-18 Jubilant Prodell Llc Pyrimidine derivatives as inhibitors of PD1/PD-L1 activation
US11833156B2 (en) 2017-09-22 2023-12-05 Jubilant Epipad LLC Heterocyclic compounds as pad inhibitors

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ID23219A (en) * 1998-04-17 2000-03-30 Lilly Co Eli SUBSTITUTED TRISICLIC
DZ2769A1 (en) * 1998-04-17 2003-12-01 Lilly Co Eli Substituted tricyclics.
CN101410366A (en) * 2006-03-30 2009-04-15 旭化成制药株式会社 Substituted bicyclic derivatives and use thereof
WO2017073060A1 (en) * 2015-10-29 2017-05-04 国立大学法人東北大学 Collagen production inhibitor
CN112955431A (en) 2018-09-06 2021-06-11 艾尼纳制药公司 Compounds useful for the treatment of autoimmune and inflammatory disorders

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB970440A (en) * 1961-08-14 1964-09-23 Merck & Co Inc Anti-inflammatory compositions comprising steroids and indole derivatives
DE1816993A1 (en) * 1967-12-27 1969-10-16 Lodzkie Zaklady Farma Process for the preparation of 1-aoyl- or 1-heteroaroyl- or 1-benzheteroaroyl-2-methyl-5-alkoxy- (or 5-aralkoxy) -indolyl-3-acetic acids
DE2337340A1 (en) * 1972-07-24 1974-02-14 Hoffmann La Roche CARBAZOLE
US3974179A (en) * 1971-06-01 1976-08-10 American Home Products Corporation 1,3,4,9-Tetrahydropyrano[3,4-b]indole-1-acetamides and derivatives
US4041169A (en) * 1975-03-05 1977-08-09 Ayerst Mckenna And Harrison Ltd. Pharmaceutical method for using pyrano-and thiopyranoindole derivatives
EP0301813A1 (en) * 1987-07-31 1989-02-01 American Home Products Corporation Naphthalenepropionic acid derivatives
US4810699A (en) * 1987-02-20 1989-03-07 American Home Products Corporation Substituted 1,3,4,9-tetrahydropyrano[3,4,-b]indole-1-acetic acids, pharmaceutical compositions containing them, and methods for treating inflammatory conditions and for analgesic purposes using them

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB970440A (en) * 1961-08-14 1964-09-23 Merck & Co Inc Anti-inflammatory compositions comprising steroids and indole derivatives
DE1816993A1 (en) * 1967-12-27 1969-10-16 Lodzkie Zaklady Farma Process for the preparation of 1-aoyl- or 1-heteroaroyl- or 1-benzheteroaroyl-2-methyl-5-alkoxy- (or 5-aralkoxy) -indolyl-3-acetic acids
US3974179A (en) * 1971-06-01 1976-08-10 American Home Products Corporation 1,3,4,9-Tetrahydropyrano[3,4-b]indole-1-acetamides and derivatives
DE2337340A1 (en) * 1972-07-24 1974-02-14 Hoffmann La Roche CARBAZOLE
US4041169A (en) * 1975-03-05 1977-08-09 Ayerst Mckenna And Harrison Ltd. Pharmaceutical method for using pyrano-and thiopyranoindole derivatives
US4810699A (en) * 1987-02-20 1989-03-07 American Home Products Corporation Substituted 1,3,4,9-tetrahydropyrano[3,4,-b]indole-1-acetic acids, pharmaceutical compositions containing them, and methods for treating inflammatory conditions and for analgesic purposes using them
EP0301813A1 (en) * 1987-07-31 1989-02-01 American Home Products Corporation Naphthalenepropionic acid derivatives

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Chemical Abstracts, volume 98, 1983, (Columbus, Ohio, US) A. Roy et al.: "Further studies on antiinflammatory activity of two potent indan-1-acetic acids", see page 26 *

Cited By (101)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5221678A (en) * 1990-07-26 1993-06-22 Merck Frosst Canada, Inc. (quinolin-2-ylmethoxy)tetrahydrocarbazoles as inhibitors of the biosynthesis of leukotrienes
EP0468785A2 (en) * 1990-07-26 1992-01-29 Merck Frosst Canada Inc. (Quinolin-2-ylmethoxy)tetrahydrocarbazoles as inhibitors of the biosynthesis of leukotrienes
EP0468785A3 (en) * 1990-07-26 1992-03-04 Merck Frosst Canada Inc. (quinolin-2-ylmethoxy)tetrahydrocarbazoles as inhibitors of the biosynthesis of leukotrienes
EP0468789A3 (en) * 1990-07-26 1992-03-11 Merck Frosst Canada Inc. (quinolin-2-ylmethoxy)heterotetrahydrocarbazoles as inhibitors of the biosynthesis of leukotrienes
US5187180A (en) * 1990-07-26 1993-02-16 Merck Frosst Canada, Inc. (quinolin-2-ylmethoxy)heterotetrahydrocarbazoles as inhibitors of the biosynthesis of leukotrienes
EP0468789A2 (en) * 1990-07-26 1992-01-29 Merck Frosst Canada Inc. (Quinolin-2-ylmethoxy)heterotetrahydrocarbazoles as inhibitors of the biosynthesis of leukotrienes
EP0535926A1 (en) * 1991-09-30 1993-04-07 Merck Frosst Canada Inc. (Hetero-arylmethoxy) indoles as inhibitors of leukotriene biosynthesis
EP0535924A1 (en) * 1991-09-30 1993-04-07 Merck Frosst Canada Inc. (Bicyclic-azaarylmethoxy)indoles as inhibitors of leukotriene biosynthesis
EP0535923A1 (en) * 1991-09-30 1993-04-07 Merck Frosst Canada Inc. (Azaarylmethoxy)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
US5190968A (en) * 1991-09-30 1993-03-02 Merck Frosst Canada, Inc. (Polycyclic-arylmethoxy) indoles as inhibitors of leukotriene biosynthesis
US5410061A (en) * 1991-10-24 1995-04-25 Lilly Industries Limited Pharmaceutical compounds
WO1993016069A1 (en) * 1992-02-13 1993-08-19 Merck Frosst Canada Inc. (azaaromaticalkoxy)indoles as inhibitors of leukotriene biosynthesis
EP0574774A1 (en) * 1992-06-17 1993-12-22 Bayer Ag Substituted benzothiazolyl- and quinoxalyl methoxy-phenyl acetic acid derivatives
WO1994001407A2 (en) * 1992-07-10 1994-01-20 American Home Products Corporation Indole and indene derivatives as pla2 and lipoxygenase inhibitors
WO1994001407A3 (en) * 1992-07-10 1994-03-03 American Home Prod Indole and indene derivatives as pla2 and lipoxygenase inhibitors
WO1994022869A1 (en) * 1993-03-29 1994-10-13 Merck Frosst Canada Inc. FURO[3,2-b]PYRIDINES AND THIENO[3,2-b]PYRIDINES 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
US5496957A (en) * 1993-09-01 1996-03-05 Virginia Commonwealth University Tryptamine analogs with 5-HT1D selectivity
DE4338770A1 (en) * 1993-11-12 1995-05-18 Matthias Dr Lehr Indole-2-alkanoic acids and their derivatives as inhibitors of phospholipase A¶2¶
US6175021B1 (en) 1994-04-01 2001-01-16 Eli Lilly And Company 1H-indole-3-glyoxylamide sPLA2 inhibitors
EP1081135A3 (en) * 1994-04-01 2003-11-05 Eli Lilly And Company 1H-indole-3-glyoxylamide spla2 inhibitors
US6433001B1 (en) 1994-04-01 2002-08-13 Eli Lilly And Company 1H-indole-3-glyoxylamide sPLA2 inhibitors
US5654326A (en) * 1994-04-01 1997-08-05 Eli Lilly And Company 1H-indole-3-glyoxylamide SPLA2 inhibitors
EP0675110A1 (en) * 1994-04-01 1995-10-04 Eli Lilly And Company 1H-Indole-3-glyoxylamide sPLA2 inhibitors
US5733923A (en) * 1994-04-01 1998-03-31 Eli Lilly And Company 1H-indole-3-glyoxylamide sPLA2 inhibitors
US5919943A (en) * 1994-04-01 1999-07-06 Eli Lilly And Company Process for preparing 1H-indole-3-glyoxylamides
US5919810A (en) * 1994-04-01 1999-07-06 Eli Lilly And Company 1H-indole-3-glyoxylamide sPLA2 inhibitors
EP1950200A1 (en) * 1994-04-01 2008-07-30 Eli Lilly And Company [[3-(2-amino-1,2-dioxoethyl)-2-ethyl-1-(phenylmethyl)-11H-indol-4-yl]oxy]acetic acid methyl ester as sPLA2 inhibitor
CN1067054C (en) * 1994-04-01 2001-06-13 伊莱利利公司 1H-indole-3-glyoxylamide spla2 inhibitors
EP1081135A2 (en) * 1994-04-01 2001-03-07 Eli Lilly And Company 1H-indole-3-glyoxylamide spla2 inhibitors
EP2341045A1 (en) * 1994-04-01 2011-07-06 Eli Lilly and Company [[3-(2-amino-1,2-dioxoethyl)-2-ethyl-1-(phenylmethyl)-11H-indol-4-yl]oxy]acetic acid (acyloxy)alkyl or ((alkoxycarbonyl)oxy)alkyl ester as SPLA2 inhibitor
US5504101A (en) * 1994-05-06 1996-04-02 Allelix Biopharmaceuticals, Inc. 5-HT-1D receptor ligands
EP0690050A1 (en) * 1994-06-28 1996-01-03 Adir Et Compagnie (Thia)cycloalkyl(b) indole derivatives as 5-lipoxygenase inhibitors
CN1054374C (en) * 1994-06-28 2000-07-12 阿迪尔公司 New (thia)cycloalkyl[b]indole compounds, process for the preparation thereof, and pharmaceutical compositions containing them
FR2721610A1 (en) * 1994-06-28 1995-12-29 Adir Novel (thia) cycloalkyl [b] indole derivatives, process for their preparation and pharmaceutical compositions containing them
US6121321A (en) * 1996-06-13 2000-09-19 Cell Pathways, Inc. Substituted methoxy benzylidene indenyl acetic and propionic acids for treating patients with precancerous lesions
US6063818A (en) * 1996-06-13 2000-05-16 Cell Pathways Inc. Substituted benzylidene indenyl formamides, acetamides and propionamides
US5998477A (en) * 1996-06-13 1999-12-07 Cell Pathways Inc. Substituted methoxy benzylidene indenyl-acetic and propionic acids for treating patients with precancerous lesions
US5965619A (en) * 1996-06-13 1999-10-12 Cell Pathways Inc. Method for treating patients having precancerous lesions with substituted indene derivatives
WO1998005637A1 (en) * 1996-08-01 1998-02-12 Merckle Gmbh Acylpyrroldicarboxylic acids and acylindoldicarboxylic acids and their derivatives and inhibitors of the cytosolic phospholipase a¿2?
US5948779A (en) * 1997-12-12 1999-09-07 Cell Pathways, Inc. Substituted condensation products of n-benzyl-3-indenyl acetamides with heterocyclic aldehydes
US6500853B1 (en) 1998-02-28 2002-12-31 Genetics Institute, Llc Inhibitors of phospholipase enzymes
US6028116A (en) * 1998-04-03 2000-02-22 Cell Pathways, Inc. Substituted condensation products of 1H-indenyl-hydroxyalkanes with aldehydes for neoplasia
US6706752B1 (en) 1999-07-19 2004-03-16 Eli Lilly And Company sPLA2 inhibitors
WO2001005761A1 (en) * 1999-07-19 2001-01-25 Eli Lilly And Company sPLA2 INHIBITORS
US6635670B2 (en) 1999-07-19 2003-10-21 Eli Lilly And Company sPLA2 inhibitors
WO2001066110A3 (en) * 2000-03-09 2002-04-25 William Louis Macias METHOD FOR THE TREATMENT OF RENAL DYSFUNCTION WITH sPLA2 INHIBITORS
WO2001066110A2 (en) * 2000-03-09 2001-09-13 Eli Lilly And Company METHOD FOR THE TREATMENT OF RENAL DYSFUNCTION WITH sPLA2 INHIBITORS
WO2002020478A1 (en) * 2000-08-24 2002-03-14 Dublin City University N-benzylindole-3-acetic acid derivatives for use in the treatment of drug resistan cancer
US6933313B2 (en) 2001-03-28 2005-08-23 Eli Lilly And Company Substituted carbazoles as inhibitors of sPLA2
WO2002079154A1 (en) * 2001-03-28 2002-10-10 Eli Lilly And Company Substituted carbazoles as inhibitors of spla2
US6797708B2 (en) 2001-12-03 2004-09-28 Wyeth Inhibitors of cytosolic phospholipase A2
US7906548B2 (en) 2001-12-03 2011-03-15 Wyeth Llc Methods for the use of inhibitors of cytosolic phospholipase A2
US7101875B2 (en) 2001-12-03 2006-09-05 Wyeth Methods for treating arthritic disorders
US7605156B2 (en) 2001-12-03 2009-10-20 Wyeth Methods for the use of inhibitors of cytosolic phospholipase A2
US6984735B2 (en) 2001-12-03 2006-01-10 Wyeth Process for making an aldehyde
US7713964B2 (en) 2001-12-03 2010-05-11 Wyeth Llc Methods for treating asthmatic conditions
US6635771B2 (en) 2001-12-03 2003-10-21 Wyeth N-benzhydryl indole compounds
GB2407318A (en) * 2003-10-23 2005-04-27 Oxagen Ltd Substituted Indol-3-yl acetic acid derivatives
US7579002B2 (en) * 2003-12-05 2009-08-25 Wisconsin Alumni Research Foundation Method for improving body weight uniformity and increasing carcass yield in animals
US7557135B2 (en) 2005-05-27 2009-07-07 Wyeth Inhibitors of cytosolic phospholipase A2
US8283373B2 (en) 2005-05-27 2012-10-09 Pfizer Inc. Inhibitors of cytosolic phospholipase A2
US8841295B2 (en) 2005-11-04 2014-09-23 Panmira Pharmaceuticals, Llc 5-lipoxygenase-activating protein (FLAP) inhibitors
US7834037B2 (en) 2005-11-04 2010-11-16 Amira Pharmaceuticals, Inc. 5-lipoxygenase-activating protein (FLAP) inhibitors
US7977359B2 (en) 2005-11-04 2011-07-12 Amira Pharmaceuticals, Inc. 5-lipdxygenase-activating protein (FLAP) inhibitors
US8710081B2 (en) 2005-11-04 2014-04-29 Panmira Pharmaceuticals, Llc 5-lipoxygenase-activating protein (FLAP) inhibitors
GB2431927B (en) * 2005-11-04 2010-03-17 Amira Pharmaceuticals Inc 5-Lipoxygenase-activating protein (FLAP) inhibitors
EA019252B1 (en) * 2008-07-23 2014-02-28 Арена Фармасьютикалз, Инк. SUBSTITUTED (1,2,3,4-TETRAHYDROCYCLOPENTA[b]INDOL-3-YL)ACETIC ACID DERIVATIVES USEFUL IN THE TREATMENT OF AUTOIMMUNE AND INFLAMMATORY DISORDERS
US9522133B2 (en) 2008-07-23 2016-12-20 Arena Pharmaceuticals, Inc. Substituted 1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid derivatives useful in the treatment of autoimmune and inflammatory disorders
JP2011529049A (en) * 2008-07-23 2011-12-01 アリーナ ファーマシューティカルズ, インコーポレイテッド Substituted 1,2,3,4-tetrahydrocyclopenta [b] indol-3-yl) acetic acid derivatives useful in the treatment of autoimmune and inflammatory disorders
JP2014144948A (en) * 2008-07-23 2014-08-14 Arena Pharmaceuticals Inc SUBSTITUTED 1,2,3,4-TETRAHYDROCYCLOPENTA[b]INDOL-3-YL)ACETIC ACID DERIVATIVES USEFUL IN TREATMENT OF AUTOIMMUNE DISORDERS AND INFLAMMATORY DISORDERS
WO2010011316A1 (en) * 2008-07-23 2010-01-28 Arena Pharmaceuticals, Inc. SUBSTITUTED 1,2,3,4- TETRAHYDROCYCLOPENTA[b]INDOL-3-YL) ACETIC ACID DERIVATIVES USEFUL IN THE TREATMENT OF AUTOIMMUNE AND INFLAMMATORY DISORDERS
CN104311472B9 (en) * 2008-07-23 2020-03-17 艾尼纳制药公司 Substituted 1,2,3, 4-tetrahydrocyclopenta [ b ] indol-3-yl acetic acid derivatives
CN104311472A (en) * 2008-07-23 2015-01-28 艾尼纳制药公司 Substituted 1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid derivatives
CN104311472B (en) * 2008-07-23 2019-12-31 艾尼纳制药公司 Substituted 1,2,3, 4-tetrahydrocyclopenta [ b ] indol-3-yl acetic acid derivatives
US9126932B2 (en) 2008-07-23 2015-09-08 Arena Pharmaceuticals, Inc. Substituted 1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid derivatives useful in the treatment of autoimmune and inflammatory disorders
US8580841B2 (en) 2008-07-23 2013-11-12 Arena Pharmaceuticals, Inc. Substituted 1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid derivatives useful in the treatment of autoimmune and inflammatory disorders
JP2016029067A (en) * 2008-07-23 2016-03-03 アリーナ ファーマシューティカルズ, インコーポレイテッド Substituted 1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid derivatives useful in treatment of autoimmune and inflammatory disorders
US8415484B2 (en) 2008-08-27 2013-04-09 Arena Pharmaceuticals, Inc. Substituted tricyclic acid derivatives as S1P1 receptor agonists useful in the treatment of autoimmune and inflammatory disorders
US9108969B2 (en) 2008-08-27 2015-08-18 Arena Pharmaceuticals, Inc. Substituted tricyclic acid derivatives as S1P1 receptor agonists useful in the treatment of autoimmune and inflammatory disorders
US8853419B2 (en) 2010-01-27 2014-10-07 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and salts thereof
US9447041B2 (en) 2010-01-27 2016-09-20 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[B]indol-3-yl)acetic acid and salts thereof
US9175320B2 (en) 2010-01-27 2015-11-03 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[B]indol-3-yl)acetic acid and salts thereof
US11149292B2 (en) 2010-01-27 2021-10-19 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[B]indol-3-yl)acetic acid and salts thereof
US11674163B2 (en) 2010-01-27 2023-06-13 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and salts thereof
US9085581B2 (en) 2010-03-03 2015-07-21 Arena Pharmaceuticals, Inc. Processes for the preparation of S1P1 receptor modulators and crystalline forms thereof
US11896578B2 (en) 2015-01-06 2024-02-13 Arena Pharmaceuticals, Inc. Methods of treating conditions related to the S1P1 receptor
US11007175B2 (en) 2015-01-06 2021-05-18 Arena Pharmaceuticals, Inc. Methods of treating conditions related to the S1P1 receptor
US10301262B2 (en) 2015-06-22 2019-05-28 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compund1) for use in SIPI receptor-associated disorders
US11091435B2 (en) 2015-06-22 2021-08-17 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3, 4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(compound1) for use in S1P1 receptor-associated disorders
US11884626B2 (en) 2015-06-22 2024-01-30 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compound1) for use in S1P1 receptor-associated disorders
US10676435B2 (en) 2015-06-22 2020-06-09 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compound 1) for use in SIPI receptor-associated disorders
US11478448B2 (en) 2017-02-16 2022-10-25 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of inflammatory bowel disease with extra-intestinal manifestations
US11534424B2 (en) 2017-02-16 2022-12-27 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of primary biliary cholangitis
US11833156B2 (en) 2017-09-22 2023-12-05 Jubilant Epipad LLC Heterocyclic compounds as pad inhibitors
US11426412B2 (en) 2017-10-18 2022-08-30 Jubilant Epipad LLC Imidazo-pyridine compounds as PAD inhibitors
US11629135B2 (en) 2017-11-06 2023-04-18 Jubilant Prodell Llc Pyrimidine derivatives as inhibitors of PD1/PD-L1 activation
US11459338B2 (en) 2017-11-24 2022-10-04 Jubilant Episcribe Llc Heterocyclic compounds as PRMT5 inhibitors
US11529341B2 (en) 2018-03-13 2022-12-20 Jubilant Prodel LLC Bicyclic compounds as inhibitors of PD1/PD-L1 interaction/activation

Also Published As

Publication number Publication date
JPH05502222A (en) 1993-04-22
BR9007790A (en) 1992-09-15
AU7740491A (en) 1991-05-31
CA2070422A1 (en) 1991-04-28
HUT63407A (en) 1993-08-30
IE903872A1 (en) 1991-05-22
KR920702345A (en) 1992-09-03
AU643996B2 (en) 1993-12-02
FI921865A (en) 1992-04-24
PT95692A (en) 1991-09-13
EP0502106A1 (en) 1992-09-09
WO1991006537A3 (en) 1991-10-17
FI921865A0 (en) 1992-04-24

Similar Documents

Publication Publication Date Title
AU643996B2 (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
US5420289A (en) Substituted indole-, indene-, pyranoindole- and tetrahydrocarbazole-alkanoic acid derivatives as inhibitors of PLA2 and lipoxygenase
EP0275667B1 (en) 3-hetero-substituted-n-benzyl-indoles
DK168534B1 (en) Quinoline derivative and pharmaceutical composition containing such derivative
US4851409A (en) 2-substituted quinoline dioic acids and pharmaceutical compositions
IE913128A1 (en) Naphthalenepropionic acid derivatives as¹anti-inflammatory/antiallergic agents
EP0318084A2 (en) Benzoheterazoles
US5218124A (en) Substituted benzoylbenzene-, biphenyl- and 2-oxazole-alkanoic acid derivatives as inhibitors of pla2 and lipoxygenase
US5021576A (en) 2-Anilino phenylacetic acid derivatives
AU647425B2 (en) Substituted benzoylbenzene-, biphenyl- and 2-oxazole- alkanoic acid derivatives as inhibitors of PLA2 and lipoxygenase
EP0301813B1 (en) Naphthalenepropionic acid derivatives
Musser et al. Substituted indole-, indene-, pyranoindole-, and tetrahydrocarbazole-alkanoic acid derivatives as inhibitors of PLA 2 and lipoxygenase
US5159085A (en) 2-anilino phenylacetic acid derivatives as inhibitors of PLA2 and lipoxygenase
US5364944A (en) Substituted benzoylbenzene-, biphenyl- and 2-oxazole- alkanoic acid compounds
RU2037481C1 (en) Derivatives of substituted benzoylbenzene-, biphenyl or 2-oxazolealkanoic acid or their pharmacologically acceptable salts, and a method of their synthesis
US5428171A (en) 2-substituted quinoline dioic acids
FI92827C (en) Process for the preparation of therapeutically useful 1-heteroaryl-3-acyl-2-oxindole derivatives
CA2090042A1 (en) Substituted indole-, indene-, pyranoindole- and tetrahydrocarbazole- alkanoic acid derivatives as inhibitors of pla2 and lipoxygenase

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AU BR CA FI HU JP KR SU

AL Designated countries for regional patents

Kind code of ref document: A2

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

AK Designated states

Kind code of ref document: A3

Designated state(s): AU BR CA FI HU JP KR SU

AL Designated countries for regional patents

Kind code of ref document: A3

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

WWE Wipo information: entry into national phase

Ref document number: 2070422

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 921865

Country of ref document: FI

WWE Wipo information: entry into national phase

Ref document number: 1991900547

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1991900547

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1991900547

Country of ref document: EP