CA2294601A1 - Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities - Google Patents

Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities Download PDF

Info

Publication number
CA2294601A1
CA2294601A1 CA002294601A CA2294601A CA2294601A1 CA 2294601 A1 CA2294601 A1 CA 2294601A1 CA 002294601 A CA002294601 A CA 002294601A CA 2294601 A CA2294601 A CA 2294601A CA 2294601 A1 CA2294601 A1 CA 2294601A1
Authority
CA
Canada
Prior art keywords
compound
mmol
formula
compounds
solution
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002294601A
Other languages
French (fr)
Inventor
Elliott S. Klein
Alan T. Johnson
Andrew M. Standeven
Richard L. Beard
Samuel J. Gillett
Tien T. Duong
Sunil Nagpal
Vidyasagar Vuligonda
Min Teng
Roshantha A. Chandraratna
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Allergan Inc
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of CA2294601A1 publication Critical patent/CA2294601A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/02Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C57/00Unsaturated compounds having carboxyl groups bound to acyclic carbon atoms
    • C07C57/46Unsaturated compounds having carboxyl groups bound to acyclic carbon atoms containing six-membered aromatic rings and other rings, e.g. cyclohexylphenylacetic acid
    • C07C57/50Unsaturated compounds having carboxyl groups bound to acyclic carbon atoms containing six-membered aromatic rings and other rings, e.g. cyclohexylphenylacetic acid containing condensed ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/16Emollients or protectives, e.g. against radiation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • 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
    • C07C233/00Carboxylic acid amides
    • C07C233/64Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings
    • C07C233/81Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/64Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings
    • C07C233/81Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups
    • C07C233/82Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom
    • C07C233/87Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by carboxyl groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by an acyclic carbon atom of a carbon skeleton containing six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C245/00Compounds containing chains of at least two nitrogen atoms with at least one nitrogen-to-nitrogen multiple bond
    • C07C245/02Azo compounds, i.e. compounds having the free valencies of —N=N— groups attached to different atoms, e.g. diazohydroxides
    • C07C245/06Azo compounds, i.e. compounds having the free valencies of —N=N— groups attached to different atoms, e.g. diazohydroxides with nitrogen atoms of azo groups bound to carbon atoms of six-membered aromatic rings
    • C07C245/10Azo compounds, i.e. compounds having the free valencies of —N=N— groups attached to different atoms, e.g. diazohydroxides with nitrogen atoms of azo groups bound to carbon atoms of six-membered aromatic rings with nitrogen atoms of azo groups bound to carbon atoms of six-membered aromatic rings being part of condensed ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C327/00Thiocarboxylic acids
    • C07C327/38Amides of thiocarboxylic acids
    • C07C327/48Amides of thiocarboxylic acids having carbon atoms of thiocarboxamide groups bound to carbon atoms of six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C63/00Compounds having carboxyl groups bound to a carbon atoms of six-membered aromatic rings
    • C07C63/33Polycyclic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C63/00Compounds having carboxyl groups bound to a carbon atoms of six-membered aromatic rings
    • C07C63/33Polycyclic acids
    • C07C63/49Polycyclic acids containing rings other than six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C63/00Compounds having carboxyl groups bound to a carbon atoms of six-membered aromatic rings
    • C07C63/66Polycyclic acids with unsaturation outside the aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C63/00Compounds having carboxyl groups bound to a carbon atoms of six-membered aromatic rings
    • C07C63/68Compounds having carboxyl groups bound to a carbon atoms of six-membered aromatic rings containing halogen
    • C07C63/72Polycyclic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C63/00Compounds having carboxyl groups bound to a carbon atoms of six-membered aromatic rings
    • C07C63/68Compounds having carboxyl groups bound to a carbon atoms of six-membered aromatic rings containing halogen
    • C07C63/74Compounds having carboxyl groups bound to a carbon atoms of six-membered aromatic rings containing halogen having unsaturation outside the aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C65/00Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups
    • C07C65/01Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups containing hydroxy or O-metal groups
    • C07C65/19Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups containing hydroxy or O-metal groups having unsaturation outside the aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C65/00Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups
    • C07C65/21Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups containing ether groups, groups, groups, or groups
    • C07C65/28Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups containing ether groups, groups, groups, or groups having unsaturation outside the aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C65/00Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups
    • C07C65/32Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups containing keto groups
    • C07C65/38Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups containing keto groups having unsaturation outside the aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/612Esters of carboxylic acids having a carboxyl group bound to an acyclic carbon atom and having a six-membered aromatic ring in the acid moiety
    • C07C69/618Esters of carboxylic acids having a carboxyl group bound to an acyclic carbon atom and having a six-membered aromatic ring in the acid moiety having unsaturation outside the six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/76Esters of carboxylic acids having a carboxyl group bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/76Esters of carboxylic acids having a carboxyl group bound to a carbon atom of a six-membered aromatic ring
    • C07C69/84Esters of carboxylic acids having a carboxyl group bound to a carbon atom of a six-membered aromatic ring of monocyclic hydroxy carboxylic acids, the hydroxy groups and the carboxyl groups of which are bound to carbon atoms of a six-membered aromatic ring
    • C07C69/90Esters of carboxylic acids having a carboxyl group bound to a carbon atom of a six-membered aromatic ring of monocyclic hydroxy carboxylic acids, the hydroxy groups and the carboxyl groups of which are bound to carbon atoms of a six-membered aromatic ring with esterified hydroxyl and carboxyl groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/76Esters of carboxylic acids having a carboxyl group bound to a carbon atom of a six-membered aromatic ring
    • C07C69/94Esters of carboxylic acids having a carboxyl group bound to a carbon atom of a six-membered aromatic ring of polycyclic hydroxy carboxylic acids, the hydroxy groups and the carboxyl groups of which are bound to carbon atoms of six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/54Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/55Acids; Esters
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/22Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • C07D277/30Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/38Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D307/54Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/58Benzo[b]pyrans, not hydrogenated in the carbocyclic ring other than with oxygen or sulphur atoms in position 2 or 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/06Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to the ring carbon atoms
    • C07D333/24Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D335/00Heterocyclic compounds containing six-membered rings having one sulfur atom as the only ring hetero atom
    • C07D335/04Heterocyclic compounds containing six-membered rings having one sulfur atom as the only ring hetero atom condensed with carbocyclic rings or ring systems
    • C07D335/06Benzothiopyrans; Hydrogenated benzothiopyrans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic System
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/0803Compounds with Si-C or Si-Si linkages
    • C07F7/081Compounds with Si-C or Si-Si linkages comprising at least one atom selected from the elements N, O, halogen, S, Se or Te
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2602/00Systems containing two condensed rings
    • C07C2602/02Systems containing two condensed rings the rings having only two atoms in common
    • C07C2602/04One of the condensed rings being a six-membered aromatic ring
    • C07C2602/10One of the condensed rings being a six-membered aromatic ring the other ring being six-membered, e.g. tetraline
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2602/00Systems containing two condensed rings
    • C07C2602/02Systems containing two condensed rings the rings having only two atoms in common
    • C07C2602/14All rings being cycloaliphatic
    • C07C2602/26All rings being cycloaliphatic the ring system containing ten carbon atoms
    • C07C2602/28Hydrogenated naphthalenes

Abstract

Aryl-substituted and aryl and (3-oxo-1-propenyl)-substituted benzopyran, benzothiopyran, 1,2-dihydroquinoline, and 5,6-dihydronaphthalene derivatives have retinoid negative hormone and/or antagonist-like biological activities. The invented RAR antagonists can be administered to mammals, including humans, for the purpose of preventing or diminishing action of RAR agonists on the bound receptor sites. Specifically, the RAR agonists are administered or coadministered with retinoid drugs to prevent or ameliorate toxicity or side effects caused by retinoids or vitamin A or vitamin A precursors. The retinoid negative hormones can be used to potentiate the activities of other retinoids and nuclear receptor agonists. For example, the retinoid negative hormone called AGN 193109 effectively increased the effectiveness of other retinoids and steroid hormones in in vitro transactivation assays. Additionally, transactivation assays can be used to identify compounds having negative hormone activity. These assays are based on the ability of negative hormones to down-regulate the activity of chimeric retinoid receptors engineered to possess a constitutive transcription activator domain.

Description

. CA 02294601 1999-12-16 REPLACEi~IENT SHEET 7 1 SYNTHESIS AND USE O~' COt'rlPt)UNDS 3~AVING iIIEGATI~
2 RETINOID HORt'~IONE AND/OR RETINOID AN'f:~GOI~1IST
3 ~CT3YiTY
Related .Applications This application is a continuation-in-part of application serial number 6 08/613,863, filed on March 11, 1996 which claims the benefit of priority under 7 35 U.S.C. ~ 119(e) of the three following U.S. applications, each of which was 8 filed as a nonprovisional application and converted to a provisional application 9 by separate petitions filed on January 31, 1996: Application No. 08/522,778, filed September 1, 1995, which became provisional application serial number 11 60/019,015; Application No. 08/522,779, filed September 1, 1995 which 12 became provisional application serial number 60/064,853; and Application No.
13 08/542,648, filed October l3, 1995 which became provisional application 14 serial number 60/020,501. The complete disclosures of these related applications is hereby incorporated herein by this reference thereto.
16 Field of the Invention 17 The present invention relates to novel compounds having retinoid 18 negative hormone and/or retinoid antagonist-like biological activities.
More 19 specifically, the invention relates to 4-aryl substituted benzopyran, 4-aryl substituted benzothiopyran, 4-aryl substituted 1,2-dihydroquinoline and 8-aryl 21 substituted 5,6-dihydronaphthalene derivatives which may also be substituted 22 by a substituted 3-oxo-1-propenyl group. These novel compounds have 23 retinoid antagonist like-activity and are useful for treating or preventing 24 retinoid and vitamin A and vitamin A precursor induced toxicity in mammals and as an adjunct to treatment of mammals with retinoids to prevent or 26 ameliorate unwanted or undesired side effects. The invention further relates to 27 the use of retinoid negative hormones for increasing the biological activities of 28 other retinoids and steroid hormones and inhibiting the basal activity of n,~acN~7Ei) SH$T
....~~....-..~.. .__...~.....-.~p_-_.. . .__,.._.u~-...~.~__. .._._._...-...w.-.~.......~..~... .~._._.~

1 unliganded retinoic acid receptors.
2 Background of the Invention 3 Compounds which have retinoid-like activity are well known in 4 the art, and are described in numerous United States and other patents and in scientific publications. It is generally known and accepted in the 6 art that retinoid-like activity is useful for treating mammals, including 7 humans, in order to cure or alleviate the symptoms associated with 8 numerous diseases and conditions.
9 Retinoids (vitamin A and its derivatives) are known to have broad l0 activities, including effects on cell proliferation and differentiation, in a 11 variety of biological systems. This activity has made retinoids useful in 12 the treatment of a variety of diseases, including dermatological disorders 13 and cancers. The prior art has developed a large number of chemical 14 compounds which have retinoid-like biological activity, and voluminous patent and chemical literature exists describing such compounds. The 16 relevant patent literature includes United States Patent Nos. 4,980,369, 17 5,006,550, 5,015,658, 5,045,551, 5,089,509, 5,134,159, 5,162,546, 18 5,234,926, 5,248,777, 5,264,578, 5,272,156, 5,278,318, 5,324,744, 19 5,346,895, 5,346,915, 5,348,972, 5,348,975, 5,380,877, 5,399,561, 5,407,937, (assigned to the same assignee as the present application) and 21 patents and publications cited therein, which particularly describe or 22 relate to chroman, thiochroman and 1,2,3,4-tetrahydroquinoline 23 derivatives which have retinoid-like biological activity. In addition, 24 several applications are pending which are assigned to the assignee of the present application, and which are directed to further compounds 26 having retinoid-like activity.
27 United States Patent Nos. 4,740,519 (Shroot et al.), 4,826,969 28 (Maignan et al.), 4,326,055 (Loeliger et al.), 5,130,335 (Chandraratna et 29 al.), 5,037,825 (Klaus et al.), 5,231,113 {Chandraratna et al.), 5,324,840 (Chandraratna), 5,344,959 (Chandraratna), 5,130,335 (Chandraratna et 1 al.), Published European Patent Application Nos. 0 176 034 A (bluest 2 et al.), 0 350 846 A (Klaus et al.), 0 176 032 A (Frickel et al.), 0 176 033 3 A (Frickel et al.), 0 253 302 A (Klaus et al.), 0 303 915 A (Bryce et al.), 4 UK Patent Application GB 2190378 A (Klaus et al.), German Patent Application Nos. DE 3715955 Al (Klaus et al.), DE 3602473 A1 (bluest 6 et al., and the articles J. Amer. Acad. Derm. 15: 756 - 764 (1986) (Sporn 7 et al.), Chem. Pharm. Bull. 33: 404-407 (1985) (Shudo et al.), J. Med 8 Chem. 31: 2182 - 2192 (1988) (Kagechika et al.), Chemistry and Biology 9 of S n~hetic Retinoids CRC Press Inc. 1990 pp. 334 - 335, 354 (Dawson l0 et al.), describe or relate to compounds which include a 11 tetrahydronaphthyl moiety and have retinoid-like or related biological 12 activity. United States Patent No. 4,391,731 (Boller et al.) describes 13 tetrahydronaphthalene derivatives which are useful in liquid crystal 14 compositions.
An article by Kagechika et al. in J. Med. Chem 32:834 ( 1989) 16 describes certain 6-(3-oxo-1-propenyl)- 1,2,3,4-tetramethyl- 1,2,3,4-17 tetrahydronaphthalene derivatives and related flavone compounds 18 having retinoid-like activity. The articles by Shudo et al. in Chem.
19 Pharm. Bull. 33:404 ( 1985) and by Jetten et al. in Cancer Research 47:3523 (1987) describe or relate to further 3-oxo-1-propenyl derivatives 21 (chalcone compounds) and their retinoid-like or related biological 22 activity.
23 Unfortunately, compounds having retinoid-like activity (retinoids) 24 also cause a number of undesired side effects at therapeutic dose levels, including headache, teratogenesis, mucocutaneous toxicity, 26 musculoskeletal toxicity, dyslipidemias, skin irritation, headache and 27 hepatotoxicity. These side effects limit the acceptability and utility of 28 retinoids for treating disease.
29 It is now general knowledge in the art that two main types of retinoid receptors exist in mammals (and other organisms). The two 1 main types or families of receptors are respectively designated as the 2 RARs and RXRs. Within each type there are subtypes: in the RAR
3 family the subtypes are designated RAR-a, RAR-~3 and RAR-y, in 4 RXR the subtypes are: RXR-a, RXB-~3 and RXR-~y . Both families of receptors are transcription factors that can be distinguished from each 6 other based on their ligand binding specificities. All-trans-RA (ATRA) 7 binds and activates a class of retinoic acid receptors (RARs) that 8 includes RAR-a, RAR-~3 and RAR-~y . A different ligand, 9-cis-RA (9C
9 RA), binds and activates both the RARs and members of the retinoid X
receptor (RXR) family.
11 It has also been established in the art that the distribution of the 12 two main retinoid receptor types, and of the several subtypes is not 13 uniform in the various tissues and organs of mammalian organisms.
14 Moreover, it is generally accepted in the art that many unwanted side effects of retinoids are mediated by one or more of the RAR receptor 16 subtypes. Accordingly, among compounds having agonist-like activity at 17 retinoid receptors, specificity or selectivity for one of the main types or 18 families, and even specificity or selectivity for one or more subtypes 19 within a family of receptors, is considered a desirable pharmacological property.
21 Relatively recently compounds have been developed in the art 22 which bind to RAR receptors without triggering the response or 23 responses that are triggered by agonists of the same receptors. The 24 compounds or agents which bind to RAR receptors withc~ut triggering a "retinoid" response are thus capable of blocking (to les~:_ : or greater 26 ex~.ent) the activity of RAR agonists in biological assays and systems.
27 More particularly, regarding the scientific and patent literature in this 28 field, published PCT Application WO 94/14777 describes certain 29 heterocyclic carboxylic acid derivatives which bind to RAR retinoid receptors and are said in the application to be useful for treatment of 1 certain diseases or conditions, such as acne, psoriasis, rheumatoid 2 arthritis and viral infections. A similar disclosure is made in the article 3 by Yoshimura et aI. JMed. Chem. 38: 3163-3173 (1995). Kaneko et al.
4 Med. Chem Res. 1:220-225 (1991); Apfel et al. Proc. Natl. Acad. Sci.
5 USA 89: 7129-7133 Augusty 1992 Cell Biology; Eckhardt et al. Toxicology 6 Letters 70:299-308 {1994); Keidel et ai. Molecular and Cellular Biology 7 14:287-298 (1994); and Eyrolles et al. J. Med. Chem. 37: 1508-1517 8 (1994) describe compounds which have antagonist like activity at one or 9 more of the RAR retinoid subtypes.
l0 In addition to undesirable side-effects of therapy with retinoid 11 compounds, there occurs occasionally a serious medical condition 12 caused by vitamin A or vitamin A precursor overdose, resulting either 13 from the excessive intake of vitamin supplements or the ingestion of 14 liver of certain fish and animals that contain high levels of the vitamin.
The chronic or acute toxicities observed with hypervitaminosis A
16 syndrome include headache, skin peeling, bone toxicity, dyslipidemias, 17 etc. In recent years, it has become apparent that the toxicities observed 18 with vitamin A analogs, i.e., retinoids, essentially recapitulate those of 19 hypervitaminosis A syndrome, suggesting a common biological cause, i.e., RAR activation. These toxicities are presently treated mainly by 21 supportive measures and by abstaining from further exposure to the 22 causative agent, whether it be liver, vitamin supplements, or retinoids.
23 While some of the toxicities resolve with time, others (e.g., premature 24 epiphyseal plate closure) are permanent.
Generally speaking, specific antidotes are the best treatment for 26 poisoning by pharmacological agents, but only about two dozen 27 chemicals or classes of chemicals out of thousands in existence have 28 specific known antidotes. A specific antidote would clearly be of value 29 in the treatment of hypervitaminosis A and retinoid toxicity. Indeed, as increasingly potent retinoids are used clinically, a specific antidote for .. .,..w.-.~...._,. -~. ~_..~...... ..r.w..._ _ . r.........~....W .,~ . . ...
... ...~-~...

1 retinoid poisoning could be life saving.
2 Summary of the Invention 3 The present invention covers compounds of Formula 1 R,.
6 / / Yt~

11 Formula 1 12 wherein X is S, O, NR' where R' is H or alkyl of I to 6 carbons, 13 or 14 X is (C(R,}z~n where R1 is independently H or alkyl of 1 to 6 carbons, and n is an integer between 0 and 2;
16 Rz is hydrogen, lower alkyl of 1 to 6 carbons, F, Cl, Br, I, CF3, 17 fluoro substituted alkyl of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 18 carbons, or alkylthio of 1 to 6 carbons;
19 R3 is hydrogen, lower alkyl of 1 to 6 carbons or F;
m is an integer having the value of 0 - 3;
21 o is an integer having the value of 0 - 3;
22 Z is -C--_C-, 23 -N = N-, 24 -N = CR,-, -CR1=N, 26 -(CR,=CR,)~>- where n' is an integer having the value 0 - 5, 27 -CO-NR,-, 28 -CS-NR,-, 29 -NR,-CO, -NR,-CS, 1 -COO-, 2 -OCO-;
3 -CSO-;
4 -OCS-;
Y is a phenyl or naphthyl group, or heteroaryl selected from a 6 group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, 7 pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrrazolyl, said phenyl and 8 heteroaryl groups being optionally substituted with one or two R2 9 groups, or l0 when Z is -(CR1=CR1)~: and n' is 3, 4 or 5 then Y represents a 11 direct valence bond between said (CR2=CR2)"~ group and B;
12 A is (CH2)q where q is 0-5, lower branched chain alkyl having 3-6 13 carbons, cycloalkyl having 3-6 carbons, alkenyl having 2-6 carbons and 1 14 or 2 double bonds, alkynyl having 2-6 carbons and 1 or 2 triple bonds;
B is hydrogen, COOH or a pharmaceutically acceptable salt 16 thereof, COORS, CONR9R,o, -CH20H, CHZOR", CH~OCOR", CHO, 17 CH{OR,2)2, CHOR,30, -CORD, CR,(OR,2)2, CR~OR,30, or tri-lower 18 alkylsilyl, where R~ is an alkyl, cycloalkyl or alkenyl group containing 1 19 to 5 carbons, R$ is an alkyl group of 1 to 10 carbons or trimethylsilylalkyl where the alkyl group has 1 to 10 carbons, or a 21 cycloalkyl group of 5 to 10 carbons, or R8 is phenyl or lower 22 alkylphenyl, R9 and R,o independently are hydrogen, an alkyl group of 1 23 to 10 carbons, or a cycloalkyl group of 5-10 carbons, or phenyl or lower 24 alkylphenyl, R" is lower alkyl, phenyl or lower alkylphenyl, R,2 is lower alkyl, and R13 is divalent alkyl radical of 2-S carbons, and 26 R,4 is (Rls)r-Phenyl, (Rls)anaphthyl, or (R,5)~ heteroaryl where 27 the heteroaryl group has 1 to 3 heteroatoms selected from the group 28 consisting of O, S and N, r is an integer having the values of 0 - 5, and 29 R,5 is independently H, F, Cl, Br, I, NOZ, N(R$)2, N(R$)CORB, NR$CON(R8)2, OH, OCORB, ORB, CN, an alkyl group having 1 to 10 ~..,.,.. _M..~~~.~_.~~.~,.,-~.".-~,.".~~.... T .. ...,.:..~.-.-...~w~-.._.-~...._.-.~."~~...~..

1 carbons, fluoro substituted alkyl group having 1 to 10 carbons, an 2 alkenyl group having 1 to IO carbons and 1 to 3 double bonds, alkynyl 3 group having 1 to 10 carbons and 1 to 3 triple bonds, or a trialkylsilyl or 4 trialkylsilyloxy group where the alkyl groups independently have 1 to 6 carbons.
6 The present invention further covers compounds of Formula 101 9 ~~e R" (~ln' ~Yc~1 ~°~~up~"
11 (~slc / ~ ' Formula 101 16 wherein X is S, O, NR' where R' is H or alkyl of 1 to 6 carbons, 17 or 18 X is (C(R1)2J~ where R, is independently H or alkyl of 1 to 6 19 carbons, and n is an integer between 0 and 2;
R2 is hydrogen, lower alkyl of 1 to 6 carbons, F, Cl, Br, I, CF3, 21 fluoro substituted alkyl of 1 to 6 carbons, OH, SH, aikoxy of 1 to 6 22 carbons, or alkylthio of 1 to 6 carbons;
23 R3 is hydrogen, lower alkyl of 1 to 6 carbons or F;
24 m is an integer having the value of 0 - 3;
o is an integer having the va!vT~9 of 0 - 3;
26 Y is a phenyl or naphthyl gr _ a or heteroaryi selected from a 27 group consisting of pyridyl, thienyi, ruryl, pyridazinyl, pyrimidinyl, 28 pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrrazolyl, said phenyl and 29 heteroaryl groups being optionally substituted with one or two R2 groups;

1 A is (CH2)q where q is 0-5, lower branched chain alkyl having 3-6 2 carbons, cycloalkyl having 3-6 carbons, alkenyl having 2-6 carbons and 1 3 or 2 double bonds, alkynyl having 2-6 carbons and 1 or 2 triple bonds;
4 B is hydrogen, COOH or a pharmaceutically acceptable salt thereof, COORS, CONR9R,o, -CH20H, CH20R", CH20COR", CHO, 6 CH(OR,2)2, CHOR,30, -CORD, CR.,(OR,2)2, CR,OR,30, or tri-lower 7 alkylsilyl, where R, is an alkyl, cycloalkyl or alkenyl group containing 1 8 to 5 carbons, R8 is an alkyl group of 1 to 10 carbons or 9 trimethylsilylalkyl where the alkyl group has 1 to 10 carbons, or a cycloalkyl group of S to 10 carbons, or R8 is phenyl or lower I 1 alkylphenyl, R9 and R,o independently are hydrogen, an alkyl group of 1 12 to 10 carbons, or a cycloalkyl group of 5-10 carbons, or phenyl or lower 13 alkylphenyl, R" is lower alkyl, phenyl or lower alkylphenyl, R,2 is lower 14 alkyl, and R,3 is divalent alkyl radical of 2-5 carbons, and R,4 is (R,5)T Phenyl, (Rls)r naphthyl, or (R,s)r- heteroaryl where 16 the heteroaryl group has 1 to 3 heteroatoms selected from the group 17 consisting of O, S and N, r is an integer having the values of 0 - 5, and 18 R,5 is independently H, F, Cl, Br, I, NO2, N(R8)Z, N(R8)CORB, 19 NRgCON(R$)2, OH, OCORB, ORB, CN, an alkyl group having 1 to 10 carbons, fluoro substituted alkyl group having 1 to 10 carbons, an 21 alkenyl group having 1 to 10 carbons and 1 to 3 double bonds, alkynyl 22 group having 1 to 10 carbons and 1 to 3 triple bonds, or a trialkylsilyl or 23 trialkylsilyloxy group where the alkyl groups independently have 1 to 6 24 carbons;
R,6 is H, lower alkyl of 1 to 6 carbons;
26 R" is H, lower alkyl of 1 to 6 carbons, OH or OCOR", and 27 p is zero or l, with the proviso that when p is 1 then there is no 28 R,~ substituent group, and m is an integer between 0 and 2.
29 The compounds of the present invention are useful for preventing certain undesired side effects of retinoids which are administered for the . _. . _. ..- ~...~ .. w-...-..~..~.-.... . _.. __.._---_._._. T ....T~ ~ .._ ..... . ~ m. .~.... .

I treatment or prevention of certain diseases or conditions. For this 2 purpose the compounds of the invention may be coadministered with 3 retinoids. The compounds of the present invention are also useful in 4 the treatment of acute or chronic toxicity resulting from overdose or 5 poisoning by retinoid drugs or Vitamin A.
6 The present invention additionally relates to the use of RAR
7 antagonists for blocking all or some RAR receptor sites in biological 8 systems, including mammals, to prevent or diminish action of RAR
9 agonists on said receptor sites. More particularly, the present invention l0 relates to the use of RAR antagonists for (a) the prevention and (b) the I 1 treatment of retinoid (including vitamin A or vitamin A precursor) 12 chronic or acute toxicity and side effects of retinoid therapy.
13 In one particular aspect of the present invention, there is 14 provided a method of treating a pathological condition in a mammal.
The conditions treated are associated with a retinoic acid receptor 16 activity. This method involves administering to the mammal a retinoid 17 antagonist or negative hormone capable of binding to one of the 18 following retinoic acid receptor subtypes: RARa, RARR and RAR,..
19 The antagonist or negative hormone is administered in an amount pharmaceutically effective to provide a therapeutic benefit against the 21 pathological condition in the mammal.
22 As an antidote to acute or chronic retinoid or vitamin A
23 poisoning the RAR antagonist can be administered to a mammal 24 enterally, i.e., intragastric intubation or food/water admixture, or parenterally, e.g., intraperitoneally, intramuscularly, subcutaneously, 26 topically, etc. The only requirement for the rout; of administration is 27 that it must allow delivery of the antagonist to the target tissue. The 28 RAR antagonist can be formulated by itself or in combination with 29 excipients. The RAR antagonist need not be in solution in the 3o formulation, e.g., in the case of enteral use.

1 As an adjunct to therapy with retinoids and in order to prevent 2 one or more side effects of the retinoid drug which is administered, the 3 RAR antagonist can similarly be administered enterally or parenterally.
4 The RAR antagonist and RAR agonist need not be administered by the same route of administration. The key is that sufficient quantities of 6 the RAR antagonist be present continuously in the tissue of interest 7 during exposure to the RAR agonist. For the prevention of retinoid 8 toxicity, it is best that the RAR antagonist be administered concurrently 9 or prior to treatment with the RAR agonist. In many situations the RAR antagonist will be administered by a different route than the 11 agonist. For example undesirable skin effects of an enterally 12 administered retinoid may be prevented or ameliorated by an RAR
13 antagonist which is administered topically.
14 Another aspect of the present invention is a method of identifying retinoid negative hormones. The method includes the following steps:
16 obtaining transfected cells containing a reporter gene transcriptionally 17 responsive to binding of a recombinant retinoid receptor, the 18 recombinant retinoid receptor having at least protein domains located 19 C-terminal to a DNA binding domain of an intact retinoid receptor, measuring a basal level of reporter gene expression in untreated 21 transfected cells, the untreated transfected cells being propagated in the 22 absence of an added retinoid, treating the transfected cells with a 23 retinoid compound to be tested for negative hormone activity, 24 measuring a level of reporter gene expression in treated cells, comparing the levels of reporter gene expression measured in treated 26 cells and untreated cells, and identifying as retinoid negative hormones 27 those retinoid compounds producing a lower level of reporter gene 28 expression in treated cells compared with the basal level of reporter 29 gene expression measured in untreated cells. In certain preferred embodiments of this method the intact receptor is an RAR-a, RAR-~3 ._..~._.._.__"~~,.~ a..~._._. T,.""w.~.~,.....~..~..,......._.~,_.~..~ ..

1 or RAR-y subtype. In other embodiments, the intact receptor is an 2 RXR-a, RXR-J3 or RXR-y subtype. The recombinant receptor can also 3 be either a recombinant RAR or RXR receptor. In some 4 embodiments, the recombinant receptor is a chimeric retinoid receptor having a constitutive transcription activator domain. Such a constitutive 6 transcription activator domain can comprise a plurality of amino acids 7 having a net negative charge or have an amino acid sequence of a viral 8 transcription activator domain, such as the herpes simplex virus VP-16 9 transcription activator domain. In embodiments in which the constitutive transcription activator domain has a net negative charge, the 11 retinoid receptor can be recombinant and have deleted therefrom a 12 DNA binding domain, such as a DNA binding domain specific for a cis-13 regulatory element other than a retinoic acid responsive element. These 14 elements include an estrogen responsive element. The transfected cell is preferably propagated in a growth medium substantially depleted of 16 endogenous retinoids, such as one that includes activated charcoal-17 extracted serum. In this method, the reporter gene can be the 18 luciferase gene, in which case, the measuring steps can involve 19 luminometry. The reporter gene can also be the (~-galactosidase gene, in which case the measuring steps would involve a /3-galactosidase assay.
21 The transfected cell can be a transfected mammalian cell, such as a 22 Green monkey cell or a human cell.
23 Another aspect of the present invention is a method of 24 potentiating a pharmacologic activity of a steroid superfamily receptor 2S agonist adminis~ °°d to a mamm This method involves 26 coadmini~4eriny the mammal c the steroid superfamily receptor 27 agonist a comp~~~~ion comprising a pharmaceutically effective dose of a 28 retinoid negative hormone to potentiate the pharmacologic activity of 29 the steroid superfamily receptor agonist. The pharmacologic activity is measurable in a reporter gene trans-activation assay in vitro, such as by 1 measuring anti-AP-1 activity. The pharmacologic activity to be 2 potentiated can be an antiproliferative activity, such as activity of the 3 type measurable in retinal pigment epithelium. The steroid superfamily 4 receptor agonist can be any of the following: a retinoid receptor agonist, a vitamin D receptor agonist, a glucocorticoid receptor agonist, 6 a thyroid hormone receptor agonist, a peroxisome proliferator-activated 7 receptor agonist or an estrogen receptor agonist. The retinoid receptor 8 agonist can be an RAR agonist, such as all-traps-retinoic acid or 13-cis 9 retinoic acid. The retinoid receptor agonist can also be an RXR
l0 agonist. A preferred vitamin D receptor agonist is 1,25-11 dihydroxyvitamin D3. A preferred glucocorticoid receptor agonist is 12 dexamethasone. A preferred thyroid hormone receptor agonist is 3,3',5-13 triiodothyronine. The retinoid negative hormone is an RAR-specific 14 retinoid negative hormone, which preferably has a dissociation constant less than or approximately equal to 30 nM. Example of the RAR-16 specific retinoid negative hormone include AGN 193109, AGN 193385, 17 AGN 193389 and AGN 193871. The composition comprising a 18 pharmaceutically effective dose of a retinoid negative hormone can be 19 coadministered at the same time as the steroid superfamily agonist and be combined prior to coadministration. These can also be 21 coadministered as separate compositions.
22 Brief Description of the Drawings 23 Figure 1 shows the chemical structure of AGN 193109.
24 Figures 2A - 2F are a series of line graphs showing that AGN
193109 inhibited ATRA-dependent transactivation at the RARs.
26 Figures 2A and 2B represent activity at the RAR-a receptor; Figures 2C
27 and 2D represent activity at the RAR-f3 receptor; Figures 2E and 2F
28 represent activity at the RAR-y receptor. In Figures 2A, 2C and ZE, 29 open squares represent retinoic acid treatment and filled circles represent AGN 193109 treatment. In Figures 2B, 2D and 2F the single 1 lines represent luciferase activity measured after treatment with 10-8 M
2 ATRA and variable concentrations of AGN 193109.
3 Figures 3A and 3B are line graphs representing luciferase activity 4 detected in CV-1 cells transfected with reporter plasmid ERE-tk-Luc and expression plasmid ER-RAR-a and stimulated with ATRA (Figure 6 3A) or AGN 193109 (Figure 3B) at various concentrations. Data points 7 represent the mean ~ SEM of three independent luciferase 8 determinations. The results of transfections carried out using different 9 amounts of co-transfected ER-RAR-a (0.05, 0.1 and 0.2 ~cg/well) are indicated in each figure.
11 Figures 4A and 4B are line graphs representing luciferase activity 12 in CV-1 cells transfected with reporter plasmid ERE-tk-Luc and 13 expression plasmid ER-RAR-~i and stimulated with ATRA (Figure 4A) 14 or AGN 193109 (Figure 4B) at various concentrations. Data points represent the mean ~ SEM of three independent luciferase 16 determinations. The results of transfections carried out using different 17 amounts of co-transfected ER-RAR-J3 {0.05, 0.1 and 0.2 ,ug/well) are 18 indicated in each figure.
19 Figures SA and SB are line graphs representing luciferase activity detected in CV-1 cells transfected with reporter plasmid ERE-tk-Luc 21 and expression plasmid ER-RAR-y and stimulated with ATRA (Figure 22 SA) or AGN 193109 (Figure SB) at various concentrations. Data points 23 represent the mean ~ SEM of three independent luciferase 24 determinations. The results of transfections carried out using different amounts of co-transfected ER-RAR-y (0.05, 0.1 and 0.2 ~cg/well) are 26 indicated in each ~ :.Jure.
27 Figure 6 shows ATRA and AGN 193109 dose responses of CV-1 28 cells cotransfected with the ERE-tk-Luc reporter plasmid and either the 29 ER-RXR-a chimeric receptor expression plasmid alone, or in combination with the RAR-y-VP-16 expression plasmid. ER-RXR-a 1 cotransfected cells were treated with ATRA (square) and AGN 193109 2 (diamond). Cells cotransfected with the combination of ER-RXR-a and 3 RAR-y-VP-16 were treated with ATRA (circle) or AGN 193109 4 (triangle).
5 Figure 7 shows a line graph representing luciferase activity 6 measurements recorded in lysates of CV-1 cells transfected with the 7 ERE-tk-Luc reporter and ER-RAR-y expression construct and then 8 treated with ATRA at 10-8 M and the test compounds at the 9 concentrations indicated on the horizontal axis. The test compounds 10 were AGN 193109 (square), AGN 193357 (open diamond), AGN
11 193385 (circle), AGN 193389 (triangle), AGN 193840 (hatched square) 12 and AGN 192870 (filled diamond).
13 Figure 8 shows a line graph representing luciferase activity 14 measurements recorded in lysates of CV-1 cells transfected with the 15 ERE-tk-Luc reporter and RAR-y-VP-16 and ER-RXR-a expression 16 constructs and then treated with the test compounds at the 17 concentrations indicated on the horizontal axis. The test compounds 18 were ATRA {open square}, AGN 193109 (open circle), AGN 193174 19 (open triangle), AGN 193199 (hatched square), AGN 193385 (hatched circle), AGN 193389 (inverted triangle), AGN 193840 (diagonally filled 21 square) and AGN 193871 (half-filled diamond).
22 Figures 9A, 9B and 9C schematically diagram a mechanism 23 whereby AGN 193109 can modulate the interaction between the RAR
24 {shaded box) and negative coactivator proteins (-) illustrated in the context of a transactivation assay. Figure 9A shows that negative z6 coactivator proteins and positive coactivator proteins (+) are in a 27 binding equilibrium with the RAR. In the absence of a ligand, basal 28 level transcription of the reporter gene results. As illustrated in Figure 29 9B, addition of an RAR agonist promotes the association of positive coactivator proteins with the RAR and results in upregulated reporter ......~.. -...~... .-.._ ~ ~."~.....w.-~. . _ ,...._.. ...~..~,-. -.......
~~..w ..--..___ .

1 gene transcription. As illustrated in Figure 9C, addition of AGN
2 193109 promotes the association of negative coactivator proteins with 3 the RAR and prevents reporter gene transcription.
4 Figure 10 is a bar graph showing the inhibition of TPA-induced Str-AP1-CAT expression as a function of AGN 191183 concentration 6 (10-1 to 10-12 M) with the AGN 193109 concentration held constant at 7 10-$ M. Results from trials conducted with AGN 191183 alone are 8 shown as hatched bars while stripped bars represent the results from 9 treatment with the combination of AGN 193109 and AGN 191183.
Figure 11 schematically diagrams a mechanism whereby AGN
11 193109 can potentiate the activities of RARs and other nuclear receptor 12 family members. As illustrated in the diagram, introduced RARs (open 13 rectangles having AB-C-DEF domains) have increased sensitivity to 14 RAR ligands in the anti-AP1 assay because the negative coactivator protein (ncp), present in limiting supply, is sequestered onto RARs 16 thereby leading to two populations: RAR+ncp and RAR-ncp. RAR-17 ncp has increased sensitivity to ligands. Non-RAR nuclear factors 18 (shaded rectangles having AB-C-DEF domains) have increased 19 sensitivity to cognate ligands because ncp has been sequestered to the RAR by the activity of AGN 193109. The modular domains of the 21 nuclear receptors are designated using standard nomenclature as "AB"
22 (ligand independent transactivation domain), "C" (DNA binding 23 domain), and "DEF" (ligand regulated transactivation domain and 24 dimerization domain.
2 ~ Figure 12 is a line graph showing the effect of AG"~ 193109 on 26 the 1,25-dihydroxyvitamin D3 dose response in CV-1 cells transtected 27 with the MTV-DR3-Luc reporter plasmid. Transfectants were treated 28 with 1,25-dihydroxyvitamin D3 (filled square), 1,25-dihydroxyvitamin D~
29 and 10-8 M AGN 193109 (filled triangle), and 1,25-dihydroxyvitamin D3 and 10-~ M AGN 193109 (filled circle).

Figure 13 is a bar graph showing the effect of AGN 193109 ( 10 2 nM) coadministration on 1,25-dihydroxyvitamin D3-mediated inhibition 3 of TPA induced Str-AP1-CAT activity. Filled bars represent inhibition 4 of CAT activity in transfected cells treated with 1,25-dihydroxyvitamin D3 alone. Open bars represent inhibition of CAT activity in transfected 6 cells treated with the combination of 1,25-dihydroxyvitamin D3 and 7 AGN 193109.
8 Figure 14 is a line graph showing the effect of AGN 193109 alone 9 and in combination with AGN 191183 on HeLa cells cotransfected with RAR-y and the RAR responsive MTV-TREp-Luc reporter construct.
11 Drug treatments illustrated in the graph are: AGN 193109 alone 12 (square), AGN 193109 in combination with AGN 191183 at 10-1 M
13 (diamond) and AGN 193109 in combination with AGN 191183 at 10-9 14 M.
Figure 15 is a line graph showing that ECE16-1 cells proliferated 16 in response to EGF (filled square) but not in response to defined 17 medium alone (open circle). Cells treated with AGN 193109 alone are 18 represented by the filled triangle. The filled circles represent results 19 obtained for cells treated with 10 nM AGN 191183 and 0 - 1000 nM
AGN 193109.
21 Figure 16 is a bar graph showing the effect of AGN 193109 on 22 the proliferation of CaSki cells in the presence or absence of the AGN
23 191183 retinoid agonist. All sample groups received 20 ng/ml of 24 epidermal growth factor (EGF) with the exception of the sample propagated in defined medium (DM) alone (open bar). Stripped bars 26 represent samples propagated in the absence of AGN 193109. Filled 27 bars represent samples propagated in the presence of 1000 nM AGN
28 193109. The concentrations of AGN 191183 used in the procedure are 29 shown on the horizontal axis.
Figure 17 is a dose response curve showing that AGN 193109 1 potentiated the antiproliferative activity of ATRA on retinal pigment 2 epithelium (RPE) cells. Samples treated with ATRA alone are 3 represented by filled squares. Samples treated with the combination of 4 ATRA and AGN 193109 (10-~ M) are represented by filled circles. The ATRA concentration used for treating the various samples is given on 6 the horizontal axis.
7 Figure 18 is a dose response curve showing that both 13-cis-RA
8 and ATRA inhibited RPE cell growth, and that AGN 193109 9 potentiated the antiproliferative activity of 13-cis-RA. The various 1 o sample treatments shown in the dose response included 13-cis-RA alone 11 (filled square), 13-cis-RA in combination with AGN 193109 (10-6 M) 12 (filled circle), 13-cis-RA in combination with AGN 193109 (10-8 M) 13 {filled triangle), and ATRA (filled diamond). The concentrations of 13-14 cis-RA and ATRA used in the sample treatments are shown on the horizontal axis.
16 Figure 19 is a dose response curve showing that AGN 193109 17 potentiated the antiproliferative activity of dexamethasone in primary 18 RPE cell cultures. The various sample treatments shown in the dose 19 response included ATRA (filled square), dexamethasone alone (filled circle), dexamethasone in combination with AGN 193109 (10-8 M) (filled 21 triangle), and dexamethasone in combination with AGN 193109 ( 10-6 M) 22 (filled diamond). The concentrations of dexamethasone and ATRA
23 used in the sample treatments are shown on the horizontal axis 24 Figure 20 is a dose response curve showing that AGN 193109 potentiated the antiproliferative activity of thyroid hormone (T3) in 26 primary RPE cell cultures. The various sample treatments shown in the 27 dose response included ATRA (filled square), T3 alone (filled circle), 28 T3 in combination with AGN 193109 {10-8 M) (filled triangle), T3 in 29 combination with AGN 193109 ( 10-6 M) (filled diamond). The concentrations of T3 and ATRA used in the sample treatments are 1 shown on the horizontal axis.
2 Detailed Description of the Invention 3 Definitions 4 For the purposes of the present invention, an RAR antagonist is defined as a chemical that binds to one or more of the RAR subtypes 6 with a Kd of less than 1 micromolar (Kd < l~cM) but which does not 7 cause significant transcriptional activation of that RAR subtypes-8 regulated genes in a receptor co-transfection assay. Conventionally, 9 antagonists are chemical agents that inhibit the activities of agonists.
l0 Thus, the activity of a receptor antagonist is conventionally measured by 1l virtue of its ability to inhibit the activity of an agonist.
12 An RAR agonist is defined as a chemical that binds to one or 13 more RAR receptor subtype with Kd of less than 1 micromol (Kd < 1 14 ~cM) and causes transcriptional activation of that RAR-subtype-regulated genes in a receptor co-transfection assay. The term "RAR
16 agonist" includes chemicals that may bind and/or activate other 17 receptors in addition to RARs, e.g., RXR receptors.
18 As used herein, a negative hormone or inverse agonist is a ligand 19 for a receptor which causes the receptor to adopt an inactive state relative to a basal state occurring in the absence of any ligand. Thus, 21 while an antagonist can inhibit the activity of an agonist, a negative 22 hormone is a ligand that can alter the conformation of the receptor in 23 the absence of an agonist. The concept of a negative hormone or 24 inverse agonist has been explored by Bond et al. in Nature 374:272 (1995). More specifically, Bond et al. have proposed that unliganded 26 X32-adrenoceptor exists in an equilibrium between an inactive 27 conformation and a spontaneously active conformation. Agonists are 28 proposed to stabilize the receptor in an active conformation.
29 Conversely, inverse agonists are believed to stabilize an inactive receptor conformation. Thus, while an antagonist manifests its activity by virtue ....,..,.. W.,~""",~ . ~,. .e. ..,.. ... ... . . T .. .. .".."~.........~ .y, .....~.""."~..~.~...

WO 98!58922 PCT/US98/13065 1 of inhibiting an agonist, a negative hormone can additionally manifest 2 its activity in the absence of an agonist by inhibiting the spontaneous 3 conversion of an unliganded receptor to an active conformation. Only a 4 subset of antagonists will act as negative hormones. As disclosed 5 herein, AGN 193109 is both an antagonist and a negative hormone. To 6 date, no other retinoids have been shown to have negative hormone 7 activity.
8 As used herein, coadministration of two pharmacologically active 9 compounds refers to the delivery of two separate chemical entities, 10 whether in vitro or in vivo. Coadministration refers to the simultaneous 11 delivery of separate agents; to the simultaneous delivery of a mixture of 12 agents; as well as to the delivery of one agent followed by delivery of 13 the second agent. In all cases, agents that are coadministered are 14 intended to work in conjunction with each other.
15 The term alkyl refers to and covers any and all groups which are 16 known as normal alkyl, branched-chain alkyl and cycloalkyl. The term 17 alkenyl refers to and covers normal alkenyl, branch chain alkenyl and 18 cycloalkenyl groups having one or more sites of unsaturation. Similarly, 19 the term alkynyl refers to and covers normal alkynyl, and branch chain 20 alkynyl groups having one or more triple bonds.
21 Lower alkyl means the above-defined broad definition of alkyl 22 groups having 1 to 6 carbons in case of normal lower alkyl, and as 23 applicable 3 to b carbons for lower branch chained and cycloalkyl 24 groups. Lower alkenyl is defined similarly having 2 to 6 carbons for normal lower alkenyl groups, and 3 to 6 carbons for branch chained and 26 cyclo- lower alkenyl groups. Lower alkynyl is also defined similarly, 27 having 2 to 6 carbons for normal lower alkynyl groups, and 4 to 6 28 carbons for branch chained lower alkynyl groups.
29 The term "ester" as used here refers to and covers any compound falling within the definition of that term as classically used in organic 1 chemistry. It includes organic and inorganic esters. Where B (of 2 Formula 1 or Formula 101) is -COON, this term covers the products 3 derived from treatment of this function with alcohols or thiols 4 preferably with aliphatic alcohols having 1-6 carbons. Where the ester is derived from compounds where B is -CH20H, this term covers 6 compounds derived from organic acids capable of forming esters 7 including phosphorous based and sulfur based acids, or compounds of 8 the formula -CH20COR11 where R,1 is any substituted or unsubstituted 9 aliphatic, aromatic, heteroaromatic or aliphatic aromatic group, l0 preferably with 1-6 carbons in the aliphatic portions.
11 Unless stated otherwise in this application, preferred esters are 12 derived from the saturated aliphatic alcohols or acids of ten or fewer 13 carbon atoms or the cyclic or saturated aliphatic cyclic alcohols and 14 acids of 5 to 10 carbon atoms. Particularly preferred aliphatic esters are those derived from lower alkyl acids and alcohols. Also preferred are 16 the phenyl or lower alkyl phenyl esters.
17 Amides has the meaning classically accorded that term in organic 18 chemistry. In this instance it includes the unsubstituted amides and all 19 aliphatic and aromatic mono- and di- substituted amides. Unless stated otherwise in this application, preferred amides are the mono- and di-21 substituted amides derived from the saturated aliphatic radicals of ten 22 or fewer carbon atoms or the cyclic or saturated aliphatic-cyclic radicals 23 of 5 to IO carbon atoms. Particularly preferred amides are those 24 derived from substituted and unsubstituted lower alkyl amines. Also preferred are mono- and disubstituted amides derived from the 26 substituted and unsubstituted phenyl or lower alkylphenyl amines.
27 Unsubstituted amides are also preferred.
28 Acetals and ketals include the radicals of the formula-CK where 29 K is (-OR)2. Here, R is lower alkyl. Also, K may be -OR~O- where R, is lower alkyl of 2-5 carbon atoms, straight chain or branched.

1 A pharmaceutically acceptable salt may be prepared for any 2 compounds in this invention having a functionality capable of forming a 3 salt, for example an acid functionality. A pharmaceutically acceptable 4 salt is any salt which retains the activity of the parent compound and does not impart any deleterious or untoward effect on the subject to 6 which it is administered and in the context in which it is administered.
7 Pharmaceutically acceptable salts may be derived from organic or 8 inorganic bases. The salt may be a mono or polyvalent ion. Of 9 particular interest are the inorganic ions, sodium, potassium, calcium, and magnesium. Organic salts may be made with amines, particularly 11 ammonium salts such as mono-, di- and trialkyl amines or ethanol 12 amines. Salts may also be formed with caffeine, tromethamine and 13 similar molecules. Where there is a nitrogen sufficiently basic as to be 14 capable of forming acid addition salts, such may be formed with any inorganic or organic acids or alkylating agent such as methyl iodide.
16 Preferred salts are those formed with inorganic acids such as 17 hydrochloric acid, sulfuric acid or phosphoric acid. Any of a number of 18 simple organic acids such as mono-, di- or tri- acid may also be used.
19 Some of the compounds of the present invention may have trans and cis (E and Z) isomers. In addition, the compounds of the present 21 invention may contain one or more chiral centers and therefore may 22 exist in enantiomeric and diastereomeric forms. The scope of the 23 present invention is intended to cover all such isomers per se, as well as 24 mixtures of cis and trans isomers, mixtures of diastereomers and racemic mixtures of enantiomers (optical isomers) as well. In the present 26 application when no specific mention is made of the configuration ~~;is, 27 trans or R or S) of a compound (or of an asymmetric carbon) then a 28 mixture of such isomers, or either one of the isomers is intended.

1 Arvl Substituted Benzopyran Benzothiopvran 1 2-Dihydroquinoline and 2 5,6-Dihvdronaphthalene Derivatives Having Retinoid Anta onist 3 Like Biological Activity 4 With reference to the symbol Y in Formula 1, the preferred compounds of the invention are those where Y is phenyl, pyridyl, thienyl 6 or furyl. Even more preferred are compounds where Y is phenyl or 7 pyridyl, and still more preferred where Y is phenyl. As far as 8 substitutions on the Y (phenyl) and Y (pyridyl) groups are concerned, 9 compounds are preferred where the phenyl group is 1,4 (para) l0 substituted by the Z and A-B groups, and where the pyridine ring is 2,5 11 substituted by the Z and A-B groups. (Substitution in the 2,5 positions 12 in the "pyridine" nomenclature corresponds to substitution in the 6-13 position in the "nicotinic acid" nomenclature.) In the preferred 14 compounds of the invention either there is no optional RZ substituent on the Y group, or the optional R2 substituent is fluoro (F).
16 The A-B group of the preferred compounds is (CHZ)~-COOH or 17 (CH2)n-COORS, where n and R8 are defined as above. Even more 18 preferably n is zero and R8 is lower alkyl, or n is zero and B is COOH
19 or a pharmaceutically acceptable salt thereof.
In the majority of the presently preferred examples of compounds 21 of the invention X is [C(R,)2j~ where n is 1. Nevertheless, compounds 22 where n is zero (indene derivatives) and where X is S or O
23 (benzothiopyran and benzopyran derivatives) are also preferred. When 24 X is [C(R,)2Jn and n is 1, then Rl preferably is alkyl of 1 to 6 carbons, even more preferably methyl.
26 The R2 group attached to the aromatic portion of the 27 tetrahydronaphthalene, benzopyran, benzothiopyran or dihydroquinoline 28 moiety of the compounds of Formula 1 is preferably H, F or CF3. R3 is 29 preferably hydrogen or methyl, even more preferably hydrogen.
Referring now to the group Z in the compounds of the invention 1 and shown in Formula 1, in a plurality of preferred examples Z
2 represents an acetylenic linkage (Z = -C=C-). However, the "linker 3 group" Z is also preferred as a diazo group (Z = -N=N-), as an olefinic 4 or polyolefinic group (Z = -(CRl=CR1)n.-), as an ester (Z = -COO-), amide (Z = -CO-NR2-) or thioamide (Z = -CS-NR2-) linkage.
6 Referring now to the R,4 group, compounds are preferred where 7 R14 is phenyl, 2-pyridyl, 3-pyridyl, 2- thienyl, and 2-thiazolyl. The R,5 8 group (substituent of the R14 group) is preferably H, lower alkyl, 9 trifluoromethyl, chlorine, lower alkoxy or hydroxy.
l0 The presently most preferred compounds of the invention are 11 shown in Table 1 with reference to Formula 2, Formula 3, Formula 4, 12 Formula 5, and Formula Sa, and in Table lA with reference to 13 Formulas Sb and Sc.

c Formula 2 Formula 3, ~a , Ru 6 ~~ ~ ~ \-' \ \ ~sR:
i i ~ v v v-7 ~ w g c~, cft, Formula 4 ~ ~ Formula 5 c 19 Formula Sa Chi 23 Ru i i 24 ~ ~ ~ N F ~ H
H
Sc 26 sb SUBSTITUTE SHEET (RULE 26j . ,_.".~ ~.~._..W.... _ ......H.._~~...~.~.. r ...~~.,..~.~ ._~T.y_ 3 Compound Rl4' Z RZ"' R8*
Formula 4 #

6 1 2 4-methylphenyl -C-_-C- H Et 7 la 2 phenyl -C=C- H Et 8 2 2 3-methylphenyl -C---C- H Et 9 3 2 2-methylphenyl -C=C- H Et 4 2 3,5-dimethylphenyl -C--__ H Et C-1 5 2 4-ethylphenyl -C=C- H Et t 12 6 2 4-t-butylphenyl -C--__ H Et C-13 7 2 4-chlorophenyl -C--__ H Et C-14 8 2 4-methoxyphenyl -C=C- H Et 9 2 4-trifluoromethylphenyl-C=C- H Et 16 10 2 2-pyridyl -C=C- H Et 17 11 2 3-pyridyl -C--__ H Et C-18 12 2 2-methyl-5-pyridyl -C--__ H Et C-19 13 2 3-hydroxyphenyl -C= C- H Et 14 2 4-hydroxy phenyl -C=C- H Et 21 15 2 5-methyl-2-thiazolyl-C=C- H Et 22 15a 2 2-thiazolyl -C---C- H Et 23 16 2 4-methyl-2-thiazolyl-C--_C- H Et 24 17 2 4,5-dimethyl-2-thiazolyl-C=C- H Et 18 2 2-methyl-5-pyridyl -C--__ H H
C-26 19 2 2-pyridyl -C=C- H H

27 20 2 3-methylphenyl -C=C- H H

28 21 2 4-ethylphenyl -C=C- H H

29 22 2 4-methoxyphenyl -C=C- H H

23 2 4-trifluoromethylphenyl-C=C- H H

31 24 2 3,5-dimethylphenyl -C--__C- H H

32 25 2 4-chlorophenyl -C--__ H H
C-33 26 2 3-pyridyl -C--__C- H H

34 27 2 2-methylphenyi -C=C- H H

28 2 3-hydroxyphenyl -C-_-C- H H

36 29 2 4-hydroxyphem' -C=C- H H

37 30 2 5-methyl-2-thin 'yl -C=C- H H

38 30a 2 2-thiazolyl -C=C- H H

39 31 2 4-methyl-2-thiazolyl-C=C- H H

32 2 4,5-dimethyl-2-thiazolyl-C=C- H H

41 33 2 5-methyl-2-thienyl -C-_C- H Et 42 33a 2 2-thienyl -C--__C- H Et 43 34 2 5-methyl-2-thienyi -C=C- H H

1 34a 2-thienyl -C=C- H H

2 3 5 2 4-methylphenyl -CONH- H Et 3 3 6 2 4-methylphenyl -CONH- H H

4 3 7 2 4-methylphenyl -COO- H Et 38 2 4-methylphenyl -COO- H
(CH,)ZSi(CH3) 6 39 2 4-methylphenyl -COO- H H

7 40 2 4-methylphenyl -CONH- F Et 8 41 2 4-methylphenyl -CONH F H

9 42 2 4-methylphenyl -CSNH- H Et l0 43 2 4-methylphenyl -CSNH- H H

11 44 2 4-methylphenyl -CH=CH- H Et 12 45 2 4-methylphenyl -CH=CH- H H

13 46a 2 4-methylphenyl -N=N- H Et 14 46b 2 4-methylphenyl -N=N- H H

47 3 4-methylphenyl -C=C- H Et 16 48 3 4-methylphenyl -C-_-C- H H

17 49 4 4-methylphenyl -C--_C- H Et 18 50 4 4-methylphenyl -C=C- H H

19 51 5 4-methylphenyl - - Et 52 5 4-methylphenyl - - H

21 60 2 4-methylphenyl -C---C- H H

22 60a 2 phenyl -C--__ C- H H

23 61 2 4-t-butylphenyl -C--__ C- H H

24 62 2 4-methylphenyl -CSNH F Et 63 2 4-methylphenyl -CSNH F H

26 64 5a 4-methylphenyl ---- - Et 27 65 5a 4-methylphenyl ---- - H

28 66 2 2-furyl -C--__C- H Et 29 67 2 2-furyl -C--__ C- H H

31 Table lA
32 Compound # Formula R14* R2* RA*

33 212 5b 4-methylphenyl -- Et 34 203 5b 4-methylphenyl -- H

221 5c 4-methylphenyl H Et 36 204 5c 4-methylphenyl H H

37 222 5c 4-methylphenyl F Et 38 205 5c 4-methylphenyl F H

1 Aryl and (3-Oxo-1-Propen~l)-Substituted Benzopyran, Benzothiopyran, 2 Dihydroquinoline and 5,6-Dih d~phthalene Derivatives Having 3 Retinoid 4 Antagonist-Like Biological Activity With reference to the symbol Y in Formula 101, the preferred 6 compounds of the invention are those where Y is phenyl, pyridyl, thienyl 7 or furyl. Even more preferred are compounds where Y is phenyl or 8 pyridyl, and still more preferred where Y is phenyl. As far as 9 substitutions on the Y (phenyl) and Y (pyridyl) groups are concerned, compounds are preferred where the phenyl group is 1,4 (para) 11 substituted by the -CR16=CR"- and A-B groups, and where the pyridine 12 ring is 2,5 substituted by the -CR,6=CR,~- and A-B groups. (Substitution 13 in the 2,5 positions in the "pyridine" nomenclature corresponds to 14 substitution in the 6- position in the "nicotinic acid" nomenclature.) In the preferred compounds of the invention there is no optional R
16 substituent on the Y group.
17 The A-B group of the preferred compounds is (CH2)n-COOH or 18 (CH2)n-COOR8, where n and R8 are defined as above. Even more 19 preferably n is zero and R8 is lower alkyl, or n is zero and B is COOH
or a pharmaceutically acceptable salt thereof.
21 In the presently preferred examples of compounds of the 22 invention X is [C(R1)2J" where n is 1. Nevertheless, compounds where 23 X is S or O (benzothiopyran and benzopyran derivatives) are also 24 preferred. When X is [C(RU)Z)" and n is 1, then R, preferably is alkyl of 1 to 6 carbons, even more preferably methyl.
26 The R2 group attached to the aromatic portion of the 27 tetrahydronaphthalene, benzopyran, benzothiopyran or dihydroquinoline 28 moiety of the compounds of Formula 101 is preferably H, F or CF3. R3 29 is preferably hydrogen or methyl, even more preferably hydrogen.
Referring now to the R~4 group, compounds are preferred where 1 R,4 is phenyl, 2-pyridyl, 3-pyridyl, 2- thienyl, and 2-thiazolyl. The R,5 2 group (substituent of the R,4 group) is preferably H, lower alkyl, 3 trifluoromethyl, chlorine, lower alkoxy or hydroxy.
4 Preferred compounds of the invention are shown in Table 2 with reference to Formula 102.
b 7 _ 14 Formula 102 16 Compund R,5' R$' 1 ~ 101 CH3 H
18 102 CH3 Et 104 H Et 22 Biological Activity, Modes of Administration 23 As noted above, the compounds of the present invention are 24 antagonists of one or more RAR receptor subtypes. This means that the compounds of the invention bind to one or more RAR receptor 26 subtypes, but do not trigger the response which is triggered by agonists 27 of the same receptors. Some of the compounds of the present invention 28 are antagonists of all three RAR receptor subtypes (RAR-a, RAR-/3 29 and RAR-y), and these are termed "RAR pan antagonists". Some others are antagonists of only one or two of RAR receptor subtypes.
SUBSTITUTE SHEET (RULE 26) _.....~~~....,~. , . .,........ .__.. w..... .

1 Some compounds within the scope of the present invention are partial 2 agonists of one or two RAR receptor subtypes and antagonists of the 3 remaining subtypes. The compounds of the invention do not bind to 4 RXR receptors, therefore they are neither agonists nor antagonists of 5 RXR.
6 Depending on the site and nature of undesirable side effects 7 which are desired to be suppressed or ameliorated, compounds used in 8 accordance with the invention may be antagonists of only one or two of 9 RAR receptor subtypes. Some compounds used in accordance with the 10 invention may be partial agonists of one or two RAR receptor subtypes 11 and antagonists of the remaining subtypes. Such compounds are, 12 generally speaking, usable in accordance with the invention if the 13 antagonist effect is on that RAR receptor subtype (or subtypes) which is 14 (are) predominantly responsible for the overdose poisoning or for the 15 undesired side effect or side effects. In this connection it is noted that, 16 generally speaking, a compound is considered an antagonist of a given 17 receptor subtype if in the below described co-tranfection assays the 18 compound does not cause significant transcriptional activation of the 19 receptor regulated reporter gene, but nevertheless binds to the receptor 20 with a Kd value of less than approximately 1 ~cM.
21 Whether a compound is an RAR antagonist and therefore can be 22 utilized in accordance with the present invention, can be tested in the 23 following assays.
24 A chimeric receptor transactivation assay which tests for agonist-25 like activity in the RAR-a, RAR-(3, RAR-~y, RXR-a receptor subtypes, 26 and which is based on work published by Feigner P. L. and Holm M.
27 Focus Vol 11, No. 2 (1989) is described in detail in published PCT
28 Application No. W094/17796, published on August 18, 1994. The latter 29 publication is the PCT counterpart of U. S. application serial no.
30 08/016,404, filed on February 11, 1993, which issued as U.S. Patent No.
1 5,455,265. PCT publication W094/17796 and the specification of U.S.
2 patent 5,455,265 are hereby expressly incorporated by reference. A
3 compound should not cause significant activation of a reporter gene 4 through a given receptor subtype (RAR-a, RAR-~i or RAR-y ) in this assay, in order to qualify as an RAR antagonist with utility in the 6 present invention.
7 A holoreceptor transactivation assay and a ligand binding assay 8 which measure the antagonist/agonist like activity of the compounds of 9 the invention, or their ability to bind to the several retinoid receptor l0 subtypes, respectively, are described in published PCT Application No.
11 W093/11755 (particularly on pages 30 - 33 and 37 - 41) published on 12 3une 24, 1993, the specification of which is also incorporated herein by 13 reference. A description of the holoreceptor transactivation assay is 14 also provided below.
Holoreceptor Transactivation Assax 16 CV 1 cells {5,000 cells/well) were transfected with an RAR
17 reporter plasmid MTV-TREp-LUC (50 ng) along with one of the RAR
18 expression vectors (10 ng) in an automated 96-well format by the 19 calcium phosphate procedure of Heyman et al. Cell 68: 397 - 406. For RXR-a and RXR-y transactivation assays, an RXR-responsive reporter 2I plasmid CRBPII-tk-LUC (50 ng) along with the appropriate RXR
22 expression vectors (10 ng) was used substantially as described by 23 Heyman et al. above, and Allegretto et al. J. Biol. Chem. 268: 26625 -24 26633. For RXR-(3 transactivation assays, an RXR-responsive reporter plasmid CPRE-tk-LUC (50 mg) along with RXR-~3 expression vector 26 (10 mg) was used as described in above. These reporters contain DRI
27 elements from human CRBPII and certain DRI elements from 28 promotor, respectively (see Mangelsdorf et al. The Retinoids: Biolo~v, 29 Chemistry and Medicine, pp. 319 - 349, Raven Press Ltd., New York and Heyman et al., cited above). A (3-galactosidase (50 ng) expression . _. _~-. ~....-.,-_.. ....... . ,...~..~... w t 1 vector was used as an internal control in the transfections to normalize 2 for variations in transfection efficiency. The cells were transfected in 3 triplicate for 6 hours, followed by incubation with retinoids for 36 hours, 4 and the extracts were assayed for luciferase and (~-galactosidase activities. The detailed experimental procedure for holoreceptor 6 transactivations has been described in Heyman et al. above, and 7 Allegretto et al. cited above. The results obtained in this assay are 8 expressed in ECso numbers, as they are also in the chimeric receptor 9 transactivation assay. The Heyman et al. Cell 68: 397 - 406, Allegretto et al. J. Biol. Chem. 268: 26625 - 26633, and Mangelsdorf et al. The 11 Retinoids: BioloQV, Chemistry and Medicine, pp. 319 - 349, Raven Press 12 Ltd., New York, are expressly incorporated herein by reference. The 13 results of ligand binding assay are expressed in Kd numbers. (See 14 Cheng et al. Biochemical Pharmacology 22: 3099-3108, expressly incorporated herein by reference.) 16 A compound should not cause significant activation of a reporter 17 gene through a given receptor subtype (RAR-a, RAR-/3 or RAR-'y) in 18 the holoreceptor transactivation assay assay, in order to qualify as an 19 RAR antagonist with utility in the present invention. Last, but not least, a compound should bind to at least one of the RAR receptor subtypes 21 in the ligand binding assay with a Kd of less than approximately 1 22 micromolar (Ka < 1 ~cM) in order to be capable of functioning as an 23 antagonist of the bound receptor subtype, provided the same receptor 24 subtype is not significantly activated by the compound.
Table 3 below shows the results of the holoreceptor 26 transactivation assay and Table 4 discloses the efficacy (in percentage) 27 in this assay of the test compound relative to all traps retinoic acid, for 28 certain exemplary compounds of the invention. Table 5 and SA show 29 the results of the ligand binding assay for certain exemplary compounds of the invention.

WO 98/58922 PCT/US98l13065 TABLE

2 Holorecep tor v Transactivation Assa 3 Compound ECso (nanomolar) #

RAR13 RARy RXRa RXR13 RXR~y 6 18 0.00 0.00 0.00 0.00 0.00 0.00 7 19 0.00 0.00 0.00 0.00 0.00 0.00 8 20 0.00 0.00 0.00 0.00 0.00 0.00 9 21 0.00 0.00 0.00 0.00 0.00 0.00 l0 22 0.00 0.00 0.00 0.00 0.00 0.00 11 23 0.00 0.00 0.00 0.00 0.00 0.00 12 24 0.00 0.00 0.00 0.00 0.00 0.00 13 25 0.00 0.00 0.00 0.00 0.00 0.00 14 26 0.00 0.00 0.00 0.00 0.00 0.00 27 0.00 0.00 0.00 0.00 0.00 0.00 16 28 0.00 0.00 0.00 0.00 0.00 0.00 17 29 0.00 0.00 0.00 0.00 0.00 0.00 18 30 0.00 0.00 0.00 0.00 0.00 0.00 19 31 0.00 0.00 0.00 0.00 0.00 0.00 32 0.00 0.00 0.00 0.00 0.00 0.00 21 34 0.00 0.00 0.00 0.00 0.00 0.00 22 36 0.00 0.00 0.00 0.00 0.00 0.00 23 39 0.00 0.00 0.00 0.00 0.00 0.00 24 41 0.00 0.00 0.00 0.00 0.00 0.00 45 0.00 0.00 0.00 0.00 0.00 0.00 26 46b 0.00 0.00 0.00 0.00 0.00 0.00 27 52 0.00 0.00 0.00 0.00 0.00 0.00 28 60 0.00 0.00 0.00 0.00 0.00 0.00 29 61 0.00 0.00 0.00 0.00 0.00 0.00 63 0.00 0.00 0.00 o.oa o.oo 0.00 31 lol o.oo o.oo o.o0 0.00 0.00 0.00 32 103 0.00 0.00 0.00 0.00 0.00 0.00 33 O.O in Table 3 indicates that the compound is less than 20 % as active 34 (efficacious) in this assay than all trans retinoic acid.

2 Transactivation Assav Efficacy l% of RA activi 3 Compound #
4 RARa RAR13 RARE RXRa RXR13 RXRy 6 18 4.00 1.00 0.00 2.00 10.00 1.0 7 19 0.00 5.0 3.0 0.0 9.0 4.0 8 20 3.0 4.0 0.00 4.00 0.00 3.0 9 21 2.00 2.00 2.00 3.00 0.00 3.00 22 0.00 0.00 2.00 1.00 0.00 2.00 11 23 0.00 6.00 3.00 1.00 0.00 4.00 12 24 3.00 7.00 4.00 1.00 0.00 3.00 13 25 2.00 3.00 3.00 5.00 0.00 3.00 14 26 1.00 6.00 0.00 2.00 0.00 3.00 27 9.00 14.00 6.00 2.00 0.00 4.00 16 28 2.00 10.00 2.00 2.00 0.00 3.00 17 29 0.00 6.00 11.00 0.00 6.00 2.00 18 30 3.00 5.00 1.00 0.00 9.00 3.00 19 31 4.00 14.00 2.00 1.00 8.00 6.00 32 0.00 2.00 2.00 1.00 0.00 2.00 21 34 3.00 5.00 2.00 1.00 0.00 3.00 22 36 1.00 5.00 0.00 1.00 7.00 2.00 23 39 1.00 7.00 9.00 2.00 0.00 1.00 24 41 3.00 5.00 6.00 1.00 0.00 3.00 45 2.00 0.00 7.00 3.00 8.00 0.00 26 46b 4.00 5.00 3.00 2.00 0.00 4.00 27 52 0.00 15.00 3.00 0.00 0.00 10.00 28 60 0.00 1.00 4.00 3.00 0.00 3.00 29 61 2.00 2.00 0.00 1.00 0.00 3.00 63 2.00 2.00 7.00 1.00 0.00 1.00 31 101 0.00 4.00 2.00 1.00 0.00 3.0 32 103 4.00 12.0 7.0 0.00 0.0 2.0 2 Ligand Bindin Assay 3 Compo und # Ka (nanomolar) 4 RARa RAR13 RARy RXRa RXRI3 RXRy 6 18 24.00 11.00 24.00 0.00 0.00 0.00 7 19 565 210 659 0.00 0.00 0.00 8 20 130.00 22.0 34.00 0.00 0.00 0.00 9 21 16 9 13 0.00 0.00 0.00 l0 22 24.0 17.0 27.0 0.00 0.00 0.00 11 23 32.00 25.00 31.00 0.00 0.00 0.00 12 24 699 235 286 0.00 0.00 0.00 13 25 50 17 20 0.00 0.00 0.00 14 26 40.00 31.00 36.00 0.00 0.00 0.00 15 27 69.00 14.00 26.00 0.00 0.00 0.00 16 28 669 77 236 0.00 0.00 0.00 17 29 234 48 80 0.00 0.00 0.00 18 30 683 141 219 0.00 0.00 0.00 19 31 370 52.00 100.00 0.00 0.00 0.00 20 32 0.00 89.00 169.00 0.00 0.00 0.00 21 34 52.00 30.00 17.00 0.00 0.00 0.00 22 36 13.00 550.00 0.00 0.00 0.00 0.00 23 39 67.00 38.00 113.00 0.00 0.00 0.00 24 41 5.1 491 725 0.00 0.00 0.00 25 45 12.0 2.80 17.0 0.00 0.00 0.00 26 46b 250 3.70 5.80 0.00 0.00 0.00 27 52 60.00 63.00 56.00 0.00 0.00 0.00 28 60 1.5 1.9 3.3 0.00 0.00 0.00 29 61 96 15 16 0.00 0.00 0.00 30 63 133 3219 0.00 0.00 0.00 0.00 31 101 750 143 637 0.00 0.00 0.00 32 103 301 273 261 0.00 0.00 0.00 34 O.O in Table 5 indicates a value greater than 1000nM.
36 Table SA
37 Ligand Binding Assay 39 Compound Kd (nanomolar) #

RARa RAR~3 RAR y 41 203 32 15 2256 1257 > 30,000 42 204 2.8 t 320 t 87 7258 2648 1.1 43 205 0.690 541112 72802720 44 retinoic 151.7 132.5 181 acid I As it can be seen from the test results summarized in Tables 3, 4 2 and S, the therein indicated exemplary compounds of the invention are 3 antagonists of the RAR receptor subtypes, but have no affinity to RXR
4 receptor subtypes. (Other compounds of the invention may be antagonist of some but not all RAR receptor subtypes and agonists of 6 the remaining RAR subtypes.) Due to this property, the compounds of 7 the invention can be used to block the activity of RAR agonists in 8 biological assays. In mammals, including humans, the compounds of the 9 invention can be coadministered with RAR agonists and, by means of pharmacological selectivity or site-specific delivery, preferentially 11 prevent the undesired effects of RAR agonists. The compounds of the 12 invention can also be used to treat Vitamin A overdose, acute or 13 chronic, resulting either from the excessive intake of vitamin A
14 supplements or from the ingestion of liver of certain fish and animals that contain high level of Vitamin A. Still further, the compounds of 16 the invention can also be used to treat acute or chronic toxicity caused 17 by retinoid drugs. It has been known in the art that the toxicities 18 observed with hypervitaminosis A syndrome (headache, skin peeling, 19 bone toxicity, dyslipidemias) are similar or identical with toxicities observed with other retinoids, suggesting a common biological cause, . 21 that is RAR activation. Because the compounds of the present 22 invention block RAR activation, they are suitable for treating the 23 foregoing toxicities.
24 The compounds of the invention are able to substantially prevent skin irritation induced by RAR agonist r~ :voids, when the compound of 26 the invention is topically coadministered ~o the skin. Similarly, 27 compounds of the invention can be administered topically to the skin, to 28 block skin irritation, in patients or animals who are administered RAR
29 agonist compounds systemically. The compounds of the invention can accelerate recovery from pre-existing retinoid toxicity, can block 1 hypertriglyceridemia caused by co- administered retinoids, and can block 2 bone toxicity induced by an RAR agonist (retinoid).
3 Generally speaking, for therapeutic applications in mammals in 4 accordance with the present invention, the antagonist compounds can be admistered enterally or topically as an antidote to vitamin A, vitamin A
6 precursors, or antidote to retinoid toxicity resulting from overdose or 7 prolonged exposure, after intake of the causative factor (vitamin A
8 precursor or other retinoid) has been discontinued. Alternatively, the 9 antagonist compounds are coadministered with retinoid drugs in accordance with the invention, in situations where the retinoid provides 11 a therapeutic benefit, and where the coadministered antagonist 12 alleviates or eliminates one or more undesired side effects of the 13 retinoid. For this type of application the antagonist may be 14 administered in a site-specific manner, for example as a topically applied cream or lotion while the coadministered retinoid may be given 16 enterally.
17 For therapeutic applications in accordance with the present 18 invention the antagonist compounds are incorporated into 19 pharmaceutical compositions, such as tablets, pills, capsules, solutions, suspensions, creams, ointments, gels, salves, lotions and the like, using 21 such pharmaceutically acceptable excipients and vehicles which per se 22 are well known in the art. For example preparation of topical 23 formulations are well described in Remington's Pharmaceutical Science, 24 Edition I7, Mack Publishing Company, Easton, Pennsylvania. For topical application, the antagonist compounds could also be 26 administered as a powder or spray, particularly in aerosol form. If the 27 drug is to be administered systemically, it may be confected as a 28 powder, pill, tablet or the like or as a syrup or elixir suitable for oral 29 administration. For intravenous or intraperitoneal administration, the antagonist compound will be prepared as a solution or suspension rv....~.u.~ ....~._~._,w. . . T._._ _... ~.~~.. ... . .._. ~~~..~.__......_.

1 capable of being administered by injection. In certain cases, it may be 2 useful to formulate the antagonist compounds by injection. In certain 3 cases, it may be useful to formulate the antagonist compounds in 4 suppository form or as extended release formulation for deposit under the skin or intramuscular injection.
6 The antagonist compounds will be administered in a 7 therapeutically effective dose in accordance with the invention. A
8 therapeutic concentration will be that concentration which effects 9 reduction of the particular condition (such as toxicity due to retinoid or l0 vitamin A exposure, or side effect of retinoid drug) or retards its 11 expansion. It should be understood that when coadministering the 12 antagonist compounds to block retinoid-induced toxicity or side effects 13 in accordance with the invention, the antagonist compounds are used in 14 a prophylactic manner to prevent onset of a particular condition, such as skin irritation.
16 A useful therapeutic or prophylactic concentration will vary from 17 condition to condition and in certain instances may vary with the 18 severity of the condition being treated and the patient's susceptibility to 19 treatment. Accordingly, no single concentration will be uniformly useful, but will require modification depending on the particularities of 21 the chronic or acute retinoid toxicity or related condition being treated.
22 Such concentrations can be arrived at through routine experimentation.
23 However, it is anticipated that a formulation containing between 0.01 24 and 1.0 milligrams of antagonist compound per mililiter of formulation will constitute a therapeutically effective concen ?on for t~~~pical 26 application. If administered systemically, an amc ~ betty- ~ 0.01 and 27 mg per kg per day of body weight would be expected to effect a 28 therapeutic result.
29 The basis of the utility of RAR antagonists for the prevention or treatment of RAR agonist-induced toxicity is competitive inhibition of 1 the activation of RAR receptors by RAR agonists. The main distinction 2 between these two applications of RAR antagonists is the presence or 3 absence of preexisting retinoid toxicity. Most of the examples 4 immediately described below relate to the use of retinoids to prevent retinoid toxicity, but the general methods described herein are 6 applicable to the treatment of preexisting retinoid toxicity as well.
7 Description of experiments demonstrating the use of RAR antagonists 8 to prevent or treat retinoid toxicity and/or side effects of retinoid drugs 9 Example 1: skin iritation induced by tonicall~p~plied agonist is treated l0 with topicall~pplied anta onist 11 The compound 4-((E)-2-(5,6,7,8-tetrahydro-5,5,8,8-12 tetramethylnaphthalen-2-yl)propen-1-yl]benzoic acid, designated AGN
13 191183, is known in the prior art as a potent RAR agonist (see for 14 example the descriptive portion and figure 2b of United States Patent No. 5,324,840). {The "AGN" number is an arbitrarily designated 16 reference number utilized by the corporate assignee of the present 17 invention for identification of compounds.) 18 4-((5,6-dihydro-5,5-dimethyl-8-(phenyl)-2-19 naphthalenyl)ethynyl]benzoic acid (AGN 192869, also designated Compound 60a) is a compound the preparation of which is described 21 below. This compound is an RAR antagonist.
22 Skin irritation induced by an RAR agonist, AGN 191183, 23 administered topically, can be blocked by an RAR antagonist, AGN
24 192869, also administered topically in hairless mice.
More particularly skin irritation was measured on a 26 semiquantitative scale by the daily subjective evaluation of skin flaking 27 and abrasions. A single number, the topical irritation score, summarizes 28 the skin irritation induced in an animal during the course of an 29 experiment. The topical irritation score is calculated as follows. The topical irritation score is the algebraic sum of a composite flaking score ._..- ~..~,.~~.~.... r 1 and a composite abrasion score. The composite scores range from 0-9 2 and 0-8 for flaking and abrasions, respectively, and take into account the 3 maximum severity, the time of onset, and the average severity of the 4 flaking and abrasions observed.
5 The severity of flaking is scored on a 5-point scale and the 6 severity of abrasions is scored on a 4- point scale, with higher scores 7 reflecting greater severity. The maximum severity component of the 8 composite scores would be the highest daily severity score assigned to a 9 given animal during the course of observation.
1 o For the time of onset component of the composite score, a score 11 ranging from 0 to 4 is assigned as follows:

13 Time to Appearance of Flaking or Abrasions 14 of Severity 2 or Greater 16 da s Time of Onset Score 24 The average severity component of the composite score is the sum of the daily flaking or abrasion scores divided by the number of 26 observation days. The first day of treatment is not counted, since the 27 drug compound has not had an opportunity to take effect at the time of 28 first treatment.
29 To calculate the composite flaking and ~hrasion scores, the average severity and time of onset scores are med and divided by 2.
31 The result is added to the maximal severity score. The composite 32 flaking and abrasion scores are then summed to give the overall topical 33 irritation score. Each animal receives a topical irritation score, and the 34 values are expressed as the mean ~ SD of the individual scores of a 1 group of animals. Values are rounded to the nearest integer.
2 Female hairless mice [CrI:SKH1-hrBR) (8-12 weeks old, n=6) 3 were treated topically for S consecutive days with acetone, AGN 191183, 4 AGN 192869, or some combination of AGN 192869 and 191183. Doses of the respective compounds are given in Table 7. Treatments are 6 applied to the dorsal skin in a total volume of 4 ml/kg ( ~ 0.1 ml). Mice 7 were observed daily and scored for flaking and abrasions up to and 8 including 3 days after the last treatment, i.e., day 8.

l0 Experimental Desi n and Results Example 1 11 Dose Dose Molar Ratio Topical 12 AGN 191183 AGN 192869 ( 192869: Irritation 13 G_rouu ~mgLkg,/~d (mg~g~) 19~ 1-183. Score A 0 0 -- 0~0 16 B 0.025 0 -- 8 ~ 2 17 C O.OZS 0.06 2:1 S + 2 18 D 0.025 0.30 10:1 2 ~ 1 19 E 0.025 1.S 50:1 1+_0 F 0 1.S -- 0 ~ 0 22 The topical irritation scores for Example 1 are given in Table 7.
23 Neither acetone (vehicle) nor AGN 192869 (antagonist) at a dose of 1.S
24 mg/kg/d (group F) caused observable topical irritation. AGN 191183, 2S the RAR agonist, caused modest topical irritation at a dose of 0.025 26 mg/kg/d. However, AGN 191183-induced topical irritation was inhibited 27 in a dose-dependent fashion by AGN 192869, with nearly complete 28 abrogation of irritation in the presence of a S0-fold molar excess of 29 AGN 192869. This demonstrates that a topical RAR antagonist blocks skin irritation caused by a topical RAR agonist. Complete blockade of 31 RAR agonist-induced skin irritation can be achieved with lower molar 32 ratios of antagonist to agonist when the RAR antagonists is more 33 potent, such as the compound 4-({S,6-dihydro-S,S- dimethyl-8-(4-_. .....__.. ..w_-,.-..~-..-...__,.,_.,.~...~.~.._ r, ..M_.._W.~-..
~.~m.~....~~...-.~~......_ 1 methylphenyl)-2- naphthalenyl)ethynyl)benzoic acid (AGN 193109, also 2 designated in this application as Compound 60.) 3 Example 2: skin iritation induced by orallX auulied agonist is blocked 4 with topically applied anta onist The potent RAR agonist AGN 191183 (4-((E)-2- (5,6,7,8-6 tetrahydro-5,5,8,8-tetramethylnaphthalen-2- yl)propen-1-yl)benzoic acid) 7 and the potent RAR antagonist 4-((5,6-dihydro-5,5-dimethyl-8-(4-8 methylphenyl)-2-naphthalenyl)ethynyl)benzoic acid (AGN 193109, 9 Compound 60) were used in this example and body weight of the experimental animals (mice) was used as a marker of systemic RAR
11 agonist exposure.
12 Groups of female hairless mice (8-12 weeks old, n=6) were 13 treated by intragastric intubation with corn oil or AGN 191183 (0.26 14 mg/kg) suspended in corn oil (5 ml/kg). Mice were simultaneously treated topically on the dorsal skin with vehicle (97.6% acetone/2.4%
16 dimethylsulfoxide) or solutions of AGN 193109 in vehicle (6 ml/kg).
17 Specific doses for the different treatment groups are give in Table 8.
18 Treatments were administered daily for 4 consecutive days. Mice were 19 weighed and graded far topical irritation daily as described in Example 1 up to and including 1 day after the last treatment. Percent body 21 weight change is calculated by subtracting final body weight (day 5) 22 from initial body weight (day 1), dividing by initial body weight, and 23 multiplying by 100%. Topical irritation scores are calculated as 24 described in Example 1.
Topical irritation scores and weight loss fo!~ ' he different groups 26 are given in Table 8. Combined treatment with tue topical and oral 27 vehicles, i.e., acetone and corn oil, respectively, caused no topical 28 irritation or weight loss. Similarly, combined treatment with the oral 29 vehicle and the topical antagonist AGN 193109 resulted in no topical irritation or weight loss. Oral AGN 191183 by itself induced substantial 1 weight loss and skin irritation. AGN 191183-induced skin irritation was 2 substantially reduced when combined with the lower dose of AGN
3 193109 and completely blocked at the higher dose of AGN 193109.
4 AGN 191183-induced weight loss was also blocked in a dose-related fashion by topical AGN 193109, but the blockade was not complete.
6 Thus, topical AGN 193109 preferentially blocked the dermal toxicity of 7 AGN 191183. Presumably, low levels of AGN 193109 were absorbed 8 systemically and thus partially blocked the weight loss induced by AGN
9 191183. However, such absorption would likely be even less in a species with less permeable skin, such as humans. Alternatively, the weight loss 11 inhibition by AGN 193109 could be due to amelioration of the AGN
12 191183 induced skin irritation.

14 Experimental Example 2 Design and Results Dose of TopicalDose of Oral % Weight Topical 16 AGN 193109 AGN 191183 Gain or 17 Irritation 18 Group ~m_gLkg(d"l ~mg~k~ Loss Score 21 B 0 0.26 (216) 81 22 C 0.12 0.26 (9 5) 1 1 23 D 0.47 0.26 (3 5) 0 1 24 E 0.47 0 33 00 26 Thus, Example 2 demonstrates that RAR antagonists 27 administered topically can be used to block preferentially the skin 28 irritation induced by an RAR agonist administered orally.
29 Example 3: topicall~pplied antagonist accelarates recovery from prexistin~ retinoid toxicity 31 In this example, weight loss is induced by topical treatment with 32 the RAR agonist AGN 191183 and then the test animals are topically 33 treated with either vehicle or the RAR antagonist AGN 193109.
_...,~. .-....... _w . T... ......~. ~...~ .. .......,....~ ...._w...

1 Female hairless mice (8-12 weeks old, n=5) were treated topically 2 with AGN 191183 (0.13 mg/kg/d) in vehicle (97.6% acetone/2.4%
3 DMSO, 4 ml/kg) daily for 2 days. Groups of these same mice (n=5) 4 were then treated topically either with vehicle or AGN 193109 in vehicle (4 ml/kg) daily for 3 consecutive days beginning on day 3. Mice 6 were weighed on days 1-5 and on day 8. Body weights are expressed as 7 the mean ~ SD. Means were compared statistically using an unpaired, 8 two-tailed t-test. Differences were considered significant at P < 0.05.

l0 Results, Example 3 11 Treatment Body Weight (g) 12 (days 3-5) vehicle 24.6 t 1.5 23.9 t 1.2 21.4 t 1.2 20.3 t 1.7 21.0 f 1.4 24.7 t 1.0 16 AGN 193109 23.9 t 1.0 23.5 t 1.2 21.4 t 0.6 22.2 t 0.7 22.8 t 0.8 25.0 t 1.1 18 The time course of body weights in Example 3 are given in Table 19 9. Body weights of both groups of mice were lowered in parallel on days 2 and 3 as a result of AGN 191183 treatment on days 1 and 2.
21 Body weights in the two groups were not significantly different on days 22 l, 2, or 3. However, AGN 193109 treatment significantly increased 23 body weight relative to vehicle treatment on days 4 and 5. These data 24 indicated that recovery from AGN 191183-induced body weight loss was accelerated by subsequent treatment with AGN 193109. Body weights 26 were not significantly different between the two groups of mice on day 27 8, indicating that full recovery was achievable in both groups given 28 sufficient time. Thus, RAR antagonists are effective in alleviating RAR
29 agonist-induced toxicity even if RAR agonist-induced toxicity precedes RAR antagonist treatment, i.e., in the RAR agonist poisoning scenario.
31 Example 4: orall~applied antagonist blocks hxpertrig_lyceridemia 32 incuded by orally coadministered retinoid agonist 33 5-((E}-2-(5,6,7,8-tetrahydro-3,5,5,8,8-pentamethylnaphthalen-2-1 yl)propen-1-yl)-2- thiophencarboxylic acid, is a known RAR/RXR pan-t agonist (see United States Patent No. 5,324,840 column 32) and is 3 designated AGN 191659. This compound was used orally to induce 4 acute hypertriglyceridemia in rats, and AGN 193109 Compound 60 was 5 coadministered orally to block the AGN 191659-induced 6 hypertriglyceridemia.
7 Male Fischer rats (6-7 weeks old, n=5) were treated by 8 intragastric incubation with corn oil (vehicle), AGN 191659, AGN
9 193109 or a combination of AGN 191659 and AGN 193109. AGN
10 191659 and AGN 193109 were given as fine suspensions in corn oil.
11 The experimental design, including doses, is given in Table 10.
12 Blood was withdrawn from the inferior vena cava under carbon 13 dioxide narcosis. Serum was separated from blood by low speed 14 centrifugation. Total serum triglycerides (triglycerides plus glycerol) 15 were measured with a standard spectrophotometric endpoint assay 16 available commercially as a kit and adapted to a 96-well plate format.
17 Serum triglyceride levels are expressed as the mean ~ SD. Means were 18 compared statistically by one-way analysis of variance followed by 19 Dunnett's test if significant differences were found. Differences were 20 considered significant at P < 0.05.
21 As shown in Table 10, AGN 191659 by itself caused significant 22 elevation of serum triglycerides relative to vehicle treatment. AGN
23 193109 by itself did not significantly increase serum triglycerides.
24 Importantly, the combination of AGN 193109 and AGN 191659 at 25 molar ratios of I:1 and 5:1 reduced serum triglycerides to levels that 26 were not signficantly different from control.

2 E~erimental Des~n and Results. Example 4 3 Group Treatment ydose), Serum Triglycerides i(mg~) A vehicle 55.0 ~ 3:1 6 B AGN 193109 ( 19.6 mg/kg) 52.4 ~ 6.3 7 C AGN 191659 (3.7 mg/kg) 122.5 ~ 27.6 8 D AGN 193109 (3.9 mg/kg) 55.7 ~ 14.7 9 + AGN 191659 (3.7 mg/kg) E AGN 193109 (19.6 mg/kg) 72.7 ~ 8.9 11 + AGN 191659 (3.7 mg/kg) 13 Example 4 demonstrates that an RAR antagonist can be used to 14 block hypertriglyceridemia induced by a coadministered retinoid.
Example 5: parenterally applied antagonist blocks bone toxicity incuded 16 b~parenterally coadministered retinoid agonist 17 Example 5 demonstrates that RAR antagonists can block bone 18 toxicity induced by an RAR agonist. In this example, AGN 193109 is 19 used to block premature epiphyseal plate closure caused by a coadministered RAR agonist, AGN 191183, in guinea pigs.
21 Groups of male Hartley guinea pigs ( ~ 3 weeks old, n=4) were 22 implanted intraperitoneally with osmotic pumps containing vehicle {20%
23 dimethylsulfoxide/80% polyethylene glycol-300), AGN 191183 (0.06 24 mg/ml), or AGN 191183 (0.06 mg/ml) in combination with AGN 193109 (0.34 mg/ml). The osmotic pumps are designed by the manufacturer to 26 deliver ~ 5 ,ul of solution per hour continuously for 14 days.
27 The animals were euthanized by carbon dioxide asphyxiation 14 28 days after implantation. The left tibia was was removed and placev in 29 10% buffered formalin. The tibias were decalcified by exposure t~
formic acid/formalin solution for 3-4 days, and paraffin sections were 31 prepared. Sections were stained with hematoxylin and eosin by standard 32 methods. The proximal tibial epiphyseal plate was examined and scored 33 as closed or not closed. Epiphyseal plate closure is defined for this 1 purpose as any interruption of the continuity of the epiphyseal growth 2 plate cartilage, i.e., replacement by bone and/or fibroblastic tissue.
3 None of the four vehicle-treated guinea pigs showed epiphyseal 4 plate closure by the end of the experiment. This was expected, since the proximal epiphyseal plate of guinea pig tibia does not normally close 6 until the animals are at least 10 months old. All four of the AGN
7 191183-treated guinea pigs showed partial or complete epiphyseal plate 8 closure. However, none of the guinea pigs treated with the combination 9 of AGN 191183 and AGN 193109 demonstrated epiphyseal plate closure. Thus, AGN 193109 at a 5-fold molar excess completely 11 blocked AGN 191183-induced bone toxicity when these compounds 12 were coadministered parenterally.
13 RAR Antagonist Compounds 14 The compounds 4-((5,6-dihydro-5,5-dimethyl-8-(4-methylphenyl)-2- naphthalenyl)ethynyl]benzoic acid (AGN 193109, 16 Compound 60) and 4-((5,6-dihydro-5,5-dimethyl- 8-(phenyl)-2-17 naphthalenyl)ethynyl)benzoic acid (AGN 192869, Compound 60a) are 18 examples of RAR antagonists which were used in the above-described 19 animal tests for blocking RAR receptors in accordance with the present invention. The compounds of the following formula (Formula 1) serve 21 as further and general examples for additional RAR antagonist 22 compounds for use in accordance with the present invention.

~4 t~1 Formula 1 SU8ST1TUTE SHEET (RULE 26) . .._....... -~.m-_....,. ... T "..... .~ .~.... ..: ...... ...........~.. ...

1 In Formula 1, X is S, O, NR' where R' is H or alkyl of 1 to 6 2 carbons, or 3 X is [C(R,)2]~ where R, is H or alkyl of 1 to 6 carbons, and n is 4 an integer between 0 or 1;
R2 is hydrogen, lower alkyl of 1 to 6 carbons, F, CF3, fluor 6 substituted alkyl of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, or 7 alkylthio of 1 to 6 carbons;
8 R3 is hydrogen, lower alkyl of 1 to 6 carbons or F;
9 m is an integer having the value of 0 - 3;
o is an integer having the value of 0 - 3;
11 Z is -C--__C-, 12 -N=N-, 13 -N=CR,-, 14 -CR, = N, -(CR,=CR,)~.- where n' is an integer having the value 0 - 5, 16 -CO-NR,-, 17 -CS-NR,-, 18 -NR,-CO, 19 -NR,-CS, -COO-, 21 -OCO-;
22 -CSO-;
23 -OCS-;
24 Y is a phenyl or naphthyl group, or heteroaryl selected from a group consisting of pyridyl, thienyl, fnr3~l, pyridazinyl, pyrimidinyl, 2b pyrazir; l, thiazolyl, oxazolyl, imidazolyl and pyrrazolyl, said phenyl and 27 heteroaryl groups being optionally substituted with one or two R2 28 groups, or 29 when Z is -(CR,=CR,)~~- and n' is 3, 4 or 5 then Y represents a direct valence bond between said (CRz=CR2)n, group and B;

1 A is (CH2}q where q is 0-5, lower branched chain alkyl having 3-6 2 carbons, cycloalkyl having 3-6 carbons, alkenyl having 2-6 carbons and 1 3 or 2 double bonds, alkynyl having 2-6 carbons and 1 or 2 triple bonds;
4 B is hydrogen, COOH or a pharmaceutically acceptable salt thereof, COOR8, CONR9R,o, -CH20H, CHZOR", CH20COR", CHO, 6 CH(OR12)2, CHOR130, -CORD, CR~(OR12)2, CR,OR,30, or tri-lower 7 alkylsilyl, where R, is an alkyl, cycloalkyl or alkenyl group containing 1 8 to 5 carbons, R8 is an alkyl group of 1 to 10 carbons or 9 trimethylsilylalkyl where the alkyl group has 1 to 10 carbons, or a l0 cycloalkyl group of 5 to 10 carbons, or R$ is phenyl or lower 11 alkylphenyl, R9 and Rlo independently are hydrogen, an alkyl group of 1 12 to 10 carbons, or a cycloalkyl group of 5-10 carbons, or phenyl or lower 13 alkylphenyl, R" is lower alkyl, phenyl or lower alkylphenyl, R,2 is lower 14 alkyl, and R1~ is divalent alkyl radical of 2-5 carbons, and R,4 is (R,5)~ phenyl, (R,s)anaphthyl, or (R,5)~ heteroaryl where 16 the heteroaryl group has 1 to 3 heteroatoms selected from the group 17 consisting of O, S and N, r is an integer having the values of 0 - 5, and 18 R,5 is independently H, F, Cl, Br, I, N02, N(R$)2, N(R$)CORB, 19 NR$CON(R8)z, OH, OCORB, ORg, CN, an alkyl group having 1 to 10 carbons, fluoro substituted alkyl group having 1 to 10 carbons, an 21 alkenyl group having 1 to 10 carbons and 1 to 3 double bonds, alkynyl 22 group having 1 to 10 carbons and 1 to 3 triple bonds, or a trialkylsilyl or 23 trialkylsilyloxy group where the alkyl groups independently have 1 to 6 24 carbons.
Synthetic Methods - Aryl Substituted Compounds 26 The exemplary RAR antagonist compounds of Formula 1 can be 27 made by the synthetic chemical pathways illustrated here. The synthetic 28 chemist will readily appreciate that the conditions set out here are 29 specific embodiments which can be generalized to any and all of the compounds represented by Formula 1.
._ ~.-...~..-~. . ... ..... . ~ . . ....._ .~w~..__.- w_.-W~..... ..

6 r 7 _ _ I AIC~3 / Brl ~o I
8 R, (Rslm R, R, \ i~lm 9 Fbmula s Fo mina 7 10 H - SiMe~
Fit ~ MpBr I 1 Et20 PdCIZiPPHg~
EZ3N! Cot/70' C

13 Ho , 'M~a i~a~o ' I i~ ~ I
15 ~ a (~)rn ~ (Rs?m 16 ~~~ ~ s Fom~uta 8 18 pTsOH
a iC2C03 MeOH

20 R.. o 21 r /
22 i~~ w_ I i~ ~ I
23 R' R' ~~ Ry a, v~~Irn 24 ~~~ ~ s Fortnult 9 Reaction Scheme 1 4 ~

PdCl2(PPh3~
6 Cul/Et3N/70~C
(R~lo 7 ~ ~(Rx)m R~ ~ Y(~)"A-B, g Formila 10 __ Formula 9 Fo rrra~la 11 rJarJ(Sa~le3n ~Fias~C

Homologs and 12 Derivatives 13 oNa Y(F5Z) ~ ~ ,(~)m R, Fom~uta 12 a Cr4z A~e 21 R14 / Y(R~) 22 , , ~ Rl4v,G
23 (R~)o \ ~ Pd(PPh3k 24 R~ R1 ~(~,m ~F
Fomwta 14 Formula 13 27 Homologs and Derivatives Reaction Scheme 1 (continued) ... _ _... _._,...~....~.,-,~, ._... _ .. .. . , _ ...~-~..m~..~ ..~~.-.~...~~.._...~...

1 Reaction Scheme 1 illustrates the synthesis of compounds of 2 Formula 1 where the Z group is an ethynyl function (-C=C-) and X is 3 [C(R,)2)n where n is 1. In other words, Reaction Scheme 1 illustrates 4 the synthesis of ethynyl substituted dihydronaphthalene derivatives of the present invention. In accordance with this scheme, a 6 tetrahydronaphtalene-1-one compound of Formula 6 is brominated to 7 provide the bromo derivative of Formula 7. The compounds of Formula 8 6 already carry the desired R,, R2 and R3 substituents, as these are 9 defined above in connection with Formula 1. A preferred example of a compound of Formula 6 is 3,4-dihydro-4,4-dimethyl-1(2H)-11 naphthalenone, which is described in the chemical literature (Arnold et 12 al. J. Am. Chem. Soc. 69: 2322 - 2325 (1947)). A presently preferred 13 route for the synthesis of this compound from 1-bromo-3-phenylpropane 14 is also described in the experimental section of the present application.
The compounds of Formula 7 are then reacted with 16 (trimethylsilyl)acetylene to provide the (trimethylsilyl)ethynyl-17 substituted 3,4-dihydro- naphthalen-1(2H)-one compounds of Formula 18 8. The reaction with (trimethylsilyl)acetylene is typically conducted 19 under heat (approximately 100°C) in the presence of cuprous iodide, a suitable catalyst, typically having the formula Pd(PPh3)2C12, an acid 21 acceptor (such as triethylamine) under an inert gas (argon} atmosphere.
22 Typical reaction time is approximately 24 hours. The 23 (trimethylsilyl)ethynyl- substituted 3,4-dihydro-naphthalen-1(2H)-one 24 compounds of Formula 8 are then reacted with base (potassium h~~droxide or potassium carbonate) in an alcoholic solvent, such as 26 r~_ Lhanol, to provide the ethynyl substituted 3,4-dihydro-1-27 naphthalen-1(2H)ones of Formula 9. Compounds of Formula 9 are 28 then coupled with the aromatic or heteroaromatic reagent X,-Y(R2)-A-29 B' (Formula 10) in the presence of cuprous iodide, a suitable catalyst, typically Pd(PPh3)2C12, an acid acceptor, such as triethylamine, under 1 inert gas (argon) atmosphere. Alternatively, a zinc salt (or other 2 suitable metal salt) of the compounds of Formula 9 can be coupled with 3 the reagents of Formula 10 in the presence of Pd(PPh3)4 or similar 4 complex. Typically, the coupling reaction with the reagent X1-Y(R2)-A-B' (Formula 10) is conducted at room or moderately elevated 6 temperature. Generally speaking, coupling between an ethynylaryl 7 derivative or its zinc salt and a halogen substituted aryl or heteroaryl 8 compound, such as the reagent of Formula 10, is described in United 9 States Patent No. 5,264,456, the specification of which is expressly incorporated herein by reference. The compounds of Formula 11 are I 1 precursors to exemplary compounds of the present invention, or 12 derivatives thereof protected in the B' group, from which the protecting 13 group can be readily removed by reactions well known in the art. The 14 compounds of Formula 11 can also be converted into further precursors to the exemplary compounds by such reactions and transformations 16 which are well known in the art. Such reactions are indicated in 17 Reaction Scheme 1 by conversion into "homologs and derivatives". One 18 such conversion employed for the synthesis of several exemplary 19 compounds is saponification of an ester group (when B or B' is an ester) to provide the free carboxylic acid or its salt.
21 The halogen substituted aryl or heteroaryl compounds of Formula 22 10 can, generally speaking, be obtained by reactions well known in the 23 art. An example of such compound is ethyl 4-iodobenzoate which is 24 obtainable, for example, by esterification of 4-iodobenzoic acid.
Another example is ethyl 6-iodonicotinoate which can be obtained by 26 conducting a halogen exchange reaction on 6-chloronicotinic acid, 27 followed by esterification. Even more generally speaking, regarding 28 derivatization of compounds of Formula 11 and/or the synthesis of aryl 29 and heteroaryl compounds of Formula 10 which can thereafter be reacted with compounds of Formula 9, the following well known and ... ._....",..._".""».,".,..~......»»~».....-...,.~»"",-."........._ T.
........u".-"....,»."","~.........~.w....»-_........_..

1 published general principles and synthetic methodology can be 2 employed.
3 Carboxylic acids are typically esterified by refluxing the acid in a 4 solution of the appropriate alcohol in the presence of an acid catalyst such as hydrogen chloride or thionyl chloride. Alternatively, the 6 carboxylic acid can be condensed with the appropriate alcohol in the 7 presence of dicyclohexylcarbodiimide and dimethylaminopyridine. The 8 ester is recovered and purified by conventional means. Acetals and 9 ketals are readily made by the method described in March, Advanced l0 Organic Chemistry, 2nd Edition, McGraw-Hill Book Company, p. 810).
11 Alcohols, aldehydes and ketones all may be protected by forming 12 respectively, ethers and esters, acetals or ketals by known methods such 13 as those described in McOmie, Plenum Publishing Press, 1973 and 14 Protecting Groups, Ed. Greene, John Wiley & Sons, 1981.
To increase the value of n in the compounds of Formula 10 16 before affecting the coupling reaction of Reaction Scheme 1 (where 17 such compounds corresponding to Formula 10 are not available from a 18 commercial source) aromatic or heteroaromatic carboxylic acids are 19 subjected to homologation by successive treatment under Arndt-Eistert conditions or other homologation procedures. Alternatively, derivatives 21 which are not carboxylic acids may also be homologated by appropriate 22 procedures. The homologated acids can then be esterified by the 23 general procedure outlined in the preceding paragraph.
24 Compounds of Formula 10, (or other intermediates or exemplary compounds) where A is an aia;z~yl group having one or core double 26 bonds can be made for exampa,, by synthetic schemes well known to the 27 practicing organic chemist; for example by Wittig and like reactions, or 28 by introduction of a double bond by elimination of halogen from an 29 alpha-halo-arylalkyl- carboxylic acid, ester or like carboxaldehyde.
Compounds of Formula 10 (or other intermediates or exemplary 1 compounds) where the A group has a triple (acetylenic) bond can be 2 made by reaction of a corresponding aromatic methyl ketone with 3 strong base, such as lithium diisopropylamide, reaction with diethyl 4 chlorophosphate and subsequent addition of lithium diisopropylamide.
5 The acids and salts derived from compounds of Formula 11 (or 6 other intermediates or exemplary compounds) are readily obtainable 7 from the corresponding esters. Basic saponification with an alkali metal 8 base will provide the acid. For example, an ester of Formula 11 (or 9 other intermediates or exemplary compounds) may be dissolved in a 10 polar solvent such as an alkanol, preferably under an inert atmosphere 11 at room temperature, with about a three molar excess of base, for 12 example, lithium hydroxide or potassium hydroxide. The solution is 13 stirred for an extended period of time, between 15 and 20 hours, 14 cooled, acidified and the hydrolysate recovered by conventional means.
15 The amide may be formed by any appropriate amidation means 16 known in the art from the corresponding esters or carboxylic acids. One 17 way to prepare such compounds is to convert an acid to an acid chloride 18 and then treat that compound with ammonium hydroxide or an 19 appropriate amine.
20 Alcohols are made by converting the corresponding acids to the 21 acid chloride with thionyl chloride or other means (J. March, Advanced 22 Organic Chemistry, 2nd Edition, McGraw-Hill Book Company), then 23 reducing the acid chloride with sodium borohydride (March, Ibid, p.
24 1124), which gives the corresponding alcohols. Alternatively, esters may 25 be reduced with lithium aluminum hydride at reduced temperatures.
26 Alkylating these alcohols with appropriate alky halides under 27 Williamson reaction conditions (March, Ibid, p. 357) gives the 28 corresponding ethers. These alcohols can be converted to esters by 29 reacting them with appropriate acids in the presence of acid catalysts or 3o dicyclohexylcarbodiimide and dimethylaminopyridine.
_.. ___ . .. _.w .. .~.......~ ~ _._...-~..~ ~...... ........A. . _. ~ .
_....... ......~.~....~.~......~~ .

1 Aldehydes can be prepared from the corresponding primary 2 alcohols using mild oxidizing agents such as pyridinium dichromate in 3 methylene chloride (Corey, E. J., Schmidt, G., Tet. Lett. 399, 1979), or 4 dimethyl sulfoxide/oxalyl chloride in methylene chloride (Omura, K., Swern, D., Tetrahedron 34: 1651 (1978)).
6 Ketones can be prepared from an appropriate aldehyde by 7 treating the aldehyde with an alkyl Grignard reagent or similar reagent 8 followed by oxidation.
9 Acetals or ketals can be prepared from the corresponding l0 aldehyde or ketone by the method described in March, Ibid, p. 810.
11 Compounds of Formula 10 (or other intermediates, or exemplary 12 compounds) where B is H can be prepared from the corresponding 13 halogenated aromatic or hetero aromatic compounds, preferably where 14 the halogen is I.
Referring back again to Reaction Scheme l, the compounds of 16 Formula 11 are reacted with sodium bis(trimethylsilyl)amide and 2-17 ~N,N-bis(trifluoromethylsulfonyl)amino-5-chloropyridine in an inert 18 ether type solvent, such as tetrahydrofuran, at low temperatures (-78°C
19 and 0°C). This is shown in Reaction Scheme 1 where the usually unisolated sodium salt intermediate is shown in brackets as Formula 12.
21 The reaction results in the trifluoromethylsulfonyloxy derivatives 22 represented in Formula 13. (Tf = S02CF3). The compounds of 23 Formula 13 are then converted to the exemplary compounds of the 24 invention, shown in Formula 14, by reaction with an organometal derivative derived from th a aryl or vteroaryl compound R,4H, such ~ :'-:at 26 the formula of the organometal derivative is R,4Met (Met stands fo.
27 monovalent metal), preferably R,QLi. (R,4 is defined as in connection 28 with Formula 1.) The reaction with the organometal derivative, 29 preferably lithium derivative of the formula R,4Li is usually conducted in an inert ether type solvent (such as tetrahydrofuran) in the presence 1 of zinc chloride (ZnCl2) and tetrakis(triphenylphosphine)-palladium(0) 2 (Pd(PPh3)4). The organolithium reagent Rl4Li, if not commercially 3 available, can be prepared from the compound R,4H (or its halogen 4 derivative R,4 X, where X, is halogen) in an ether type solvent in accordance with known practice in the art. The temperature range for 6 the reaction between the reagent R,4Li and the compounds of Formula 7 13 is, generally speaking in the range of approximately -78°C to 50°C.
8 The compounds of Formula 14 can be converted into further homologs 9 and derivatives in accordance with the reactions discussed above.
The intermediate 7-bromo-tetrahydronaphthalene-1- one 11 compounds of Formula 7 shown in Reaction Scheme 1 can also be 12 converted with a Grignard reagent of the formula R,QMgBr (R,4 is 13 defined as in connection with Formula 1) to yield the tertiary alcohol of 14 Formula 15. The tertiary alcohol is dehydrated by treatment with acid to provide the 3,4-dihydro-7-bromonaphthalene derivatives of Formula 16 16, which serve as intermediates for the synthesis of additional 17 compounds of the present invention (see Reaction Schemes 6, 7, and 8).
...... ~ .. ~..,-~ . ~"~,... .. , . ... ~ ....~ ~-~..,.....-._..._ . .

3 r / : / r 4 \ ~ Brc(~ho(~)2oo2H
HX ~ Fomxla'18 (R~ \
~n &ue (gym Fomuta 17 Fomxila 19 H;/e O 'Men O
/ H =--~;Me~ / ( r 11 ~° ~ ~ (fro Rj~ PdG2(PPh3~ ' 12 Fom~ula21 Cul/Et,3Nl70'C
Fomiula20 lC2C03 14 MeOH
O ~A
16 / o Y(r~) / PdCl2(PPh3~
17 (~° \ ~ Cul/Et3N/70'C (~ /
(~>~, \ i I 8 X~'Y(Rs)'A--B' (Rikn Fom~ula22 19 Formula 10 t. NaN(SiMe3~
Homologs and ,~F /'g. C
Derivatives 21 2, ci R" Y(~ Tf Y(Ri) 24 Rl~a / / ( Pd(PPh3)4 (~) / /
2 5 ~ ~ THF ! 50° C
26 Fomwta25 ~ Formula24 27 Homolo9s and Dsrivativ~s Reaction Scheme 2 SUBSTITUTE SHEET (RULE 26) 1 Referring now to Reaction Scheme 2 a synthetic route to those 2 compounds is disclosed where with reference to Formula 1 X is S, O or 3 NR' and the Z group is an ethynyl function (-C---C-). Starting material 4 for this sequence of the reaction is a bromophenol, bromothiophenol or bromoaniline of the structure shown in Formula 17. For the sake of 6 simplifying the present specification, in the ensuing description X can be 7 considered primarily sulfur as for the preparation of benzothiopyran 8 derivatives. It should be kept in mind, however, that the herein 9 described scheme is also suitable, with such modifications which will be readily apparent to those skilled in the art, for the preparation of 11 benzopyran (X = O) and dihydroquinoline (X = NR') compounds of 12 the present invention. Thus, the compound of Formula 17, preferably 13 para bromothiophenol, para bromophenol or para bromoaniIine is 14 reacted under basic condition with a 3-bromo carboxylic acid of the Formula 18. In this reaction scheme the symbols have the meaning 16 described in connection with Formula 1. An example for the reagent of 17 Formula 18 where R3 is hydrogen, is 3-bromopropionic acid. The 18 reaction with the 3-bromocarboxyiic acid of Formula 18 results in the 19 compound of Formula 19. The latter is cyclized by treatment with acid to yield the 6-bromothiochroman-4-one derivative (when X is S) or 6-21 bromochroman derivative (when X is 0) of Formula 20. The bromo 22 compounds of Formula 20 are then subjected to substantially the same 23 sequence of reactions under analogous conditions, which are described 24 in Reaction Scheme 1 for the conversion of the bromo compounds of Formula 7 to the compounds of the invention. Thus, briefly 26 summarized here, the bromo compounds of Formula 20 are reacted 27 with (trimethylsilyl)acetylene to provide the 6- (trimethylsilyl)ethynyl-28 substituted thiochroman-4-one or chroman-4-one compounds of 29 Formula 21. The 6- (trimethylsilyl)ethynyl- substituted thiochroman-4-one compounds of Formula 21 are then reacted with base (potassium 1 hydroxide or potassium carbonate) to provide the ethynyl substituted 6-2 ethynyl substituted thiochroman-4-ones of Formula 22. Compounds of 3 Formula 22 are then coupled with the aromatic or heteroaromatic 4 reagent X1-Y(R2)-A-B' (Formula 10) under conditions analogous to 5 those described for the analogous reactions of Reaction Scheme 1, to 6 yield the compounds of Formula 23.
7 The compounds of Formula 23 are then reacted still under 8 conditions analogous to the similar reactions described in Reaction 9 Scheme 1 with sodium bis(trimethylsilyl)amide and 2-(N,N-10 bis(trifluoromethylsulfonyl)amino]-S-chloropyridine to yield the 4-11 trifluoromethylsulfonyloxy benzothiopyran or benzopyran derivatives 12 represented in Formula 24. The compounds of Formula 24 are then 13 converted to compounds shown in Formula 25, by reaction with an 14 organometal derivative derived from the aryl or heteroaryl compound 15 R,4H, as described in connection with Reaction Scheme 1.
16 Similarly to the use of the intermediate 7-bromo-17 tetrahydronaphthalene-1-one compounds of Formula 7 of Reaction 18 Scheme 1, the intermediate 6-bromothiochroman- 4-one compounds of 19 Formula 20 can also be used for the preparation of further compounds 20 within the scope of the present invention, as described below, in 21 Reaction Schemes 6, 7 and 8. The compounds of Formula 25, can also 22 be converted into further homologs and derivatives, in reactions 23 analogous to those described in connection with Reaction Scheme 1.

O TMS
O
6 / ~ H - =:Me, /
\ ~ PdCl2(PPh3yZ (F~o \
7 (Ra)o Cul/ Et3N l70'C
8 Fomula26 ~~~2~

MeOH

O PdG2(PPh3~ p Cull E1;3N/70'C
14 (Ri)o \ ~ X; Y(Ftz)-A-H' \
Fom~ula 10 Fom~ula29 Fomx~la28 17 1. NaN(SiMe3~
TNF / 78' C
18 2~ '~~~~ _ 19 ~''' 21 p~ Yes) Rl4ZnC1 22 ~ / Pd(PPh3)4 23 two ~~ T~tF / sa' c t~~
24 Fom~la30 Fomula3l Homdo~s arid 26 Derivatives Reaction Scheme 3 SUBSTtTUTE SHEET (RULE 26) _..__~..~.~..~~~~wH . ...~...~ ._ . .. , .....,~ _. ~e..4 ..~.~.w..~.~.~w_.

1 Reaction Scheme 3 discloses a synthetic route to compounds 2 where, with reference to Formula 1, X is (C(Rj)2~n, n is 0 and the Z
3 group is an ethynyl function (-C--_C-). In accordance with this scheme, 4 a 6- bromo-2,3-dihydro-1H-inden-1-one derivative of Formula 26 is reacted in a sequence of reactions (starting with reaction with 6 trimethylsilylacetylene) which are analogous to the reactions described 7 above in connection with Reaction Schemes 1 and 2, to provide, 8 through intermediates of the formulas 27 - 30, the indene derivatives of 9 Formula 31. In a preferred embodiment within the scope of Reaction Scheme 3, the starting material is 6-bromo-2,3-dihydro-3,3-dimethyl-1H-11 inden-1-one that is available in accordance with the chemical literature 12 (See Smith et al. Org. Prep. Proced. Int. 1978 10, 123-131). Compounds 13 of Formula 26, such as 6-bromo-2,3-dihydro-3,3-dimethyl-1H-inden-1-14 one, can also be used for the synthesis of still further exemplary compounds for use in the present invention, as described below.

~. ~ I R~a~a /
3 ~ '~' ~ ~ . ots',arisomsr 2 Ct03 4 ~' /~~~" HOAc i ArtO
Fomuia32 Fomufa33 HOCH2CH20Y~
pTaOH I a R' off o 0 / ~R, Rl4MgBr 11 ~ .~ ~ .~E
I2 ~ ~(~~ El2o R~ R, 13 Fom~ula 35 Fom~uta34 pTsOH
a 17 ~ ~ c~~» cH
R,. o ~ R,. R, ~ Formula 37 ( ~ / /
R
19 ~ ~ ~ r~aH ITHF W
~(~~n 2 DIBAL-H ~ ~ (~~m R~ Ft, 21 Fommta 36 Fom,uta 38 O ii, 23 Formula 39 24 I.DA I THF
R,. R, R, Homologs and E (~ / / I ~ ~ w 2'7 Derivatives R, ~ ~~~(~M, 28 R, R, 29 Fomails~to Reaction Scheme 4 '..~_..._.~ ~...".~w.... ._ . , . .._'..._-..~.._..,~ ~.m.'._._..

1 Referring now to Reaction Scheme 4 a synthetic route to 2 exemplary compounds is disclosed where, with reference to Formula 1, 3 Z is -(CR,=CR,)"~ , n' is 3, 4 or 5 and Y represents a direct valence 4 bond between the (CR1=CR,)~, group and B. This synthetic route is described for examples where the X group is [C(R1)z~~ and n is 1 6 (dihydronaphthalene derivatives). Nevertheless, it should be understood 7 that the reactions and synthetic methodology described in Reaction 8 Scheme 4 and further ensuing schemes, is also applicable, with such 9 modifications which will be readily apparent to those skilled in the art, l0 to derivatives where X is is S, O, NR' (benzothiopyran, benzopyran or 11 dihydroquinoline derivatives) or [C(R,)z~n and n is 0 (indene 12 derivatives).
13 In accordance with Reaction Scheme 4, a 1,2,3,4-14 tetrahydronaphthalene derivative of Formula 32 is reacted with an acid chloride (R,COCI) under Friedel Crafts conditions, and the resulting 16 acetylated product is oxidized, for example in a Jones oxidation 17 reaction, to yield a mixture of isomeric 6- and 7- acetyl-I(2H)-18 naphthalenone derivatives of Formula 33. In a specific preferred 19 example of this reaction, the starting compound of Formula 32 is 1,2,3,4-tetrahydro-1,1- dimethylnaphthalene (a known compound) which 21 can be prepared in accordance with a process described in the 22 experimental section of the present application. The 7-acetyl-1(2H)-23 naphthalenone derivative of Formula 33 is reacted with ethylene glycol 24 in the presence of acid to protect the oxo function of the exocyclic ketone moiety, as a ket;~.l derivative of Formula 34. The ketal of 26 Formula 34 is thereafter reacted with a Grignard reagent of the formula 27 R,4MgBr (the symbols are defined as in connection with Formula 1), to 28 yield the tertiary alcohol of Formula 35. Thereafter the dioxolane 29 protective group is removed and the tertiary alcohol is dehydrated by treatment with acid to provide the 3,4- dihydro-7-acetylnaphthalene 1 derivatives of Formula 36. The ketone function of the compounds of 2 Formula 36 is subjeceted to a Horner Emmons {or analogous) reaction 3 under strongly alkaline conditions with a phosphonate reagent of 4 Formula 37, to yield, after reduction, the aldehyde compounds of 5 Formula 38. Still another Horner Emmons (or analogous) reaction 6 under strongly alkaline conditions with a reagent of Formula 39 7 provides compounds of Formula 40. The latter can be converted into 8 further homologs and derivatives in accordance with the reactions 9 described above. A specific example of the Horner Emmons reagent of 10 Formula 37 which is used for the preparation of a preferred compound 11 is diethylcyanomethylphosphonate; an example of the Horner Emmons 12 reagent of Formula 39 is diethyl-(E)-3- ethoxycarbonyl-2-13 methylallylphosphonate.

._.._.... _ .. _. _ ............._,.. y...~wa ~ . .e~..~.,..~m ._ . . T.._.~ .
....-.~, ~ ~~ ...,.... . .m.. ....,s / O
6 ~ \ I H~ ~ / I ~ s ~(~kr1 H2S04 ~xltn 7 R' R' R, R, Fontula6 Formula 41 H2/Pd EtOAe 11 O ~ O
12 / ~Y~ HOAc (~ /
13 ~~° \ i \
ON-Y(F~)-A-$ (R=~
R R ~~ Formula 43 R~ R~
14 > >
Fo~~'~'1 Formula 42 16 1. NaN(SiMe3y2 THF I~78' C
17 2. c s Tt ~A'e R,.
/ / ~Y~) Rl4ZnCl / / ( iY ) 21 (R~° ~ Pd(PPh3)1 (tea \ ---~ \
22 R ~~ hIFISO'c ~ ~ t Fomula 45 Fomuta 16 Homdogs and Deriva~ves Reaction Scheme 5 1 Reaction Scheme 5 discloses a synthetic process for preparing 2 compounds where the Z group is an azo group (-N=N-). As in 3 Reaction Scheme 4 this process is described for examples where the X
4 group is [C(R,)2]~ and n is 1 (dihydronaphthalene derivatives).
Nevertheless, it should be understood that the synthetic methodology 6 described is also applicable, with such modifications which will be 7 readily apparent to those skilled in the art, to all azo compounds for use 8 in the invention, namely to derivatives where X is is S, O, NR' 9 (benzothiopyran, benzopyran or dihydroquinoline derivatives) or [C(R,)2]n and n is 0 (indene derivatives). Thus, a nitro group is 11 introduced into the starting compound of Formula 6 under substantially 12 standard conditions of nitration, to yield the 3,4-dihydro-7-nitro-1(2H)-13 naphthalenone derivative of Formula 41. The latter compound is 14 reduced to the 3,4-dihydro-7-amino-1(2H)-naphthalenone derivative of Formula 42 and is thereafter reacted with a nitroso compound of the 16 formula ON-Y(R2)-A-B (Formula 43) under conditions normally 17 employed (glacial acetic acid) for preparing azo compounds. The 18 nitroso compound of Formula 43 can be obtained in accordance with 19 reactions known in the art. A specific example for such compound, which is used for the synthesis of a preferred compound is ethyl 4-21 nitrosobenzoate. The azo compound of Formula 44 is thereafter 22 reacted with sodium bis(trimethylsilyl)amide and 2-[N,N-23 bis(trifluoromethylsulfonyl)amino]-5-chloropyridine to yield the 4-24 trifluoromethylsulfonyloxy derivatives represented in Formula 45. The compounds of Formula 45 are then converted to the azo compounds 26 shown in Formula 46, by reaction with an organometalic derivative 27 derived from the aryl or heteroaryl compound R,4H. These latter two 28 reactions, namely the conversion to the 4-trifluoromethylsulfonyloxy 29 derivatives and subsequent reaction with the organometal derivative, have been described above in connection with Reaction Schemes 1, 2 m.._..~_ r 1 and 3, and are employed in several presently preferred synthetic 2 processes leading to exemplary RAR antagonist compounds.

R~
r t. t-BuLi / / THF / 78' ~ / /
9 (~Jo ~ i \ 2. ~ - \
R )m 'f~(R~M~
R, 11 Fom~ula l fi Fo m~ula 47 12 _ 13 ~-Y(~)-A~
Formula 48 14 (Xy = NRl or O) 16 R,. o .e / / Y'' s' 12 Hornotogs and Derivatives \
18 . . ~ Rl~~~~)m 19 Fommm ~a Reaction Scheme 6 26 Reaction Scheme 6 discloses a presently preferred synthetic 27 process for the preparation of compounds where, with reference to 28 Formula 1, the Z group is COO- or CONR, (R1 is preferably H).
29 These ester and amide derivatives are prepared from the 3,4-dihydro-7-bromonaphthalene derivatives of Formula 16, which can be obtained as 1 described in Reaction Scheme 1. Thus, the compounds of Formula 16 2 are reacted with strong base, such as t-butyllithium, in an inert ether 3 type solvent, such as tetrahydrofuran, at cold temperature, and carbon 4 dioxide (COz) is added to provide the 5,6- dihydro-2-naphthalenecarboxylic acid derivatives of Formula 47. Compounds of 6 Formula 47 are then reacted with compounds of the formula X2 Y(R2)-7 A-B (Formula 48) where X2 reperesent an OH or an NR, group, the R, 8 preferably being hydrogen. Those skilled in the art will recognize that 9 the compounds of Formula 48 are aryl or heteroaryl hydroxy or amino l0 derivatives which can be obtained in accordance with the state-of-the-11 art. The reaction between the compounds of Formula 47 and Formula 12 48 can be conducted under various known ester or amide forming 13 conditions, such as coupling of the two in the presence of 1-(3 14 dimethylaminopropyl)-3- ethylcarbodiimide hydrochloride and 4 dimethylaminopyridine. Alternatively, the compounds of Formula 47 16 can be converted into the corresponding acid chloride for coupling with 17 the compounds of Formula 48 in the presence of base. The amide or 18 ester compounds of Formula 49 can be converted into further homologs 19 and derivatives, as described above. Although Reaction Scheme 6 is described and shown for the example where the X group of Formula 1 21 is (C(R,)2Jn and n is 1 (dihydronaphthalene derivatives), the herein 22 described process can be adapted for the preparation of benzopyran, 23 benzothiopyran, dihydroquinoline and indene derivatives as well.
24 Compounds of the present invention where with reference to Formula 1, Z is -OCO-, NR,CO, as well as the corresponding thioester 26 and thioamide analogs, can be prepared from the intermediates derived 27 from the compounds of Formula 16 where the bromo function is 28 replaced with an amino or hydroxyl group and in accordance with the 29 teachings of United States Patent Nos. 5,324,744, the specification of which is expressly incorporated herein by reference.
__....._.~_-~....-..-,-~.,.-~.u.-~-.~_ .... , ....... .-...,.~~. ~_.....

o ~) ~I t w Br HO
10 ~ ) ~~ ~ Br b i i Br a CGiH
11 ~ \ ) r''~. \ ) w compound c Compound n Compound x a c 16 ~' Compound 75: X ~ Y~ H Compound l6: X-Ys 11 1 g Compound 40: X= H, Y= F Compound 41: X= N, Y= F
Com pound 212: X = Y r F Canpocr~d Z03: X= Y= F

(a) Mg° ! 4-bromotoluene I THF I 1 h (62%); (b) p-TsOH~ H~0 I benzene I
80 °C I 2h (81 %); (c) t 21 BuLi I THF I -78 °C I COZ, then HCt I -78 °C to rt (75%)' (d) EDC / DMAP ! DMF I 4-H=NC,H.COzEt (10, 7496), Or 2-F~-HZNC6H,COZEt (t1, 50%), Or 2,6~ii8uoro-4-HZNC6HZCOzEt ('t2, 19%); (e) 22 NaOH I MeOH I Hz0 (1, 73%; 2, 72%; 3, 70%).

Reaction Scheme 6A

1 Reaction Scheme 6A discloses a synthetic route to specific 2 preferred dihydronaphthalene compounds of the invention where with 3 reference to Formula 1 the Z group is CONH (amide derivatives).
4 Although the nature of the reagents utilized in this specific synthetic route is indicated in the scheme itself, and is described in detail in the 6 description of the specific examples, the following explanation is also 7 provided here. 3,4-Dihydro- 4,4-dimethyl-1(2H)-naphthalenone is 8 brominated in the presence of aluminum chloride to give 3,4-9 dihydro-4,4-dimethyl-7-bromo-1(2H)-naphthalenone (Compound B) 1 o which is the first compound shown in the scheme. Compound B is 11 reacted with a Grignard reagent obtained from 4-bromotoluene to give 12 1,2,3,4-tetrahydro-1-hydroxy-1-(4-methylphenyl)-4,4-dimethyl-7-13 bromonaphthalene (Compound C) that is dehydrated by heating with 14 para-toluene sulfonic acid to provide 3,4-dihydro-1-(4-methylphenyl)-4,4-dimethyl-7- bromonaphthalene (Compound D). Compound D is treated 16 with t-butyllithium to cause a metal halogen exchange, and the resulting 17 organolithium compound is quenched with carbon dioxide to yield 5,6-i 8 dihydro-5,5-dimethyl-8-(4-methylphenyl)-2-naphthalenecarboxylic acid 19 (Compound K). Compound K is condensed with ethyl 4-aminobenzoate, ethyl 2-fluoro-4-aminobenzoate or ethyl 2,6-difluoro-4-21 aminobenzoate, respectively, in the presence of 1-(3-22 dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC)and of 23 4-dimethylaminopyridine (DMAP) to provide 24 ethyl4-j[(5,6-dihydro-5,5-dimethyl-8-(4-methylphenyl)-2-naphthalenyl)carbonylJaminoJ-benzoate (Compound 35), ethyl 2-fluoro-26 4-[[(5,6-dihydro-5,5-dimethyl-8-(4-methylphenyl)-2-27 naphthalenyl)carbonyl]amino]-benzoate (Compound 40) and ethyl 2,6-28 difluoro-4-jj(5,6-dihydro-5,5-dimethyl-8-(4-methylphenyl)-2-29 naphthalenyl)carbonylJaminoJ-benzoate (Compound 212), respectively.
Saponification of these ethyl esters, provided the free carboxylic acids, 1 4-[[(S,6-dihydro-S,S-dimethyl-8-(4-methyiphenyl)-2-2 naphthalenyl)carbonyl]aminoJ-benzoic acid {Compound 36), 2-fluoro-4-3 (((S,6-dihydro-S,S-dimethyl-8-(4-methylphenyl)-2-4 naphthalenyl)carbonyl]aminoJ-benzoic acid (Compound 41) and 2,6-difluoro-4-([(S,6-dihydro-S,S-dimethyl-8-(4-methylphenyl)-2-6 naphthalenyl)carbonyl]amino]-benzoic acid (Compound 203), 7 respectively.

° o a 11 ~ I ~ I ~" I -~-.. ~ I pEi o~ o~
12 ~mPo~ 213 Caanpo~a~d 21a Cornpourp 275 Compound 216 1 S ~ ~ \ I ~oF~ ~ I oE~ , ~.
r Compound 217 17 Compound 218 Compouraa Zt9: R = Et ~j Compound ZZO: R = H ~9 19 ~~ ;off-, n 22 ~v~nd 221: x = H compo~r~d 2oa: x = H
Compoutd 2ZI: x= F Compound 20s: x= F

(a) MeMgCI I THF I then H~&~, / H20 / '100 °C ('!00%); (b) CH~COCI !
AICI, I CH2C1 (54%); (~) NaOC1 / NaOH I dioxane I 65 °C then EtOH I f =~O, I d (100%); (d) Br2 I
HOAc (100%); (e) CtOj 26 HOAc / AGzO (76%); (~ p-tolyl-MgBr / THF then p-TsOH°HZO I toluene I
D (25°~); (g) NaOH l EtOH / H=O (98%); (h) EDC I DMAP I DMF / 2-F~4-H=NCgH3COZEt (21, 63%), 012,6~1nu0ro~4-27 HzNC6H2CO~Et (22, 33°~); (e) NaOH I EtOH I H=O (~t, 86%; 5, 7096).

Reaction Scheme 6B

1 Reaction Scheme 6B discloses a synthetic route to specific 2 preferred benzopyran (chromene) compounds of the invention where 3 with reference to Formula 1 the Z group is also CONH (amide 4 derivatives). In accordance with this scheme, dihydrocoumarine (Compound 213) is subjected to a Grignard reaction with methyl 6 magnesium chloride followed by heating a tertiary alcohol intermediate 7 with acid, to provide 2,2-dimethylchroman (Compound 214).
8 Compound 214 is converted into 6-acetyl-2,2-dimethylchroman 9 (Compound 215) in a Friedel-Crafts reaction, and the latter is oxidized 1 o with NaOCI in the presence of sodium hydroxide and thereafter 11 esterified to give ethyl 2,2-dimethylchroman-6-carboxylate (Compound 12 216). Compound 216 is treated with bromine in glacial acetic acid to 13 give ethyl 8-bromo-2,2-dimethylchroman-6-carboxylate (Compound 217).
14 Compound 217 is oxidized with chromium trioxide to form a ketone function in the 4-position of the chroman ring and yield ethyl $-bromo-16 2,2-dimethyl-4-chromanone-6-carboxylate (Compound 218). Reaction of 17 Compound 218 with the Grignard reagent made from 4-bromotoluene, 18 and subsequent heating with p-TSOH to complete dehydration of the 19 intermediate tertiary alcohol, provides ethyl 8-bromo-2,2-dimethyl-4-2o tolyl-4(21-chroman-6-carboxylate (Compound 219). Saponification of 21 Compound 219 with base gives the corresponding free carboxylic acid, 22 8-bromo-2,2-dimethyl-4-tolyl-4(2F~-chroman-6-carboxylic acid 23 (Compound 220). Compound 220 is condensed with ethyl 2-fluoro-4-24 aminobenzoate and ethyl 2,6-difluoro-4-aminobenzoate, in the presence of 1-(3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC) 26 and of 4-dimethylaminopyridine (DMAP) to provide ethyl 2-fluoro-4-27 [[8-bromo-2,2-dimethyl-4-tolyl-6-chromanyl)carbonyl]amino]-benzoate 28 (Compound 221) and ethyl 2,6-difluoro-4-[[8-bromo-2,2-dimethyl-4-29 tolyl-6-chromanyl)carbonyl]amino]-benzoate (Compound 222), respectively. Saponification of Compounds 221 and 222 yields the 1 corresponding free carboxylic acids, 2-fluoro-4-((8-bromo-2,2-dimethyl-4-2 tolyl-6-chromanyl)carbonyl]amino]-benzoic acid (Compound 204) and 3 2,6-difluoro-4-[[8-bromo-2,2-dimethyl-4-tolyl-6-4 chromanyl)carbonyl]amino]-benzoic acid (Compound 205), respectively.

R" 1. Mg I 1'HF
r 2. Zn A
~a~o / / I 3. Ni(O~/THF (~ ~
11 ~ --~_.~. .
12 ~ q, )m X, Y(F~?-A-8 R ~(~kn Formula 10 ' R' 13 Fom~ula 16 ~ FomulaSO
14 Homologs and Derivatives Reaction Scheme 7 21 Reaction Scheme 7 discloses a presently preferred synthetic 22 process for the preparation of compounds where with reference to 23 Formula 1, Z is -(CR,=CRl)~: and n' is 0. These compounds of 24 Formula 50 can be obtained in a coupling reaction between compounds of Fc~rrnula 16 and a Grignard reagent derive' from the halo 26 compounds of Formula 10. The coupling rea _on is typically conducted 27 in the presence of a zinc salt and a nickel (Ni(0)) catalyst in inert ether 28 type solvent, such as tetrahydrofuran. The compounds of Formula 50 29 can be converted into further homologs and derivatives, as described above.

Pd2(Pph3)2(Ck)2 ~
2 / ~ ~ E13N I P(o-McPh)3 / Y( 3 (Ralo CH~CH=Y(R~-A-=B \
4 R, ~ Formula 51 R, R, S Formula 7 Formula 52 6 1. NaN(SiMeg)2 7 THF I T8' C
2. c ~c~o 10 R,~ ~A ~ Tf Y(R:) RI,~ZrC1 / / Y(Rz) 11 ~ / / ~ ~ Pd(~ (Ra)o .\
12 (R~~" THF I sa' c R )m R, R, , 13 ~~~ $4 Fortnuta53 16 Nomolcps and Derivatives 17 Reaction Scheme 8 18 Referring now to Reaction Scheme 8 a presently preferred 19 synthetic process is disclosed for the preparation of compounds where Z
is -(CR1=CR,)~: and n' is 1. More particularly, Reaction Scheme 8 21 discloses the presently preferred process for preparing those compounds 22 which are dihydronaphtalene derivatives and where the Z group 23 represents a vinyl (-CH=CH-) function. However, the generic 24 methodology disclosed herein can be extended, with such modifications which will be apparent to those skilled in the art, to the analogous 26 benzopyran, benzothiopyran, dihydroquinoline compounds, and to 27 compounds where the vinyl group is substituted. Thus, in accordance 28 with Reaction Scheme 8 the 7-bromo-1(2H)-naphthalenone derivative of 29 Formula 7 is reacted with a vinyl derivative of the structure -CH2=CH-3o Y(R2)-A-B (Formula 51) in the presence of a suitable catalyst, typically 1 having the formula Pd(PPh3), an acid acceptor (such as triethylamine) 2 under an inert gas (argon) atmosphere. The conditions of this reaction 3 are analogous to the coupling of the acetylene derivatives~of Formula 9 4 with the reagent of Formula 10 (see for example Reaction Scheme 1), and this type of reaction is generally known in the art as a Heck 6 reaction. The vinyl derivative of Formula 51 can be obtained in 7 accordance with the state of the art, an example for such a reagent used 8 for the synthesis of a preferred compound to be used in the invention is 9 ethyl4-vinylbenzoate.
The product of the Heck coupling reaction is an ethenyl 11 derivative of Formula 52, which is thereafter converted into compounds 12 used in the present invention by treatment with sodium 13 bis(trimethylsilyl)amide and 2-(N,N-bis{trifluoromethylsulfonyl)amino]-5-14 chloropyridine to yield the 4-trifluoromethylsulfonyloxy derivatives of Formula 53, and subsequent reaction with an organometal derivative 16 derived from the aryl or heteroaryl compound R,4H, as described above.
17 The resulting compounds of Formula 54 can be converted into further 18 homologs and derivatives.
19 The compounds of Formula 54 can also be obtained through synthetic schemes which employ a Wittig or Horner Emmons reaction.
21 For example, the intermediate of Formula 33 (see Reaction Scheme 4) 22 can be reacted with a triphenylphosphonium bromide (Wittig) reagent 23 or more preferably with a diethylphosphonate (Homer Emmons) 24 reagent of the structure (EtO),PO-CH2--Y(R2)-A-B, as described for analogous Horner Emmons ~ ~trt~ns in x'nited ~aates Patent No.
26 5,324,840, the specification ofnigh is incorporated herein by reference.
27 The just mentioned Horner Emmons reaction provides intermediate 28 compounds analogous in structure to Formula 52, and can be converted 29 into compounds of Formula 54 by the sequence of reactions described in Reaction Scheme 8 for the compounds of Formula 52.

1 Synthetic Methods - 1 and 3-Oxy-1-Propenyl~ Substituted 2 Comyounds 3 The exemplary RAR antagonist compounds of Formula 101 can 4 be made by the synthetic chemical pathways illustrated here. The synthetic chemist will readily appreciate that the conditions set out here 6 are specific embodiments which can be generalized to any and all of the 7 compounds represented by Formula 101.

g ~'1r~' 1. Na3 .
R,s~2~ R,. _ ~Jo I tRa)o 2 G03 ~ ~ dkatona 1 ~ R, R, HOArJAr~O R, R, O
11 FORMULA 103 FpRAiULA 104 HOCHZCHZOH
13 p-TsOH

o (R,)m 16 ~~ 0H (~)m ( R,. R~~_ (~o I R,.

THF or Et20 18 R, R, O~ R, R, 0, 2~ 1. ~ Nald(SiMe3y~lt'HFf-78'C
z 1 ~ t~.~.
( 22 ' H ""~' H~ 2 ~~lt~4X/R-ti/ifiE
23 ~ znaz/1MF
7 Pc(o) /htF / sc ~c ~
R» ~a~n R,~Y(Rs)~A-B R" (~)rn z6 27 «)° / ~ I R~~ FORMULA 108 / Rm Y ) (~o I I
NaOH / EtOH
2g R~ R~ 0 ~ R,s R, O
29 FORMULA 107 HOn1010~5 and FORMULA 109 Derivatives Reaction Scheme 101 SUBSTITUTE SHEET (RULE 26) . ....._.~.. ... .~..._~.~~..~.W~.~...,__~~ ~. .. t.... . .... ~..,._....

1 Reaction Scheme 101 illustrates the synthesis of compounds of 2 Formula 101 where X is ~C(Rl)2~~, n is 1, p is zero and R~~ is H or lower 3 alkyl. In other words, Reaction Scheme 101 illustrates the synthesis of 4 compounds of the invention which are 3,4-dihydronaphthalene derivatives. In accordance with this scheme, a tetrahydronaphthalene 6 compound of Formula 103 which is appropriately substituted with the 7 R3 and R2 groups (as these are defined in connection with Formula 101 }
8 serves as the starting material. A preferred example of a compound of 9 Formula 103 is 1,3,3,4-tetrahydro-1,1-dimethyl-naphthalene, which is 1 o described in the chemical literature (Mathur et al. Tetrahedron, 1985, 11 41:1509. A presently preferred route for the synthesis of this compound 12 from 1-bromo-3-phenylpropane is also described in the experimental 13 section of the present application.
14 The compound of Formula 103 is reacted in a Friedel Crafts type reaction with an acid chloride having the structure R,6CHZCOCI (R,6 is 16 defined as in connection with Formula 101) and is thereafter oxidized 17 with chromium trioxide in acetic acid to provide the isomeric 6 and 7 18 acyl-3,4-dihydro-1(2H)-naphthalenone derivatives. Only the 6-acyl 19 derivative which is of interest from the standpoint of the present invention, is shown by structural formula (Formula 104) in Reaction 21 Scheme 101. In the preparation of the presently preferred compounds 22 of this invention the R, groups represent methyl, RZ, R3 and R,6 are H, 23 and therefore the preferred intermediate corresponding to Formula 104 24 is 3,4-dihydro-4,4-dimethyl-6-acetyl-1(2H)- naphthaienone.
The exocyclic ketone funLtion of the compound of Formula 104 is 26 thereafter protected as a ketal, for example by treatment with ethylene 27 glycol in acid, to provide the 1,3-dioxolanyl derivative of Formula 105.
28 The compound of Formula 105 is then reacted with a Grignard reagent 29 of the formula Rl4MgBr (R14 is defined as in connection with Formula 101 ) to give the 1,2,3,4- tetrahydro-1-hydroxy-naphthalene derivative of 1 Formula 106. The exocyclic ketone function of the compound of 2 Formula 106 is then deprotected by treatment with acid and dehydrated 3 to give the compound of Formula 107.
An alternate method for obtaining the compounds of Formula 107 from the compounds of Formula 105 is by reacting the compounds 6 of Formula 105 with sodium bis(trimethylsilyl)amide and 2-[N,N-7 bis(trifluoromethylsulfonyl)amino]-5-chloropyridine (Tf = SOZCF3) in an 8 inert ether type solvent, such as tetrahydrofuran, at low temperatures 9 (-78°C and 0°C). This reaction proceeds through a sodium salt intermediate which is usually not isolated and is not shown in Reaction 11 Scheme 101. The overall reaction results in a trifluoromethylsulfonyloxy 12 derivative, which is thereafter reacted with an organometal derivative 13 derived from the aryl or heteroaryl compound R,4H, such that the 14 formula of the organometal derivative is R,4Met (Met stands for monovalent metal), preferably R,4Li. (R,4 is defined as in connection 16 with Formula 101.) The reaction with the organometal derivative, 17 preferably lithium derivative of the formula R,4Li is usually conducted 18 in an inert ether type solvent (such as tetrahydrofuran) in the presence 19 of zinc chloride {ZnCl2) and tetrakis(triphenylphosphine)-palladium(0) {Pd(PPh3)4). The organolithium reagent R,4Li, if not commercially 21 available, can be prepared from the compound R,4H (or its halogen 22 derivative R,4-XI where Xl is halogen) in an ether type solvent in 23 accordance with known practice in the art. The temperature range for 24 the reaction between the reagent R,4Li and the trifluoromethylsulfonyloxy derivative is, generally speaking, in the range 26 of approximately -78°C to 50°C.
27 The compounds of the invention are formed as a result of a 28 condensation between the ketone compound of Formula 107 and an 29 aldehyde or ketone of Formula 108. In the preparation of the preferred exemplary compounds of the invention the reagent of Formula 108 is 4-.........,"w_,w,~, "o~,v,.,... . T . .,._._..w~_ -~~~r.... _.w......_....

1 carboxybenzaldehyde (R,.,-H). Examples of other reagents within the 2 scope of Formula 108 and suitable for the condensation reaction and for 3 the synthesis of compounds within the scope of the present invention 4 (Formula 101) are: 5-carboxy-pyridine-2-aldehyde, 4- carboxy-5 pyridine-2-aldehyde, 4-carboxy-thiophene-2- aldehyde, 5-carboxy-6 thiophene-2-aldehyde, 4-carboxy- furan-2-aldehyde, 5-carboxy-furan-2-7 aldehyde, 4- carboxyacetophenone, 2-acetyl-pyridine-5-carboxylic acid, 2-8 acetyl-pyridine-4-carboxylic acid, 2-acetyl- thiophene-4-carboxylic acid, 2-9 acetyl-thiophene-5- carboxylic acid, 2-acetyl-furan-4-carboxylic acid, and l0 2-acetyl-furan-5-carboxylic acid. The latter compounds are available in 11 accordance with the chemical literature; see for example Decroix et al., 12 J. Chem. Res.(S), 4: 134 (1978); Dawson et al., J. Med. Chem. 29:1282 13 ( 1983); and Queguiner et al., Bull Soc. Chimique de France No. 10, pp.
14 3678 - 3683 (1969). The condensation reaction between the compounds 15 of Formula 107 and Formula 108 is conducted in the presence of base 16 in an alcoholic solvent. Preferably, the reaction is conducted in ethanol 17 in the presence of sodium hydroxide. Those skilled in the art will 18 recognize this condensation reaction as an aldol condensation, and in 19 case of the herein described preferred examples (condensing a ketone 20 of Formula 107 with an aldehyde of Formula 108) as a Claisen-Schmidt 21 reaction. (See March: Advanced Organic Chemistry: Reactions, 22 Mechanisms, and Structure, pp. 694 - 695 McGraw Hill (1968). The 23 compounds of Formula 109 are within the scope of the present 24 invention, and can also be subjected to further transformations resulting 25 in additional compounds of the invention. Alternatively, the A-B group 26 of Formula 108 may be a group which is within the scope of the 27 invention, as defined in Formula 101, only after one or more synthetic 28 transformations of such a nature which is well known and within the 29 skill of the practicing organic chemist. For example, the reaction 30 performed on the A-B group may be a deprotection step, homologation, 1 esterification, saponification, amide formation or the like.
2 Generally speaking, regarding derivatization of compounds of 3 Formula 109 and/or the synthesis of aryl and heteroaryl compounds of 4 Formula 108 which can thereafter be reacted with compounds of Formula 107, the following well known and published general principles 6 and synthetic methodology can be employed.
7 As indicated above, carboxylic acids are typically esterified by 8 refluxing the acid in a solution of the appropriate alcohol in the 9 presence of an acid catalyst such as hydrogen chloride or thionyl l0 chloride. Alternatively, the carboxylic acid can be condensed with the 11 appropriate alcohol in the presence of dicyclohexylcarbodiimide and 12 dimethylaminopyridine. The ester is recovered and purified by 13 conventional means. Acetals and ketals are readily made by the method 14 described in March, Advanced Organic Chemistry, 2nd Edition, McGraw-Hill Book Company, p. 810). Alcohols, aldehydes and ketones 16 all may be protected by forming respectively, ethers and esters, acetals 17 or ketals by known methods such as those described in McOmie, 18 Plenum Publishing Press, 1973 and Protecting Groups, Ed. Greene, 19 John Wiley & Sons, 1981.
To increase the value of n in the compounds of Formula 108 21 before affecting the condensation reaction of Reaction Scheme 101 22 (where such compounds corresponding to Formula 108 are not available 23 from a commercial source) aromatic or heteroaromatic carboxylic acids 24 may be subjected to homologation (while the aldehyde group is protected) by successive treatment under Arndt- Eistert conditions or 26 other homologation procedures. Alternatively, derivatives which are not 27 carboxylic acids may also be homologated by appropriate procedures.
28 The homologated acids can then be esterified by the general procedure 29 outlined in the preceding paragraph.
Compounds of Formula 108, (or other intermediates or of the ..-_~..~...... . _.... r ........._.......... .......,. ....~._.

1 invention, as applicable) where A is an alkenyl group having one or 2 more double bonds can be made for example, by synthetic schemes well 3 known to the practicing organic chemist; for example by Wittig and like 4 reactions, or by introduction of a double bond by elimination of halogen from an alpha-halo-arylalkyl- carboxylic acid, ester or like 6 carboxaldehyde. Compounds of Formula 108 (or other intermediates or 7 of the invention, as applicable) where the A group has a triple 8 (acetylenic) bond can be made by reaction of a corresponding aromatic 9 methyl ketone with strong base, such as lithium diisopropylamide, reaction with diethyl chlorophosphate and subsequent addition of 11 lithium diisopropylamide.
12 The acids and salts derived from compounds of Formula 109 (or 13 other intermediates or compounds of the invention, as applicable) are 14 readily obtainable directly as a result of the condensation reaction, or from the corresponding esters. Basic saponification with an alkali metal 16 base will provide the acid. For example, an ester of Formula 109 (or 17 other intermediates or compounds of the invention, as applicable) may 18 be dissolved in a polar solvent such as an alkanol, preferably under an 19 inert atmosphere at room temperature, with about a three molar excess of base, for example, lithium hydroxide or potassium hydroxide. The 21 solution is stirred for an extended period of time, between 15 and 20 22 hours, cooled, acidified and the hydrolysate recovered by conventional 23 means.
24 The amide may be formed by any appropriate amidation means knows ~ the art from the corresponding esters or carboxylic acids. One 26 way to . .:pare such compounds is to convert an acid to an acid chloride 27 and then treat that compound with ammonium hydroxide or an 28 appropriate amine.
29 Alcohols are made by converting the corresponding acids to the acid chloride with thionyl chloride or other means (J. March, Advanced WO 98!58922 PCTNS98/13065 1 Organic Chemistry, 2nd Edition, McGraw-Hill Book Company), then 2 reducing the acid chloride with sodium borohydride (March, Ibid, p.
3 1124), which gives the corresponding alcohols. Alternatively, esters may 4 be reduced with lithium aluminum hydride at reduced temperatures.
Alkylating these alcohols with appropriate alky halides under 6 Williamson reaction conditions (March, Ibid, p. 357) gives the 7 corresponding ethers. These alcohols can be converted to esters by 8 reacting them with appropriate acids in the presence of acid catalysts or 9 dicyclohexylcarbodiimide and dimethylaminopyridine.
l0 Aldehydes can be prepared from the corresponding primary 11 alcohols using mild oxidizing agents such as pyridinium dichromate in 12 methylene chloride (Corey, E. J., Schmidt, G., Tet. Lett., 399, 1979), or 13 dimethyl sulfoxide/oxalyl chloride in methylene chloride (Omura, K., 14 Swern, D., Tetrahedron, 34:1651 (197$)).
Ketones can be prepared from an appropriate aldehyde by 16 treating the aldehyde with an alkyl Grignard reagent or similar reagent 17 followed by oxidation.
18 Acetals or ketals can be prepared from the corresponding 19 aldehyde or ketone by the method described in March, Ibid, p. 810.

...o.y.._ T . . w".""."., ,."-.~~.~"...~~...._ OCH3 RIRtZ~
9 (R~o \ ~ (~o CH2d2 O R, R, 12 Cro3 HOAc 13 Ac2o 14 ~, off o / OCH3 R~4MgBr / OCHg (~0 1 E--~-16 ~ THF or Et20 (~° \
17 a' R, R, 1 g FORMULA 1 t3 FORMULA 112 19 H~
21 R,.
1. BBrg R"
OCH3 ~ta2 ~ ~ ~ O~CE't2Rm 22 (~o ~ (~ 'Io 2. a,6cH2coa w o 23 R, ~, py,;~,e R, Reaction Scheme 102 SU6STlTUTE SHEET (RULE 26) R,.
6 .~ ~ ' o~~~,. _ ~ ~ i ~ off (~ t'o 7 ~ O (Friss ~c ~ ( ~2~s narran msnt) R, ~ O
R, R, 9°

11 cWta,i~,-w pyridne 13 ~.a 14 R'. o~Y(~) OH R,.
i IS (R~c ~A~ KOH ~ ~ O
Y(R=) ~ PY~ (Ra)o ~ ~ C
16 ~R'.
R~ ~ O OH R~
R. O

19 HZ so,, HOAc 21 R,.
,A
22 i i O Yes) Homologs and )o ( ~ Derivatives 24 R~ R, o 2S FORMULA 11s Reaction Scheme 102 (cont'd) SUBSTITUTE SHEET (RULE 26) ....~,..~~~, ~..wr.w . 1 _._.a......~ ...~....,.e..~__.__~...~r..w.....

1 Referring now to Reaction Scheme 102, a synthetic route to those 2 compounds of the invention is described in which, with reference to 3 Formula 101 p is zero, R2 in the aromatic portion of the condensed ring 4 structure is OH and Rl~ is OH. Those skilled in the art will readily recognize that these compounds are (3-diketones in the enol form.
6 Reaction Scheme 102 also describes a synthetic route to those 7 compounds of the invention where p is 1. Those skilled in the art will 8 readily recognize that the latter compounds are flavones. Thus, in 9 accordance with this scheme a 1,2,3,4- tetrahydro-6-methoxynaphthalene-1-one derivative of Formula 110 is reacted with 11 dialkyl zinc (R,Zn) in the presence of titanium tetrachloride in a 12 suitable solvent such as CH2Cl2 to replace the oxo function with the 13 geminal dialkyl group RIR,, to yield a compound of Formula 111, where 14 R~ is lower alkyl. In preferred embodiments of the compounds of the invention which are made in accordance with Reaction Scheme 102 the 16 R3 group is hydrogen and R, are methyl. Accordingly, the dialkyl zinc 17 reagent is dimethyl zinc, and the preferred starting material of Formula 18 110 is 1,2,3,4- tetrahydro-6-methoxynaphthalene-1-one. The latter 19 compound is commercially available, for example from Aldrich Chemical Company. The 1,2,3,4-tetrahydro-1,2- dialkyl-6-methoxy 21 naphthalene derivative of Formula 111 is thereafter oxidized with 22 chromium trioxide in acetic acid and acetic anhydride to give a 1,2,3,4-23 tetrahydro- 3,4-dialkyl-7-methoxy naphthalen-1-one derivative of 24 Formula 112. The ketone compound of Formula 112 is reacted with a Grignard reagent (R,4MgBr, R~4 is defined as in connection with 26 Formula 101) to yield a 1- hydroxy-1-aryl-3,4-dihydro-3,4-dialkyl-7-27 methoxy naphthalene derivative of Formula 113. The hydroxy 28 compound of Formula 113 is subjected to elimination by heating, 29 preferably in acid, to yield the dihydronaphthalene compound of Formula 114. The methyl group is removed from the phenolic methyl 1 ether function of the compound of Formula 114 by treatment with 2 boron tribromide in a suitable solvent, such as CH2C12, and therafter the 3 phenolic OH is acylated with an acylating agent that introduces the 4 R,6CH2C0 group, to give a compound of Formula 115. In the preferred embodiment R,6 is H, and therefore the acylating agent is 6 acetyl chloride or acetic anhydride. The acetylation reaction is 7 conducted in a basic solvent, such as pyridine. The acylated phenol 8 compound of Formula 115 is reacted with aluminum chloride at 9 elevated temperature, causing it to undergo a Fries rearrangement and l0 yield the 1-aryl-3,4-dialkyl-3,4-dihydro-6-acyl-7-hydroxy-naphthalene 11 compound of Formula 116. The phenolic hydroxyl group of the 12 compound of Formula 116 is acylated with an acylating agent (such as 13 an acid chloride) that introduces the CO-Y(R2)-A-B group to yield a 14 compound of Formula 117. In the acid chloride reagent CI-CO-Y(R2)-A-B (or like acylating reagent) the symbols Y, R2 and A-B have the 16 meaning defined in connection with Formula 101. In the preparation of 17 a preferred compound of the invention in accordance with this scheme 18 this reagent is CICOC6H4COOEt (the half ethyl ester half acid chloride 19 of terephthalic acid).
The compound of Formula 117 is reacted with strong base, such 21 as potassium hydroxyde in pyridine, to yield, as a result of an 22 intramolecular Claisen condensation reaction, a compound of Formula 23 118. The compounds of Formula 118 are within the scope of the 24 invention and of Formula 101, where there is an OH for the RZ
substituent in the aromatic portion of the condensed ring moiety and 26 R,~ is OH. In connection with the foregoing reaction (intramolecular 27 Claisen condensation) and the previously mentioned Fries 28 rearrangement it is noted that these probable reaction mechanisms are 29 mentioned in this description for the purpose of fully explaining the herein described reactions, and for facilitating the work of a person of ., .,_....,",...--.M.-..,.-...,. ..T,.......,.,.".. ,-...,.-..." ._....._ ",".~ .-_..." _.... ...

1 ordinary skill in the art to perform the herein described reactions and 2 prepare the compounds of the invention. Nevertheless, the present 3 inventors do not wish to be bound by reaction mechanisms and theories, 4 and the herein claimed invention should not be limited thereby.
The compounds of Formula 118 are reacted with strong acid, 6 such as sulfuric acid, in a suitable protonic solvent, such as acetic acid, 7 to yield the flavone compounds of Formula 119. The compounds of 8 Formula I19 are also compounds of the invention, within the scope of 9 Formula 101 where p is 1. Both the compounds of Formula 118 and Formula 119 can be subjected to further reactions and transformations 11 to provide further homologs and derivatives, as described above in 12 connection with Reaction Scheme 101. This is indicated in Reaction 13 Scheme 102 as conversion to homologs and derivatives.

S
H _ 6 ~~ BrC~Ry~CH(R~~02 HO C

C \ /
7 ~ w / Ru b~ ~1= s FORMULA 120 FORMULA t22 I 0 CH3SOgH
II
12 0 (~hn Pay?a2 0 (~Im 13 'R,e~l~2 (~o ~ Oslo 14 Wac~sr \ / R,~
O oxidation I$ FORMULA 124 FORMULA 123 p-TsOH

20 0 (~Im R, off (~>m R M Br 21 (R,)o ! Ru ~ (R~o ~ R,~
THE or E120 22 0~ o~
23 FORMULA 125 FORMULA t26 Reaction Scheme 103 SUBSTITUTE SHEET (RULE 26) ... . .....~...d.~.~..~.. ... T...._. . .,".....w..m_ ._.......w~..d......-..~..... .

R" OH ~kn R" (R~
S

6 (~o ' ~ R'e -"~ (Ra)o ( R,e 7 p O
g FORMULA 126 FORMULA 127 10 a,~ ~c~~-a NaOH I EtOH

14 R,. (~)m 15 Homologs and ~ R" Y(Rz) Derivatives 16 . R,e 21 Reaction Scheme 103 (cont'd) 22 Referring now to Reaction Scheme 103 a synthetic route is shown 23 leading to those compounds of the invention where, with reference to 24 Formula 101 X is S, O or NR', p is zero and R,~ is H or lower alkyl.
However, by applying the : :~°~eric principles of synthesis shown in 26 Reactic ;icheme 102 the presently shown synthetic process can be 27 modified or adapted by those of ordinary skill in the art to also obtain 28 compounds of the invention where X is S, O or NR' and p is 1, or 29 where X is S, O or NR' and p is zero, the R2 group in the aromatic portion of the condensed ring moiety is OH and R1~ is OH.
SUBSTITUTE SHEET (RULE 26) 1 The starting compound of Reaction Scheme 103 is a phenol, 2 thiophenol or aniline derivative of Formula 120. In the presently 3 preferred compounds of the invention the RZ and R16 groups are both 4 hydrogen, and the preferred starting compounds of Formula 120 are 3-ethenyl-thiophenol or 3-ethenyl-phenol which are known in the chemical 6 literature (Nuyken, et al. Polym. Bull (Berlin) 11:165 (1984). For the 7 sake of simplifying the present specification, in the ensuing description 8 X can be considered primarily sulfur as for the preparation of 9 benzothiopyran derivatives of the present invention. It should be kept in mind, however, that the herein described scheme is also suitable, with 11 such modifications which will be readily apparent to those skilled in the 12 art, for the preparation of benzopyran (X = O) and dihydroquinoline 13 (X = NR') compounds within the scope of the present invention. Thus, 14 the compound of Formula 120 is reacted under basic condition with a 3-bromo carboxylic acid of the Formula 121. In this reaction scheme the 16 symbols have the meaning described in connection with Formula 101.
17 An example for the reagent of Formula 121 where R3 is hydrogen, is 3-18 bromopropionic acid. The reaction with the 3-bromocarboxylic acid of 19 Formula 121 results in the compound of Formula 122. The latter is cyclized by treatment with acid to yield the 7-ethenyl-thiochroman-4-one 21 derivative (when X is S) or 7-ethenyl-chroman derivative (when X is 0) 22 of Formula 123. The 7-ethenyl- thiochroman-4-one or 7-ethenyl-23 chroman-4-one derivative of Formula 123 is oxidized in the presence of 24 Pd(II)CI2 and CuCl2 catalysts to provide the corresponding 7-acyl (ketone) compound of Formula 124. Those skilled in the art will 26 recognize the latter reaction as a Wacker oxidation. The exocyclic 27 ketone group of the compound of Formula 124 is protected in the form 28 of a ketal, for example by treatment with ethylene glycol in acid, to 29 provide the 1,3-dioxolanyl derivative of Formula 125. Thereafter the compound of Formula 125 is subjected to a sequence of reactions ..._..._ ., _....._._,_-...~-_.~."..._..~...M_. _ T....._. .~..~"nm~,w ,."-",w,.."...~,.~.,...w_~

1 analogous to those described for the compounds of Formula 105 in 2 Reaction Scheme 101. Thus, the 1,3- dioxolanyl derivative of Formula 3 125 is reacted with a Grignard reagent of the formula R,4MgBr to give 4 the tertiary alcohol of Formula 126, which is thereafter dehydrated in acid to provide the benzothiopyran (X is S), benzopyran (X is O) or 6 dihydroquinoline (X is NR') derivative of Formula 127. The ketone 7 compound of Formula 127 is then reacted in the presence of base with 8 the reagent of Formula 108 in an aldol condensation (Claisen-Schmidt) 9 reaction to provide compounds of the invention of Formula 128. The l0 compounds of Formula 128 can be converted into further homologs and 11 derivatives, as described above in connection with Reaction Schemes 12 101 and 102.
13 Specific Examples 14 2-hydroxy-2-methyl-5-phen~pentane To a mixture of magnesium turnings 13.16 g (0.541 mol) in 200 16 ml of anhydrous Et20 was added 100.0 g (0.492 mol) of 1-bromo-3-17 phenyl propane as a solution in 100 ml of Et20. After of 5-10 ml of the 18 solution had been added, the addition was stopped until the formation 19 of the Grignard reagent was in progress. The remaining bromide was then added over 1 hour. The Grignard reagent was stirred for 20 21 minutes at 35°C and then 31.64 g (0.541 mol) of acetone was added 22 over a 45 minute period. The reaction was stirred overnight at room 23 temperature before being cooled to 0°C and acidified by the careful 24 addition of 20% HCI. The aqueous layer was extracted with Et20 (3 x 200 ml) and the combined organic layers washed with ~,~rater, and 26 saturated aqueous NaCI before being dried over MgS a. Removal of 27 the solvent under reduced pressure and distillation of the residue 28 afforded 63.Og (72%) of the product as a pale-yellow oil, by 99-102°C /
29 0.5 mm Hg. 1H NMR (CDC13): 8 7.28-7.18 (5H, m), 2.63 (2H, t, J =
7.5 Hz), 1.68 (2H, m), 1.52 (2H, m), 1.20 (6H, s).

1 1,2.3,4-tetrahydro-1 1-dimethylnaphthalene 2 A mixture of P205 {55.3 g, 0.390 mol) in 400 ml of 3 methanesulfonic acid was heated to 105°C under argon until all of the 4 solid had dissolved. The resulting solution was cooled to room temperature and 2-hydroxy-2-methyl-S-phenylpentane (63.0 g, 0.354 6 mol) added slowly with stirring. After 4 hours the reaction was 7 quenched by carefully pouring the solution onto 1 L of ice. The 8 resulting mixture was extracted with Et20 (4 x 125 ml)and the combined 9 organic layers washed with water, saturated aqueous NaHCO3, water, and saturated aqueous NaCI before being dried over MgS04.
11 Concentration of the solution under reduced pressure, followed by 12 distillation afforded 51.0 g (90%) of the product as a clear colorless oil, 13 bp. 65-67°C / 1.1 mmHg. 1H NMR (CDCl3): 8 7.32 (1H, d, J = 7.4 14 Hz), 7.16-7.05 (3H, m), 2.77 (2H, t, J = 6.3 Hz), 1.80 (2H, m), 1.66 (2H, m), 1.28 (6H, s).
16 3,4-dihvdro-4,4-dimethyl-1(2H1-naphthalenone (Compou I7 A solution of 350 ml of glacial acetic acid and 170 ml of acetic 18 anhydride was cooled to 0°C and Cr03, 25.0 g (0.25 mol) carefully 19 added in small portions. The resulting mixture was stirred for 30 minutes before 120 ml of benzene was added. 1,2,3,4-tetrahydro-1,1-21 dimethylnaphthalene was added slowly as a solution in 30 ml of 22 benzene. Upon completing the addition the reaction was stirred for 4 23 hours at 0°C. The solution was diluted with H20 (200 ml) and extracted 24 with Et20 (5 x 50 ml). The combined organic layers were washed with water, saturated aqueous NaC03, and saturated aqueous NaCI, before 26 being dried over MgS04. Removal of the solvents under reduced 27 pressure, and distillation afforded 16.0 g (74%) of the product as a pale-28 yellow oil, by 93-96°C / 0.3 mm Hg 1H NMR (CDC13): 8 8.02 {1H, dd, 29 J=1.3,7.8Hz),7.53(lH,m),7.42(lH,d,J=7.9Hz),7.29(lH,m), 2.74 (2H, t, J = 6.8 Hz), 2.02 (2H, t, J = 6.8 Hz), 1.40 (6H, s).

1 3,4-dihydro-4,4-dimethyl-7-bromo-1{2H1-naphthalenone (Compound B) 2 A 100 ml three-necked flask, fitted with an efficient reflux 3 condenser and drying tube, and addition funnel, was charged with a 4 mixture of AlCl3 9.Sg (71.4 mmol) and 3 ml of CH2C12. The 3,4-dihydro-4,4-dimethyl-I(2H)-naphthalenone (S.Og, 28.7 mmol), was added 6 dropwise with stirring (Caution: Exothermic Reaction') to the mixture 7 at room temperature. Bromine, 5.5 g {34.5 mmol), was then added very 8 slowly, and the resulting mixture stirred for 2 hours at room 9 temperature. (Note: if stirring stops, the mixture can be warmed to 70°C
until stirring resumes.) The reaction was then quenched by the slow 11 addition of ice-cold 6M HCI. The mixture was extracted with Et20 and 12 the combined organic layers washed with water, saturated aqueous 13 NaHC03, and saturated NaCI, before being dried over MgS04.
14 Removal of the solvent under reduced pressure, and distillation of the residue afforded 5.8 g {80%) of the product as a pale-yellow oil which 16 solidified on standing, bp: 140°C / 0.4 mm Hg. 1H NMR (CDCl3): 8 17 8.11(lH,d,J=3.OHz),7.6I(lH,dd,J=3.0,9.OHz),7.31(lH,d,J
18 = 9.0 Hz), 2.72 {2H, t, J = 6.0 Hz), 2.01 (2H, t, J = 6.0 Hz), 1.28 (6H, 19 s).
1,2,3,4-tetrahydro-1-hydroxy-~4-meth l~phen.l~l-4,4-dimeth, 21 bromonaphthalene (Compound C) 22 To a mixture of magnesium turnings (648.0 mg, 27.0 mmol) in 25 23 ml of THF was added a solution of 4- bromotoluene (5.40 g, 31.8 24 mmol) in 10 ml of THF in two portions. The reaction was initiated by the addition of 2 ml of the solution, followed h-- the slow addition of the 26 remaining solution via an addition funnel. The mixture was stirred at 27 room temperature for 1 hour, and then the solution was transferred to a 28 second flask using a canula. To the resulting Grignard reagent was 29 added 4.0 g (15.9 mmol) of 3,4-dihydro- 4,4-dimethyl-7- bromo-1(2H)-naphthalenone (Compound B) as a solution in 15 ml of THF. The WO 98/58922 PCT/(3598/13065 1 resulting solution was heated to reflux overnight, cooled to room 2 temperature, and the reaction quenched by the careful addition of ice-3 cold 10% HCI. Extraction with Et20 was followed by washing of the 4 combined organic layers with H20 and saturated aqueous NaCI, then 5 drying over MgS04. Removal of the solvent under reduced pressure 6 provided an oil which afforded the product as a colorless solid after 7 column chromatography (hexanes / EtOAc , 96 : 4). 1H NMR (CDC13):
8 8 7.36 (1H, dd, J = 2.1, 7.6 Hz), 7.26 (3H, m), 7.12 (3H, s), 2.34 (3H, 9 s}, 2.24-2.04 (2H, m), 1.81 (1H, m), 1.55 (1H, m), 1.35 (3H, s), 1.30 (3H, 1 o s).
11 3,4-dihydro-1- 4-meth~phenyl)-4 4-dimethyl-7- bromona~hthalene 12 (Compound D~
13 A flask equipped with a Dean-Stark trap was charged with 3.4 g 14 of (9.85 mmol) of 1,2,3,4-tetrahydro-1-hydroxy-1-(4-methylphenyl)-4,4-15 dimethyl-7-bromonaphthalene (Compound C) and 40 ml of benzene. A
16 catalytic amount of p-toluenesulfonic acid monohydrate was added and 17 the resulting solution heated to reflux for 2 hours. Upon cooling to 18 room temperature, Et20 was added and the solution washed with H20, 19 saturated aqueous NaHC03, and saturated aqueous NaCI then dried 20 over MgS04. Removal of the solvents under reduced pressure, and 21 column chromatography (100% hexane / silica gel) afforded the title 22 compound as a colorless solid. 1H NMR (CDC13): 8 7.32 (1H, dd, J =
23 2.1, 8.2 Hz), 7.21 (SH, m), 7.15 (1H, d, J = 2.1 Hz}, 5.98 (1H, t, J = 4.7 24 Hz), 2.40 (3H, s), 2.32 (2H, d, J = 4.7 Hz), 1.30 (6H, s).
25 7-Ethvnyl-3,4-dihydro-4 4-dimeth~~phthalen-1(2H)-one Compound E) 26 To a solution (flushed for 15 minutes with a stream of argon) of 7 27 g (27.6 mmol) of 3,4-dihydro-4,4- dimethyl-7-bromo-1(2H)-28 naphthalenone (Compound B) in 150 ml of triethylamine was added 29 0.97 g (1.3 mmol) of bis(triphenylphosphine)palladium(II) chloride and 30 0.26 g (1.3 mmol) of cuprous iodide. The solution mixture was flushed 1 with argon for 5 minutes and then 39 ml (36.6 mmol) of 2 (trimethylsilyl)acetylene was added. The reaction mixture was sealed in 3 a pressure tube and placed in a preheated oil bath (100°C) for 24 hours.
4 The reaction mixture was then filtered through Celite, washed with EtzO
and the filtrate concentrated in vacuo to give crude 7-6 (trimethylsilyl)ethynyl-3,4-dihydro- 4,4-dimethylnaphthalen-1(2H)-one.
7 To a solution of this crude TMS-acetylenic compound in 50 ml of 8 methanol was added 0.6 g (4.3 mmol) of K2C03. The mixture was 9 stirred for 8 hours at ambient temperature and then filtered. The l0 filtrate was concentrated in vacuo, diluted with Et20, washed with water, 11 10% HCl and brine, dried over MgS04 and concentrated in vacuo.
12 Purification by column chromatography (silica, 10% EtOAc-hexane) 13 yielded the title compound as a white solid. PMR (CDC13) : 8 1.39 (6H, 14 s), 2.02 (2H, t, J = 7.0 Hz), 2.73 (2H, t, J = 7.0 Hz), 3.08 (1H, s), 7.39 (lH,d,J=8.2Hz),7.61(lH,dd,J=1.8,8.2Hz),8.14(lH,d,J=9 16 1.8 Hz).
17 Ethyl-4-iodobenzoate 18 To a suspension of 10 g (40.32 mmol) of 4-iodobenzoic acid in 19 100 mi absolute ethanol was added 2 ml thionyl chloride and the mixture was then heated at reflux for 3 hours. Solvent was removed in 21 vacuo and the residue was dissolved in 100 ml ether. The ether solution 22 was washed with saturated NaHC03 and saturated NaCI solutions and 23 dried (MgS04). Solvent was then removed in vacuo and the residue 24 Kugelrohr distilled (100°C; 0.55 mm) to give the title compound as a colorless oil, PMR (CDCl3): 8 1.42 (3H, t, J " 7 Hz), 4,4 (2H, q, J ~ 7 26 Hz), 7.8 (4H).
27 6-iodonicotinic acid 28 Sodium iodide (20.59 g, 137.40 mmol) was cooled to -78°C under 29 argon and then hydriodic acid (97.13 g, 759.34 mmol) was added. The cooling bath was removed and the suspension was stirred for S minutes.

1 To this mixture was added 6-chloronicotinic acid (22.09 g, 140.20 mmol) 2 and the resulting mixture was slowly warmed to ambient temperature 3 with stirring. The mixture was heated to reflux at 125°C for 24 hours, 4 cooled to ambient temperature and poured into acetone (500 ml) at 0°C. The yellow solid was collected by filtration and washed with 200 6 ml of 1N aqueous NaHS03 solution. Recrystallization from methanol 7 (crystals were washed with ethyl ether) afforded the title compound as 8 white crystals: mp 177-179°C [lit. mp 187-192, Newkome et al.
9 "Reductive Dehalogenation of Electron-Poor Heterocycles: Nicotinic Acid Derivatives" J. Org. Chem. 51: 953-954 (1986). 1H NMR (DMSO-11 d6): 8 8.81 (1H, dd, J = 0.8, 2.4 Hz), 8.01 (1H, dd, J = 0.8, 8.2 Hz), 12 7.91(lH,dd,J=2.4,8.2Hz).
13 Ethyl6-iodonicotinoate 14 To a suspension of 6-iodonicotinic acid (23.38 g, 94.20 mmol) in dichloromethane (100 ml) was added a solution of 1-(3-16 dimethylaminopropyl)-3- ethylcarbodiimide hydrochloride (19.86 g, 103.6 17 mmol) in dichloromethane (250 mI). To this mixture was added ethanol 18 (12.40 g, 269.27 mmol) followed by dimethylaminopyridine (1.15 g, 9.41 19 mmol). The mixture was heated at 50°C for 24.5 hours, concentrated in vacuo, and diluted with water (200 ml) then extracted with ethyl ether 21 (550 ml). The combined organic phases were washed with saturated 22 aqueous NaCI, dried (MgS04) and concentrated to a yellow solid.
23 Purification by flash chromatography (silica, 10% EtOAc-hexane) 24 afforded the title compound as white needles: mp 48-49°C; 1H NMR
(CDCi3): 8 8.94 (1H, d, J = 2.1 Hz), 7.91 (1H, dd, J = 2.1, 8.2 Hz), 26 7.85 (1H, d, J = 8.2 Hz), 4.41 (2H, q, J = 7.1 Hz), 1.41 (3H, t, J = 7.1 27 Hz).
28 Eth~4-.y(5.6,7,8-tetrahydro-5 5-dimethyl-8-oxo-2-29 naphthalenyl)etl~nvllbenzoate {Compound F) To a solution of 4 g (21.7 mmol) of 7-ethynyl-3,4-dihydro-4,4-1 dimethylnaphthalen-1(2H)-one (Compound E ) flushed for 15 minutes 2 with a stream of argon, and 6 g (21.7 mmol) of ethyl 4-iodobenzoate in 3 100 ml of triethylamine was added 5 g (7.2 mmol) of 4 bis(triphenylphosphine)palladium(II) chloride and 1.4 g (7.2 mmol) of cuprous iodide. The mixture was flushed with argon for 5 minutes and 6 then stirred at ambient temperature for 18 hours. The reaction mixture 7 was filtered through Celite and the filtrate was concentrated in vacuo.
8 Purification by flash chromatography (silica, 10 % EtOAc-hexane) 9 yielded the title compound as a white solid. PMR (CDCl3) : 8 1.41 (3H, 1 o t, J = 7.2 Hz), 1.41 (6H, s), 2.04 (2H, t, J = 6.5 Hz), 2.76 (2H, t, J =
11 6.5 Hz), 4.40 (2H, q, J = 7.2 Hz), 7.44 (1H, d, J = 8.2 Hz), 7.59 (2H, d, 12 J = 8.4 Hz), 7.68 (1H, dd, J = 1.8, 8.2 Hz), 8.04 (2H, d, J = 8.4 Hz), 13 8.15 (1H, d, J = 1.8 Hz).
14 Eth~y(5,6-dihydro-5,5-dimeth~(trifluoromethylsulfonyl,~oxy-2-naphthalen~leth~~]benzoate (Compound G) 16 To a cold solution (-78°C) of 291.6 mg (1.59 mmol) of sodium 17 bis(trimethylsily)amide in 5.6 ml of THF was added a solution of 500.0 18 mg (1.44 mmol) of ethyl 4- ((5,6,7,8-tetrahydro-5,5-dimethyl-8-oxo-2-19 naphthalenyl)ethynyl]benzoate (Compound F) in 4.0 ml of THF. The reaction mixture was stirred at -78°C for 35 minutes and then a solution 21 of 601.2 mg (1.59 mmol) of 5-chloro(2-bis-triflouromethylsulfonyl)imide 22 in 4.0 ml of THF was added. After stirring at -78°C for 1 hour, the 23 solution was warmed to 0°C and stirred for 2 hours. The reaction was 24 quenched by the addition of saturated aqueous NH4C1. The mixture was extracted with EtOAc (50 ml) and the combined organic layers 26 were washed with 5% aqueous Na~iH, water, an:~ brine. The organic 27 phase was dried over Na2S04 and then concentrated in vacuo to a 28 yellow oil. Purification by column chromatography (silica, 7% EtOAc-29 hexanes) yielded the title compound as a colorless solid. 1H NMR
(CDCl3): 8 8.04 (2H, dd, J = 1.8, 8.4 Hz), 7.60 (2H, dd, J = 1.8, 8.4 1 Hz), 7.51 (2H, m), 7.32 (iH, d, J = 8.0 Hz), 4.40 (2H, q, J = 7.1 Hz), 2 6.02 (1H, t, J = 5.0 Hz), 2.44 (2H, d, J = 5.0 Hz), 1.43 (3H, t, J = 7.1 3 Hz), 1.33 (6H, s).
4 Ethyl 4-[(5,6-dihydro-5 5-dimethv~4-methXlphen,~~ll-2-nanhthalenylleth~~]benzoate~Compound 1) 6 A solution of 4-lithiotoluene was prepared by the addition of 7 189.9 mg ( 1.74 ml, 2.96 mmol) of t-butyl lithium ( 1.7M solution in 8 hexanes) to a cold solution (-78°C) of 253.6 mg ( 1.482 mmol) of 4-9 bromotoluene in 2.0 ml of THF. After stirring for 30 minutes a l0 solution of 269.4 mg (1.977 mmol) of zinc chloride in 3.0 ml of THF
11 was added. The resulting solution was warmed to room temperature, 12 stirred for 30 minutes, and added via cannula to a solution of 472.9 mg 13 (0.988 mmol) of ethyl 4-((5,6-dihydro-5,5-dimethyl-8-14 (trifluoromethylsulfonyl)oxy-2-naphthalenyl)ethynyl]benzoate (Compound G) and 50 mg (0.04 mmol) of 16 tetrakis(triphenylphosphine)palladium(0) in 4.0 ml of THF. The 17 resulting solution was heated at 50°C for 45 minutes, cooled to room 18 temperature and diluted with sat. aqueous NH4C1. The mixture was 19 extracted with EtOAc (40 ml) and the combined organic layers were 2o washed with water and brine. The organic phase was dried over Na~S04 21 and concentrated in vacuo to a yellow oil. Purification by column 22 chromatography (silica, 5% EtOAc-hexanes) yielded the title compound 23 as a colorless solid. 1H NMR (d6-acetone): s 1.35 (6H, s), 1.40 (3H, t, J
24 = 7.1 Hz), 2.36 (2H, d, J = 4.7 Hz), 2.42 (3H,s), 4.38 (2H, q, J = 7.1 Hz), 5.99 (1H, t, J = 4.7 Hz), 7.25 (SH, m), 7.35 (2H, m), 7.52 (2H, d, J
26 = 8.5 Hz), 7.98 (2H, d, J = 8.5 Hz).
27 Eth~((5.6-dihydro-5 S-dimethyl-8-phenyl-2-28 naphthalene, ethynyl~benzoate ,(Compound lal 29 Employing the same general procedure as for the preparation of ethyl 4-((5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-1 naphthalenyl)ethynylJbenzoate (Compound 1), 203.8 mg {0.43 mmol) of 2 ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-3 naphthalenyl)ethynylJbenzoate (Compound G) was converted into the 4 title compound (colorless solid) using 58.2 mg (0.36 ml, 0.69 mmol) of phenyllithium ( 1.8M solution in cyclohexane/Et20), 116.1 mg (0.85 6 mmol) of zinc chloride and 13.8 mg (0.01 mmol) of 7 tetrakis(triphenylphosphine)palladium(0). PMR (CDCl3): 8 1.36 (6H, 8 s), 1.40 (3H, t, J = 7.lHz), 2.37 (2H, d, J = 4.7 Hz), 4.38 (2H, q, J =
9 7.1 Hz), 6.02 (1H, t, J = 4.7 Hz), 7.20 (1H, d, J = 1.5 Hz), 7.27 (1H, m), 7.39 (6H, m), 7.52 (2H, d, J = 8.2 Hz}, 7.98 (2H, d, J = 8.2 Hz).
11 Ethyl 4-j(5,6-dihydro-5 5-dimeth.~(3-meth, l~phenyll-2-12 naphthalenyl)eth~yllbenzoate (Compound 2) 13 Employing the same general procedure as for the preparation of 14 ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-naphthalenyl)ethynyl]benzoate (Compound 1), 250.0 mg (0.522 mmol) 16 of ethyl 4-((5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-1'7 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the 18 title compound (colorless solid) using 284.8 mg (2.090 mmol) of zinc 19 chloride, 24 mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) in 2.0 ml of THF, and 3-methylphenyl lithium (prepared by adding 201.2 21 mg (1.86 ml, 3.14 mmol) of tert-butyllithium (1.7M solution in pentane) 22 to a cold solution (- 78°C) of 274.0 mg (1.568 mmol} of 3-23 methylbromobenzene in 2.0 ml of THF). 1H NMR (CDC13): 8 7.99 24 (2H, d, J = 8.4 Hz), 7.51 (2H, d, J = 8.4 Hz), 7.39-7.14 (7H, m), 5.99 (1H, t, J = 4.7 : z), 4.37 (2H, q, J = 7.1 Hz), 2.60 (3H, s), 2.35 (2H, d, 26 J = 4.7 Hz), 1.39 (3H, t, J = 7.1 Hz), 1.34 (6H, s).
27 Ethyl 4-[(5,6-dihydro-5,5-dimethyl-~2-meth, l~uhenyll-2-28 naphthalenylleth~yl~benzoate !Compound 3) 29 Employing the same general procedure as for the preparation of ethyl4-[(5,6-dihydro-5,5-dimethyl-8-(4-methylphenyl)-2-1 naphthalenyl)ethynyl]benzoate (Compound 1), 200.0 mg (0.4I8 mmol) 2 of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-3 naphthalenyl)ethynyl]benzoate {Compound G) was converted into the 4 title compound (colorless solid) using 199.4 mg (1.463 mmol) of zinc chloride, 24 mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) 6 in 4.0 ml of THF, and 2-methylphenyl lithium (prepared by adding 133.9 7 mg ( 1.23 ml, 2.09 mmol) of tert-butyilithium ( 1.7M solution in pentane) 8 to a cold solution (- 78°C) of 178.7 mg ( 1.045 mmol) of 2-9 methylbromobenzene in 2.0 ml of THF). 1H NMR (CDCl3): 8 7.97 l0 (2H, d, J = 8.4 Hz), 7.50 (2H, d, J = 8.4 Hz), 7.49-7.19 (6H, m), 6.8I
11 (1H, d, J = 1.6 Hz), 5.89 {1H, t, J = 4.5 Hz), 4.36 {2H, q, J = 7.1 Hz), 12 2.43-2.14 (2H, dq, J = 3.7, 5.4 Hz), 2.15 (3H, s), I.39-1.34 (9H, m).
13 Ethyl 4-[~5,6-dihydro-5 5-dimethyl-8-(3 5-dimeth~phen~)-2-14 naphthalen~leth~n~]benzoate (Compound 4) Employing the same general procedure as for the preparation of 16 ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-1'7 naphthalenyl)ethynyl]benzoate (Compound 1), 250.0 mg (0.522 mmol) 18 of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-19 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title compound (colorless solid) using 249.0 mg ( 1.827 mmol) of zinc 21 chloride, 24 mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) 22 in 2.0 ml of THF, and 3,5-dimethylphenyl lithium (prepared by adding 23 167.7 mg ( 1.54 ml, 2.62 mmol) of tert- butyllithium ( 1.7M solution in 24 pentane) to a cold solution (-78°C) of 249.0 mg (1.305 mmol) of 3,5-dimethylbromobenzene in 2.0 ml of THF). 1H NMR (CDCl3): 8 7.98 26 (2H, d, J = 8.4 Hz), 7.52 (2H, d, J = 8.4 Hz), 7.40-7.33 (2H, m), 7.20 27 (IH, d, J = 1.6 Hz), 7.00 (1H, s), 6.97 (2H, s), 5.97 (1H, t, J = 4.8 Hz), 28 4.37 (2H, q, J = 7.1 Hz), 2.36 (6H, s), 2.34 (2H, d, J = 4.8 Hz), I.39 29 3H, t, J = 7.1 Hz), I.37 (6H, s).
Ether[~5,6-dihydro-5,5-dimethyl-8-i(4-ethylnhenvll-2-......_...,...,.~. ~...~.-».~..._.....-..~._-.",....-.~._-~.~..~.-..~..~...-.~.

1 naphthalenyl~~nylJbenzoate~Compound 5) 2 Employing the same general procedure as for the preparation of 3 ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-4 naphthalenyl)ethynylJbenzoate (Compound 1), 250.0 mg (0.522 mmol) of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-6 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the 7 title compound (colorless solid) using 249.0 mg (1.827 mmol) of zinc 8 chloride, 24 mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) 9 in 2.0 ml of THF, and 4-ethylphenyl lithium (prepared by adding 167.7 l0 mg (i.54 ml, 2.62 mmol) of tert-butyllithium (1.7M solution in pentane) 11 to a cold solution (-78°C) of 244.0 mg ( 1.305 mmol) of 4-12 ethylbromobenzene in 2.0 ml of THF). 1H NMR (CDCI~): 8 7.99 (2H, 13 d, J = 8.4 Hz), 7.51 (2H, d, J = 8.4 Hz), 7.42 - 7.24 (7H, m), 5.99 (1H, 14 t,J=4.7Hz),4.37(2H,q,J=7.lHz),2.71(2H,q,J=7.6Hz),2.35 (2H, d, J = 4.7 Hz), 1.39 ( 3H, t, J = 7.1 Hz), 1.34 {6H, s).
16 Ethyl4-[(5,6-dihydro-5,5-dimeth~~4~11-dimethyleth~~phenyy-2-17 naphthalenyl)ethynyllbenzoate (Compound 6) 18 Employing the same general procedure as for the preparation of 19 ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(2- thiazolyl)-2-naphthalenyl)ethynyl)benzoate (Compound 1), 250.0 mg (0.52 mmol) of 21 ethyl4-[(5,6-dihydro-5,5-dimethyl-8-(trifluoromethylsulfonyl)oxy-2-22 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the 23 title compound (colorless solid) using 142.4 mg (1.045 mmol) of zinc 24 chloride and 4-tert- butylphenyl lithium (prepared by adding 100.6 mg {0.97 ml, 1.57 mmoi) of tert-butylljthium (1.5M solution in pentane) t~~
26 a cold solution (-78°C) of 167.0 mg (0.78 mmol) of 4-tert-27 butyibromobenzene in I.0 ml of THF) . 1H NMR (CDCl3): 8 7.99 (~
28 d, J = 8.4 Hz), 7.55 (2H, d, J = 8.4 Hz), 7.28-7.45 (7H, m), 6.02 (1H, t, 29 J = 4.9 Hz), 4.38 (2H, q, J = 7.2 Hz), 2.36 (2H, d, J = 4.9 Hz), 1.59 (3H, s), 1.40 (3H, t, J = 7.2 Hz), 1.39 (9H, s), 1.35 (6H, s).

1 Ethyl 4-((S,6-dihydro-5 S-dimethyl-8-(4-chlorophenYl)-2-2 nanhthalen~leth~n~J~benzoate (Compound 7) 3 Employing the same general procedure as for the preparation of 4 ethyl 4-[(S,6-dihydro-S,S-dimethyl-8-(4- methylphenyl)-2-naphthalenyl)ethynylJbenzoate (Compound 1), 250.0 mg (O.S22 mmol) 6 of ethyl 4-[(5,6-dihydro-S,S- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-7 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the 8 title compound (colorless solid) using 249.0 mg ( 1.827 mmol) of zinc 9 chloride, 24 mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) in 2.0 ml of THF, and 4-chlorophenyl lithium (prepared by adding 167.7 11 mg (1.54 ml, 2.62 mmol) of tert-butyllithium (1.7M solution in pentane) 12 to a cold solution (- 78°C) of 252.4 mg (1.305 mmol) of 4-chloro-1-13 bromobenzene in 2.0 ml of THF). 1H NMR (CDC13): 8 7.98 (2H, d, J
14 = 8.4 Hz), 7.53 (2H, d, J = 8.4 Hz), 7.40-7.27 {6H, m), 7.12 (1H, d, J =
1.6 Hz), 6.00 (1H, t, J = 4.8 Hz), 4.37 (2H, q, J = 7.1 Hz), 2.35 (2H, d, 16 J = 4.8 Hz), 1.40 (2H, t, J = 7.I Hz), 1.34 (6H, s).
17 Ethvl 4-x(5,6-dihydro-S S-dimethy~4-methoxXphenyll-2-18 naohthalen~leth~yl~benzoate (Compound 81 19 Employing the same general procedure as for the preparation of 2o ethyl 4-[(5,6-dihydro-S,S-dimethyl-8-(4- methylphenyl)-2-21 naphthalenyl)ethynyl)benzoate (Compound 1), 250.0 mg (O.S22 mmol) 22 of ethyl 4-((S,6-dihydro-S,S- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-23 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the 24 title compound (colorless solid) using 249.0 mg (1.827 mmol) of zinc chloride, 24 mg {0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) 26 in 2.0 ml of THF, and 4-methoxyphenyl lithium (prepared by adding 27 167.7 mg (1.54 ml, 2.62 mmol) of tent-butyllithium (1.7M solution in 28 pentane) to a cold solution (-78°C) of 244. I mg ( 1.305 mmol) of 4-29 methoxy-1-bromobenzene in 2.0 ml of THF). 1H NMR (CDCl3): 8 7.98 (2H, d, J = 8.S Hz), 7.52 (2H, d, J = 8.6 Hz), 7.40-7.21 (SH, m), 6.95 _.~..~~.._~._..~.-."-.w--w...w.~ .. r...~.. . .. .,... _ ~.-_......~.... .

1 (2H, d, J = 8.7 Hz), 5.97 (1H, t, J = 4.7 Hz), 4.37 (2H, q, J = 7.1 Hz), 2 4.34 (3H, s), 2.34 (2H, d, J = 4.7 Hz), 1.39 (3H, t, J = 7.1 Hz), 1.34 3 (6H, s).
4 Ether((5,6-dihydro-5,5-dimethvl-8-(4-trifluorometh~phen~l-2-naphthalenylyeth~~]benzoate (Compound 9) 6 Employing the same general procedure as for the preparation of 7 ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-8 naphthalenyl)ethynyl]benzoate (Compound 1), 250.0 mg (0.522 mmol) 9 of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-naphthalenyl)ethynyl]benzoate (Compound G) was converted into the 11 title compound (colorless solid) using 249.0 mg (1.827 mmol) of zinc 12 chloride, 24 mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) 13 in 2.0 ml of THF, and 4-trifluoromethylphenyl lithium (prepared by 14 adding 167.7 mg (1.54 ml, 2.62 mmoI) of tert-butyllithium (1.7M
solution in pentane) to a cold solution (-78°C) of 296.6 mg (1.305 16 mmol) of 4- trifluoromethylbromobenzene in 2.0 ml of THF). 1H
17 NMR (CDCl3): 8 7.98 (2H, d, J = 8.5 Hz), 7.67 (2H, d, J = 8.3 Hz), 18 7.54 - 7.36 (6H, m), 7.10 (1H, d, J = 1.6 Hz), 6.06 (IH, t, J = 4.8 Hz), 19 4.37 (2H, q, J = 7.1 Hz), 2.38 (2H, d, J = 4.8 Hz), 1.39 (3H, t, J = 7.1 Hz), 1.35 (6H, s).
21 Ether((5,6-dihydro-5,5-dimethyl-8~2 ~yrid,1~)-2-22 naphthalenyllethyn~~benzoate (Compound 10) 23 Employing the same general procedure as for the preparation of 24 ethyl4-((5,6-dihydro-5,5-dimethyl-8-(4-methylphenyl)-2-naphthalenyl)ethynyl]benzoate (Compound 1), 250.0 m-~ '0.52 mmol) of 2~ ethyl4-((5,6~dihydro-5,5-dimethyl-8-y:rifluoromethylsu. yl)oxy-2-27 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the 28 title compound (colorless solid) using 142.4 mg (1.045 mmol) of zinc 29 chloride and 2-lithiopyridine (prepared by the addition of 100.6 mg (0.97 ml, 1.57 mmol) of tert-butyllithium (1.SM solution in pentane) to 1 a cold solution (-78°C) of 123.8 mg (0.784 mmol) of 2-bromopyridine in 2 1.0 ml of THF). 1H NMR (d6-acetone): 8 8.64 (1H, m), 7.99 (2H, d, J
3 =8.SHz),7.85(lH,ddd,J=1.8,7.7,9.SHz),7.58(2H,d,J=8.4 4 Hz), 7.50 (1H, d, J = 7.7 Hz), 7.47 (2 H, d, J = 1.1 Hz), 7.35 (2H, m), 6.32 (1H, t, J = 4.8 Hz), 4.34 (2H, q, J = 7.2 Hz), 2.42 (2H, d, J = 7.4 6 Hz), 1.35 (3H, t, J = 7.0 hz), 1.35 (6H, s).
7 Ethyl 4-[(5.6-dihydro-5 5-dimethyl-8-(3-pvridy11~2-8 nanhthalen~leth~~rl,]benzoate (Compound 11) 9 Employing the same general procedure as for the preparation of ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-11 naphthalenyl)ethynyl]benzoate (Compound 1), 170.0 mg (0.35 mmol) of 12 ethyl4-[(5,6-dihydro-5,5-dimethyl-8-(trifluoromethylsulfonyl)oxy-2-13 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the 14 title compound (colorless solid) using 142.4 mg (1.045 mmol) of zinc chloride and 3-lithiopyridine (prepared by the addition of 100.2 mg 16 (0.92 ml, 1.56 mmoI) of tert-butyllithium (1.SM solution in pentane) to 17 a cold solution (-78°C) of 123.8 mg (0.784 mmol) of 3-bromopyridine in 18 1.0 ml of THF). 1H NMR (CDCI3): 8 8.63-8.61 (2H, dd, J = 1.7 Hz), 19 7.99 2H, d, J = 8.4 Hz), 7.67 (1H, dt, J = 7.9 Hz), 7.52 (2H, d, J = 8.4 Hz), 7.43-7.34 (3H, m), 7.10 (1H, d, J = 1.6 Hz), 6.07 (1H, t, J = 4.7 21 Hz), 4.37 (2H, q, J = 7.1 Hz), 2.40 (2H, d, J = 4.7 Hz), 1.390 (3H, t, J
22 = 7.1 Hz), 1.36 (6H, s).
23 Ethvl 4-((5,6-dihydro-5 5-dimeth~(2-methyl-5-p r~idyl)-2-24 naphthalenyl)ethyn~~benzoate (Compound 12) Employing the same general procedure as for the preparation of 26 ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenlyl)-2-27 naphthalenyl)ethynyl]benzoate (Compound 1), 250.0 mg (0.522 mmol) 28 of ethyl 4-[(5,6- dihydro-5,5-dimethyl-8-(trifluoromethylsulfonyl)oxy-2-29 naphthalenyl)ethynyl]benzoate {Compound G) was converted into the title compound (colorless solid) using 142.4 mg ( 1.045 mmol) of zinc . ..a.....w..~...m-.....~.. ~w. ~.W .. ... . . .. . ......_~....~.w~~,.~.-...~....~.~,..-..~.~~....~._.

1 chloride and 2-methyl-5- lithiopyridine (prepared by the addition of 2 100.5 mg (0.92 ml, i.57 mmol) of tert-butyllithium (1.7 M solution in 3 pentane) to a cold solution (-78°C) of 134.8 mg (0.784 mmol) of 2-4 methyl-5-bromopyridine in 1.0 ml of THF). 1H NMR (CDC13): b 8.50 (lH,d,J=2.2Hz),7.99(2H,d,J=8.3Hz),7.56(lH,dd,J=2.3,8.0 6 Hz), 7.53 (2H, d, J = 8.4 Hz), 7.43 (1H, dd, J = 2.3, 8.0 Hz), 7.37 (2H, 7 d,J=8.OHz),7.21(lH,d,J=8.lHz),7.11(lH,d,J=l.SHz),6.04 8 (1H, t, J = 4.7 Hz), 4.38 (2H, q, J = 7.2 Hz), 2.63 (3H, s), 2.38 (2H, d, 9 J = 4.6 Hz), 1.40 (3H, t, J = 7.1 Hz}, 1.35 (6H, s).
Ether[~5,6-dihydro-5,5-dimethyl-8-y-((2.2- dimeth,1~-11 dimeth lsy iloxyyphenyly-2- naphthalenyl, eth~yl~benzoate (Compound H}
12 Employing the same general procedure as for the preparation of 13 ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-14 naphthalenyl)ethynyl)benzoate (Compound G), 150.0 mg (0.314 mmol) of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-16 naphthalenyl)ethynyl)benzoate (Compound G) was converted into the 17 title compound (colorless solid) using 150.0 mg (1.10 mmol) of zinc 18 chloride, 24 mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) 19 in 2.0 ml of THF, and 3-((2,2-dimethylethyl)dimethylsiloxy)phenyl lithium (prepared by adding 100.2 mg (0.92 ml, 1.564 mmol) of tert-21 butyllithium (1.7M solution in pentane) to a cold solution (-78°C) of 22 226.0 mg (0.787 mmol) of 3-((2,2-23 dimethylethyl)dimethylsiloxy)bromobenzene in 2.0 ml of THF). 1H
24 NMR (CDC13): 8 ?.98 (2H, d, J = 8.4 Hz), 7.51 (2H, d, J = 8.4 Hz), 7.40-7.22. {4H, m}, 6.95 ~ , d, J = 7. Hz), 6.84-6.82 {2H, m), 6.00 26 {1H, t, J = 4.7 H~~), 4.3 vi, q, J = ';.1 Hz), 2.35 (2H, d, J = 4.7 Hz), 27 1.39 (3H, t, J = 7.1 Hz), r.34 (3H, s), 0.99 (9H, s), 0.23 (6H, s).
28 Ethyl4-[{5,6-dih~dro-5,5-dimethyl-8-(4-({2,2-dimethyleth~~
29 dimeth ls~xylphen~)-2- naphthalene eth~yllbenzoate (Compound I1 Employing the same general procedure as for the preparation of 1 ethyl 4-((5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-2 naphthalenyl)ethynyl]benzoate (Compound 1), 210.0 mg (0.439 mmol) 3 of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-4 naphthalenyl)ethynyl~benzoate (Compound G) was converted into the title compound (colorless solid) using 209.0 mg (1.53 mmol) of zinc 6 chloride, 24 mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) 7 in 2.0 ml of THF, and 4-((2,2-dimethylethyl)dimethylsiloxy)phenyl 8 lithium (prepared by adding 140.3 mg (1.30 ml, 2.19 mmol) of tert-9 butyllithium (1.7M solution in pentane) to a cold solution (-78°C) of l0 315.0 mg (1.09 mmol) of 4-((2,2-11 dimethylethyl)dimethylsiloxy)bromobenzene in 2.0 ml of THF). 1H
12 NMR (CDCI3): 8 7.98 (2H, d, J = 8.4 Hz), 7.51 (2H, d, J = 8.4 Hz), 13 7.39-7.25 (3H, m), 7.21 (2H, d, J = 8.5 Hz), 5.87 (2H, d, J = 8.5 Hz), 14 5.96(lH,t,J=4.7Hz),4.37(2H,q,J=7.lHz),2.33(2H,d,J=4.7 Hz), 1.39 {3H, t, J = 7.1 Hz), 1.33 (6H, s), 1.01 (9H, s), 0.25 (6H, s).
16 Ethyl 4-[(5,6-dihydro-5 5-dimethyl-8=(3-hydroxyphenyl)-2-17 naphthalen~)ethvn~~benzoate (Compound 13) 18 To a solution of ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(3-((2,2-19 dimethylethyl)-dimethylsiloxy)- phenyl)-2-naphthalenyl)ethynyl]benzoate (Compound H) 60.0 mg (0.114 mmol) in 1.0 ml of THF at room 21 temperature was added 91.5 mg (0.35 ml, 0.35 mmol) of 22 tetrabutylamonium flouride (1 M solution in THF). After stirring 23 overnight, the solution was diluted with EtOAc and washed with H20 24 and saturated aqueous NaCI, before being dried over MgS04. Removal of the solvents under reduced pressure, followed by column 26 chromatography (4:1, Hexanes:EtOAc) afforded the title compound as a 27 colorless solid. 1H NMR (CDC13): 8 7.98 (2H, d, J = 7.8 Hz), 7.52 28 (2H, d, J = 8.3 Hz), 7.39 - 7.21 (4H, m), 6.93 (1H, d, J = 7.5 Hz), 6.84 29 (1H, d, 7.1 Hz), 6.83 (1H, s), 6.01 1H, t, J = 4.7 Hz), 4.91 (1H, s), 4.39 (2H, q, J = 7.1 Hz), 2.35 (2H, d, J = 4.7 Hz), 1.39 (3H, t, J = 7.1 Hz), ..... "-.-.- .,.w~ .. ~..~~_ .~-.w~w..v..,...~.w_..~..~....~......

1 1.34 (6H, s).
2 Ethyl4-[(5,6-dihydro-5,5-dimethyl-8-(4-h~droxyphen~L2-3 naphthalen~lleth~nyl]benzoate i(Com~ound 14~
4 To a solution of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(4-((2,2-dimethylethyl)- dimethylsiloxy)phenyl)-2-naphthalenyl}ethynyl]benzoate 6 (Compound I) 50.0 mg (0.095 mmol) in 1.0 ml of THF at room 7 temperature was added 73.2 mg (0.29 ml, 0.29 mmol) of 8 tetrabutylamonium fluoride (1 M solution in THF). After stirring 9 overnight, the solution was diluted with EtOAc and washed with H20 and saturated aqueous NaCI, before being dried over MgS04. Removal 11 of the solvents under reduced pressure, followed by column 12 chromatography (4:1, Hexanes:EtOAc) afforded the title compound as a 13 colorless solid. 1H NMR (CDCl3): b 7.98 (2H, d, J = 8.2 Hz), 7.52 14 (2H, d, J = 8.3 Hz), 7.41 - 7.20 (5H, m), 6.88 (2H, d, J = 8.4 Hz), 5.96 (lH,t,J=4.5Hz),4.37(2H,q,J=7.lHz),2.34(2H,d,J=4.5Hz), 16 1.39 (3H, t, J = 7.1 Hz), 1.34 (6H, s).
17 Ethyl 4-[~5,6-dihydro-5,5-dimeth~(5-methylthiazol-2-. ly 1-2-18 na~hthalenyhynyll benzoate (Compound 15) 19 Employing the same general procedure as for the preparation of 2o ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-21 naphthalenyl)ethynyl]benzoate (Compound 1), 264.0 mg (0.552 mmol) 22 of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-23 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the 24 title compound (colorless solid) using 150.0 mg (1.10 mmol} of zinc chloride, 14 mg (0.012 mmol) of 26 tetrakis(triphenylphosphine)palladium(0) in 4.0 ml of THF, and 5-27 methylthiazol-2-yl lithium (prepared by adding 53.2 mg (0.53 ml, 0.83 28 mmol) of n- butyllithium (1.55 M solution in hexanes) to a cold solution 29 (-78°C) of 82.0 mg (0.83 mmol} of 5-methylthiazole in 5.0 ml of THF).
1H NMR (CDC13): 8 7.99 (2H, d, J = 7.8 Hz), 7.88 (1H, d, J = 1.5 1 Hz), 7.55 (2H, d, J = 7.8 Hz), 7.54 (1H, s), 7.45 (1H, dd, J = 1.5, 8.0 2 Hz),7.35(lH,d,J=7.9Hz),6.48(lH,t,J=4.8Hz),4.38(2H,q,J
3 = 7.1 Hz), 2.51 (3H, s), 2.38 (2H, d, J = 4.8 Hz), 1.40 (3H, s), 1.32 (6H, 4 s).
Ethvl 4-[l5,6-dihvdro-5 5-dimethyl-8-(2-thiazolyll-2-6 naphthalenyl ethYnyl)benzoate {Compound 15a) 7 A solution of 2-lithiothiazole was prepared by the addition of 41.2 8 mg (0.42 ml, 0.63 mmol) of n-butyl- lithium (1.SM solution in hexanes) 9 to a cold solution (-78 °C) of 53.4 mg (0.63 mmol) of thiazole in 1.0 ml l0 of THF. The solution was stirred at for 30 minutes and then a solution 11 of 113.9 mg (0.84 mmol) of zinc chloride in 1.5 ml of THF was added.
12 The resulting solution was warmed to room temperature, stirred for 30 13 minutes and then the organozinc was added via cannula to a solution of 14 200.0 mg (0.42 mmol) of ethyl 4-[(5,6- dihydro-5,5-dimethyl-8-(trifluoromethylsulfonyl)oxy-2- naphthalenyl)ethynyl]benzoate 16 (Compound G) and 12.4 mg (0.01 mmol) of 17 tetrakis(triphenylphosphine)palladium(0) in 1.5 ml of THF. The 18 resulting solution was heated at 50°C for 45 minutes, cooled to room 19 temperature and diluted with sat. aqueous NH4Cl. The mixture was extracted with EtOAc (40 ml) and the combined organic layers were 21 washed with water and brine. The organic phase was dried over Na2S0~
22 and concentrated in vacuo to a yellow oil. Purification by column 23 chromatography (silica, 20% EtOAc-hexanes) yielded the title 24 compound as a colorless oil. PMR (CDCl3): 8 1.35 (6H, s), 1.40 (3H, t, J = 7.1 Hz), 2.42 (2H, d, J = 4.8 Hz), 4.38 (2H, q, J = 7.1 Hz), 6.57 26 (lH,t,J=4.8Hz),7.33(lH,d,J=3.3Hz),7.36(lH,d,J=8.OHz), 27 7.46(lH,dd,J=1.7,8.1Hz),7.55{2H,d,J=8.4Hz),7.87{lH,d,J
28 = 1.7 Hz), 7.92 (1H, d, J = 3.3 Hz), 8.00 (2H, d, J = 8.4 Hz).
29 Ethvl 4-[~5.6-dihydro-5 5-dimethyl-8-{4-methylthiazol-2-,~~1)-2-naphthalen~)eth~n~] benzoate (Compound 16}

1 Employing the same general procedure as for the preparation of 2 ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-3 naphthalenyl)ethynyl]benzoate (Compound 1), 295.0 mg (0.617 mmol) 4 of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-{trifluoromethylsulfonyl)oxy-2-naphthalenyl)ethynyl]benzoate (Compound G) was converted into the 6 title compound (colorless solid} using 168.0 mg (1.23 mmol) of zinc 7 chloride, 16 mg (0.014 mmol) of 8 tetrakis(triphenylphosphine)palladium(0) in 6.0 ml of THF, and 4-9 methylthiazol-2-yl lithium (prepared by adding 59.6 mg (0.60 ml, 0.93 mmol) of n- butyllithium (1.55 M solution in hexanes) to a cold solution ~ 1 (-78°C) of 92.0 mg (0.93 mmol) of 4-methylthiazole in 6.0 ml of THF).
12 1H NMR (CDC13): 8 8.00 (2H, d, J = 8.4 Hz), 7.80 (1H, d, J = 1.7 13 Hz), 7.55 (2H, d, J = 8.4 Hz), 7.45 (1H, dd, J = 1.7, 8.0 Hz), 7.35 (1H, 14 d, J = 8.0 Hz), 6.87 (1H, s), 6.52 (1H, t, J = 4.7 Hz), 4.37 (2H, q, J =
7.2 Hz), 2.54 (3H, s), 2.39 (2H, d, J = 4.7 Hz), 1.40 (3H, t, J = 7.2 Hz), 16 1.33 (3H, s).
17 Ethyl4-((5,6-dihydro-5,5-dimeth~(4,5-dimethylthiazol-2-~)-2-18 naphthalene eth~yll benzoate (Compound 17}
19 Employing the same general procedure as for the preparation of ethyl4-[(5,6-dihydro-5,5-dimethyl-8-(4-methylphenyl)-2-21 naphthalenyl)ethynyl)benzoate (Compound 1), 200.0 mg (0.418 mmol) 22 of ethyl 4-((5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-23 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the 24 title compound (colorless solid) using 110.0 mg (0.84 mmol) of zinc chloride, 12 mg (0.011 mmol) of 26 tetrakis(triphenylphosphine)palladium(0) in 2.0 ml of THF, and 4,5-27 dimethylthiazol-2-yl lithium (prepared by adding 40.2 mg (0.39 ml, 0.63 28 mmol) of n- butyllithium (1.55 M solution in hexanes) to a cold solution 29 (-78°C) of 71.0 mg (0.63 mmol) of 4,5-dimethylthiazole in 2.0 ml of THF). 1H NMR (CDCl3): 8 8.00 (2H, d, J = 8.4 Hz), 7.82 (1H, d, 3 =

1 1.7 Hz), 7.54 (2H, d, J = 8.4 Hz ), 7.43 (1H, dd, J = 1.7, 8.0 Hz), 7.33 2 91H, d, J = 8.0 Hz), 6.45 (1H, t, J = 4.9 Hz), 4.38 (2H, q, J = 7.1 Hz), 3 2.41 (3H, s), 2.40 (3H, s), 2.37 (2H, d, J = 4.9 Hz), 1.40 (3H, t, J = 7.1 4 Hz), 1.32 {6H, s).
4-~(5,6-Dihydro-5,5-dimeth~8~2-methyl-5-pyrid,~~l)-2-6 naphthalenyl)eth~mvl]benzoic acid (Compound 18) 7 A solution of 81.7 mg (0.194 mmol) of ethyl 4- ((5,6-dihydro-5,5-8 dimethyl-8-(2-methyl-5-pyridyl)-2- naphthalenyl)ethynyl]benzoate 9 (Compound 12) and 40.7 mg (0.969 mmol) of LiOH-H20 in 3 ml of THF/water (3:1, v/v), was stirred overnight at room temperature. The 11 reaction was quenched by the addition of saturated aqueous NH4C1 and 12 extracted with EtOAc. The combined organic layers were washed with 13 water and brine, dried over Na2S04 and concentrated in vacuo to give 14 the title compound as a colorless solid. 1H NMR (d6-DMSO): 8 8.41 (1H, d, J = 1.9 Hz), 7.90 (2H, d, J = 8.3 Hz), 7.63 (1H, dd, J = 2.3, 7.9 16 Hz), 7.59 (2H, d, J = 8.3 Hz), 7.49 (2H, m), 7.33 (1H, d, J = 7.8 Hz), 17 6.95(lH,s),6.11(lH,t,J=4.SHz),2.52(3H,s),2.37(2H,d,J=4.6 18 Hz), 1.31 (6H, s).
19 4-((5,6-Dihydro-5,5-dimethyl-8-.(2-p, rid ly_)-2-naphthalenvl eth,~n~)benzoic acid (Compound 19) 21 A solution of 80.0 mg (0.196 mmol) of ethyl 4- [(5,6-dihydro-5,5-22 dimethyl-8-(2-pyridyl)-2- naphthalenyl)ethynyl]benzoate (Compound 10) 23 and 20.6 mg (0.491 mmol) of LiOH-H20 in 3 ml of THF/water (3:1, 24 v/v), was stirred overnight at room temperature. The reaction was quenched by the addition of saturated aqueous NH4Cl and extracted 26 with EtOAc. The combined organic layers were washed with water and 27 brine, dried over Na2S04 and concentrated in vacuo to give the title 28 compound as a colorless solid. 1H NMR (d6-DMSO): 8 8.64 (1H, m), 29 7.94 {2H, d, J = 8.3 Hz), 7.87 (1H, dt, J = 1.7, 7.8 Hz), 7.58 {2H, d, J
= 8.3 Hz), 7.50 (1H, d, J = 8.2 Hz), 7.47 (2H, s), 7.37 ( 1H, m), 7.25 .._,_.-...~~.-,...-....,~"~".~,~.- ..~.,u.._..~ .. . ~........".~.,._..,~.~.~
w ~w"...w.,~.~.~~.~....~_. _ .......__.~

1 (1H, s), 6.30 (1H, t, J = 4.6 Hz), 2.39 (2H, d, J = 4.6 Hz), 1.31 (6H, s).
2 4-[~5,6-Dihydro-5,5-dimethyl-8-(3-meth l~phenyll-2-3 naphthalene ethyn~]benzoic acid (Compound 20) 4 To a solution of ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(3-methylphenyl)-2-naphthalenyl)ethynyl]benzoate (Compound 2) 30.Omg 6 (0.071 mmol) in 3 ml of EtOH and 2 ml of THF was added 28.0 mg 7 (0.70 mmol, 0.7 ml) of NaOH (1.0 M aqueous solution). The solution 8 was heated to 50°C for 2 hours, cooled to room temperature, and 9 acidified with 10% HCI. Extraction with EtOAc, followed by drying l0 over Na2S04, and removal of the solvents under reduced pressure I1 afforded the title compound as a colorless solid. 1H NMR {DMSO): 8 12 7.90 (2H, d, J = 8.5 Hz), 7.59 (2H, d, J = 8.5 Hz), 7.46 (2H, s), 13 7.32-7.13 (4H, m), 7. I0 (1H, s), 6.98 (1H, t, J = 4.5 Hz), 2.34 (3H, s), 14 2.31 (2H, d, J = 4.5 Hz), 1.30 (6H, s).
4-[(5,6-Dihydro-5.5-dimethyl-8-(4~eth~phenyll-2-16 naphthalenyl)eth~~]benzoic acid (Compound 21) 17 To a solution of ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(4-18 ethylphenyl)-2-naphthalenyl)ethynyl]benzoate (Compound 5) 47.Omg 19 (0.108 mmol) in 3 mi of EtOH and 2 ml of THF was added 28.0 mg (0.70 mmol, 0.7 ml) of NaOH (1.0 M aqueous solution). The solution 21 was heated to 50°C for 2 hours, cooled to room temperature, and 22 acidified with 10% HCI. Extraction with EtOAc, followed by drying 23 over Na2S04, and removal of the solvents under reduced pressure 24 afforded the title compound as a colorless solid. 1H NMR (DMSO): 8 7.90 (21-i, d, J = 8.3 Hz), 7.59 (2H, d, J = 8.3 Hz), 7.46 (2H, s), 26 7.29-7.21 (4H, m), 7.02 (1H, s), 6.U1 (1H, t, J = 4.5 Hz), 2.64 (2H, q, J
27 = 7.5 Hz), 2.33 (2H, d, J = 4.5 Hz), 1.29 (6H, s), 1.22 (3H, t, J = 7.5 28 Hz).
29 4-[(5,6-Dihydro-5,5-dimeth 1-~8-,(4-methoxxphenyl)-2 naphthalen~lethvnyllbenzoic acid (Compound 22) 1 To a solution of ethyl 4-((5,6-dihydro-5,5-dimethyl-8-(4-2 methoxyphenyl)-2-naphthalenyl)ethynyl]benzoate (Compound 8) 80.0 mg 3 (0.183 mmol) in 3 ml of EtOH and 2 ml of THF was added 40.0 mg 4 (1.00 mmol, 1.0 ml) of NaOH (1.0 M aqueous solution). The solution was heated to 50°C for 2 hours, cooled to room temperature, and 6 acidified with 10% HCI. Extraction with EtOAc, followed by drying 7 over Na2S04, and removal of the solvents under reduced pressure 8 afforded the title compound as a colorless solid. 1H NMR (DMSO): 8 9 7.90 (2H, d, J = 8.3 Hz), 7.60 (2H, d, J = 8.3 Hz), 7.45 (2H, s), 7.24 (2H, d, J = 8.6 Hz), 7.02-6.89 (3H, m), 5.98 {1H, t, J = 4.4 Hz), 3.79 t 1 (3H, s), 2.31 (2H, d, J = 4.7 Hz), 1.29 (6H, s).
12 4-f (5,6-Dihydro-5 5-dimeth~l-8-(4-trifluoromethylphenyll-2-13 nanhthalenyl)eth~~]benzoic acid Compound 23) 14 To a solution of ethyl 4-((5,6-dihydro-S,5-dimethyl-8-(4-trifluoromethylphenyl)- -2-naphthalenyl)ethynyl]benzoate (Compound 9) 16 70.0 mg (0.148 mmol) in 3 ml of EtOH and 2 ml of THF was added 17 60.0 mg (1.50 mmol, 1.50 ml) of NaOH (1.0 M aqueous solution). The 18 solution was heated to 50°C for 2 hours, cooled to room temperature, 19 and acidified with 10% HCI. Extraction with EtOAc, followed by drying over NaZS04, and removal of the solvents under reduced pressure 2t afforded the title compound as a colorless solid. 1H NMR (DMSO): 8 22 7.90 (2H, d, J = 8.3 Hz), 7.80 (2H, d, J = 8.1 Hz), 7.61-7.47 (6H, m), 23 6.97 (2H, s), 6.16 (1H, t, J = 4.5 Hz), 2.37 (2H, d, J = 4.6 Hz), 1.30 24 (6H, s).
4-[(5,~, 6DihYdro-5 5-dimeth~-8-(3 5-dimeth,~~lphenyll-2-26 nanhthalen~, eth~yllbenzoic acid (Compound 241 27 To a solution of ethyl 4-((5,6-dihydro-5,5-dimethyl-8-(3,5-28 dimethylphenyl)-2-naphthalenyl)ethynyl]-benzoate (Compound 4) 90.0 29 mg (0.207 mmol) in 3 ml of EtOH and 2 ml of THF was added 48.0 mg (1.20 mmol, 1.20 ml) of NaOH (1.0 M aqueous solution). The solution 1 was heated to 50°C for 2 hours, cooled to room temperature, and 2 acidified with 10% HCI. Extraction with EtOAc, followed by drying 3 over NaZS04, and removal of the solvents under reduced pressure 4 afforded the title compound as a colorless solid. 1H NMR (DMSO): 8 7.90 (2H, d, J = 8.2 Hz), 7.59 (2H, d, J = 8.2 Hz), 7.45 (2H, s), 7.00 6 (lH,s),6.97(lH,s),5.97(lH,t,J=4.SHz),2.31(2H,d,J=4.SHz), 2.30 (6H, s), 1.29 (6H, s).
8 4-[{5.6-Dihydro-5,5-dimeth~{4-chloro~hen,1~)-2-9 naphthalenvllethyn~]benzoic acid Compound 25~
To a solution of ethyl 4-[{5,6-dihydro-5,5-dimethyl-8-(4-11 chlorophenyl)-2-naphthalenyl)ethynyl)benzoate (Compound 7) 80.0 mg 12 (0.181 mmol) in 3 ml of EtOH and 2 ml of THF was added 48.0 mg 13 (1.20 mmol, 1.20 ml) of NaOH (1.0 M aqueous solution). The solution 14 was heated to 50°C for 2 hours, cooled to room temperature, and acidified with 10% HCI. Extraction with EtOAc, followed by drying 16 over Na2S04, and removal of the solvents under reduced pressure 17 afforded the title compound as a colorless solid. 1H NMR {DMSO): 8 18 7.90 {2H, d, J = 8.3 Hz), 7.60 (2H, d, J = 8.3 Hz), 7.51-7.48 (4H, m), 19 7.34 (2H, d, J = 8.4 Hz), 6.97 (1H, s), 6.07 (1H, t, J = 4.5 Hz), 2.34 (2H, d, J = 4.6 Hz), 1.29 (6H, s).
2Z 4-(f5,6-Dihydro-5.5-dimeth~-8~3-pyrid~)-2-22 naphthalenyl ethyn~]benzoic acid~Compound 26) 23 To a solution of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(3-24 pyridyl)-2-naphthalenyl)ethynyl)benzoate (Comps ~ wd 11) 45.0 mg (0.110 mmol) in 3 ml of EtOH and 2 ml of TE '-~ was :;d 48.0 mg (1.20 26 mmol, 1.20 ml) of NaOH (1.0 M aqueous solut~~n). The solution was 27 heated to 50°C for 2 hours, cooled to room temperature, and acidified 28 with 10% HCI. Extraction with EtOAc, followed by drying over 29 Na2S04, and removal of the solvents under reduced pressure afforded the title compound as a colorless solid. 1H NMR (DMSO): b 8.60 1 (lH,d,J=4.6Hz),8.55(lH,s),7.90(2H,d,J=8.3Hz),7.76(lH,d, 2 J = 7.5 Hz), 7.60 (2H, d, J = 8.3 Hz), 7.51-7.46 (3H, m), 6.94 (1H, s), 3 6.14(lH,t,J=4.5Hz),2.37(2H,d,J=4.5Hz),1.31(6H,s).
4 4-[15,6-Dihydro-5,5-dimethy~2-methy_lphenyl)-2-nanhthalenvl)eth~~jbenzoic acid ~Com ound 27~
6 To a solution of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(2-7 methylphenyl)-2-naphthalenyl)ethynylj- benzoate (Compound 3) 80.0 mg 8 (0.190 mmol) in 3 ml of EtOH and 2 ml of THF was added 60.0 mg 9 ( 1.50 mmol, 1.50 ml) of NaOH ( 1.0 M aqueous solution). The solution l0 was heated to 50°C for 2 hours, cooled to room temperature, and 11 acidified with 10% HCI. Extraction with EtOAc, followed by drying 12 over Na2S04, and removal of the solvents under reduced pressure 13 afforded the title compound as a colorless solid. 1H NMR (DMSO): 8 14 7.89(2H, d, J = 8.4 Hz), 7.57 (2H, d, J = 8.4 Hz), 7.46 (2H, s), 7.29-7.14 (4H, m), 6.59 (1H, s), 5.90 (1H, t, J = 4.7 Hz), 2.39 (2H, m), 16 2.60 (3H, s), 1.39 (3H, s), 1.29 (3H, s).
17 4-[(5,6-Dihydro-5,5-dimeth 1-~8-(3-h~xyphen~)-2-18 naphthalenvl eth~n~jbenzoic acid (Compou 19 To a solution of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(3-((2,2-dimethylethyl)- dimethylsiloxy)phenyl)-2-naphthalenyl)ethynyljbenzoate 21 (Compound H) 40.0 mg (0.076 mmol) in 3 ml of EtOH and 2 ml of 22 THF was added 40.0 mg (1.00 mmol, 1.00 ml) of NaOH (1.0 M aqueous 23 solution). The solution was heated to SO°C for 2 hours, cooled to room 24 temperature, and acidified with 10% HCI. Extraction with EtOAc, followed by drying over Na2S04, and removal of the solvents under 26 reduced pressure afforded the title compound as a colorless solid. 1H
27 NMR (d6-acetone): s 7.90 (2H, d, J = 8.3 Hz), 7.49 (2H, d, J = 8.4 28 Hz), 7.35 (2H, s), 7.15-7.07 (2H, m), 6.77-6.69 (3H, m), 5.92 (1H, t, J =
29 4.7 Hz), 2.25 (2H, d, J = 4.7 Hz), 1.23 (6H, s).
4-(~5,6-Dihydro-S,5-dimethyl-8-(4-hydroxyphen,~~l)-2-...__....-._~.. ... ._ .._.-w....~._.~ ,..~..... ......-.."~..~..,.W.-.~ . .
.T..._.~..~..m.~..m .... _~.... u.. w-~...~

1 naphthalen~l~ethYn,~~l]benzoic acid lCompound 29) 2 To a solution of ethyl 4-((5,6-dihydro-5,5- dimethyl-8-(4-((2,2-3 dimethylethyl)- dimethylsiloxy)phenyl)-2-naphthalenyl)ethynyl]benzoate 4 (Compound I) 75.0 mg (0.143 mmol) in 3 ml of EtOH and 2 ml of THF
was added 60.0 mg {1.50 mmol, 1.50 ml) of NaOH (1.0 M aqueous 6 solution). The solution was heated to 50°C for 2 hours, cooled to room 7 temperature, and acidified with 10% HCI. Extraction with EtOAc, 8 followed by drying over Na2S04, and removal of the solvents under 9 reduced pressure afforded the title compound as a colorless solid. 1H
1 o NMR (d6-acetone): 8 8.01 (2H, d, J = 8.3 Hz), 7.59 (2H, d, J = 8.4 11 Hz), 7.45 (2H, s), 7.20-7.17 (3H, m), 6.92-6.89 (2H, m), 5.97 (1H, t, J =
12 4.7 Hz), 2.35 (2H, d, J = 4.7 Hz), 1.34 (6H, s).
13 4-({5,6-Dihydro-5,5-dimeth~-8~5-methylthiazol-2 ~yl)-2-14 naphthalen, l~leth~~~benzoic acid lCompound 30) To a solution of ethyl 4-(5,6-dihydro-5,5-dimethyl-8-(5-16 methylthiazol-2-yl)-2-naphthalenyl]ethynylbenzoate (Compound 15) (100 17 mg, 0.23 mmol) and 4 ml of EtOH at room temperature was added 18 aqueous NaOH (1 ml, 1 M, 1 mmol). The resulting solution was 19 warmed to 50°C for 1 hour and concentrated in vacuo. The residue was suspended in a solution of CH2C12 and ether (5:1) and acidified to pH 5 21 with 1M aqueous HCI. The layers were separated and the organic layer 22 was washed with brine, dried (Na2S04), filtered and the solvents 23 removed under reduced pressure to give the title compound as a white 24 solid. 1H NMR (d6-DMSO): 8 7.96 (1H, d, J = 1.7 Hz), 7.95 (2H, d, J
-~- ~.0 Hz), 7.65 (2H, d, J = 8.0 Hz), i.64 (1H, s), 7.53 (1H, dd, J = 1.7, 26 8.~ Hz), 7.46 {1H. , J = 8.0 H~,~, 6.59 ( 1H, t, J = 4.5 Hz), 2.50 (3H, 27 s), 2.39 (2H, d, J = 4.5 Hz), 1.27 (6H, s).
28 4-((5,6-dihydro-5,5-dimethy~2-thiazolyl)T2-29 naphthalen, l~leth~~]benzoic acid Compound 30a) A solution of 33.9 mg (0.08 mmol) of ethyl 4-((5,6-dihydro-5,5-1 dimethyl-8-(2-thiazolyl)-2- naphthalenyl)ethynyl]benzoate (Compound 2 15a) and 8.5 mg (0.20 mmol) of LiOH-H20 in 3 ml of THF/water (3:1, 3 v/v), was stirred overnight at room temperature. The reaction was quenched by the addition of sat. aqueous NH4C1 and extracted with EtOAc. The combined organic layers were washed with water and 6 brine, dried over Na2SOQ and concentrated in vacuo to give the title 7 compound as a colorless solid. PMR (d6 DMSO): 8 1.29 (6H, s), 2.42 8 (2H, d, J = 4.6 Hz), 6.68 (1H, t, J = 4.6 Hz), 7.51 (2H, m), 7.62 (2H, d, 9 J = 8.2 Hz), 7.77 (1H, d, J = 3.3 Hz), 7.93 (2H, d, J = 8.2 Hz), 7.98 l0 (lH,d,J=3.3Hz).
11 4-y(5,6-Dihydro-5,5-dimethyl-8-(4-methylthiazol-2-yll-2-12 naphthalene ethynyl) benzoic acid (Compound 31) 13 To a solution of ethyl 4-X5,6-dihydro-5,5-dimethyl-8-(4-14 methylthiazol-2-yl)-2-naphthalenyl]ethynylbenzoate (Compound 16) (145.0 mg, 0.34 mmol) and 4 ml of EtOH at room temperature was 16 added aqueous NaOH ( 1 ml, 1 M, 1 mmol). The resulting solution was 17 warmed to 50°C for 1 hour and concentrated in vacuo. The residue was 18 suspended in a solution of CH2C12 and ether (5:1) and acidified to pH 5 19 with 1M aqueous HCI. The layers were separated and the organic layer was washed with brine, dried (Na2S04), filtered and the solvents 21 removed under reduced pressure to give the title compound as a white 22 solid. 1H NMR (d6-DMSO): 8 7.94 (2H, d, J = 8.1 Hz), 7.87 (1H, d, J
23 = 1.6 Hz), 7.63 (2H, d, J = 8.3 Hz), 7.50 (1H, dd, J = 1.6, 8.1 Hz), 7.45 24 (lH,d,J=8.lHz),7.27(lH,s),6.58(lH,t,J=4.8Hz),2.43(3H,s), 2.37 (2H, d, J = 4.8 Hz), 1.26 (6H, s).
26 4-j(5,6-Dihydro-5.5-dimethyl-814,5-dimethylthiazol-2-vl)-2-27 naphthalenylleth~~] benzoic acid (Compound 32~
28 To a solution of ethyl 4-(5,6-dihydro-5,5-dimethyl-8-(4,5-29 dimethylthiazol-2-yl)-2-naphthalenyl]ethynylbenzoate (Compound 17) (58.0 mg, 0.13 mmol) and 4 ml of EtOH at room temperature was ..,..:n.~,.."_~...~....n.. ._..T._~ww... ......_ .._....

1 added aqueous NaOH (1 ml, 1 M, i mmol). The resulting solution was 2 warmed to 50°C for 1 hour and concentrated in vacuo. The residue was 3 suspended in a solution of CH2C12 and ether (5:1) and acidified to pH 5 4 with 1M aqueous HCI. The layers were separated and the organic layer was washed with brine, dried (NaZS04), filtered and the solvents 6 removed under reduced pressure to give the title compound as a white 7 solid. 1H NMR (d6-DMSO): 8 7.94 (2H, d, J = 8.4 Hz), 7.86 (1H, d, J
8 - 1.6 Hz), 7.61 (2H, d, J = 8.3 Hz), 7.50 ( 1H, dd, J = 1.6, 8.0 Hz), 7.45 9 (1H, d, J = 8.0 Hz), 6.51 (1H, t, J = 4.9 Hz), 2.37 (3H, s), 2.36 (2H, d, 1 o J = 4.6 Hz), 2.32 (3H, s), 1.26 (6H, s).
11 Ethyl 4-[(5,6-dihydro-5 5-dimethyl-8-{5-methyl-2-thienYl)-2-naphthalenyl) 12 ethynyll benzoate (Compound 33,~
13 Employing the same general procedure as for the preparation of 14 ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-{4- methylphenyl)-2-naphthalenyl)ethynyl]benzoate (Compound 1), 170.0 mg (0.366 mmol) 16 of ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(trifluoromethylsulfonyl)oxy-2-17 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the 18 title compound (colorless solid) using 202.0 mg (1.48 mmol) of zinc 19 chloride, 24 mg (0.022 mmol) of tetrakis(triphenylphosphine)palladium(0) in 2.0 ml of THF, and 5-21 methyl-2-lithiothiophene (prepared by adding 58.6 mg (0.36 ml, 0.915 22 mmol) of n- butyllithium (2.5 M solution in hexanes) to a cold solution 23 (-78°C) of 89.8 mg (0.915 mmol) of 2-methylthiophene in 2.0 ml of 24 THF). 1H NMR (CDCl3): 8 8.00 (2H, d, J = 8.3 Hz), 7.59 (1H, d, J =
1.7 Hz), 7.55 (2H, d, J = 8.2 Hz), 7.4:' !1H, dd, J = 1.7, 8.0 Hz), 7.35 26 (1H, d, J = 8.0 H~. ~, 6.87 (1F1, d, J = ~ ..~ Hz), 6.74 (1H, d, J = 2.8 Hz), 27 6.15 (1H, t, J = 4.8 Hz), 4.38 (2H, q, J = 7.1 Hz), 2.52 (3H, s), 2.32 28 (2H, d, J = 4.8 Hz). 1.40 (3H, t, 7.1 Hz), 1.32 (6H, s).
29 Eth~4-~f 5,6-dihydro-5 5-dimethyl-8-(2-thienylT
naphthalenyl)ethynyllbenzoate (Compound 33a) 1 Employing the same general procedure as for the preparation of 2 ethyl 4-((5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-3 naphthalenyl)ethynyl]benzoate (Compound 1), 250.0 mg (0.52 mmol) of 4 ethyl 4-[{5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-naphthalenyl)ethynyl]benzoate (Compound G) was converted into the 6 title compound (colorless solid) using 186.8 mg (1.37 mmol) of zinc 7 chloride 37.1 mg (0.03 mmoI) of 8 tetrakis(triphenylphosphine)palladium(0) and 2-lithiothiophene 9 (prepared by the addition of 65.9 mg (0.69 ml, 1.03 mmol) of n-l0 butyllithium (1.SM solution in hexane) to a cold solution (-78 oC) of 11 86.5 mg (1.03 mmol) of thiophene in 1.0 ml of THF). PMR (CDC13): 8 12 1.33 {6H, s), 1.36 (3H, t, J = 7.1 Hz), 2.38 (2H, d, J = 4.7 Hz), 4.34 13 (2H,q,J=7.2Hz),6.25(lH,t,J=4.7Hz),7.13(2H,m),7.47(4H, 14 m), 7.62 ( 2H, d, J = 8.5 Hz), 8.00 (2H, d, J = 8.5 Hz).
4-[(5,6-Dihydro-5,5-dimethy~5-methyl-2-thien,~~l)-2-16 naphthalenyllethynvll benzoic acid (Compound 34~
17 To a solution of ethyl 4-[5,6-dihydro-5,5-dimethyl-8-(5-methyl-2-18 thienyl)-2-naphthalenyl]ethynylbenzoate (Compound 33) {35.0 mg, 0.082 19 mmol) in 2 ml of EtOH and 1 ml THF at room temperature was added aqueous NaOH (1 ml, 1 M, 1 mmol). The resulting solution was stirred 21 at room temperature overnight and then acidified with 10% HCI.
22 Extraction with EtOAc, followed by drying over Na2S04, and removal of 23 the solvents under reduced pressure afforded the title compound as a 24 colorless solid. 1H NMR (d6-acetone): 8 8.03 (2H, d, J = 8.6 Hz), 7.63 (2H, d, J = 8.6 Hz), 7.54-7.48 (3H, m), 6.89 (1H, m), 6.18 (1H, t, J =
26 4.7 Hz), 2.49 (3H, s), 2.35 (2H, d, J = 4.7 Hz), 1.32 (6H, s).
27 4-(~5,6-dihydro-5,5-dimethyl-8-(2-thiefll-~phthalenyl)eth~yl]benzoic 28 acid (Compound 34a) 29 Employing the same general procedure as for the preparation of 4-[{5,6-dihydro-5,5-dimethyl-8-(2- thiazolyl)-2-1 naphthalenyl)ethynyl]benzoic acid (Compound 30a), 70.0 mg (0.17 2 mmol) of ethyl 4-[(5,6- dihydro-5,5-dimethyl-8-(2-thienyl)-2-3 naphthalenyl)ethynyl]benzoate (Compound 33a) was converted into the 4 title compound (colorless solid) using 17.8 mg {0.42 mmol) of LiOH in H20. PMR (db DMSO): 8 1.27 (6H, s), 2.33 (2H, d, J = 4.9 Hz), 6.23 6 (1H, t, J = 4.9 Hz), 7.14 (2H, m), 7.38 - 7.56 (4H, m), 7.61 (2H, d, J =
7 8.3 Hz), 7.92 (2H, d, J = 8.3 Hz).
8 5,6-Dihydro-5,5-dimeth~4-methylphenyl)-2-nanhthalenecarbo , lic 9 acid (Compound K) 1 o A solution of 3,4-dihydro-1-(4-methylphenyl)-4,4- dimethyl-7-11 bromonaphthalene (Compound D) {250.0 mg, 0.764 mmol) in 2.0 ml of 12 THF was cooled to -78°C and 1.0 ml of t-butyllithium ( 1.68 mmol, 1.7 13 M solution in pentane) was added slowly. After stirring for 1 hour at 14 -78°C gaseous C02 (generated by evaporation of Dry-Ice, and passed though a drying tube} was bubbled through the reaction for 1 hour.
16 The solution was then allowed to warm to room temperature and the 17 reaction was quenched by the addition of 10% HCI. Extraction with 18 EtOAc was followed by washing the combined organic layers with H20 19 and saturated aqueous NaCI, and drying over MgS04. Removal of the solvents under reduced pressure and washing of the solid with hexanes 21 afforded the title compound as a colorless solid. 1H NMR (CDCl3): b 22 7.94 ( 1 H, dd, J = 1.8, 8.1 Hz), 7.76 ( 1 H, d, J = 1. 8 Hz), 7.45 ( 1 H, d, J
23 = 8.1 Hz), 7.24 (4H, m), 6.01 (1H, t, J = 4.7 Hz), 2.40 (3H, s), 2.36 24 (2H, d, J = 4.7 Hz), 1.35 (6H, s).
Ethyl 4-[[(5,6-dihydro-5 5-dimeth~-8-y4-~-~eth~phen~l-2-26 naphthalenyl carbons]amino)-benzoate :~:ompound 27 A solution of 170.0 mg (0.58 mmol) 5,6-dihydro-5,5-dimethyl-8-(4-28 methylphenyl)-2-naphthalenecarboxylic acid (Compound K) 115.0 mg 29 (0.70 mmol) of ethyl 4-aminobenzoate, 145.0 mg {0.76 mmol) of 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, and 92.4 mg 1 (0.76 mmol) of 4-dimethylaminopyridine in 6.0 ml of DMF was stirred 2 overnight at room temperature. Ethyl acetate was added and the 3 resulting solution washed with H20, saturated aqueous NaHC03, and 4 saturated aqueous NaCI, then dried over MgS04. After removal of the solvent under reduced pressure, the product was isolated as a colorless 6 solid by column chromatography (10 to 15% EtOAc/hexanes). 1H
7 NMR (CDCl3): 8 8.02 (2H, d, J = 8.7 Hz), 7.72 (2H, m), 7.65 (2H, d, J
8 = 8.7 Hz), 7.52 (1H, d, J = 1.8 Hz), 7.48 (1H, d, J = 8.0 Hz), 7.25 (4H, 9 m), 6.15 (1H, t, J = 4.9 Hz), 4.36 (2H, q, J = 7.1 Hz), 2.40 (3H, s), 2.38 (2H, d, J = 4.9 Hz), 1.39 (3H, t, J = 7.1 Hz), 1.37 (6H, s).
11 4-[[j5.6-Dihydro-5,5-dimethyl-8-(4-methylphen~l-2-t2 naphthalenyllcarbonyl]amino]-benzoic acid (Compound 36) 13 To a solution of 26.5 mg (0.06 mmol) ethyl 4[[(5,6-dihydro-5,5-14 dimethyl-8-(4-methylphenyl)-2- naphthalenyi)carbonyl]amino]-benzoate (Compound 35) in 3.0 ml EtOH and 4.0 ml of THF was added 240.1 16 mg NaOH (6.00 mmol, 3.0 ml of a 2M aqueous solution). After stirring 17 at room temperature for 72 hours, the reaction was quenched by the 18 addition of 10% HCI. Extraction with EtOAc, and drying of the 19 organic layers over MgS04, provided a solid after removal of the solvent under reduced pressure. Crystallization from CH3CN afforded the title 21 compound as a colorless solid. 1H NMR (d6-DMSO): 8 10.4 {1H, s), 22 7.91-7.81 (SH, m), 7.54 (1H, d, J = 8.1 Hz), 7.45 (1H, d, J = 1.7 Hz), 23 7.23 (4H, s), 6.04 (1H, t, J = 4.7 Hz), 2.35 (SH, s), 1.33 (6H, s).
24 Ethyl 4-[[(5,6-dihYdro-S,5-dimeth~-8-(4-meth ~~l-phen~~2-naphthalenyl)carbons]oxyl-benzoate ~~Compound 371 26 A solution of 25.0 mg (0.086 mmol) 5,6-dihydro-5,5-dimethyl-8-(4-27 methylphenyl)-2-naphthalenecarboxylic acid {Compound K) 17.5 mg 28 (0.103 mmol) of ethyl 4-hydroxybenzoate, 21.4 mg (0.112 mmol) of 1-(3-29 dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, and 12.6 mg (0.103 mmol) of 4-dimethylaminopyridine in 2.0 ml of DMF was stirred 1 overnight at room temperature. Ethyl acetate was added and the 2 resulting solution washed with H20, saturated aqueous NaHC03, and 3 saturated aqueous NaCI, before being dried over MgS04. After removal of the solvent under reduced pressure, the product was isolated by column chromatography as a pale-yellow solid (10% EtOAc/hexanes).
6 1H NMR (CDC13): 8 8.08 (2H, d, J = 8.1 Hz), 8.05 (1H, dd, J = 1.8, 7 8.1 Hz), 7.89 (1H, d, J = 1.8 Hz), 7.50 (2H, d, J = 8.1 Hz), 7.22 (SH, 8 m), 6.05 (1H, t, J = 4.7 Hz), 4.37 (2H, q, J = 7.1 Hz), 2.39 (2H, d, J =
9 4.7 Hz), 2.38 (3H, s), 1.39 (3H, t, J = 7.1 Hz), 1.37 (6H, s).
2-Trimethylsilylethyl 4-(~(5 6-dihydro-5 5-dimeth~l-8-(4-meth~phen~)-2-11 naphthalenyllcarbonylloxyl-benzoate Compound 38) 12 A solution of 93.5 mg (0.320 mmol) 5,6-dihydro-S,5-dimethyl-8-(4-13 methylphenyl)-2-naphthalenecarboxylic acid (Compound K) 76.0 mg 14 (0.319 mmol) of 2-trimethylsilylethyl-4-hydroxybenzoate, 80.0 mg (0.417 mmol) of 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, 16 and 51.0 mg (0.417 mmol) of 4-dimethylaminopyridine in 4.0 ml of 17 DMF was stirred overnight at room temperature. Ethyl acetate was 18 added and the resulting solution washed with H20, saturated aqueous 19 NaHC03, and saturated aqueous NaCI, before being dried over MgS04.
After removal of the solvent under reduced pressure, the product was 21 isolated as a colorless solid by column chromatography (5%
22 EtOAc/hexanes). 1H NMR (CDC13): 8 8.08 (2H, d, J = 8.8 Hz), 8.05 23 (1H, dd, J = 1.8, 8.1 Hz), 7.50 (1H, d, J = 8.1 Hz), 7.26- 7.18 (6H, m), 24 6.05 (1H, t, J = 4,7 Hz), 4.42 (2H, t, J = 8.4 Hz), 2.40 (2H, d, J = 4.7 Hz), 2.39 (3H, s), I.38 (6H, s), 0.09 (9H, s).
4-(~~5,6-Dihydro-5 5-dimethyl-8-i(4-meth l~nyl)-2-27 naphthalenyl carbonyl]ox~]-benzoic acid (Compound 39~
28 A solution of 110.0 mg (0.213 mmol) 2- trimethylsilylethyl 4(((5,6-29 dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-naphthalenyl)carbonyl]oxy]-benzoate (Compound 38) and 167.3 mg of tetrabutylammonium flouride 1 (0.640 mmol, 0.64 ml of a 1M solution in THF) in 2.0 ml THF was 2 stirred at room temperature for 22 hours. Ethyl acetate was added and 3 the resulting solution washed with H20 and saturated aqueous NaCI
4 then dried over MgS04. Removal of the solvents under reduced pressure and washing of the residual solid with EtOAc and CH3CN
6 afforded the title compound as a colorless solid. 1H NMR (d6-7 acetone): b 8.10 (2H, d, J = 8.8 Hz), 8.06 (1H, dd, J = 2.0, 8.1 Hz), 8 7.82(lH,d,J=1.9Hz),7.64(lH,d,J=8.lHz),7.35(2H,d,J=8.6 9 Hz), 7.25 (4H, m), 6.08 (1H, t, J = 4.7 Hz), 2.42 (2H, d, J = 4.7 Hz), 2.35 (3H, s), 1.39 (6H, s).
11 Ethvl 2-fluoro-4-x((5,6-dihydro-5 5-dimethyl-8-(4-meth, l~phen,~)-2-12 naphthalenYl carbon~~amino~-benzoate Compound 40) 13 A solution of 115.0 mg (0.41 mmol) 5,6-dihydro-S,5-dimethyl-8-(4-14 methylphenyl)-2-naphthalenecarboxylic acid (Compound K) 89.0 mg (0.49 mmol) of ethyl 2-fluoro-4-aminobenzoate, 102.0 mg (0.53 mmol) 16 of 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, and 17 65.0 mg (0.53 mmol) of 4-dimethylaminopyridine in 5.0 ml of DMF was 18 stirred at 50°C for 1 hour and then overnight at room temperature.
19 Ethyl acetate was added and the resulting solution washed with H20, saturated aqueous NaHC03, and saturated aqueous NaCI, before being 21 dried over MgS04. After removal of the solvent under reduced 22 pressure, the product was isolated as a colorless solid by column 23 chromatography (20% EtOAc/hexanes). 1H NMR (CDC13): 8 7.96 24 (1H, s), M.89 (1H, t, J = 8.4 Hz), 7.70 (2H, m), 7.52 (1H, d, J = 1.9 Hz), 7.45 (1H, d, J = 8.1 Hz), 7.23 (5H, m), 6.04 (1H, t, J = 4.8 Hz), 26 4.36 (2H, q, J = 7.1 Hz), 2.38 (3H, s), 2.35 (2H, d, J = 4.8 Hz), 1.39 27 (3H, t, J = 7.1 Hz), 1.36 (6H, s).
28 2-Fluoro-4-([,(5,6-dihydro-5 5-dimeth~-8-(4-meth~yhen~)-2-29 naphthalene)carbon~~amino)-benzoic acid Compound 41,) To a solution of 41.6 mg (0.091 mmol) ethyl 2- fluoro-4-(((5,6-...-. ..~...~... . ..-. .~... a . ~...~~.... ...w..u.. _........w ....am_ .~

1 dihydro-5,5-dimethyl-8- (4-methylphenyl)-2-2 naphthalenyl)carbonyl]amino]- benzoate (Compound 40) in 2.0 ml 3 EtOH and 2.0 ml of THF was added 40.0 mg NaOH ( 1.00 mmol, 1.0 ml 4 of a 1 M aqueous solution). After stirring at room temperature for overnight, the reaction was quenched by the addition of 10% HCI.
6 Extraction with EtOAc, and drying of the organic layers over MgS04, 7 provided a solid after removal of the solvent under reduced pressure.
8 Crystallization from CH3CN afforded the title compound as a pale-9 yellow solid. 1H NMR (d6-acetone): 8 9.84 (1H, s), 7.94-7.83 (3H, m), l0 7.64 (1H, dd, J = 2.0 Hz), 7.53 (2H, d, J = 8.1 Hz), 7.23 (4H, s), 6.04 ~ 1 (1H, t, J = 4.7 Hz), 2.38 (2H, d, J = 4.7 Hz), 2.36 (3H, s), 1.35 (6H, s).
12 Ethyl 4[[(5,6-dihydro-5 5-dimethyl-8-i8-i(4-meth'~lphen,~~I)- 2-13 naphthalenyl)thiocarbonyl]amino]-benzoate (Compound 42) 14 A solution of 110.0 mg (0.25 mmol) ethyl 4-[[(5,6-dihydro-5,5-dimethyl-8-(4-methylphenyl)- 2-naphthalenyl)carbonyl]amino]-benzoate 16 (Compound 35) and 121.0 mg (0.30 mmol) of [2,4-bis(4-17 methoxyphenyl)- 1,3-dithia-2,4-diphosphetane-2,4-disulfide] (Lawesson's 18 Reagent) in 12.0 ml of benzene was refluxed overnight. Upon cooling 19 to room temperature, the mixture was filtered and the filtrate concentrated under reduced pressure. The title compound was isolated 21 by column chromatography (10 to 25% EtOAc / hexanes) as a yellow 22 solid. 1H NMR (CDCI3): 8 8.92 (1H, s), 8.06 (2H, t, 3 = 8.5 Hz), 23 7.88-7.70 (3H, m), 7.42 (2H, d, J = 8.1 Hz), 7.18 (4H, m), 6.03 (1H, t, J
24 = 4.7 Hz), 4.37 (2H, q, J = 7.1 Hz), 2.38 (3H, s), 2,36 (2H, d, J = 4.7 Hz), 1.56 (3H, t, J = 7.1 Hz), 1.35 (6H, s).
26 4-[[(5,6-Dihydro-5,5-dimethyl-8-(4-meth, l~pheny~-2-27 naphthalenyl)thiocarbonvllaminol-benzoic acid (ComDOUnd 43 28 To a solution of 84.0 mg (0.184 mmol) ethyl 4[[(5,6-dihydro-5,5-29 dimethyl-8-(4-methylphenyl)-2- naphthalenyl)thiocarbonyl]amino]-benzoate (Compound 42) in 2.0 ml EtOH and 2.0 ml of THF was added 1 60.0 mg NaOH ( 1.50 mmol, 1.5 ml of a 1 M aqueous solution). After 2 stirring at room temperature overnight, the reaction was quenched by 3 the addition of 10% HCI. Extraction with EtOAc, and drying of the 4 organic layers over MgS04, provided a solid after removal of the solvent under reduced pressure. Crystallization from CH3CN afforded the title 6 compound as a yellow solid. 1H NMR (d6-acetone): 8 10.96 (1H, s), 7 8.05 (4H, m), 7.72 (1H, dd, J = 2.0, 8.0 Hz), 7.54 (1H, s), 7.46 (1H, d, J
8 = 8.1 Hz), 7.20 (4H, m), 6.04 (1H, t, J = 4.7 Hz), 2.38 (2H, d, J = 4.7 9 Hz), 2.33 (3H, s), 1.35 (6H, s).
2-acetyl-6-bromonaphthalene (Compound L) 11 To a cold (10°C) mixture of 44.0 g (0.212 mol) of 2-12 bromonaphthalene and 34.0 g (0.255 mol) of aluminum chloride in 400 13 ml of nitrobenzene was added 21.0 g (267 mmol) of acetyl chloride.
14 The mechanically stirred reaction mixture was warmed to room temperature, and heated to 40°C for 18 hours. After cooling to 0°C in 16 an ice bath, the reaction was quenched by the addition of 12M HCl (70 17 ml). The layers were separated and the organic phase was washed with 18 water and dilute aqueous Na2C03. Kugelrohr distillation, followed by 19 recrystallization from 10% EtOAc-hexane yielded 23 g of the title compound as a tan solid. 1H NMR (CDC13): 8 8.44 (1H, br s), 21 8.04-8.10 (2H, m), 7.85 (1H, d, J = 8.5 Hz), 7.82 (1H, d, J = 8.8 Hz), 22 7.64(lH,d,J=8.8Hz),2.73(3H,s).
23 6-bromo-2-n~hthalenecarboxylic acid (Compound M) 24 To a solution of sodium hypochlorite (62 ml, 5.25% in water (w/w), 3.6 g, 48.18 mmol) and sodium hydroxide (6.4 g, 160.6 mmol) in 26 50 ml of water was added a solution of 2-acetyl-6-bromonaphthalene 27 (Compound L) 4 g, (16.06 mmol) in 50 ml of 1,4-dioxane. The yellow 28 solution was heated to 70°C in an oil bath for 2 hours, cooled to 29 ambient temperature, and extracted with ethyl ether (2 x 50 ml). The aqueous layers were diluted with NaHS03 solution (until KI indicator _.~W.w.,~~...~~... ~.~_........ .. _ w ....._...~~~~...ru~ a . . . . ....u. ..-....... ..

1 solution remained colorless) and then acidified (pH <2) with 1N
2 sulfuric acid to give a white precipitate. The mixture was extracted with 3 ethyl ether, and the combined organic phase washed with saturated 4 aqueous NaCI, dried (MgS04) and concentrated to give 3.54 g (88%) of the title compound as a solid. 1H NMR (DMSO-d6): 8 8.63 (iH, br s), 6 8.32 (1H, d, J = 2.0 Hz), 8.10 (1H, d, J = 8.8 Hz), 8.00-8.05 (2H, m), 7 7.74(lH,dd,J=2.0,8.8Hz).
8 Ethyl 6-bromo-2-naphthalenecarbo,1~(Com~ound N) 9 To a solution of 6-bromo-2-naphthalenecarboxylic acid (Compound M) 3.1 g, (12.43 mmol) in ethanol (30 ml, 23.55 g, 511.0 11 mmol) was added 18M sulfuric acid (2 ml). The solution was refluxed 12 for 30 minutes, cooled to room temperature, and the reaction mixture 13 partitioned between pentane (100 ml) and water (100 ml). The aqueous 14 phase was extracted with pentane (100 ml) and the combined organic layers washed with saturated aqueous NaCI (100 ml), dried (MgS04), 16 and concentrated to yield an off-white solid. Purification by flash 17 chromatography (silica, 10% EtOAc-hexane) afforded the title 18 compound as a white solid. 1H NMR (CDCl3): 8 8.58 (1H, br s), 8.10 19 (lH,dd,J=1.7,9Hz),8.06{lH,d,J=2Hz),7.83(lH,d,J=9Hz), 7.80 (1H, d, J = 9 Hz), 7.62 (1H, dd, 3 = 2, 9 Hz).
21 Eth 1~(E)-4-~2-(5,6,7,8-tetrahydro-5 5-dimethyl-8-oxo-2-22 naphthalene ethenyll-benzoate (Compound O) 23 To a solution of 520.0 mg (2.00 mmol) of 3,4- dihydro-4,4-24 dimethyl-7-bromo-1(2H)-naphthalenone (Compound B) and 510.0 m~
(2.90 mmol) of ethyl 4- vinyibenzoate in 4.0 ml of triethylamine 26 (degassed by sparging with argon for 25 minutes), was added 124.0 mg 27 (0.40 mmol) of tris(2-methylphenyl) phosphine, followed by 44.0 mg 28 (0.20 mmol) of palladium(II)acetate. The resulting solution was heated 29 to 95°C for 2.5 hours, cooled to room temperature, and concentrated under reduced pressure. Purification by column chromatography (10%

1 EtOAc / hexanes} afforded the title compound as a colorless solid. 1H
2 NMR (CDC13): 8 8.19 (1H, d, J = 2.0 Hz), 8.03 (2H, d, J = 8.4 Hz), 3 7.69 (1H, dd, J = 2.0, 8.2 Hz), 7.57 (2H, d, J = 8.4 Hz), 7.45 (1H, d, J
4 = 8.2 Hz), 7.20 (2H, s), 4.39 (2H, q, J = 7.1 Hz), 2.76 (2H, t, J = 6.5 Hz), 2.04 (2H, t, J = 6.5 Hz), 1.41(3H, t, J = 7.1 Hz, and 6H, s).
6 Ethyl (E)-4-[~5,6-dihydro-5 5-dimethyl-8-,{trifluoromethylsulfon,~~l)ox ~-~2-7 naphthalen~lethenyl]- benzoate (Compound P) 8 To a cold (-78°C) solution of 440.0 mg (2.40 mmol) of sodium 9 bis(trimethylsilyl)amide in 10.0 ml of THF was added 700.0 mg (2.00 l0 mmol) of ethyl (E)-4-[2- (5,6,7,8-tetrahydro-5,5-dimethyl-8-oxo-2 11 naphthalenyl)ethenyl]-benzoate (Compound O) as a solution in 25.0 ml 12 of THF. After stirring at -78°C for 1.5 hours, 960.0 mg (2.40 mmol) of 13 2[N,N- bis(trifluoromethylsulfonyl)amino]-S-chloropyridine was added in 14 one portion. After 30 minutes the solution was warmed to 0°C and stirred for 3 hours. The reaction was quenched by the addition of 16 saturated aqueous NH4Cl, and extracted with EtOAc. The combined 17 extracts were washed with 5% aqueous NaOH, dried (Na~S04), and the 18 solvents removed under reduced pressure. The title compound was 19 isolated as a colorless solid by column chromatography (7% EtOAc /
hexanes). 1H NMR (CDCl3): 8 8.04 (1H, d, J = 8.4 Hz), 7.57 (2H, d, J
21 =8.4Hz),7.52(lH,s),7.49(lH,d,J=8.OHz),7.33{lH,d,J=8.0 22 Hz),7.20(lH,d,J=16.4Hz),7.10(lH,d,J=16.4Hz),6.00(lH,t,J
23 = 4.9 Hz), 4.39 (2H, q, J = 7.1 Hz), 2.43 (2H, d, J = 4.9 Hz), 1.41 (3H, 24 t, J = 7.1 Hz), 1.32 (6H, s).
Ethyl (E)-4-(~5,6-dihydro-5 5-dimeth-y~4- methylphen,~}-2-26 naphthalenyl}ethenyll-be~ nzoate~Compound 441 27 A solution of 4-lithiotoluene was prepared at -78°C by the 28 addition of 130.7 mg of t-butyllithium (2.04 mmol; 1.20 ml of a 1.7M
29 solution in pentane) to a solution of 374.5 mg ( 2.20 mmol) of 4-bromotoluene in 2.5 ml of THF. After 30 minutes a solution of 313.4 ,.. .. .. ... .. T . . ....... ._ .._"....,.~...,..~..".-..-.-.. ........

1 mg (2.30 mmol) of ZnCl2 in 2.0 ml of THF was added. The resulting 2 solution was warmed to room temperature, stirred for 1.25 hour and 3 then added via canula to a solution of 285.0 mg (0.590 mmol) of ethyl 4 (E)-4-[2- (5,6-dihydro-5,5-dimethyl-8- (trifluoromethylsulfonyl)oxy-2-naphthalenyl)ethenyl]- benzoate (Compound P) and 29.0 mg (0.025 6 mmol) of tetrakis(triphenylphosphine)palladium(0) in 2.0 ml of THF.
7 The resulting solution was stirred at room temperature for 1 hour and 8 then at 55°C for 2 hours. Upon cooling to room temperature the 9 reaction was quenched by the addition of saturated aqueous NH4CI.
l0 The mixture was extracted with EtOAc, and the combined extracts were 11 washed with 5% aqueous NaOH, saturated aqueous NaCI, and dried 12 over Na2S04 before being concentrated under reduced pressure. The 13 title compound was isolated by column chromatography (10% EtOAC
14 hexanes) as a colorless solid. 1H NMR (CDC13): 8 7.96 (2H, d, J = 8.1 Hz), 7.47 (2H, d, J = 8.1 Hz), 7.43-7.16 (7H, m), 7.07 (1H, d, J = 16.3 16 Hz), 6.93 (1H, d, J = 16.3 Hz), 5.97 (1H, t, J = 4.7 Hz), 4.39 (2H, q, J
17 = 7.0 Hz), 2.41 (3H, s), 2.33 (1H, d, J = 4.7 Hz), I.38 (3H, t, J = 7.0 18 Hz), 1.33 (6H, s).
19 (El-4-[2-(5,6-DihYdro-5 5-dimethyl-8-(4-methylphenyl)-2-naphthalenyl)ethen~]-benzoic acid Compound 45 21 To a solution of 65.0 mg (0.190 mmol) of ethyl (E)-4-[2-(5,6 22 dihydro-5,S-dimethyl-8-{4-methylphenyl)- 2-naphthalenyl)ethenyl]
23 benzoate (Compound 44) in 4.0 ml of THF was added 30.0 mg of LiOH
24 (0.909 mmol, 1.0 ml of a 1.1M solution) and 1.0 ml of MeOH. The solution was heated to 55°C for 3 hours, cooled to room temperature 26 and concentrated under reduced pressure. The residue was dissolved ~:.
27 H,O and extracted with hexanes. The aqueous layer was acidified to 28 pH 1 with 10% HCl, and extracted with Et20. The combined organic 29 layers were washed with saturated aqueous NaCI, diluted with EtOAc to give a clear solution, and dried over Na2S04. The solvents were 1 removed under reduced pressure to give the title compound as a 2 colorless solid. 1H NMR (d6-DMSO): 8 7.86 (2H, d, J = 8.4 Hz), 7.66 3 (2H,d,J=8.4Hz),7.58(lH,dd,J=1.7,8.1Hz),7.41(lH,d,J=8.1 4 Hz), 7.28 ( 1H, d, J = 16.5 Hz), 7.23 (4H, s), 7.08 ( 1 H, d, J = 1.7 Hz), 7.07 (1H, d, J = 16.5 Hz), 5.97 (1H, t, J = 4.6 Hz), 2.35 {3H, s), 2.31 6 (1H, d, J = 4.6 Hz), 1.29 (6H, s).
7 Ethyl4-[2-{1,1-dimethy~4-methylphenYl)-5-indenXl)ethxn~~benzoate 8 !Compound 47) 9 A solution of 32.0 mg (0.187 mmol) of 4- bromotoluene in 1.0 ml THF was cooled to -78°C and 24.0 mg of t-butyllithium (0.375 mmol, 11 0.22 ml of a 1.7 M solution in pentane) was slowly added. The yellow 12 solution was stirred for 30 minutes at which time 29.8 mg (0.219 mmol) 13 of ZnCl2 was added as a solution in 1.0 ml THF. The resulting solution 14 was warmed to room temperature and after 30 minutes added to a second flask containing 29.0 mg (0.062 mmol) of ethyl 4-[2- (1,1-16 dimethyl-3-(triouoromethylsulfonyl)oxy-5-indenyl) ethynyl)benzoate 17 (Compound FF) and 2.9 mg (0.003 mmol) of 18 tetrakis(triphenylphosphine)palladium (0) in 1.0 ml THF. The resulting 19 solution was warmed to SO°C for 1 hour and then stirred at room temperature for 4 hours. The reaction was quenched by the addition of 21 saturated aqueous NH4Cl, and then extracted with Et20. The combined 22 organic layers were washed with water, saturated aqueous NaCI, and 23 dried over MgS04 before being concentrated under reduced pressure.
24 The title compound was isolated as a colorless oil by column chromatography (10% Et20 / hexanes). 1H NMR (300 MHz, CDC13): 8 26 8.03 (2H, d, J = 8.5 Hz), 7.66 (1H, s), 7.58 (2H, d, J = 8.5 Hz), 7.50 27 (2H, d, J = 8.0 Hz), 7.46 (1H, d, J = 7.9 Hz), 7.38 (1H, d, 3 = 7.7 Hz), 28 7.28 (2H, d, J = 9 Hz), 6.43 (1H, s), 4.40 (2H, q, J = 7.2 Hz), 2.43 (3H, 29 s), 1.41 (3H, t; + 6H, s).
4-[2-(1,1-dimeth,Yl-3-(4-methylphen~, -Lindenyl)ethyn,~~benzoic acid r,...... ~ ~_. .... _ ,....

1 f Compound 48) 2 To a solution of 10.0 mg (0.025 mmol) of ethyl 4- [2-( 1,1-3 dimethyl-3-(4-methylphenyl)-5- indenyl)ethynyl]benzoate (Compound 4 47) in 0.5 ml THF/H20 (3:1 v/v) was added 5.2 mg (0.12 mmol) LiOH
H20. After stirring at room temperature for 48 hours the solution was 6 extracted with hexanes and the aqueous layer was acidified with 7 saturated aqueous NH4C1. Solid NaCI was added and the resulting 8 mixture extracted with EtOAc. The combined organic layers were dried 9 (Na~S04) and concentrated under reduced pressure to give the title compound as a colorless solid. 1H NMR (300 MHz, db-DMSO): 8 7.95 11 {2H, d, J = 8.3 Hz), 7.65 (2H, d, J = 8.3 Hz), 7.57 (2H, m), 7.49 (3H, I2 m), 7.30 (2H, d, J = 7.9 Hz), 6.61 (1H, s), 2.36 (3H, s), 1.36 (6H, s).
I3 ~4-bromothiophenoxy~yropionic acid 14 To a solution of 1.44 g (35.7 mmol) of NaOH in 20.0 ml degassed H20 (sparged with argon) was added 6.79 g (35.7 mmol) of 4-16 bromothiophenol. The resulting mixture was stirred at room 17 temperature for 30 minutes. A second flask was charged with 2.26 g 18 (1b.3 mmol) of KZC03 and 15 ml of degassed H20. To this solution was 19 added (in portions) 5.00 g (32.7 mmol) of 3- bromopropionic acid. The resulting potassium carboxylate solution was added to the sodium 21 thiolate solution, and the resulting mixture stirred at room temperature 22 for 48 hours. The mixture was filtered and the filtrate extracted with 23 benzene, and the combined organic layers were dicarded. The aqueous 24 layer was acidified with 10% HCl and extracted with EtOAc. The combined organic layers were washed with saturated aqueous NaCI, 26 dried over MgS04, and concentrated under reduced pressure. The 27 resulting solid was recrystallized from Et20 - hexanes to give the title 28 compound as off-white crystals. 1H NMR (CDCl3): 8 7.43 (2H, d, J =
29 8.4 Hz), 7.25 (2H, d, J = 8.4 Hz), 3.15 (2H, t, J = 7.3 Hz), 2.68 (2H, t, J = 7.3 Hz).

1 2,3-dihydro-6-bromo~4H)-1-benzothio~yran-4-one, 2 A solution of 3.63 g ( 13.9 mmol) of 3-(4-3 bromothiophenoxy)propionic acid in 60 ml methanesulfonic acid was 4 heated to 75°C for 1.5 hours. After cooling to room temperature the solution was diluted with H20 and extracted with EtOAc. The 6 combined organic layers were washed with 2N aqueous NaOH, H20, 7 and saturated aqueous NaCI and then dried over MgS04. Removal of 8 the solvent under reduced pressure afforded a yellow solid from which 9 the product was isolated by column chromatography (3% EtOAc-l0 hexanes) as a pale-yellow solid. 1H NMR (CDCl3): 8 8.22 (1H, d, J =
11 2.1 Hz), 7.48 1H, dd, J = 2.1,8.3 Hz), 7.17 (1H, d, J = 8.5 Hz), 3.24 12 (2H, t, J = 6.4 Hz), 2.98 (2H, t, J = 6.7 Hz).
13 2,3-dihydro-6-(2-trimethylsilvleth~nyl~(4H)-1-benzothio~yran-4-one 14 A solution of 1.00 g {4.1 i mmol) 2,3-dihydro-6- bromo-(4H)-1-benzothiopyran-4-one and 78.3 mg (0.41 mmol) CuI in 15.0 ml THF
16 and 6.0 ml Et2NH was sparged with argon for 5 minutes. To this I7 solution was added 2.0 ml (1.39 g, 14.2 mmol) of 18 (trimethylsilyl)acetylene followed by 288.5 mg (0.41 mmol) of 19 bis(triphenylphosphine)palladium(II) chloride. The resulting dark solution was stirred at room temperature for 3 days and then filtered 21 through a pad of Celite, which was washed with EtOAc. The filtrate 22 was washed with H20 and saturated aqueous NaCI before being dried 23 over MgS04. The title compound was isolated as an orange oil by 24 column chromatography (4% EtOAc - hexanes). 1H NMR (CDC13): 8 8.13(lH,d,J=1.9Hz),7.36(lH,dd,J=2.1,8.2Hz),7.14(lH,d,J
26 = 8.2 Hz), 3.19 (2H, d, J = 6.3 Hz), 2.91 (2H, d, J = 6.3 Hz), 0.21 (9H, 27 s).
28 2,3-dihydro-6-eth,~yl-(4H)-1-benzothio~~an-4-one 29 A solution containing 600.0 mg (2.25 mmol) of 2,3- dihydro-6-(2-trimethylsilylethynyl)-(4H)-1- benzothiopyran-4-one and 100.0 mg (0.72 ~,-,_..,.~.. ~,..."~.,-...-... T .. . ,.,...,.,_ .., ~._,",~.~.w.~ _ _ .

1 mmol) K2C03 in 15 ml MeOH was stirred at room temperature for 20 2 hours. The solution was diluted with H20 and extracted with Et20. The 3 combined organic layers were washed with H20 and saturated aqueous 4 NaCI before being dried over MgS04. Removal of the solvents under reduced preesure afforded the title compound as an orange solid. 1H
6 NMR (CDCl3): 8 8.17 (1H, d, J = 1.8 Hz), 7.40 (1H, dd, J = 1.8, 8.2 7 Hz), 7.19 (1H, d, J = 8.2 Hz), 3.22 (2H, t, J = 6.3 Hz), 3.08 (1H, s) 8 2.94 (2H, t, J = 6.3 Hz).
9 Ether[2-(6-(2,3-dih dy ro-(4H1-1-benzothiop r on~l~)eth~Xllbenzoate 11 A solution of 405.0 mg {2.15 mmol) 2,3-dihydro-6- ethynyl-12 (4H)-1-benzothiopyran-4-one and 594.0 mg {2.15 mmol) of ethyl 4-13 iodobenzoate in 15 ml Et3N and 3 ml THF was sparged with argon for 14 15 minutes. To this solution was added 503.0 mg (0.72 mmol) of bis{triphenylphosphine)palladium(II) chloride and 137.0 mg (0.72 mmol) 16 CuI. This solution was stirred for 20 hours at room temperature and 17 then filtered through a pad of Celite, which was washed with EtOAc.
18 Removal of the solvents under reduced pressure afforded a brown solid.
19 Column chromatography (3% EtOAc-hexanes) afforded the title compound as an orange solid. 1H NMR (d6 acetone): 8 8.15 (1H, d, J
21 = 2.0 Hz), 8.02 (2H, d, J = 8.5 Hz), 7.69 (2H, d, J = 8.5 Hz), 7.61 (1H, 22 dd, J = 2.1, 8.3 Hz), 7.40 ( 1H, d, J = 8.2 Hz), 4.35 (2H, q, J = 7.1 Hz), 23 3.40 (2H, t, J = 6.3 Hz), 2.96 (2H, t, J = 6.3 Hz), 1.37 (3H, t, J = 7.1 24 Hz).
Ethvl4-~2-~6-(4-(trifluorornethylsulfonyllox~(2H)-1-benzothiop,~ra 26 nyl~leth,~,~~1)benzoate 27 To a solution of 221.9 mg ( 1.21 mmol) of sodium 28 bis(trimethylsilyl)amide in 3.0 ml THF cooled to -78°C was added 370.0 29 mg ( 1.10 mmol) of ethyl 4-[2-(6-(2,3- dihydro-(4H)-1-benzothiopyran-4-onyl))ethynyl)benzoate in 4.0 ml THF. After 30 minutes, a solution of 1 2-[N,N- bis(trifluoromethylsulfonyl)amino]-5-chloropyridine in 4.0 ml 2 THF was slowly added. The reaction was slowly warmed to room 3 temperature and after 5 hours quenched by the addition of saturated 4 aqueous NH4C1. The mixture was extracted with EtOAc, and the combined organic layers were washed with 5% aqueous NaOH, H20, 6 and saturated aqueous NaCI before being dried over MgS04. Removal 7 of the solvents under reduced pressure, followed by column 8 chromatography (4% EtOAc-hexanes) afforded the title compound as a 9 pale-yellow solid. 1H NMR (db acetone): 8 8.12 (2H, d, J = 8.5 Hz), 7.66 (2H, d, J = 8.5 Hz), 7.56 (1H, d, J = 1.7 Hz), 7.49 (1H, dd, J =
11 1.7,8,1Hz),7.40(lH,d,J=8.lHz),6.33{lH,t,J=5.7Hz),4.35 12 (2H, q, J = 7.1 Hz), 3.82 (2H, d, J = 5.7 Hz), 1.37 (3H, t, J = 7.1 13 Hz).Eth~[~~4~4-metl~rlphen 1~)-(2H1-1-14 benzothiop r~anyll)eth~n~~benzoate (Compound 49) To a solution of 120.8 mg (0.70 mmol) of 4- bromotoluene in 2.0 16 ml THF at -78°C was added 88.4 mg ( 1.38 mmol, 0.81 ml of a 1.7 M
17 solution in pentane) of t-butyllithium. After 30 minutes a solution of 18 131.6 mg (0.97 mmol) ZnCl2 in 2.0 ml THF was added and the reulting 19 pale-yellow solution warmed to room temperature. Stirring for 40 minutes was followed by addition of this solution to a second flask 21 containing 129.2 mg (0.28 mmol) of ethyl 4-[2-(6-(4-22 (trifluoromethylsulfonyl)oxy-(2H)-1-benzothiopyranyl))ethynyl]benzoate, 23 14.0 mg (0.012 mmol) tetrakis(triphenylphosphine)palladium (0), and 24 2.0 ml THF. The resulting solution was heated to 50°C for 5 hours, cooled to room temperature, and quenched by the addition of saturated 26 aqueous NH4CI. The mixture was extracted with EtOAc, and the 27 combined organic layers were washed with H20 and saturated aqueous 28 NaCI, then dried {MgS04) and concentrated to an orange oil. The title 29 compound was isolated as a colorless solid by column chromatography (3 to 5% EtOAc-hexanes). 1H NMR (db acetone): 8 7.98 (2H, d, J =

1 8.3 Hz), 7.58 (2H, d, J = 8.2 Hz), 7.44-7.38 (2H, m), 7.26-7.15 (SH, m), 2 6.14(lH,t,J=5.8Hz),4.34(2H,q,J=7.lHz),3.53(2H,d,J=5.8 3 Hz), 2.37 (2H, s), 1.35 (3H, t, J = 7.1 Hz).
4 4-f2-(6-(4-(4-methyl hen~~2H)-1-benzothiopyran~) eth~Yl] benzoic acid (Compound 50) 6 To a solution of 29.0 mg (0.07 mmol) ethyl 4-[2-(6-(4-(4-7 methylphenyl)-(2H)-1- benzothiopyranyl))ethynyl ]benzoate (Compound 8 49) in 2.0 ml THF and 2.0 ml EtOH was added NaOH (160.0 mg, 4.00 9 mmol, 2.0 ml of a 2 M aqueous solution). The resulting solution was l0 stirred at 35°C for 2 hours, and then cooled to room temperature and 11 stirred an additional 2 hours. The reaction was quenched by the 12 addition of IO% aqueous HCI and extracted with EtOAc. The 13 combined organic layers were washed with HBO and saturated aqueous 14 NaCI, and dried over Na2S04. Removal of the solvents under reduced pressure afforded a solid which was washed with CH;CN and dried 16 under high vacuum to give the title compound as a pale-yellow solid.
17 1H NMR (d6-DMSO): 8 7.90 (2H, d, J = 8.4 Hz), 7.59 (2H, d, J = 8.4 18 Hz), 7.40 (4H, m), 7.25-7.13 (4H, m), 7.02 (1H, d, J = 1.7 Hz), 6.I1 19 (1H, t, J = 5.7 Hz), 3.54 (2H, d, J = 5.7 Hz), 2.34 (3H, s).
3,4-Dihvdro-4,4-dimethyl-7-acet~-1(2H)-naphthalenone (Compound RL
21 and 3,4-dihydro-4 4-dimeth 1-~~l- 1(2Hl-naphthalenone (Compound 23 To a cold (0°C) mixture of aluminum chloride (26.3 g, 199.0 24 mmols) in dichloromethane (55 ml) was added acetylchloride (15 g, 192 mmols) and 1,2,3,4-tetrahydro-1,1-dimethylnaphthalenP (24.4g, 152 26 mmols) ~n dichloromethane (20 ml) over 20 minutes. T'he reaction 27 mixture was warmed to ambient temparature and stirred for 4 hours. Ice 28 (200 g) was added to the reaction flask and the mixture diluted with 29 ether (400 ml). The layers were separated and the organic phase washed with 10% HCl (50 ml), water (50 ml), 10% aqueous sodium 1 bicarbonate, and saturated aqueous NaCI (50 ml) before being dried 2 over MgS04. Ths solvent was removed by distillation to afford a yellow 3 oil which was dissolved in benzene (SO ml).
4 To a cold (0°C) solution of acetic acid (240 ml) and acetic anhydride (120 ml) was added chromiumtrioxide (50 g, 503 mmols) in 6 small portions over 20 minutes under argon. The mixture was stirred for 7 30 mins at 0°C and diluted with benzene ( 120 ml). The benzene solution 8 prepared above was added with stirring via an addition funnel over 20 9 minutes. After 8 hours, the reaction was quenched by careful addition of isopropanol (50 ml) at 0°C, followed by water ( 100 ml). After 15 11 minutes, the reaction mixture was diluted with ether ( 1100 ml) and 12 water (200 ml), and then neutralized with solid sodium bicarbonate (200 13 g). The ether layer was washed with water (100 ml), and saturated 14 aqueous NaCI (2 x 100 ml), and dried over MgS04. Removal of the solvent under reduced pressure afforded a mixture of the isomeric 16 diketones which were separated by chromatography ( 5% EtOAc /
17 hexanes). (Compound R): 1H NMR (CDC13) : 8 8.55 (1H, d, J = 2.0 18 Hz), 8.13 (1H, dd, J = 2.0, 8.3 Hz), 7.53 (1H, d, J = 8.3 Hz), 2.77 (2H, 19 t, J = 6.6 Hz), 2.62 (3H, s), 2.05 (2H, t, J = 6.6 Hz), 1.41 (6H, s).
(Compound S): 1H NMR (CDCl3) : 8 8.10 (1H, d, J = 8.1 Hz), 8.02 21 (1H, d, J = 1.6 Hz), 7.82 (1H, dd, J = 1.6, 8.1 Hz), 2.77 (2H, t, J = 7.1 22 Hz), 2.64 (3H, s), 2.05 (2H, t, J = 7.1 Hz), 1.44 {6H, s).
23 3,4-Dihydro-4.4-dimeth~-7-i(2-(~ -methyl-1 3-dioxolanyllTl lf,2H1-24 naphthalenone (Compound T~
A mixture of 3,4-dihydro-4,4-dimethyl-7-acetyl- 1(2H)-26 naphthalenone (Compound R) (140.0 mg, 0.60 mmol), ethylene glycol 27 (55.0 mg, 0.90 mmol), p- toluenesulfonic acid monohydrate (4 mg) and 28 benzene (25 ml) was refluxed using a Dean-Stark apparatus for 12 29 hours. The reaction was quenched by the addition of 10% aqueous sodium bicarbonate, and extracted with ether (2 x 75 ml}. The 1 combined organic layers were washed with water (5 ml), and saturated 2 aqueous NaCI (S ml), and dried over MgS04. Removal of the solvent 3 under reduced pressure afforded the title compound as an oil. 1H
4 NMR(CDC13):8 8.13(lH,d,J=2.OHz),7.64(lH,dd,J=2.0,8.2 Hz), 7.40 (1H, d, J = 8.2 Hz), 3.97- 4.10 (2H, m), 3.70-3.83 (2H, m), 6 2.73 (2H, t, J = 6.5 Hz), 2.01 (2H, t, J = 6.5 Hz), 1.64 (3H, s), 1.39 7 (6H, s).
8 1.2,3,4-Tetrahydro-1-hydro~4-meth ly_phenyl)-4 4-dimethyl-7-(2-(2-9 methyl-1,3-dioxolan~llnaphthalene (Compound U~
To a solution of 195.4 mg (1.00 mmol) p-11 tolulylmagnesiumbromide (1.0 ml; 1M solution in ether) in 2 ml THF
12 was added a solution of 3,4-dihydro-4,4- dimethyl-7-(2-(2-methyl-1,3-13 dioxolanyl))-1(2H)- naphthalenone (Compound T) 135.0 mg, 0.52 14 mmol) in 5 ml THF. The solution was refluxed for 16 hours, cooled to room temperature, and diluted with ether (50 ml). The solution was 16 washed with water (5 ml), saturated aqueous NH4C1 (5 ml), and dried 17 over MgS04. Removal of the solvents under reduced pressure and 18 column chromatography (5% EtOAc / hexanes) afforded the title 19 compound as a solid. 1H NMR (CDC13) : 8 7.37 (2H, d), 7. 21 (1H, s), 7.13 (2H, d, J = 8.5 Hz), 7.08 (2H, d, J = 8.5 Hz), 3.88- 3.99 (2H, m), 21 3.58-3.75 (2H, m), 2.34 (3H, s), 2.12-2.30 (2H, m), 1.79-1.90 (1H, m), 22 1.57 (3H, s), 1.48-1.58 (1H, m), 1.38 (3H, s), 1.31 (3H, s).
23 3,4-Dihydro-1-(4-meth~phen~)-4 4-dimethyl-7-acetylnaphthalene 24 (Compou A mixture of 1,2,3,4-tetrahydro-1-hydroxy-1-(4- methylphenyl)-26 dimethy-'-(2-(2-methyl-1,3- dioxolanyl))naphthalene (Compound U) 27 130.0 mg (0.38 mmol), p-toluenesulfonic acid monohydrate (4 mg) and 28 benzene (5 ml) was refluxed for 16 hours. Upon cooling to room 29 temperature, the reaction mixture was diluted with ether (100 ml) and washed with 10% aqueous sodium bicarbonate, water, and saturated 1 aqueous NaCI. The organic layer was dried over MgS04 and the 2 solvents were removed under reduced pressure to give the title 3 compound as a solid. 1H NMR (CDCl3) : 8 7.83 (1H, dd, J = 1.8,8.0 4 Hz), 7.66 (1H, d, J = 1.8 Hz), 7.45 (1H, d, J = 8.0 Hz), 7.25 (2H, d, J
= 8.5 Hz), 7.22 (2H, d, J = 8.5 Hz), 6.03 (1H, t, J = 6.3Hz), 2.47 (3H, 6 s), 2.41 (3H, s), 2.37 {2H, d, J = 6.3Hz), 1.36 (6H, s).
7 (E~ 3-(5,6-dihydro-5,5-dimethyl-8-~(4-meth~phen~, -2-naphthalenyll-2-8 butenenitrile (Compound W~
9 To a slurry of NaH (48.0 mg, 2.00 mmol) in THF (6 ml), was to added diethylcyanomethylphosphonate (450.0 mg, 2.50 mmol). After 40 11 mins, a solution of 3,4-dihydro- 1-(4-methylphenyl)-4,4-dimethyl-7-12 acetylnaphthalene (Compound V) 95.0 mg, (0.33 mmol) in THF (4 ml) 13 was added. The mixture was stirred for 16 hours, diluted with ether (100 14 ml), and washed with water, and saturated aqueous NaCl before being dried over MgS04. Removal of the solvents under reduced pressure, 16 and column chromatography (3% EtOAc / hexanes) afforded the title 17 compound as a solid. 1H NMR (CDCl3): 8 ?.39 (1H, d, J = 1H), 7.32 18 (1H, dd, J = 2.0, 8.lHz), 7.20-7.25 (4H, brs), 7.15 (1H, d, J = 2.0 Hz), 19 6.03(lH,t,J=6.OHz),5.44(lH,s),2.42(3H,s),2.36(2H,d,J=6.0 Hz), 2.35 (3H, s), 1.35 (6H, s}.
21 jE)-3-(5,6-dihydro-5.5-dimethyl-8~4-meth~phen~l-2-naphthalenvll-2-22 butenal (Compound Xl 23 To a cold solution (-78°C) of (E)-3-(5,6-dihydro- 5,5-dimethyl-8-24 (4-methylphenyl)-2-naphthalenyl}-2- butenenitrile (Compound W) 84.0 mg, 0.29 mmol) in dichloromethane (4 ml) was added 0.50 ml (0.50 26 mmol) of diisobutylaluminumhydride (1M solution in dichloromethane).
27 After stirring for 1 hour, the reaction was quenched at -78°C by adding 28 2-propanol (1 ml) diluted with ether (100 ml). Upon warming to room 29 temperature, the solution was washed with water, 10% HCI, and saturated aqueous NaCI. The organic layer was dried over MgS04 and _. ~._.w.~..,~~,...w,m ..,....~..,~ _ T,..a.._ ....-a...~..~.-.-_..w..~"--.-.~_.".....~__..~, ..

1 the solvent removed under reduced pressure to give the title compound 2 as an oil. 1H NMR (CDC13) : 8 10.12 (1H, d, J = 7.9 Hz), 7.43 (2H, 3 s), 7.19-7.28 (SH, m), 6.27 (1H, d, J = 7.9 Hz}, 6.03 (1H, t, J = 4.8 Hz), 4 2.47 (3H, s), 2.42 (3H, s), 2.37 (2H, d, J = 4.8 Hz), 1.37 (6H, s).
Ethyl (E,E,E -3-methyl-7-i(5,6-dihydro-5 5-dimethyl-8-l4-6 meth~phenyl)-2-naphthalen~ll-2 4 6-octatrienoate (Compound Sl) 7 To a cold (-78°C) solution of diethyl-(E)-3- ethoxycarbonyl-2-8 methylallylphosphonate [prepared in accordance with J. Org. Chem. 39:
9 821 (1974)) 264.0 mg, (1.00 mmol) in THF (2 ml) was added 26.0 mg (0.41 mmol, 0.65 ml)of n-butyllithium in hexanes (1.6 M solution) 11 followed immediately by the addition of (E)-3-(5,6- dihydro-5,5-12 dimethyl-8-(4-methylphenyl)-2-naphthalen- yl)-2-butenal (Compound X}
13 82.0 mg, 0.26 mmol) in THF (3 ml). After 1 hour, the reaction mixture 14 was diluted with ether (60 mi), washed with water (S ml), saturated aqueous NaCI (5 ml) and dried over MgS04. After removal of the 16 solvents under reduced pressure, the title compound was isolated as an 17 oil by column chromatography (5% EtOAc / hexanes, followed by 18 HPLC using 1% EtOAc / hexanes). 1H NMR (acetone-d6) : 8 19 7.36-7.43 (2H, m), 7.18-7.27 {4H, m), 7.17 (1H, d, J = 1.7 Hz), 7.08 (1H, dd, J = 11.2, 15.2 Hz), 6.46 (1H, d, J = 11.2 Hz), 6.38 (1H, d, J =
21 15.2Hz),5.98(lH,t,J=4.7Hz),5.78(lH,s),4.10(2H,q,J=7.1 22 Hz), 2.35 (3H, s), 2.33 (3H, s), 2.32 (2H, d, J = 4.7 Hz), 2.12 (3H, s), 23 1.31 (6H, s), 1.22 (3H, t, J= 7.1 Hz).
24 (E,E,E)-3-meth~(5,6-dih~rdro-5 5-dimethy~4-methyl~henYl)-2-naphthal~°n~)-2,4,6-octatrienoic acid (Compound 52) 26 To a solution of ethyl (E,E,E)-3-methyl-7-(5,6r dihydro-5,5-27 dimethyl-8-(4-methylphenyl)-2- naphthalenyl)-2,4,6-octatrienoate 28 (Compound 51) 85.0 mg, 0.20 mmol) in THF (lml) and methanol (1m1) 29 was added 12.0 mg (0.50 mmol) of LiOH (0.5 ml, 1M solution). The mixture was stirred for 6 hours, diluted with ether (60 ml), acidified with 1 10% HC1 (1 ml). The solution was washed with water, and saturated 2 aqueous NaCI, before being dried over MgS04. Removal of the 3 solvents under reduced pressure afforded the title compound as a solid, 4 which was purified by recrystallization from acetone. 1H NMR
(acetone-d6) : 8 7.35-7.45 (2H, m), 7.19-7.28 (4H, m), 7.17 (1H, d, J =
6 l.BHz), 7.09 {1H, dd, J = 11.5, 15.1 Hz), 6.48 (1H, d, J = 11.5 Hz), 6.42 7 (1H, d, J = 15.1Hz),5.99 (1H, t, J = 4.7 Hz),5.82 (1H, s), 2.36 (3H, s), 8 2.33 (2H, d, J = 4.7Hz), 2.32 (3H, s), 2.13 (3H, s), 1.32 (6H, s).
9 3,4-dihydro-4,4-dimethyl-7-nitro-ly2Hl-naphthalenon~Comyound Y) l0 To 1.7 ml (3.Og, 30.6 mmol, 18M) H2S04 at -5°C (ice-NaCI bath) 11 was slowly added 783.0 mg (4.49 mmol) of 3,4-dihydro-4,4-12 dimethyl-1(2H)-naphthalenone. A solution of 426.7 mg (6.88 mmol, 13 0.43 ml, 16M) HN03, and 1.31g (0.013 mol, 0.74 ml, 18 M) H2S04 was 14 slowly added. After 20 minutes, ice was added and the resulting mixture extracted with EtOAc. The combined extracts were 16 concentrated under reduced pressure to give a residue from which the 17 title compound, a pale yellow solid, was isolated by column 18 chromatography (10% EtOAC / hexanes). 1H NMR (CDC13) : 8 8.83 19 (lH,d,J=2.6Hz),8.31(lH,dd,J=2.8,8.9Hz),7.62{lH,d,J=8.7 Hz), 2.81 (2H, t, J = 6.5 Hz), 2.08 (2H, t, J = 6.5 Hz), 1.45 (6H, s).
21 3~4-dihydro-4 4-dimethyl-7-amino-l~2Hl-naphthalenone (Compound Z) 22 A solution of 230.0 mg {1.05 mmol) 3,4-dihydro-4,4-dimethyl-7-23 nitro-1{2H)-naphthalenone (Compound Y) in 5.0 ml of EtOAc was 24 stirred at room temperature with a catalytic amount of 10% Pd-C under 1 atm of H2 for 24 hours. The catalyst was removed by filtration 26 through a pad of Celite, and the filtrate concentrated under reduced 27 pressure to give the title compound as a dark green oil. 1H NMR
28 (CDC13) : 8 7.30 (1H, d, J = 2.7 Hz), 7.22 (1H, d, J = 8.4 Hz), 6.88 29 (1H, dd, J = 2.7, 8.5 Hz), 2.70 (2H, t, J = 6.6 Hz), 1.97 (2H, t, J = 6.6 3o HZ), 1.34 (6H, s).

1 Ethvl 4-(i(5,6,7,8-tetrahydro-5 5-dimethyl-8-oxo-2-naphthalen,1)~ az~ol-_ 2 benzoate (Compound AAA
3 To a solution of 198.7 mg (1.05 mmol) 3,4-dihydro-4,4-dimethyl-7-4 amino-1(2H)-naphthaIenone (Compound Z) in 5.0 ml glacial acetic acid was added 180.0 mg ( 1.00 mmol) of ethyl 4-nitrosobenzoate. The 6 resulting solution was stirred overnight at room temperature, and then 7 concentrated under reduced pressure. The product was isolated from 8 the residual oil as a red solid, by column chromatography (15% EtOAc 9 - hexanes). 1H NMR (CDC13) : 8 8.57 (1H, d, J = 2.0 Hz), 8.19 (2H, l0 d, J = 8.4 Hz), 8.07 (1H, d, J = 8.0 Hz), 7.94 (2H, d, J = 8.4 Hz), 7.58 11 (1H, d, J = 8.6 Hz), 4.41 (2H, q, J = 7.1 Hz), 2.79 (2H, t, J = 6.6 Hz), I2 2.07 (2H, t, J = 7.02 Hz), 1.44 (6H, s), 1.42 (3H, t, 3 = 7.1 Hz).
I3 Ethvl 4-f 15,6-dihydro-5,5-dimethyl=8-(trifluoromethylsulfon ly_)ox 14 naphthalenyl, azo]- benzoate (Compound BB), To a solution of 90.4 mg sodium bis(trimethylsilyl)amide (0.48 I6 mmol, 0.48 ml of a 1.0 M THF solution) in 2.0 ml THF at -78°C, was 17 added 153.0 mg (0.437 mmol) of ethyl 4-((5,6,7,8-tetrahydro-5,5-18 dimethyl-8-oxo-2-naphthalenyl)azo]-benzoate (Compound AA) in 2.0 ml 19 THF. The dark red solution was stirred at -78°C for 30 minutes and then 204.0 mg (0.520 mmol) of 2-(N,N-21 bis(trifluoromethylsuifonyl)amino]-5-chloropyridine was added as a 22 solution in 2.0 ml THF. The reaction mixture was allowed to warm to 23 room temperature and after 3 hours it was quenched by the addition of 24 HBO. The organic layer was concentrated to a red oil under reduced p:c.ssure. The product way isolated by column chromatography (25%
26 EtOAc / hexanes) as a red oil. 1H NMR (CDCl3) : 8 8.21 (2H, d, J =
27 8.6 Hz), 7.96 (2H, d, J = 8.6 Hz), 7.94 (2H, m), 7.49 (1H, d, J = 8.2 28 Hz), 6.08 (1H, t, J = 2.5 Hz), 4.42 (2H, q, J = 7.1 Hz), 2.49 (2H, d, J =
29 4.8 Hz), 1.44 (3H, t, J = 7.1 Hz), 1.38 (6H, s).
Ethyl 4-(15,6-dihydro-5 5-dimethyl- 8-(4-meth~phenyl 1 naphthalen~rl)azo~-benzoate (Compound 46a~
2 A solution of 4-lithiotoluene was prepared by the addition of 62.9 3 mg (0.58 ml, 0.98 mmol) of t-butyl lithium {1.7 M solution in pentane) 4 to a cold solution (-78°C) of 84.0 mg (0.491 mmol) of 4-bromotoluene in 1.0 ml of THF. After stirring for 30 minutes a solution of 107.0 mg 6 (0.785 mmol) of zinc chloride in 2.0 ml of THF was added. The 7 resulting solution was warmed to room temperature, stirred for 30 8 minutes, and added via cannula to a solution of 94.7 mg (0.196 mmol) 9 of ethyl 4-[(5,6-dihydro-5,5-dimethyl-8- (trifluoromethylsulfonyl)oxy-2-l0 naphthalenyl)azo]- benzoate (Compound BB) and 25 mg (0.02 mmol) of 11 tetrakis(triphenylphosphine)palladium{0) in 2.0 ml of THF. The 12 resulting solution was heated at 50°C for 1.5 hours, cooled to room 13 temperature and diluted with sat. aqueous NH4C1. The mixture was 14 extracted with EtOAc (40 ml) and the combined organic layers were washed with water and brine. The organic phase was dried over 16 NaZS04, concentrated in vacuo, and the title compound isolated as a red 17 solid by column chromatography (25% EtOAc-hexanes) 1H NMR
18 (CDC13) : 8 8.21 (2H, d, J = 8.6 Hz), 7.96 (2H, d, J = 8.6 Hz), 7.94 19 (2H, m), 7.49 (1H, d, J = 8.2 Hz), 6.08 (1H, t, J = 2.5 Hz), 4.42 (2H, q, J = 7.1 Hz), 2.49 (2H, d, J = 4.8 Hz), 1.44 {3H, t, J = 7.1 Hz), 1.38 21 (6H, s).
22 4-[j5,6-dihydro-5 5-dimethyl-8-(4-methvlnhen ly_1-2~naphthalenyl)azol 23 benzoic acid (Compound 46b1 24 To a solution of ethyl 4-[(5,6-dihydro-S,5- dimethyl-8-(4-methylphenyl)-2-naphthalenyl)azo]- benzoate (Compound 46a) 16.5 mg, 26 0.042 mmol) in THF (2 ml) and ethanol ( iml) was added 80.0 mg (2.00 27 mmol) of NaOH (2.0 ml, 1M aqueous solution). The mixture was stirred 28 for 12 hours at room temperature, acidified with 10% HCI, and 29 extracted with EtOAc. The combined organic layers were washed with water, and saturated aqueous NaCI, then dried over MgS04. Removal ~...~....~.w~-.~.-.~~.,.. . .. ~.t...~.,-. ..~.._-_......~ ..~~.~.,a,.m..__. .
_. .

1 of the solvents under reduced pressure, and recrystallization of the 2 residue from EtOAC / hexane, afforded the title compound as a red 3 solid. 1H NMR (acetone-d6) : 8 8.19 (2H, d, J = 8.4 Hz), 7.92 (2H, d, 4 J=8.5hz),7.88(2H,dd,J=2.1,6.1Hz),7.66(lH,s),7.64{2H,d,J
= 2.3 Hz), 7.28 (4H, d, J = 3.0 Hz), 6.09 (1H, t, J = 2.5 Hz), 2.42 (2H, 6 d, J = 4.8 Hz), 2.39 (3H, s), 1.40 (6H, s).
7 6-(2-Trimethylsil~)eth~yl-2,3-dihydro-3,3-dimethyl-1H-inden-1-one 8 (Compound CC~
9 To a solution of 815.0 mg (3.41 mmol) 6-bromo-2,3- dihydro-3,3-dimethyl-1H-inden-1-one (See Smith et al. Org. Prep. Proced. Int. 1978 11 10 123-131) in 100 ml of degassed Et3N (sparged with argon for 20 min) 12 was added 259.6 mg (1.363 mmol) of copper(I) iodide, 956.9 mg (1.363 13 mmol) of bis(triphenylphosphine)palladium(II)chloride, and 3.14 g 14 (34.08 mmol) of (trimethylsilyl)acetylene. This mixture was heated at 70°C for 42 hours, cooled to room temperature, and filtered through a 16 pad of silica gel and washed with ether. The filtrate was washed with 17 water, 1 M HCI, water, and finaly with saturated aqueous NaCI before 18 being dried over MgS04. Concentration of the solution under reduced 19 pressure, followed by column chromatography (silica gel; 10% Et20 -hexanes) afforded the title compound as a brown oil. 1H NMR (300 21 MHz, CDC13): 8 7.79(1H, d, J = 1.4 Hz), 7.69 (1H, dd, J = 1.6, 8.3 22 Hz), 7.42 (1H, d, J = 8.5 Hz), 2.60 (2H, s), 1.41 (6H, s), 0.26 (9H, s).
23 6-Eth~yl-2,3-dihydro-3 3-dimethyl-1H-inden-1-one (Compound DD) ' 24 To a solution of 875.0 mg (3.41 mmol) 6-(2 trimethylsilyl)ethynyl-2,3-dihydro-3,3-dimethyl-1H-inde.r,-1-one 26 (Compound CC) in 28 ml of MeOH, was added 197. ~ nlg ( 1.43 mmol) 27 of K2C03 in one portion. After stirring for 6 hours at room 28 temperature the mixture was filtered though a pad of Celite and the 29 filtrate concentrated under reduced pressure. The residual oil was 3o placed on a silica gel column and eluted with 5% EtOAc-hexanes to 1 give the title product as a colorless oil. 1H NMR (300 MHz, CDC13): 8 2 7.82 (1H, s), 7.72 (1H, dd, J = 1.6, 7.8 Hz), 7.47 (1H, d, J = 8.4 Hz), 3 3.11 (1H, s), 2.61 (2H, s), 1.43 (6H, s).
4 Ethvl4-(2-(5,6-dihydro-5,5-dimethyl-7-oxo-2-indenvlleth~n~~benzoate (Compound EE) 6 A solution of 280.0 mg (1.520 mmol) 6-ethynyl-2,3- dihydro-3,3-7 dimethyl-1H-inden-1-one (Compound DD) and 419.6 mg (1.520 mmol) 8 ethyl 4-iodobenzoate in S ml Et3N was sparged with argon for 40 9 minutes. To this solution was added 271.0 mg (1.033 mmol) of triphenylphosphine, 53.5 mg (0.281 mmol) of copper(I) iodide, and 53.5 11 mg (0.076 mmol) of bis(triphenylphosphine)palladium(II) chloride. The 12 resulting mixture was heated to reflux for 2.5 hours, cooled to room 13 temperature, and diluted with Et20. After filtration through a pad of 14 Celite, the filtrate was washed with H20, 1 M HCI, H20, and saturated aqueous NaCI, then dried over MgS04, and concentrated under reduced 16 pressure. The title compound was isolated as a pale-yellow solid by i7 column chromatography (15% EtOAc-hexanes). 1H NMR (300 MHz, 18 d6-acetone): 8 8.05 (2H, d, J = 8.6 Hz), 7.87 (1H, dd, J = 1.4, 8.1 Hz), 19 7.75 (2H, m), 7.70 (2H, d, J = 8.5 Hz), 4.36 (2H, q, J = 7.1 Hz), 2.60 zo (2H, s), 1.45 (6H, s), 1.37 (3H, t, J = 7.1 Hz).
21 Ethvl4-(2-(1,1-dimeth~(trifluoromethyl-sulfonvlloxy-5-22 inden~)ethynyllbenzoate (Compound FF) 23 A solution of 88.0 mg (0.48 mmol) of sodium 24 bis(trimethylsilyl)amide in 0.5 ml THF was cooled to -78°C and 145.0 mg (0.436 mmol) of ethyl 4-(2-(5,6-dihydro-5,5-dimethyl-7-oxo-2-26 indenyl)ethynyl]benzoate (Compound EE) was added as a solution in 27 1.0 ml THF. After 30 minutes 181.7 mg (0.480 mmol) of 2-(N,N-28 bis(trifluoromethansuifonyl)amino)-5-chloro-pyridine was added as a 29 solution in 1.0 ml THF. The reaction was allowed to slowly warm to room temperature and quenched after 5 hours by the addition of ....m.~.~.-..-,~...._.~... . . _...~ .._.__..e~.~~w _.. ~.~..~.~m.._~~..-_ 1 saturated aqueous NH4C1. The mixture was extracted with EtOAc, and 2 the combined organic layers washed with 5% aqueous NaOH, HZO, and 3 saturated aqueous NaCI, then dried (MgS04) and concentrated under 4 reduced pressure. The product was isolated as a colorless solid by column chromatography (10% Et20-hexanes). 1H NMR (300 MHz, d6-6 acetone): 8 8.05 (2H, d, J = 8.3 Hz), 7.69 (2H, d, J = 8.4 Hz), 7.63 7 (2H, s), 7.55 (1H, s), 4.36 (2H, q, J = 7.1 Hz), 1.44 (6H, s), 1.37 (3H, t, 8 J=7.lHz).
9 4-[,(5,6-dihydro-5.5-dimethy~4-meth~~hen~l-2-l0 naphthalenylleth r~nyl]benzoic acid (Compound 60) 11 A solution of 142.6 mg (0.339 mmol) of ethyl 4- [(5,6-dihydro-5,5-12 dimethyl-8-(4-methylphenyl)-2- naphthalenyl)ethynyl]benzoate 13 (Compound 1) and 35.6 mg (0.848 mmol) of LiOH-H20 in 12 ml of 14 THF/water (4:1, v/v), was stirred overnight at room temperature. The reaction mixture was extracted with hexanes, and the hexane fraction 16 extracted with 5% aqueous NaOH. The aqueous layers were combined 17 and acidified with 1M HCI, and then extracted with EtOAc and Et20.
18 The combined organic layers were dried over Na2S04 and concentrated 19 in vacuo to give the title compound as a colorless solid. 1H NMR (db-DMSO): 8 7.91 (2H, d, J = 8.4 Hz), 7.60 (2H, d, J = 8.4 Hz), 7.47 21 (2H, s), 7.23 (4H, q, J = 8.1 Hz), 7.01 (1H, s), 6.01 (1H, t, J = 4.6 Hz), 22 2.35 (3H, s), 2.33 (2H, d, J = 4.8 Hz), 1.30 (6H, s).
23 4-[(5,6-dihydro-5,5-dimethyl-8-phen.l-~a~hthalen~~ethynyl]benzoic 24 acid ,Compound 60a) Employing the same general procedure as for the preparation of 26 4-((5,6-dihydro-5,5-dimethyl-8-(2- thiazolyl)-2-27 naphthalenyl)ethynyl]benzoic acid (Compound 30a), 27.0 mg (0.07 28 mmol) of ethyl 4-[(5,6- dihydro-5,5-dimethyl-8-phenyl-2-29 naphthalenyl)ethynyl]benzoate (Compound la) was converted into the title compound (colorless solid) using 5.9 mg (0.14 mmol) of LiOH in 1 H20. PMR (db DMSO): 8 1.31 (6H, s), 2.35 (2H, d, J = 4.5 Hz), 6.05 2 (lH,t,J=J=J=4.SHz),7.00(lH,s),7.33(2H,d,J=6.2Hz),7.44 3 (4H, m), 7.59 (2H, d, J = 8.1 Hz), 7.90 (2H, d, J = 8.1 Hz).
4 4-((5,6-Dihydro-5.5-dimethvl-8-(4-(11-dimethylethyl)phenvlT
naphthalene eth~nyl]benzoic acid (Compound 61) 6 A solution of 80.0 mg (0.173 mmol) of ethyl 4- ((5,6-dihydro-5,5-7 dimethyl-8-(4-(1,1- dimethylethyl)phenyl)-2-8 naphthalenyl)ethynyl]benzoate (Compound 6) and 18.1 mg (0.432 9 mmol) of LiOH-H20 in 6 ml of THF/water (3:1, v/v), was stirred l0 overnight at room temperature. The reaction mixture was extracted 11 with hexanes, and the remaining aqueous layer acidified with 1M HCI, 12 and then extracted with EtOAc. The combined organic layers were 13 dried over Na2S04 and concentrated in vacuo to give the title compound 14 as a colorless solid. 1H NMR (db DMSO): 8 7.82 (2H, d, J = 8.2 Hz), 7.44(6H,m),7.25(2H,d,J=8.3Hz),7.02(lH,s),6.01(lH,t,J=4.6 16 Hz), 2.32 (2H, d, J = 4.7 Hz), 1.32 (9H, s), 1.29 (6H, s).
17 Ethvl 2-fluoro-4-(y(5,6-dih d~"5,5-dimethyl-8-(4-meth3rlphenyl~2-18 naphthalene)thiocarbon~]amino]-benzoate (Compound 62) 19 A solution of 54.4 mg (0.119 mmol) ethyl 2-fluoro-4-(((5,6-dihydro-5,5-dimethyl-8-(4-methylphenyl)-2-21 naphthalenyl)carbonyl]amino]-benzoate (Compound 40) and 57.7 mg 22 (0.143 mmol) of (2,4-bis(4- methoxyphenyl)-1,3-dithia-2,4-23 diphosphetane-2,4- disulfide] (Lawesson's Reagent) in 12.0 ml of 24 benzene was refluxed overnight. Upon cooling to room temperature, the mixture was filtered and the filtrate concentrated under reduced 26 pressure. The title compound was isolated by column chromatography 27 (10 to 25% EtOAc / hexanes) as a yellow solid. 1H NMR (CDCl3): 8 28 9.08(lH,s),7.92(lH,brs),7.90(lH,t,J=8.2Hz),7.66(lH,dd,J=
29 2.0, 6.0 Hz), 7.38 (3H, m), 7.18 (4H, m), 6.01 (1H, t, J = 4.7 Hz), 4.35 (2H, q, J = 7.1 Hz), 2.36 (3H, s), 2.33 (2H, d, J = 4.7 Hz), 1.38 (3H, t, .. ....~.-...~,.., ..~.~"~ ~, ~.m .A-~..... , . . .... T.. ... ~ ~~....y, ~, N
_~. .~-...~..~"-_.. .. .

1 J = 7.1 Hz), 1.33 (6H, s).
2 2-fluoro-4-[[~5,6-dihydro-5,5-dimethyl-8-l4-methylphen 1~1-2-3 naphthalenyl)thiocarbonyl]amino]-benzoic acid (Compound 63~
4 To a solution of 46.5 mg (0.098 mmol) ethyl 2- fluoro-4-[[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-6 naphthalenyl)thiocarbonyl]amino]- benzoate (Compound 62) in 1.0 ml 7 EtOH and 1.0 ml of THF was added 55 mg NaOH (1.4 mmol) and 1.0 8 ml of H20. After stirring at room temperature for overnight EtOAc was 9 added, and the reaction quenched by the addition of 10% HCI.
Extraction with EtOAc was followed by washing of the combined 11 organic layers with H20, saturated aqueous NaCI, and drying over 12 MgS04. Removal of the solvent under reduced pressure provided a 13 solid which after crystallization from CH3CN afforded the title 14 compound as a pale-yellow solid. 1H NMR (db acetone): 8 11.05 (1H, s),8.02(lH,m),7.99(lH,t,J=8.3Hz),7.75(lH,m),7.69(lH,dd,J
16 =2.0,6.1Hz),7.52(lH,s),7.46(lH,d,J=8.lHz),7.21(4H,m),6.04 17 (IH, t, J = 4.8 Hz), 2.37 (2H, d, J = 4.8 Hz), 2.33 (3H, s), 1.36 (6H, s).
18 Ethyl 5',6'-dih~dro-5',5'-dimethyl-8'-(4-methyluhenyll~,[2 2'-19 binaphthalene]-6-carbo lx~ate (Compound 64) A solution of 3,4-dihydro-1-(4-methylphenyl)-4,4- dimethyl-7-21 bromonaphthalene Compound D) 0.45 g, 1.40 mmol) and THF (2.1 ml) 22 was added to magnesium turnings (0.044 g, 1.82 mmol) at room 23 temperature under argon. Two drops of ethylene dibromide were added, 24 and the solution, which slowly became cloudy and yellow, was heated to reflux for 1.5 hours. In a second flask was added zinc chloride (0.210 g, 26 1.54 mmol), which was melted under high vacuum, cooled to room 27 temperature and dissolved in THF (3 ml). The Grignard reagent was 28 added to the second flask and, after 30 minutes at room temperature, a 29 solution of ethyl 6-bromo-2- naphthalinecarboxylate (Compound N}
0.293 g, (1.05 mmol) and THF (2 ml) were added. In a third flask was 1 prepared a solution of Ni(PPh3)4 and THF as follows: To a solution of 2 NiCl2(PPh3)2 {0.82 g, 1.25 mmol) and PPh3 (0.66 g, 2.5 mmol) in THF
3 (3.5 ml) was added a 1M solution of diisobutylaluminum hydride and 4 hexanes (2.5 ml, 2.5 mmol), and the resulting solution diluted with THF
to a total volume of 15 ml and stirred at room temperature for 15 6 minutes. Three 0.60 ml aliquots of the Ni(PPh3)4 solution were added 7 at 15 minutes intervals to the second flask. The resulting suspension 8 was stirred at room temperature for 2 hours. The reaction was 9 quenched by the addition of 5 ml 1N aqueous HCl and stirred for 1 l0 hour before extracting the products with ethyl acetate. The organic 11 layers were combined, washed with brine, dried (MgS04), filtered and 12 the solvent removed in-vacuo. The residue was crystalized from hexanes 13 to give 130 mg of pure material. The mother liquor was concentrated 14 under reduced pressure and the residue purified by silica gel chromatography (95:5- hexanes:ethyl acetate) to give an additional 170 16 mg of the title compound (overall yield = 300 mg, 64 %) as a colorless 17 solid. 1H NMR (CDCl3) 8 8.57 (s, 1H), 8.05 (dd, 1H, J = 1.7, 8.0 Hz), 18 7.84-7.95 (overlapping d's, 3H), 7.66 {dd, 1H, J = 1.7, 8.5 Hz), 7.58 (dd, 19 1H,J=2.0,8.OHz),7.48(d,lH,J=8.OHz),7.43{d,lH,J=2.OHz), 7.32 (d, 2H, J = 8.0 Hz), 7.21 {d, 2H, J = 8.0 Hz), 6.04 (t, 1H, J = 4.8 21 Hz), 4.44 (q, 2H, J = 7.1 Hz), 2.40 (s, 3H), 2.39 (d, 2H, J = 4.8 Hz), 22 1.45 (t, 3H, J = 7.1 Hz), 1.39 (s, 6H).
23 5',6'-Dihydro-5',5'-dimethyl-8'-(4-methylphenYl~ X212'-binaphthalene]-6-24 carboxylic acid yCompou A solution of ethyl 5',6'-dihydro-5',5'-dimethyl- 8'-(4-26 methylphenyl)-(2,2'-binaphthalene]-6-carboxylate (Compound 64) 0.19 g, 27 0.43 mmol), EtOH (8 ml) and 1N aqueous NaOH (2 ml) was heated to 28 60°C for 3 hours. The solution was cooled to 0°C and acidified with 1N
29 aqueous HCI. The product was extracted into ethyl acetate, and the organic layers combined, washed with water, brine, dried (MgS04), A....__w,..-.,~,..~.-."...,.~~.w. ~.-... . T . .. ........ ~...,~. ..h m. ~
a.".~..... ~---..~.. _ .. .

1 filtered and the solvent removed in-vacuo. The residue was 2 recrystalized from THF/ethyl acetate at 0°C to give 35 mg of pure 3 material. The mother liquor was concentrated under reduced pressure 4 and the residue purified by silica gel chromatography (100% ethyl acetate) to give an additional 125 mg of the title compound (overall 6 yield = 160 mg, 90 %) as a colorless solid. 1H NMR (DMSO-d6) 8 7 8.57 (s, 1H), 8.11 (d, 1H, J = 8.7 Hz), 7.96-7.82 (overlapping d's, 8 3H),7.65 (d, 2H, J = 7.6 Hz), 7.50 {d, 1H, J = 7.9 Hz), 7.28 (s, 1H), 9 7.26(d,2H,J=8.3Hz),7.21(d,2H,J=8.3Hz),6.01(t,lH,J=4.5 1 o Hz), 3.34 (br s, 1H), 2.31 (s, 3H), 2.31 (d, 2H, J = 4.5 Hz), 1.31 (s, 6H).
1 I Ether[(5,6-dihydro-5 5-dimethyl-8-(2-furylT
12 naphthalenyl, eth~~l)benzoate (Compound 66) 13 Employing the same general procedure as for the preparation of 14 ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-naphthalenyl)ethynyl]benzoate (Compound 1), 250.0 mg (0.52 mmol) of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-17 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the 18 title compound (colorless solid) using 142.4 mg (1.045 mmol) of zinc 19 chloride, 24.1 mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) and 2-lithiofuran (prepared by 21 the addition of 53.4 mg (0.52 ml, 0.78 mmol) of n-butyllithium (1.SM
22 solution in hexane) to a cold solution (-78°C) of 53.4 mg (0.784 mmoI) 23 of furan in 1.0 ml of THF). PMR (CDC13): 8 1.32 (6H, s), 1.41 (3H, t, J
24 = 7.1 Hz), 2.35 (2H, d, J = 5.0 Hz), 4.39 (2H, q, J = 7.1 Hz), 6.41 (1H, t,J=S.OHz),6.50(2H,s),7.36(lH,d,J=8.OHz),7.45(lH,dd,J=
26 1.7, 8.0 Hz), 7.49 (1H, s), 7.57 (2H, d, J = 8.2 Hz), 7.63 (1H, d, J = 1.7 27 Hz), 8.02 (2H, d, J = 8.2 Hz).
28 4-[(5,6-dihydro-5 5-dimethyl-8-(2-fuyl)-T2-naphthalenylT ~Yl]benzoic 29 acid ,Compound 67~
Employing the same general procedure as for the preparation of 1 4-[(5,6-dihydro-5,5-dimethyl-8-(2- thiazolyl)-2-2 naphthalenyl)ethynyl]benzoic acid (Compound 30a), ethyl 4-[(5,6-3 dihydro-5,5-dimethyl-8- (2-furyl)-2-naphthalenyl)ethynyl)benzoate 4 (Compound 66) was converted into the title compound (colorless solid) using 16.0 mg (0.38 mmol) of LiOH in H20. PMR (d6-DMSO): 8 1.26 6 (6H, s), 2.33 ( 2H, d, J = 4.9 Hz), 6.41 {1H, t, J = 4.9 Hz), 6.60 (2H, '7 m), 7.45-7.53 (3H, m), 7.64 (2H, d, J = 8.3 Hz), 7.75 (1H, d, J = 1.6 8 Hz), 7.93 (2H, d, J = 8.3 Hz).
9 3 4-dih~dro-4 4-dimethvl-7-acetyl-1(2Hl-naphthalenone~Compound 100C1 and 3,4-dihydro-4,4-dimethyl-6-acet~-1(2Hl-naphthalenone 11 {Compound 100D) 12 To a cold (0° C) mixture of aluminum chloride (26.3 g, 199.0 13 mrnols) in dichloromethane (55 ml) was added acetylchloride (15 g, 192 14 mmols) and 1,2,3,4- tetrahydro-1,1-dimethylnaphthalene (24.4g, 152mmols) in dichloromethane (20 ml) over 20 minutes. The reaction 16 mixture was warmed to ambient temparature and stirred for 4 hours. Ice 17 (200 g) was added to the reaction flask and the mixture diluted with 18 ether (400 ml). The aqueous and organic layers were separated and the 19 organic phase was washed with 10% HCI (50 ml), water (50 ml), 10%
aqueous sodium bicarbonate, and saturated aqueous NaCI (50 ml) and 21 then dried over MgS04. The solvent was removed by distillation to 22 afford a yellow oil which was dissolved in benzene (50 ml).
23 To a cold (0° C) solution of acetic acid (240 ml) and acetic 24 anhydride (120 ml) was added chromium trioxide {50 g, 503 mmols) in small portions over 20 minutes under argon. The mixture was stirred for 26 30 minutes at 0° C and diluted with benzene (120 ml). The benzene 27 solution prepared above was added with stirring via an addition funnel 28 over 20 minutes. After 8 hours, the reaction was quenched by the 29 careful addition of isopropanol (50 ml) at 0° C, followed by water (100 ml). After 15 minutes, the reaction mixture was diluted with ether (1100 _ .. __.~..._....~.,..,~..-~.....~..W~.. . ~.~.~_~.-,.~_ _ .._-~..~..~..._w.~~_..

1 ml) and water (200 ml), and then neutralized with solid sodium 2 bicarbonate (200 g). The ether layer was washed with water ( 100 3 ml),and saturated aqueous NaCI (2 x 100 ml), and dried over MgS04.
4 Removal of the solvent under reduced pressure afforded a mixture of the isomeric diketones which were separated by chromatography ( 5%
6 EtOAc / hexanes). {Compound 100C): 1H NMR (CDCl3) : 8 8.55 7 (lH,d,J=2.OHz),8.13(lH,dd,J=2.0,8.3Hz),7.53(lH,d,J=8.3 8 Hz), 2.77 (2H, t, J = 6.6 Hz), 2.62 (3H, s), 2.05 (2H, t, J = 6.6 Hz), 9 1.41 (6H, s). (Compound 100D): 1H NMR (CDCl3) : 8 8.10 (1H, d, J
l0 = 8.1 Hz), 8.02 (1H, d, J = 1.6 Hz), 7.82 (1H, dd, J = 1.6, 8.1 Hz), 2.77 11 (2H, t, J = 7.1 Hz), 2.64 (3H, s), 2.05 (2H, t, J = 7.1 Hz), 1.44 (6H, s).
12 3,4-dihydro-4,4-dimethyl-6-(2-(2-methyl-1,3-dioxolanyl~~2H~
13 naphthalenone (Compound 100E~
14 A solution of 1.80 g (8.34 mmol) of a 1:5 mixture of 3,4-dihydro-4,4-dimethyl-7-acetyl-1(2H)- naphthalenone (Compound 100C);
16 and 3,4-dihydro-4,4- dimethyl-6-acetyl-1(2H)-naphthalenone (Compound 17 100D) in 50 ml benzene was combined with 517.7 mg (8.34 mmol) of 18 ethylene glycol and 20.0 mg (0.11 mmol) of p- toluenesulfonic acid 19 monohydrate. The resulting solution was heated to reflux for 18 hours, cooled to room temperature, and concentrated under reduced pressure.
21 The title compound was isolated by column chromatography (10%
22 EtOAc - hexanes) as a colorless oil. 1H NMR (CDC13) : 8 8.01 {1H, d, 23 J = 8.2 Hz), 7.51 (1H, s), 7.43 (1H, dd, J = 1.7, 6.4 Hz), 4.07 (2H, m), 24 3.79 (2H, m), 2.74 (2H, t, J = 6.5 Hz), 2.04 (2H, t, J = 7.1 Hz), 1.67 3H, s), 1.46 (6H, s).
26 1,2,3,4-tetrahydro-1-hydr ~r-1-(4-meth~phenyl)-4,4-dimeth,1-~6-(2-(2-27 methyl-1,3-dioxolan, l~llnaphthalene (Compound 100F) 28 To a solution of 496.2 mg (2.54 mmol) p-29 tolyimagnesiumbromide in 20 ml THF (2.54 ml; 1M solution in ether) was added a solution of 3,4-dihydro-4,4-dimethyl-6-(2-(2-methyl-1,3-1 dioxolan- yl))-1{2H)-naphthalenone (Compound 100E, 200.0 mg, 0.769 2 mmol) in THF {5 ml). The solution was refluxed for 16 hours, cooled to 3 room temperature, and washed with water, saturated aqueous NH4C1, 4 and dried over MgS04. Removal of the solvents under reduced pressure and column chromatography (10% EtOAc / hexanes) afforded 6 the title compound as a colorless solid. 1H NMR (CDC13) : 8 7.49 7 (1H, d, J = 1.7 Hz), 7.19 (2H, m), 7.10 (2H, d, J = 7.9 Hz), 7.04 (1H, 8 d, J = 8.2 Hz), 4.05 (2H, m), 3.80 (2H, m), 2.34 (3H, s), 2.21 (1H, m), 9 2.10 (1H, m), 1.88 (1H, m), 1.65 (3H, s), 1.54 (1H, m), 1.39 (3H, s), 1.33 (3H, s).
11 3 4-dih dy ro-1-~(4-meth~phen~)-4,4-dimethxl-6-acetylna~hthalene 12 {Compound 100G) 13 A solution of 1,2,3,4-tetrahydro-1-hydroxy-1-(4-14 methylphenyl)-4,4-dimethyl-6-(2-(2-methyl-1,3- dioxolanyl))naphthalene (Compound 100F 160.0 mg, 0.52 mmol), p-toluenesulfonic acid 16 monohydrate (4 mg) and 30 ml benzene was refluxed for 12 hours.
17 After cooling to room temperature, the reaction mixture was diluted 18 with ether (100 ml) and washed with 10% aqueous sodium bicarbonate, 19 water, and saturated aqueous NaCI. The organic layer was dried over MgS04 and the solvents were removed under reduced pressure to give 21 the title compound, which was isolated by column chromatography 22 (10% EtOAc-hexanes) as a yellow oil . 1H NMR (CDC13) : 8 7.97 (1H, 23 d, J = 1.8 Hz), 7.67 (1H, dd, J = 1.7, 6.4 Hz), 7.22 (4H, s), 7.13 (1H, d, 24 J = 8.1 Hz), 6.10 (1H, t, J = 4.5 Hz), 2.59 ( 3H, s), 2.40 (3H, s), 2.38 (2H, d, J = 4.7 Hz), 1.38 (6H, s).
26 4-(3-oxo-3-(7.8-dih, d~{4-methylphenvll-8,8-dimeth, 27 naphthaien,ill-1-propen~~l~-benzoic acid (Compound 1 28 To a solution of 78.7 mg (0.272 mmol) 3,4-dihydro- 1-(4-29 methylphenyl)-4,4-dimethyl-6-acetylnaphthalene (Compound 100G) in 4.0 ml of MeOH was added 53.1 mg (fl.354 mmol) of 4-carboxy ._~.._...,~...-,.~~.-~,..~,., .. . .. _ .~~w~,.~. ~ ..~w.e... ..w..a.~..w. ._ 1 benzaldehyde, and 80. mg (2.00 mmol; 2.0 ml of 1M aqueous NaOH).
2 The resulting solution was stirred at room temperature for 12 hours, 3 concentrated under reduced pressure, and the residual oil dissolved in 4 EtOAc. The solution was treated with 10% HCI, and the organic layer was washed with H20, and saturated aqueous NaCI, then dried over 6 Na2S04. Removal of the solvents under reduced pressure gave the title 7 compound as a colorless solid which was purified by recrystallization 8 from CH3CN. 1H NMR (acetone-d6) : 8 8.00 (7H, m), 7.83 (1H, d, J =
9 15.6Hz),7.24(4H,s),7.13(lH,d,J=8.lHz),6.12(lH,t,J=4.5 Hz), 2.42 (2H, d, J = 4.8 Hz), 2.38 (3H, s), 1.41 (6H, s).
11 3,4-dihvdro-1-nhenyl-4 4-dimeth 1-6-acetylnaphthalene (Compound 12 100H~
13 To a solution of 508.0 mg (1.95 mmol) of 3,4- dihydro-4,4-14 dimethyl-6-(2-(2-methyl-1,3-dioxolanyl))- 1(2H)-naphthalenone (Compound 100E) in 10 ml of THF was added 496.2 mg (2.54 mmol;
16 2.54 ml of a 1 M solution in Et20) of phenylmagnesium bromide. The 17 resulting solution was heated to reflux for 8 hours, H20 was added and 18 heating continued for 30 minutes. The THF was removed under 19 reduced pressure and the aqueous residue was extracted with EtOAc.
The combined organic layers were dried (MgS04), concentrated under 21 reduced pressure, and the title compound isolated from the residue by 22 column chromatography {10% EtOAc - hexanes) as a colorless oil. 1H
23 NMR (CDCl3) : 8 7.97 (1H, d, J = 1.8 Hz), 7.67 {1H, dd, J = 2.1, 8.0 24 Hz),7.34(SH,m),7.10(lH,d,J=8.lHz),6.12(lH,d,J=4.6Hz), 2.59 (3H, s), 2.39 (2H, d, J = 4.8 Hz), 1.38 (6H, s).
26 4-(3-oxo-3- 7,8-dihydro-5-phenyl-8 8-dimethyl-2-naphthalen,~~l)-1-27 propen~l~-benzoic acid (Compound 103) 28 To a solution of 115.0 mg (0.42 mmol) of 3,4- dihydro-1-29 phenyl-4,4-dimethyl-6-acetylnaphthalene (Compound 100H) and 65.0 mg (0.43 mmol) of 4-formyl- benzoic acid in 5.0 ml EtOH and 1.0 ml THF, 1 was added 120.0 mg (3.00 mmol; 3.0 ml of a 1 M aqueous solution) of 2 NaOH. The resulting yellow solution was stirred at room temperature 3 for 12 hours. The solution was acidified with 6% aqueous HCI and 4 extracted with EtOAc. The combined organic layers were dried (MgS04), concentrated under reduced pressure, and the title 6 compounds was isolated by column chromatography (50% EtOAc -7 hexanes) as a pale yellow solid. 1H NMR (CDCl3) : 8 8.13 (2H, d, J =
8 7.7 Hz), 8.04 (1H, s), 7.81 (1H, d, J = 15.5 Hz), 7.75 (3H, m), 7.60 (1H, 9 d,J=15.5Hz),7.35(SH,m),7.14(lH,d,J=8.lHz),6.I5(lH,t,J=
l0 4.2 Hz), 2.41 (2H, d, J = 4.2 Hz), 1.41 (6H, s).
11 Ethvl 2.6-difluoro-4-((~(5,6-dihydro-5,5-dimethyl-8-(4-methylphen ly')-2-12 naphthalenylycarbonyl)amino~benzoate (Compound 212 13 Using the same procedure as for the synthesis of ethyl 4-(((5,6-14 dihydro-5,5-dimethyl-8-(4-methylphenyl)-2-naphthalenyl)carbonyl]amino]-benzoate (Compound 35), the title 16 compound {Compound 212) was obtained from the reaction of 5,6-1'7 dihydro-5,5-dimethyl-8-(4-methylphenyl)-2-naphthalenecarboxylic acid 18 (Compound K) with ethyl 2,6-difluoro-4-aminobenzoate in the presence 19 of 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC) and 4-dimethylaminopyridine (DMAP) in DMF, in 19% yield as a 21 colorless solid. 1H NMR 8 7.82 (b, 1H), 7.68 (1H, dd, J = 2.1, 7.1 Hz), 22 7.50(lH,d,J=2.lHz),7.47(lH,d,J=7.lHz),7.28(2H,d,J=9.8 23 Hz), 7.23 (4H, m), 6.06 (1H, t, J = 4.8 Hz), 4.39 (2H, q, J = 4.1 Hz), 24 2.40 (3H, s), 2.37 {2H, d, J = 4.8 Hz), 1.38 (3H, t, J = 4.1 Hz), 1.36 (6H,s).
26 2 6-Difluoro-4-(j(5,6~dihydro: 5~5-dimethy~4-meth ly uhen~l-2-27 naphthalenyllcarbonyl)aminol-benzoic acid (Compound 203) 28 Using the same procedure as for the synthesis of 4-(((5,6-29 dihydro-5,5-dimethyl-8-(4-methylphenyl)-2-naphthalenyl)carbonyl)amino)-benzoic acid (Compound 36), the title ... _....._._.-.~..~.__.... . _ _. . ~......_.-~.~. _. _ ___..-.. m~...__... _ 1 compound (Compound 203) was obtained in 70% yield, as a solid by 2 reaction of ethyl 2,6-difluoro-4-[[(5,6-dihydro-5,5-dimethyl-8-(4-3 methylphenyl)-2-naphthalenyl)carbonyl)amino)-benzoate (Compound 4 212) with NaOH in EtOH and THF. ~H NMR 8 7.88 (1H, bs), 7.67 (lH,dd,J1=2.1,7.1Hz),7.50(lH,d,J=2.lHz),7.47(lH,d,J=
6 7.1 Hz), 7.30 (2H, d, J = 10.3 Hz), 7.21 (4H, m), 6.05 (1H, t, J = 4.8 7 Hz), 2.39 (3H, s), 2.37 (2H, d, J = 4.8 Hz), 1.36 (6H, s); MS (EI) m/e 8 447, 403, 275; HRMS found 447.1604 (M), calcd for C2~H2~F2N03 9 447.1646 {M). Anal. (C2~H23F2N03 ~ 1.6 H20) C, H, N.
l0 2s2-Dimethylchroman ( Compound 214 11 To a solution of dihydrocoumarin (Compound 213, S.0 g, 33.78 12 mmol) in dry THF (50 mL) at 0 ~C was added dropwise methyl 13 magnesium chloride (33.7 ml, 3M solution in THF, 0.1 mol). The 14 reaction mixture was slowly warmed to room temperature and stirred overnight. Extraction (Et20), drying (Na2S04) and concentration under 16 reduced pressure afforded a diol (6.25 g) as a colorless solid. The diol 17 was combined with 15% aqueous H2S04 (50 mL) in benzene (15 mL) 18 and the resulting mixture heated at 105 ~C for 2 hours. Upon cooling 19 to room temperature the mixture was extracted with Et20, dried (Na2S04), and concentrated under reduced pressure to give 6.0 g 21 (100%) of the title compound as a light yellow liquid. 'H NMR 8 7.08 22 (2H, m), 6.80 (2H, m), 2.79 (2H, t, J = 6.8 Hz), 1.81 (2H, t, J = 6.8 23 Hz), 1.35 (6H, s).
24 b-Acetyl-2,2-dimethylchroman ~Com~ound 215) To a solutiwn of 2,2-dimethylchroman (Compound 214, 15 g, 26 19.4 mmol) in dry CH2C12 (100 mL) was added acetyl c:hlo: pie (1.52 ml, 27 21.3 mmol) followed by A1C13 (2.84 g, 21.3 mmol). The reaction 28 mixture was stirred at room temperature for 30 minutes and then 29 poured into ice water. Extraction (CH2C12), drying (Na2S04), and concentration under reduced pressure afforded a yellow oil. Column WO 98/58922 PCT/US98/130b5 1 chromatography (EtOAc : hexanes 1:9) yielded 2.15 g (54%) of the 2 title compound as a yellow solid. 1H NMR 8 7.73 (2H, m), 6.80 (1H, d, 3 J = 8.3 Hz}, 2.83 (2H, t, J = 6.9 Hz), 2.54 (3H, s), 1.84 (2H, t, J = 6.9 4 Hz), 1.36 (6H, s). Anal. (C,3H,6O2} C, H.
Ethyl 2,2-dimethylchroman-6-carbo Ix~(Compound 216) 6 A solution of 6-acetyl-2,2-dimethylchroman (Compound 215, 7 2.15 g, 10.5 mmol) and NaOH (2.0 g, 50 mmol) in dioxane (50 mL) and 8 bleach (50 mL, 5.25% NaOCI) was heated at 65 ~C for 4 days. Upon 9 cooling to room temperature Na2S03 was added until an aliquot of this solution remained colorless color when treated with of one drop of a 11 solution of I2 in CC14. The solution was acidified with H2S04 (pH 4) 12 and extracted with ether. The combined organic layers were dried 13 (Na,S04) and concentrated under reduced pressure. Column 14 chromatography (EtOAc : hexanes 1:3) afforded 1.98 g (92%) of 2,2-dimethylchroman-6-carboxylic acid as a light tan solid. 1H NMR 8 7.78 16 (2H, m), 6.82 (1H, d, J = 8.3 Hz), 2.83 (2H, t, J = 6.7, Hz), 1.85 (2H, t, 17 J = 6.7 Hz), 1.37 {6H, s).
18 To a solution of this solid (1.64 g, 7.95 mmol) in EtOH (50 mL) 19 was added HzS04 (0.5 mL). The mixture was heated overnight at 95 ~C, cooled to room temperature and concentrated under reduced pressure.
21 Column chromatography (EtOAc : hexanes, 1:5) afforded 1.91 g (100%) 22 of the title compound as a colorless solid. 1H NMR b 7.79 (2H, m), 23 6.79 (2H, d, J = 8.3 Hz), 4.34 (2H, q, J = 7.2 Hz), 2.82 (2H, t, J = 6.7 24 Hz), 1.84 {2H, t, J = 6.7 Hz), 1.38 (3H, t, J = 7.2 Hz), 1.36 {6H, s).
Anal (C,4H,803 ~ 0.25 H20) C, H.
26 Ethyl 8-bromo-2.2-dimethylchroman-6-carbo lx~e i(Compound 217) 27 To a solution of ethyl 2,2-dimethylchroman-6-carboxylate 28 (Compound 216, 500 mg, 2.14 mmol) in HOAc (4 mL) was added Brz 29 (0.11 mL, 2.14 mmol). The reaction mixture was stirred at room temperature overnight and then a stream of air was passed through the ~..r~...~.~...._... _-.._...._....~.w.....~.... _ ~..w~..~.......a._._ .__.,w.-w..-~.-_W..~..

1 reaction mixture to remove the excess Br2. Removal of the solvent 2 under reduced pressure and column chromatography using the residue 3 (EtOAc:hexanesl:9) afforded 741 mg (100%) of the title compound as 4 an oil. 'H NMR b 8.05 (1H, d, J = 2.2 Hz), 7.73 (1H, d, J = 2.2 Hz), 4.34 (2H, q, J =7.1 Hz), 2.84 (2H, t, J = 6.7 Hz), 1.85 (2H, t, J = 6.7 6 Hz), 1.40 (6H, s), 1.38 (3H, t, J = 7.2 Hz). Anal (C,4H"BrO~ ~ 0.2 7 H20) C, H.
8 Ethvl 8-bromo-2,2-dimethyl-4-chromanone-6-carbo lx ~~ate (Compound to To a solution of HOAc {15 mL) and Ac20 (7.5 mL) at 0 ~C was 11 added Cr03 (1.18 g, 12 mmol) in small portions. This solution was 12 stirred for 15 minutes, diluted with benzene { 10 mL), and a solution of 13 ethyl 8-bromo-2,2-dimethylchroman-6-carboxylate (Compound 217, 741 14 mg, 2.38 mmol) in benzene (5 mL) was slowly added over several minutes. After stirring at 0 ~C for 4 hours, the reaction mixture was 16 poured over ice, extracted with EtOAc, the combined organic layers 17 were dried {Na2S04), and concentrated under reduced pressure to give 18 an oil. Column chromatography (EtOAc : hexanes, 1:9) yielded 589 mg 19 (76%) of the title compound as a colorless solid. IH NMR 8 8.5 (1H, d, 2o J = 2.1 Hz), 8.40 (1H, d, J = 2.1 Hz), 4.37 (2H, q, J =7.1 Hz), 2.80 21 (2H, s), 1.54 (6H, s), 1.40 (3H, t, J = 7.1 Hz); Anal. (C,4H,SBr04) C, H, 22 N.
23 Ethvl 8-bromo-2,2-dimeth ~~l-4~4-methYl~henyl)-4(2H~-chroman-6-24 carboxylate (Compound 219 To a solution of ethyl 8-bromo-'?,2-dimethyl-4-chromanone-6-26 ~;arboxylate (Compound 218, 589 mg, 1.81 mmol) in THF (20 mL) 27 under argon gas at -78 ~C was added p-tolylmagnesium bromide (2.16 28 ml, 2.72 mmol, 1 M solution in ether). The reaction mixture was slowly 29 warmed to room temperature and stirred overnight. Saturated NH4Cl solution was added to the reaction at 0 ~C and the resulting mixture 1 extracted with ether. The combined organic layers were washed with 2 saturated aqueous NaCI, dried over Na2S04, and concentrated under 3 reduced pressure. Column chromatography (EtOAc:hexanes, 1:3) 4 afforded 297 mg of a mixture of the title compound and the intermediate tertiary alcohol as an oil. This mixture was combined with 6 p-TsOH (20 mg) in dry toluene (15 mL). and heated at 110 ~C for 30 7 minutes. Upon cooling to room temperature the solvent was removed 8 under reduced pressure and the product 182 mg (25%) was isolated by 9 column chromatography (EtOAc : hexane 1:3). 'H NMR 8 8.1 (1H, d, J
= 1.7 Hz), 7.68 (1H, d, J = 1.7 Hz), 7.22 (4H, s), 5.67 (1H, s), 4.29 (2H, 11 q, J = 7.1 Hz), 2.41(3H, s), 1.56 (6H, s), 1.33 (3H, t, J = 7.1 Hz); MS
12 (EI) m/e 402, 400, 387, 385, 359, 357. Anal (CZ,Hz,Br03 ~ 0.45 H,O) C, 13 H.
14 8-Bromo-2,2-dimethyl-4-(4-methylphenvl~2Hl-chroman-6-carboxylic acid (Compound 220) 16 To a solution of ethyl 8-bromo-2,2-dimethyl-4-(4-methylphenyl)-17 4(21-chroman-6-carboxylate (Compound 219, I89 mg, 0.51 mmol) in 18 EtOH (15 mL) and THF (10 mL) was added 20% aqueous NaOH (2 19 mL). The reaction mixture was warmed to 45 ~C for 2 hours, cooled to room temperature and acidified to pH 4 with 10% aqueous HCI.
21 Extraction (EtOAc), drying (MgS04), and concentration under reduced 22 pressure afforded the title compound {171 mg) (98%) as a colorless 23 solid. 1H NMR 8 8.15 (d, J = 2.0 Hz, 1H), 7.71 (d, J = 2.0 Hz, 1H), 24 7.21 (m, 4H), 5.69 (s, 1H), 2.42(s, 3H), 1.58 (s, 6H).
Ethvl 2-fluoro-4-~~(8-bromo-2 2-dimethyl-4 ~4-meth~phenyl)-6-26 chromanyl) carbon~~amino~-benzoate~Compound 221) 27 To a solution of 8-bromo-2,2-dimethyl-4-(4-methylphenyl)-4(2I~-28 chroman-6-carboxylic acid (Compound 220, 100 mg, 0.29 mmol) in 29 CHZC12 (8 mL) was added DMAP (87 mg, 0.69 mmol), ethyl 4-amino-2-fluoro benzoate (53 mg, 0.29 mmol) and EDC (72 mg, 0.37 mmol). The ...__-....a. _ , ~. _ ..m~~. ~. _ .- ....~..~~_._~..,..._.

1 reaction mixture was stirred at room temperature overnight and then 2 concentrated to dryness. Column chromatography (EtOAc : hexanes, 3 1:3) afforded 98 mg (63%) of the title compound as a colorless solid.
4 IHNMR87.99(lH,bs),7.89(lH,t,J=7.SHz),7.88(lH,d,J=2.1 Hz),7.65{lH,dd,J=12.8,2.1Hz),7.25(lH,dd,J=7.SHz,2.lHz), 6 7.19 (4H, s), 5.70 (1H, s), 4.36 {2H, q, J = 7.2 Hz), 2.38 (3H, s), 1.56 7 (6H, s), 1.39 (3H, t, J = 7.2 Hz). MS (EI) m/e 537, 537, 524, 526, 357, 8 355, 312. Anal (C28H25BrFN04) C, H, N.
9 2-Fluoro-4-j[j8-bromo-2,2-dimethyl-4-~4-meth l~phenyl)-6-l0 chromanyl)carbons]amino]-benzoic acid {Compound 204) 11 To a solution of ethyl 2-fluoro-4-[[{8-bromo-2,2-dimethyl-4-(4-12 methylphenyl)-6-chromanyl)carbonyl]amino]-benzoate (Compound 221, 13 135 mg, 0.25 mmol) in EtOH (5 mL) was added 20% aqueous NaOH (2 14 mL). The reaction mixture was stirred at room temperature overnight and acidified to pH 4 with 10% aqueous HCI. The EtOH was removed 16 under reduced pressure and ethyl acetate and water were added to the 17 residue. The organic layer was separated and washed with saturated 18 NaHC03, saturated aqueous NaCI, and dried over MgS04.
19 Concentration under reduced pressure afforded 110 mg (86%)of the title compound as a colorless solid. 1H NMR 8 (acetone-db) 8.09 (1H, 21 d, J = 2.1 Hz), 7.91 (1H, t, J = 7.5 Hz), 7.68 (1H, d, J = 2.1 Hz), 7.83 22 (1H, dd, J= 12.8, 2.1 Hz), 7.50 (1H, dd, J = 7.5 Hz, 2.1 Hz), 7.27 (4H, 23 s), 5.87 (1H, s), 2.37 (3H, s), 1.56 (6H, s); MS (EI) m/e 511, 509, 496, 24 494 , 452, 450, 357, 355, 312. Anal. (C26H21BrFNO4 ~ 0.6 H20) C, H, N.
26 ~.__~y12,6-difluoro-4~1(8-bromo-2.2-dimethvl-4-(4-methvlnhenvll-6-27 chroman)carbonylla_, mino]-benzoate i(Compound 222) 28 Using the same procedure as for the synthesis of ethyl 2-fluoro-29 4-[[(8-bromo-2,2-dimethyl-4-(4-methylphenyl)-6-chromanyl) carbonyl]amino]-benzoate (Compound 221), the reaction of 8-bromo-1 2,2-dimethyl-4-(4-methylphenyl)-4{21~-chroman-6-carboxylic acid 2 (Compound 220, 100 mg, 0.29 mmol) with ethyl 2,6-difluoro-4-3 aminobenzoate in the presence of DMAP and EDC in CH2C12 4 afforded 53 mg (33%) of the title compound as a colorless oil. ~H
NMR 8 7.87 (1H, d, J = 2.1 Hz), 7.47 (1H, d, J = 2.1 Hz), 7.28 (2H, d, 6 J = 8.7 Hz), 7.19 (4H, m), 5.70 (1H, s), 4.38 (2H, q, 3 = 7.2 Hz), 2.41 7 (3H, s), 1.57 (6H, s), 1.38 (3H, t, J = 7.2 Hz). MS (EI) m/e 557, 555, 8 542, 540, 448, 446. Anal. (C28H24BrF2NO4) C, H, N.
9 2,6-Difluoro-4-(~(8-bromo-2 2-dimethyl-4-~4-meth,~~iphen~y-6-chroman)carbon~~amino~-benzoic acid yCompound 205 11 Using the same procedure as for the synthesis of 2-fluoro-4-(((8-12 bromo-2,2-dimethyl-4-(4-methylphenyl)-6-chromanyl)carbonyl]amino]-13 benzoic acid {Compound 204), treatment of ethyl 2,6-difluoro-4-(((8-14 bromo-2,2-dimethyl-4-(4-methylphenyl)-6-chroman)carbonyl]amino]-benzoate (Compound 222, 53 mg, 0.1 mmol) with aqueous NaOH in 16 EtOH afforded 35 mg {70%) of the title compound as a colorless solid.
17 ~H NMR 8 (acetone-d6) 9.93 (1H, bs), 8.07 (1H, d, J = 2.1 Hz), 7.66 18 ( 1 H, d, J = 2.1 Hz), 7.54 (2H, d, J = 9.9 Hz), 7.27 (4H, s), 5.88 ( 1 H, s), 19 2.38 (3H, s), 1.56 (6H, s). MS (EI) m/e 514, 512, 485, 483, 470, 468.
Anal. (C26H2oBrF2N04) C, H, N.
21 Method of Potentiating Nuclear Receptor A og nists 22 Overview and introduction 23 We have discovered that a subset of retinoid antagonists which 24 exhibit negative hormone activity can be used for potentiating the biological activities of other retinoids and steroid receptor superfamily 26 hormones. These other retinoids and steroid receptor superfamily 27 hormones can be either endogenous hormones or pharmaceutical 28 agents. Thus, for example, when used in combination with a retinoid 29 negative hormone, certain activities of pharmaceutical retinoid agonists can be rendered more active in eliciting specific biological effects.
. .... . ...._..... , ..~....-.-... _.

1 Advantageously, this combination approach to drug administration can 2 minimize undesirable side effects of pharmaceutical retinoids because 3 lower dosages of the pharmaceutical retinoids can be used with improved effectiveness.
More particularly, we have discovered that AGN 193109, a 6 synthetic retinoid having the structure shown in Figure 1, exhibits 7 unique and unexpected pharmacologic activities. AGN 193109 exhibits 8 high affinity for the RAR subclass of nuclear receptors without 9 activating these receptors or stimulating transcription of retinoid responsive genes. Instead, AGN 193109 inhibits the activation of RARs 11 by retinoid agonists and therefore behaves as a retinoid antagonist.
12 Additionally, we have discovered that retinoid negative hormones 13 can be used without coadministration of a retinoid agonist or steroid 14 hormone to control certain disease symptoms. More specifically, the retinoid negative hormone disclosed herein can down-regulate the high 16 level basal transcription of genes that are responsive to unliganded 17 RARs. If, for example, uncontrolled cellular proliferation results from 18 the activity of genes responsive to unliganded RARs, then that gene 19 activity can be reduced by the administration of a retinoid negative hormone that inactivates RARs. Consequently, cellular proliferation 21 dependent on the activity of unliganded RARs can be inhibited by the 22 negative hormone. Inhibition of unliganded RARs cannot be achieved 23 using conventional antagonists.
24 Significantly, we have discovered that AGN 193109 can both repress RAR basal activity and can sometimes potentiate the activities 26 of other retinoid and steroid receptor superfamily hormone agonists. In 27 the context of the invention, a hormone agonist is said to be potentiated 28 by a negative hormone such as AGN 193109 if, in the presence of the 29 negative hormone, a reduced concentration of the agonist elicits substantially the same quantitative response as that obtainable with the 1 agonist alone. The quantitative response can, for example, be measured 2 in a reporter gene assay in vitro. Thus, a therapeutic retinoid that elicits 3 a desired response when used at a particular dosage or concentration is 4 potentiated by AGN 193109 if, in combination with AGN 193109, a lower dosage or concentration of the therapeutic retinoid can be used to 6 produce substantially the same effect as a higher dosage or 7 concentration of the therapeutic retinoid when that therapeutic retinoid 8 is used alone. The list of agonists that can be potentiated by 9 coadministration with AGN 193109 includes RAR agonists, vitamin D
receptor agonists, glucocorticoid receptor agonists and thyroid hormone 11 receptor agonists. More particularly, specific agonists that can be 12 potentiated by coadministration include: ATRA, 13-cis retinoic acid, the 13 synthetic RAR agonist AGN 191183, 1,25-dihydroxyvitamin D3, 14 dexamethasone and thyroid hormone (3,3',5-triiodothyronine). Also disclosed herein is a method that can be used to identify other 16 hormones that can be potentiated by coadministration with AGN
17 193109.
18 Thus, AGN 193109 behaves in a manner not anticipated for a 19 simple retinoid antagonist, but as a negative hormone that can potentiate the activities of various members of the family of nuclear 21 receptors. We also disclose a possible mechanism that can account for 22 both negative hormone activity and the ability of AGN 193109 to 23 potentiate the activities of other nuclear receptor ligands. This 24 mechanism incorporates elements known to participate in retinoid-dependent signalling pathways and additionally incorporates a novel 26 negative regulatory component.
27 Those having ordinary skill in the art will appreciate that RARs, 28 which are high affinity targets of AGN 193109 binding, are transcription 29 factors that regulate the expression of a variety of retinoid responsive genes. Cis-regulatory DNA binding sites for the RARs have been _._.. ~_...w ~..... _ ~ , .. _.. ._... _ ... .._...._._.,~.~.....

1 identified nearby genes that are transcriptionally regulated in a retinoid-2 dependent fashion. RAR binding to such DNA sites, known as retinoic 3 acid response elements (RAREs), has been well defined. Importantly, 4 the RAREs bind heterodimers consisting of one RAR and one RXR.
The RXR component of the heterodimer functions to promote a high 6 affinity interaction between the RAR/RXR heterodimer and the RARE
7 (Mangelsdorf et al. The Retinoid Receptors in The Retinoids: BioloQV, 8 Chemistr~r and Medicine, 2nd edition, eds. Sporn et aL, Raven Press, 9 Ltd., New York 1994, the disclosure of which is hereby incorporated by reference).
11 As detailed below, our findings related to the negative hormone 12 activity of AGN 193109 are consistent with a mechanism involving the 13 interaction of a putative Negative Coactivator Protein (NCP) with the 14 RAR. According to the proposed mechanism, this interaction is stabilized by AGN 193109.
16 Our results further indicated that AGN 193109 can modulate 17 intracellular availability of NCP for interaction with nuclear receptors 18 other than RARs that are occupied by AGN 193109. It follows that 19 AGN 193109 can potentiate transcriptional regulatory pathways involving nuclear receptors that share with the RARs the ability to bind 21 the NCP. In this regard, AGN 193109 exhibits the ability to modulate a 22 variety of nuclear receptor pathways, an activity that would not be 23 predicted for a conventional retinoid antagonist. Accordingly, AGN
24 193109 is useful as an agent for potentiating the activity of nuclear receptor ligands, including both endogenous hormones and prescribed 26 therapeutics. This specific embodiment illustrates the more general 27 principle that any nuclear receptor negative hormone will potentiate the 28 activity of other nuclear receptors that competitively bind the NCP.
29 Although other materials and methods similar or equivalent to those described herein can be used in the practice or testing of the I present invention, the preferred methods and materials are now 2 described. General references for methods that can be used to perform 3 the various nucleic acid manipulations and procedures described herein 4 can be found in Molecular Cloning. A Laboratory Manual (Sambrook et al. eds. Cold Spring Harbor Lab Publ. 1989) and Current Protocols in 6 Molecular Biology (Ausubel et al. eds., Greene Publishing Associates and 7 Wiley-Interscience 1987). The disclosures contained in these references 8 are hereby incorporated by reference. A description of the experiments 9 and results that led to the creation of the present invention follows.
Example 6 describes the methods used to demonstrate that AGN
11 193109 bound each of three RARs with high affinity but failed to 12 activate retinoid dependent gene expression.
13 Example 6 14 AGN 193109 Binds RARs With High Affinity But Does'Not Transactivate Retinoid-Devendent Gene Expression t 6 Human RAR-a, RAR-~i and RAR-y receptors were separately 17 expressed as recombinant proteins using a baculovirus expression system 18 essentially according to the method described by Allegretto et al. in J.
19 Biol. Chem. 268:26625 (1993). The recombinant receptor proteins were separately employed for determining AGN 193109 binding affinities 21 using the [3H]-ATRA displacement assay described by Heyman et al. in 22 Cell 68:397 (1992). Dissociation constants (Kds) were determined 23 according to the procedure described by Cheng et al. in Biochemical 24 Pharmacology 22:3099 (1973).
AGN 193109 was also tested for its ability to transactivate RARs 26 in CV-1 cells transiently cotransfected with RAR expression vectors and 27 a retinoid responsive reporter gene construct. Receptor expression 28 vectors pRShRAR-a (Giguere et al. Nature 330:624 (1987)), pRShRAR-29 ~i (Benbrook et al. Nature 333:669 (1988)) and pRShRAR-~y (Ishikawa et al. Mol. Endocrinol. 4:837 (1990)) were separately cotransfected with 1 the OMTV-TREp-Luc reporter plasmid. Use of this luciferase reporter 2 plasmid has been disclosed by Heyman et al. in Cell 68:397 (1992). The 3 OMTV-TREp-Luc plasmid is essentially identical to the OMTV-TREp-4 CAT reporter construct described by Umesono et al. in Nature 336:262 ( 1988), except that the chloramphenicol acetyltransferase (CAT) 6 reporter gene was substituted by a polynucleotide sequence encoding 7 firefly luciferase. Transfection of green monkey CV-1 cells was carried 8 out using the calcium phosphate coprecipitation method described in 9 Molecular Cloning: A Laboratory Manual (Sambrook et al. eds. Cold Spring Harbor Lab Publ. 1989). CV-1 cells were plated at a density of 11 4 X 104/well in 12 well multiwell plates and transiently transfected with 12 a calcium phosphate precipitate containing 0.7 ~cg of reporter plasmid 13 and 0.1 ~cg of receptor plasmid according to standard laboratory 14 procedures. Cells were washed after 18 hours to remove the precipitate and refed with Dulbecco's modified Eagle's medium (DMEM) {Gibco), 16 containing 10% activated charcoal extracted fetal bovine serum (Gemini 1'7 Bio-Products). Cells were treated with vehicle alone (ethanol) or AGN
18 193109 (10-9 to 10-6 M) for 18 hours. Cell lysates were prepared in 0.1 19 M KP04 (pH 7.8), 1.0% TRITON X-100, 1.0 mM DTT, 2 mM EDTA.
Luciferase activity was measured as described by de Wet et al. in Mol.
2I Cell. Biol. 7:725 (1987), using firefly luciferin (Analytical Luminescence 22 Laboratory) and an EG&G Berthold 96-well plate luminometer.
23 Reported luciferase values represented the mean ~ SEM of triplicate 24 determinations.
The results presented it 'f able 11 indicated that AGN 193109 26 bound each of RAR-a, RAR-~G and RAR-y with l~~gh affinity, but did 27 not activate retinoid-dependent gene expression. More specifically, 28 AGN 193109 bound each of the three receptors with Kd values in the 2 29 - 3 nM range. Despite this tight binding, AGN 193109 failed to activate gene expression when compared with inductions stimulated by ATRA.

1 Accordingly, the half-maximal effective concentration of AGN 193109 2 (ECSO) was unmeasurable. Although not presented in the Table, we also 3 found that AGN 193109 had no measurable affinity for the RXRs.

AGN 193109 Binding and Transactivation of the RARs 6 RAR-a RAR-~3 RAR-y 7 ECSO (nM) No Activity No Activity No Activity 8 I~, (nM) 2 2 3 l0 Example describes the methods used to demonstrate that AGN

11 193109 is an antagonist of ATRA dependent gene expression.
12 Example 7 13 AGN 193109-Dependent Inhibition of RAR Transactivation 14 b~ ATRA
The ability of AGN 193109 to antagonize ATRA mediated RAR
16 activation was investigated in CV-1 cells cotransfected by the calcium 17 phosphate coprecipitation method of Sambrook et al. (Molecular 18 Cloning: A Laboratory Manual Cold Spring Harbor Lab Publ. 1989).
19 Eukaryotic expression vectors pRShRAR-a (Giguere et al. Nature 330:624 (1987)), pRShRAR-(3 (Benbrook et al. Nature 333:669 (1988)) 21 and pRShRAR-y (Ishikawa et al. Mol. Endocrinol. 4:837 (1990)) were 22 cotransfected with the 0-MTV-Luc reporter plasmid described by 23 Hollenberg et al. (Cell 55:899 (1988)). Notably, the reporter plasmid 24 contained two copies of the TRE-palindromic response element.
Calcium phosphate transfections were carried out exactly as described in 26 Example 6. Cells were dosed with vehicle alone (ethanol), ATRA (10-9 27 to 10-6 M), AGN 193109 (10-9 to 10-6 M), or 10-$ M ATRA in 28 combination with AGN 193109 (10-9 to 10-6 M) for 18 hours. Cell 29 lysates and luciferase activity measurements were also performed as in 3o Example 6.
~w~ . _ T ..

1 The results of these procedures are presented in Figures 2A
2 through 2F where luciferase values represent the mean ~ SEM of 3 triplicate determinations. More specifically, the results presented in 4 Figures 2A, 2C and 2E indicated that stimulation of transfected cells with ATRA led to dose responsive increases in luciferase activity. This 6 confirmed that ATRA activated each of the three RARs in the 7 experimental system and provided a comparative basis for detecting the 8 activity of an antagonist. The graphic results presented in Figures 2B, 9 2D and 2F indicated that cotreatment of transfected cells with 10 nM
ATRA and increasing concentrations of AGN 193109 led to an 11 inhibition of luciferase activity. In particular, equal doses of AGN
12 193109 and ATRA gave greater than 50% inhibition relative to ATRA
13 alone for all three RAR subtypes. Comparison of the ATRA dose 14 response in the presence of different concentrations of AGN 193109 indicated that ATRA was competitively inhibited by AGN 193109.
16 Notably, the horizontal axes on all of the graphs shown in Figure 2 17 represents the log of the retinoid concentration. These results proved 18 that AGN 193109 was a potent RAR antagonist.
19 We next performed experiments to elucidate the mechanism underlying the antagonist activity of AGN 193109. Those having 21 ordinary skill in the art will appreciate that nuclear receptor activation is 22 believed to involve a conformational change of the receptor that is 23 induced by ligand binding. Indeed, the results of protease protection 24 assays have confirmed that nuclear hormone agonists and antagonists cause receptor proteins to adopt different conformations (Keidel et al.
26 l~i~l. Cell. Biol. 14:287 (1994); ~ .n et al. J. Bcul. Chem. 267:19513 27 (1992)). We used such an assay to determine if AGN 193109 and 28 ATRA caused RAR-a to adopt different conformations. AGN 193583, 29 an RAR-a-selective antagonist, was included as a positive control that is known to confer an antagonist-specific pattern of protease sensitivity.

1 Example 8 describes one method that was used to detect 2 conformational changes in RAR-a resulting from AGN 193109 binding.
3 As presented below, the results of this procedure unexpectedly indicated 4 that AGN 193109 led to a pattern of trypsin sensitivity that was substantially identical to that induced by ATRA, an RAR agonist, and 6 unlike that induced by a model RAR antagonist. This finding suggested 7 that AGN 193109 possessed properties distinct from other retinoid 8 antagonists.
Example $
l0 Protease Protection Anal, 11 A plasmid constructed in the vector pGEM3Z (Pharmacia) and 12 containing the RAR-a cDNA (Giguere et al. Nature 330:624 (1987)), 13 was used in connection with the TNT-coupled reticulocyte lysate in vitro 14 transcription-translation system (Promega) to prepare [35S]-methionine labeled RAR-«. Limited proteolytic digestion of the labeled protein 16 RAR-a was carried out according to the method described by Keidel et 17 al. in Mol. Cell. Biol. 14:287 (1994). Aliquots of reticulocyte lysate 18 containing [35S]-methionine labeled RAR-a were incubated with either 19 ATRA, AGN 193583 or AGN 193109 on ice for 45 minutes in a total volume of 9 ~,1. The retinoid final concentration for all trials was 100 21 nM for ATRA and AGN 193109, and 1000 nM for AGN 193583. The 22 difference between the final concentrations of the retinoids was based 23 on the approximate 10-fold difference in relative affinities of ATRA and 24 AGN 193109 (having Kd at RAR-a of 2 and 10 nM, respectively) and AGN 193583 (having Kd at RAR-a of ? 100 nM). After ligand binding, 26 1 ,ul of appropriately concentrated trypsin was added to the mixture to 27 give final concentrations of 25, 50 or 100 ~.~,g/ml. Samples were 28 incubated at room temperature for 10 minutes and trypsin digestion 29 stopped by addition of SDS-sample buffer. Samples were subjected to polyacrylamide gel electrophoresis and autoradiographed according to _.~..~.~-_W. . ._ _. ..r.e~.-.~~....w. .

1 standard procedures.
2 Both the agonist and antagonist led to distinct patterns of trypsin 3 sensitivity that were different from the result obtained by digestion of 4 the unliganded receptor. Autoradiographic results indicated that trypsin concentrations of 25, 50 and 100 ~cg/ml completely digested the 6 radiolabeled RAR-a in 10 minutes at room temperature in the absence 7 of added retinoid. Prebinding of ATRA led to the appearance of two 8 major protease resistant species. Prebinding of the RAR-a-selective 9 antagonist AGN 193583 gave rise to a protease resistant species that l0 was of lower molecular weight than that resulting from ATRA
11 prebinding. This result demonstrated that a retinoid agonist and 12 antagonist led to conformational changes detectable by virtue of altered 13 trypsin sensitivities. Surprisingly, prebinding of AGN 193109 gave rise 14 to a protease protection pattern that was indistinguishable from that produced by prebinding of ATRA.
16 The results presented above confirmed that AGN 193109 bound 17 the RAR-a and altered its conformation. Interestingly, the nature of 18 this conformational change more closely resembled that which resulted 19 from binding of an agonist (ATRA) than the alteration produced by antagonist (AGN 193583) binding. Clearly, the mechanism of AGN
21 193109 dependent antagonism was unique.
22 We considered possible mechanisms that could account for the 23 antagonist activity of AGN 193109. In particular, we used a standard 24 gel shift assay to test whether AGN 193109 perturbed RAR/RXR
heterodimer formation or inhibited the interaction between RAR and 26 its cognate DNA binding site.
27 Example 9 describes a gel electrophoretic mobility-shift assay 28 used to demonstrate that AGN 193109 neither inhibited RAR/RXR
29 dimerization nor inhibited binding of dimers to a target DNA.

1 Example 9 2 Gel Shift Anal Isis 3 In vitro translated RAR-a was produced essentially as described 4 under Example 8, except that 35S-labeled methoinine was omitted. In vitro translated RXR-a was similarly produced using a 6 pBluescript(II)(SK)-based vector containing the RXR-a cDNA
7 described by Mangelsdorf, et al. in Nature 345:224-229 (1990) as the 8 template for generating in vitro transcripts. The labeled RAR-a and 9 RXR-a, alone or in combination, or prebound with AGN 193109 ( 10-6 l0 M) either alone or in combination, were allowed to interact with an 11 end-labeled DR-5 RARE double-stranded probe having the sequence 12 5'-TCAGGTCACCAGGAGGTCAGA-3' (SEQ ID NO:1). The binding 13 mixture was electrophoresed on a non-denaturing polyacrylamide gel 14 and autoradiographed according to standard laboratory procedures. A
single retarded species appearing on the autoradiograph that was 16 common to all the lanes on the gel represented an undefined probe-17 binding factor present in the reticulocyte lysate. Only the RAR/RXR
18 combination gave rise to a retinoid receptor-specific retarded species.
19 Neither RAR alone nor RXR alone bound the probe to produce this shifted species. The presence of AGN 193109 did not diminish this 21 interaction.
22 These results indicated that AGN 193109 did not substantially 23 alter either the homo- or hetero-dimerization properties of RAR-a.
24 Further, AGN 193109 did not inhibit the interaction of receptor dimers with a DNA segment containing the cognate binding site.
26 In view of the unique properties which characterized AGN
27 193109, we proceeded to investigate whether this antagonist could 28 additionally inhibit the activity of unliganded RARs. The 29 receptor/reporter system used to make this determination 3o advantageously exhibited high level constitutive activity in the absence _..__ ..~...._... ~....~..-o . . . . _ _W ~.~..__ 1 of added retinoid agonist. More specifically, these procedures employed 2 the ER-RAR chimeric receptor and ERE-tk-Luc reporter system. The 3 ERE-tk-Luc plasmid includes the region -397 to -87 of the estrogen 4 responsive 5'-flanking region of the Xenopus vitellogenin A2 gene, described by Klein-Hitpass, et al. in Cell 46:1053-1061 (1986), ligated 6 upstream of the HSV thymidine kinase promoter and luciferase reporter 7 gene of plasmid tk-Luc. The ER-RAR chimeric receptors consisted of 8 the estrogen receptor DNA binding domain fused to the "D-E-F"
9 domain of the RARs. Those having ordinary skill in the art appreciate this "D-E-F" domain functions to bind retinoid, to provide a retinoid 11 inducible transactivation function and to provide a contact site for 12 heterodimerization with RXR. Thus, luciferase expression in this 13 reporter system was dependent on activation of the transfected chimeric 14 receptor construct.
Example 10 describes the method used to demonstrate that AGN
16 193109 inhibited basal gene activity attributable to unliganded RARs.
17 These procedures were performed in the absence of added retinoid 18 agonist. The results presented below provided the first indication that 19 AGN 193109 exhibited negative hormone activity.
Example 10 21 Repression of Basal Gene Activity of a Retinoid-Regulated 22 Reporter in Transiently Cotransfected Cell Lines 23 CV-1 cells were co-transfected with the ERE-tk-Luc reporter 24 plasmid and either ER-RAR-a, ER-RAR-R or ER-RAR-y expression plasmids. The ERE-tk-Luc plasmid contained the estrogen-responsive 26 promoter element of the Xercopus laevis vitellogenin A2 gene and was 27 substantially identical to the reporter plasmid described by Klein-Hitpass 28 et al. in Cell 46:1053 (1986), except that the CAT reporter gene was 29 substituted by a polynucleotide sequence encoding luciferase. The ER-RAR-a, ER-RAR-/3 and ER-RAR-y chimeric receptor-encoding 1 polynucleotides employed in the co-transfection have been described by 2 Graupner et al. in Biochem. Biophys. Res. Comm. 179:1554 (1991).
3 These polynucleotides were ligated into the pECE expression vector 4 described by Ellis et al. in Cell 45:721 (1986) and expressed under transcriptional control of the SV-40 promoter. Calcium phosphate 6 transfections were carried out exactly as described in Example 6 using 7 0.5 ~cg/well of reporter plasmid and either 0.05 ,ug, 0.10 pug or 0.2 ~cg/well 8 of receptor plasmid. Cells were dosed with vehicle alone (ethanol), 9 ATRA (10-9 to 10-6 M), or AGN 193109 (10-9 to 10-6 M) for 18 hours.
l0 Cell lysates and luciferase activity measurements were performed as 1 I described in Example 6.
12 The results presented in Figures 3A, 4A and 5A confirmed that 13 ATRA strongly induced luciferase expression in all transfectants. Basal 14 level expression of luciferase for the three transfected chimeric RAR
isoforms ranged from approximately 7,000 to 40,000 relative light units 16 (rlu) and was somewhat dependent on the amount of receptor plasmid 17 used in the transfection. Thus, the three chimeric receptors were 18 activatable by ATRA, as expected. More specifically, all three receptors 19 bound ATRA and activated transcription of the luciferase reporter gene harbored on the ERE-tk-Luc plasmid.
21 Figures 3B, 4B and 5B present AGN 193109 dose response curves 22 obtained in the absence of any exogenous retinoid agonist.
23 Interestingly, ER-RAR-a (Figure 3B) was substantially unaffected by 24 AGN 193109, while the ER-RAR-~3 and ER-RAR-y chimeric receptors (Figures 4B and 5B, respectively) exhibited an AGN 193109 dose 26 responsive decrease in luciferase reporter activity.
27 We further investigated the negative hormone activity of AGN
28 193109 by testing its ability to repress gene expression mediated by a 29 chimeric RAR-y receptor engineered to possess a constitutive transcription activator domain. More specifically, we used a 1 constitutively active RAR-y chimeric receptor fused to the acidic 2 activator domain of HSV VP-16, called RAR-y-VP-16, in two types of 3 luciferase reporter systems. The first consisted of the ERE-tk-Luc 4 reporter cotransfected with ER-RARs and ER-RXR-a. The second utilized the MTV-TREp-Luc reporter instead of the ERE-tk-Luc 6 reporter.
7 Example 11 describes the method used to demonstrate that AGN
8 193109 could suppress the activity of a transcription activator domain of 9 an RAR. The results presented below proved that AGN 193109 could suppress RAR-dependent gene expression in the absence of an agonist 11 and confirmed that AGN 193109 exhibited negative hormone activity.
12 Example 11 13 Repression of RAR-VP-16 Activity in Transiently 14 Transfected Cells CV-1 cells were transiently cotransfected according to the calcium 16 phosphate coprecipitation technique described under Example 6 using 17 0.5 ,ug/well of the ERE-tk-Luc luciferase reporter plasmid, 0.1 ,ug/well of 18 the ER-RXR-a chimeric reporter expression plasmid, and either 0 ~cg or 19 0.1 ~,g/well of the RAR-y-VP-16 expression plasmid. The chimeric receptor ER-RXR-a consisted of the hormone binding domain (amino 21 acids 181 to 458) of RXR-a (Mangelsdorf, et al. Nature 345:224-229 22 (1990)) fused to the estrogen receptor DNA binding domain (Graupner, 23 et al. Biochem. Biophys. Res. Comm. 179:1554 (1991)) and was expressed 24 from the SV-40 based expression vector pECE described by Ellis, et al.
in Cell 45:721 (1986). RAR-y-VP-16 is identical to the VP16RAR-yl 26 expression plasmid described by Nagpal et al. in EMBO J. 12:2349 27 (1993), the disclosure of which is hereby incorporated by reference, and 28 encodes a chimeric protein having the activation domain of the VP-16 29 protein of HSV fused to the amino-terminus of full length RAR-~y. At eighteen hours post-transfection, cells were rinsed with phosphate 1 buffered saline (PBS) and fed with DMEM (Gibco-BRL) containing 2 10% FBS (Gemini Bio-Products) that had been extracted with charcoal 3 to remove retinoids. Cells were dosed with an appropriate dilution of 4 AGN 193109 or ATRA in ethanol vehicle or ethanol alone for 18 hours, then rinsed with PBS and lysed using 0.1 M KP04 (pH 7.8), 1.0%
6 TRITON X-100, 1.0 mM DTT, 2 mM EDTA. Luciferase activity was 7 measured according to the method described by de Wet, et al. in Mol.
8 Cell. Biol. 7:725 (1987), using firefly luciferin (Analytical Luminescence 9 Laboratory) and an EG&G Berthold 96-well plate luminometer.
Luciferase values represented the mean ~ SEM of triplicate 11 determinations.
12 As shown in Figure 6, CV-1 cells transfected with the ERE-tk-13 Luc reporter construct and the ER-RAR-a chimeric expression plasmid 14 exhibited a weak activation of luciferase activity by ATRA, likely due to isomerization of ATRA to 9C-RA, the natural ligand for the RXRs 16 (Heyman et al. Cell 68:397 (1992). Cells transfected with the same 17 mixture of reporter and chimeric receptor plasmids but treated with 18 AGN 193109 did not exhibit any effect on Iuciferase activity. As AGN
19 193109 does not bind to the RXRs, this latter result was expected. CV-1 cells similarly transfected with the ERE-tk-Luc reporter but with 21 substitution of an ER-RAR chimeric receptor expression plasmid for 22 ER-RXR-a exhibited a robust induction of luciferase activity following 23 ATRA treatment.
24 In contrast, inclusion of the RAR-'y-VP-16 expression plasmid with the ER-RXR-a and ERE-tk-Luc plasmids in the transfection 26 mixture resulted in a significant increase in the basal luciferase activity 27 as measured in the absence of any added retinoid. This increase in 28 basal luciferase activity observed for the ER-RXR-a/RAR-y-VP-16 29 cotransfectants, when compared to the result obtained using cells transfected with ER-RXR-a alone, indicated that recombinant ER-_-.,~._,--~. _w_... .. _ . _.m_.T~m~..._-.,~..u.~... .~.. ~.,4.-.._.a~_".~...~ .

1 RXR-a and RAR-y-VP-16 proteins could heterodimerize. Interaction 2 of the heterodimer with the cis-regulatory estrogen responsive element 3 led to a targeting of the VP-16 activation domain to the promoter 4 region of the ERE-tk-Luc reporter. Treatment of such triply transfected cells with ATRA led to a modest increase of luciferase 6 activity over the high basal level. However, treatment of the triple 7 transfectants with AGN 193109 resulted in a dose dependent decrease 8 in luciferase activity. Importantly, Figure 6 shows that AGN 193109 9 treatment of cells cotransfected with ER-RXR-a and RAR-y-VP-16 led l0 to repression of luciferase activity with maximal inhibition occurring at 11 approximately 10-8 M AGN 193109.
12 Our observation that AGN 193109 repressed the constitutive 13 transcriptional activation function of RAR-y-VP-16 in the presence of 14 an RXR was explained by a model wherein binding of AGN 193109 to the RAR induced a conformational change in the RAR which stabilizes 16 a negative conformation that facilitates the binding of a trans-acting 17 negative coactivator protein. When the AGN 193109/RAR complex is 18 bound by the NCP, the RAR is incapable of upregulating transcription 19 of genes that are ordinarily responsive to activated RARs. Our model further proposes that the intracellular reservoir of NCP is in limiting 21 concentration in certain contexts and can be depleted by virtue of AGN
22 193109 stimulated complexation with RARs.
23 The results presented in Figure 6 additionally indicated that even 24 at 10-6 M AGN 193109, the ER-RXR-a and RAR-y-VP-16 proteins could interact to form heterodimers competent for activating 26 transcription of the reporter gene. More specifically, cells transfected 27 with ER-RXR-a and RAR-y-VP-16 and treated with AGN 193109 at a 28 concentration (10-8-10-6 M) sufficient to provide maximal inhibition, gave 29 luciferase activity readings of approximately 16,000 rlu. Conversely, cells transfected only with ER-RXR-a and then treated with AGN

1 193109 at a concentration as high as 10-6 M exhibited luciferase 2 expression levels of only approximately 8,000 rlu. The fact that a higher 3 level of luciferase activity was obtained in cells that expressed both ER-4 RXR-a and RAR-y-VP-16, even in the presence of 10-6 M AGN 193109 demonstrated the persistence of an interaction between the two 6 recombinant receptors. The repression of RAR-y-VP-16 activity by 7 AGN 193109 suggested that modulation of NCP interaction can be 8 codominate with VP-16 activation. Accordingly, we realized that it may 9 be possible to modulate the expression of genes which are not ordinarily to regulated by retinoids in an AGN 193109 dependent manner.
1 l Candidates for AGN 193109 regulatable genes include those that 12 are activated by transcription factor complexes which consist of non-13 RAR factors that associate or heterodimerize with RARs, wherein the 14 non-RAR factor does not require an RAR agonist for activation. While stimulation with an RAR agonist may have substantially no effect on the 16 expression of such genes, administration with AGN 193109 can promote 17 formation of inactive transcription complexes comprising AGN
18 193109/RAR/NCP. Consequently, addition of the AGN 193109 retinoid 19 negative hormone can down-regulate transcription of an otherwise retinoid-insensitive gene.
21 This same mechanism can account for the observation that AGN
22 193109 can repress the activity of the tissue transglutaminase (TGase) 23 gene in HL-60 cells. A retinoid response element consisting of three 24 canonical retinoid half sites spaced by 5 and 7 base pairs has been identified in the transcription control region of this gene. While TGase 26 can be induced by RXR-selective agonists, it is not responsive to RAR-27 selective agonists. The TGase retinoid response element is bound by an 28 RAR/RXR heterodimer (Davies et al. in Press). Interestingly, AGN
29 193109 is able to repress TGase activity induced by RXR agonists. This AGN 193109 mediated repression can be accounted for by the ability of . ,.. . . ... . ~...._..ry.--.~..._._.-..__....~~_.~.-w~.-..

1 this negative hormone to sequester NCPs to the RAR component of the 2 heterodimer, thereby repressing the activity of the associated RXR.
3 We have also obtained results which support conclusions identical 4 to those presented under Example 11 by employing RAR-y-VP-16 and expression constructs and the MTV-TREp-Luc reporter plasmid 6 instead of the RAR-~y-VP-16 and ER-RXR-a expression constructs in 7 combination with the ERE-tk-Luc reporter plasmid. Consistent with 8 the results presented above, we found that RAR-y-VP-16 activity at the 9 MTV-TREp-Luc reporter was inhibited by AGN 193109. Therefore, AGN 193109 repressed RAR-y-VP-16 activity when this chimeric 11 receptor was directly bound to a retinoic acid receptor response element 12 instead of indirectly bound to an estrogen response element in the 13 promoter region of the reporter plasmid. These findings demonstrated 14 that an assay for identifying agents having negative hormone activity need not be limited by the use of a particular reporter plasmid.
16 Instead, the critical feature embodied by an experimental system useful 17 for identifying retinoid negative hormones involves detecting the ability 18 of a compound to repress the activity of an RAR engineered to contain 19 a constitutive transcription activation domain.
Generally, retinoid negative hormones can be identified as the 21 subset of retinoid compounds that repress within a transfected cell the 22 basal level expression of a reporter gene that is transcriptionally 23 responsive to direct or indirect binding by a retinoid receptor or a 24 chimeric receptor that includes at least the domains of the retinoid receptor located C-terminal to the DNA binding domain of that 26 receptor. This approach has been adapted tc~ screening method useful 27 for identifying retinoid negative hormones. In the various embodiments 28 of the invented screening method, the structure of the receptor for 29 which a negative hormone is sought is variable. More specifically, the retinoid receptor can be either of the RAR or the RXR subtype. The 1 receptor can optionally be engineered to include a constitutive 2 transcription activator domain. The retinoid receptor used to screen for 3 negative hormones optionally contains a heterologous DNA binding 4 domain as a substitute for the DNA binding domain endogenous to the native receptor. However, when a second receptor is used in the 6 screening method, and where the second receptor can dimerize with the 7 retinoid receptor for which a negative hormone is sought, then that 8 retinoid receptor may not require a DNA binding domain because it can 9 be linked to the transcription control region of the reporter gene l0 indirectly through dimerization with the second receptor which is itself 11 bound to the transcription control region.
12 In the practice of the screening method, the ability of a 13 compound to repress the basal expression of a reporter is typically 14 measured in an in vitro assay. Basal expression represents the baseline level of reporter expression in transfected cells under conditions where 16 no exogenously added retinoid agonist is present. Optionally, steps may 17 be taken to remove endogenous retinoid ligands from the environment 18 of the transfected cells via procedures such as charcoal extraction of the 19 serum that is used to culture cells in vitro.
Examples of reporter genes useful in connection with the 21 screening method include those encoding luciferase, beta galactosidase, 22 chloramphenicol acetyl transferase or cell surface antigens that can be 23 detected by immunochemical means. In practice, the nature of the 24 reporter gene is not expected to be critical for the operability of the method. However, the transcriptional regulatory region of the reporter 26 construct must include one or more cis-regulatory elements that are 27 targets of transcription factors for which negative hormones are being 28 sought. For example, if one desires to identify RAR negative 29 hormones, then the transcriptional regulatory region of the reporter construct could contain a cis-regulatory element that can be bound by .,.~~.~.r.W....~_..~._.~_ . ~...~yy-~. ........~4_~... . ......

1 an RAR-containing protein. In this example, there should be 2 correspondence between the DNA binding domain of the RAR and the 3 cis-regulatory element of the transcriptional regulatory region of the 4 reporter construct. Thus, if a chimeric RAR having a constitutive transcription activator domain and a DNA binding domain that can bind 6 cis-regulatory estrogen responsive elements is employed in the screening 7 method, then the transcriptional regulatory region of the reporter 8 construct should contain an estrogen responsive element.
9 Examples of cis-regulatory elements that directly bind retinoid l0 receptors (RAREs) useful in connection with the reporter assay are 11 disclosed by Mangelsdorf et al. in The Retinoid Receptors in The 12 Retinoids: Biolo~y, Chemistry and Medicine, 2nd edition, eds. Sporn et 13 al., Raven Press, Ltd., New York { 1994), the disclosure of which has 14 been incorporated by reference hereinabove. Examples of cis-regulatory elements that indirectly bind chimeric receptors include DNA binding 16 sites for any DNA binding protein for which the DNA binding domain 17 of the protein can be incorporated into a chimeric receptor consisting of 18 this DNA binding domain attached to a retinoid receptor. Specific 19 examples of heterologous DNA binding domains that can be engineered into chimeric receptors and that will recognize heterologous cis-21 regulatory elements include those recognizing estrogen responsive 22 elements. Thus, the retinoid receptor portion of a chimeric receptor 23 useful in connection with the screening method need not contain the 24 DNA binding of the retinoid receptor but must contain at least the ligand binding domain of the retinoid receptor.
26 A further example of indirect retinoid receptor binding to the cis-27 regulatory element includes the use of a protein that can bind the cis-28 regulatory element and dimerize with a retinoid receptor. In this case, 29 the retinoid receptor associates with the cis-regulatory element only by association with the protein responsible for DNA binding. An example 1 of such a system would include the use of a fusion protein consisting of 2 a heterologous DNA binding domain fused to an RXR, containing at 3 least the domain of the RXR responsible for dimerization with RARs.
4 Cointroduced RARs can dimerize with such a fusion protein bound to the cis-regulatory element. We anticipate that any cis-regulatory 6 element-binding protein that dimerizes with RARs to result in an 7 indirect association of the RAR with the cis-regulatory element will also 8 be suitable for use with the negative hormone screening method.
9 In a preferred embodiment of the screening method, retinoid negative hormones are identified as those retinoids that repress basal 11 expression of an engineered RAR transcription factor having increased 12 basal activity. Although not essential for operability of the screening 13 method, the engineered RAR employed in the following Example 14 included a constitutive transcription activating domain. Use of this chimeric receptor advantageously provided a means by which the basal 16 expression of a reporter gene could be elevated in the absence of any 17 retinoid. Although we have employed transient transfection in the 18 procedures detailed above, stably transfected cell lines constitutively 19 expressing the chimeric receptor would also be useful in connection with 2o the screening method.
21 As disclosed in the following Example, a chimeric retinoid 22 receptor having a constitutive transcription activator domain was 23 heterodimerizable with a second receptor engineered to contain a DNA
24 binding domain specific for an estrogen responsive cis-regulatory element. In this case the chimeric retinoid receptor having a 26 constitutive transcription activator domain associates with the cis-27 regulatory region controlling reporter gene expression indirectly via 28 binding to a second receptor that binds a DNA target sequence. More 29 particularly, the second receptor was engineered to contain a DNA
binding domain that recognized an estrogen responsive element.
T..........u.."~~.,m..,~, ...-.~e~m".~_...._~....._.~

1 Advantageously, the reporter gene having an estrogen responsive 2 element in the upstream promoter region was unresponsive to retinoid 3 agonists in the absence of the transfected chimeric receptor having the 4 constitutive transcription activator domain. Accordingly, all reporter gene activity was attributed to the transfected receptors. The 6 combination use of the estrogen responsive element DNA binding 7 domain and the estrogen responsive element cis-regulatory element are 8 intended to be illustrative only. Those having ordinary skill in the art 9 will realize that other combinations of engineered receptors having l0 specificity for non-RARE cis-regulatory elements will also be useful in 11 the practice of the invented screening method.
12 Cells useful in connection with the screening method will be 13 eukaryotic cells that can be transfected. The cells may be animal cells 14 such as human, primate or rodent cells. We have achieved very good results using CV-1 cells, but reasonably expect that other cultured cell 16 lines could also be used successfully. Any of a number of conventional 17 transfection methods known in the art can be used to introduce an 18 expression construct encoding the chimeric retinoid receptor having a 19 constitutive transcription activator domain.
The constitutive transcription activator domain will consist of a 21 plurality of amino acids which will likely have an overall acidic character 22 as represented by a negative charge under neutral pH conditions. For 23 example, the constitutive transcription activator domain may have an 24 amino acid sequence which is also found in viral transcription factors.
One example c~f a viral transcription factor having a constitutive 26 transcription activator domain is tt:~ herpes simplex virus 16. However, 27 other viral or synthetic transcription activator domains would also be 28 useful in the construction of expression constructs encoding the chimeric 29 retinoid receptor having a constitutive transcription activator domain.
As described below, we have developed a generalized screening 1 method useful for identifying retinoid negative hormones. This 2 screening method provides a means for distinguishing simple antagonists 3 from negative hormones. Table 12 lists several retinoid compounds 4 which exhibit potent affinity for RAR-y yet, with the exception of ATRA, did not transactivate this receptor in a transient cotransfection 6 transactivation assay. We therefore tested these compounds to 7 determine which were RAR-y antagonists and which, if any, of these 8 antagonists exhibited negative hormone activity.
9 Example 12 describes the method used to identify retinoid l0 compounds that were antagonists, and the subset of antagonists that 11 exhibited negative hormone activity.
12 Example 12 13 Assay for Retinoid Negative Hormones 14 4 X 104 CV-1 cells were transfected by the calcium phosphate coprecipitation procedure described in Molecular Cloning: A Laboratory 16 Manual (Sambrook et al. eds. Cold Spring Harbor Lab Publ. 1989) using 17 0.5 ~,g ERE-tk-Luc reporter plasmid and 0.1 ~cg ER-RAR-y (Graupner 18 et al. Biochem. Biophys. Res. Comm. 179:1554 (1991)) chimeric 19 expression plasmid. After 18 hours, cells were rinsed with PBS and fed with DMEM (Gibco-BRL) containing 10% activated charcoal extracted 21 FBS (Gemini Bio-Products). Cells were treated with 10-8 M ATRA in 22 ethanol or ethanol alone. In addition, ATRA treated cells were treated 23 with 10-9, 10-8, 10-~ or 10-6 M of the compounds listed in Table 12. After 24 18 hours, cells were rinsed in PBS and lysed in 0.1 M KP04 (pH 7.8), 1.0% TRITON X-100, 1.0 mM DTT, 2 mM EDTA. Luciferase 26 activities were measured as described by deWet et al. in Mol. Cell. Biol.
27 7:725 (1987).
.._ . __. ......_......-__~....~. -,._.a.._ ._._ .r 2 Compound Kd (nM) @ RAR-yQ ECso (nM) @ RAR-yb 4 AGN 193109 6 na (Compound 60) 6 AGN 193174 52 na 7 {Compound 34a) 8 AGN 193199 30 na 9 AGN 193385 25 na (Compound 23) lI AGN 193389 13 na 12 (Compound 25) 13 AGN 193840 40 na 14 AGN 193871 30 na I5 (Compound 50) 1 ~ ° Relative affinity (Kd) determined by competition of 3H-18 ATRA binding to baculovirus expressed RAR-y and application of the 19 Cheng-Prussof equation.
b ECSO measured in CV-1 cells transiently cotransfected with 21 OMTV-TREp-Luc and RS-RAR-y. "na" denotes no activity.

23 As indicated by the results presented in part in Figure 7 and in 24 Table 12, with the exception of ATRA, all of the compounds listed in Table 12 were retinoid antagonists at RAR-y.
26 The RAR-y antagonists identified in Table 12 were next screened 27 to determine which, if any, were also r void negative hormones. 4 X
28 104 CV-1 cells were transfected accords .b to the calcium phosphate 29 procedure described in Molecular Cloning: A Laboratory Manual (Sambrook et al. eds. Cold Spring Harbor Lab Publ. 1989) using 0.5 ,ug 31 ERE-tk-Luc reporter plasmid and 0.1 ~,g ER-RXR-a (Graupner et al.
32 Biochem. BiophyS. ReS. Comm. 179:1554 (1991)) and 0.2 ~.cg RAR-y-VP-1 16 (Nagpal et al. EMBO J. 12:2349 (1993)) chimeric expression 2 plasmids. After 18 hours, cells were rinsed with PBS and fed with 3 DMEM (Gibco-BRL) containing 10% activated charcoal extracted FBS
4 (Gemini Bio-Products). Cells were treated with 10-9, 10-8, 10-' or 10-6 M
of each of the compounds listed in Table 12. Treatment with ethanol 6 vehicle alone served as the negative control. After 18 hours, cells were 7 rinsed in PBS and lysed in 0.1 M KP04 (pH 7.8), 1.0% TRITON X-100, 8 1.0 mM DTT, 2 mM EDTA. Luciferase activities were measured as 9 previously by deWet et al. in Mol Cell. Biol. 7:725 (1987).
l0 As shown in Figure 8, the retinoid antagonists of Table 12 could 11 be separated into two classes by virtue of their effect on the constitutive 12 transcription activation function of the RAR-'y-VP-16 chimeric retinoid 13 receptor. One group, which included AGN 193174, AGN 193199 and 14 AGN 193840, did not repress RAR-y-VP-16 activity even though they were ATRA antagonists. In contrast AGN 193109, AGN 193385, AGN
16 193389 and AGN 193871 exhibited a dose dependent repression of 17 RAR-'y-VP-16 constitutive activity. Therefore, while the compounds of 18 both groups were RAR-~y antagonists, only those of the second group 19 exhibited negative hormone activity. This assay advantageously distinguished retinoid negative hormones from simple retinoid 21 antagonists.
22 The foregoing experimental results proved that AGN 193109 met 23 the criteria that define a negative hormone. More specifically, the 24 results presented under Example 11 demonstrated that AGN 193109 had the capacity to exert inhibitory activity at the RARs even in the 26 absence of exogenously added retinoid ligands. As such, this compound 27 possessed biological activities that did not depend upon blockade of the 28 interaction between the RARs and agonists such as ATRA and AGN
29 191183. These findings led us to conclude that AGN 193109 stabilized interactions between RARs and NCPs. As diagrammed in Figure 9, ..._.w.,.-...-...~....,,~..~. _..._ _.T..~".~ .-...~ -... a.. ....~~w-.-... ..

1 NCP/RAR/PCP interactions exist in an equilibrium state. An agonist 2 serves to increase PCP interactions and decrease NCP interactions, 3 while an inverse agonist or negative hormone stabilizes NCP and 4 decreases PCP interactions. As indicated previously, our experimental results suggested that the intracellular availability of NCP for other 6 receptors can be modulated by AGN 193109 administration. More 7 specifically, we discovered that AGN 193109 can promote complexation 8 of NCP with RARs, thereby reducing the intracellular reservoir of NCP
9 available for interaction with transcription factors other than the RARs.
l0 We next examined the effect of AGN 193109 on agonist-mediated 11 inhibition of AP-1 dependent gene expression. In Endocr. Rev. 14:651 12 (1993), Pfhal disclosed that retinoid agonists can down-regulate gene 13 expression by a mechanism that involved inhibition of AP-1 activity.
14 We postulated that AGN 193109 could have had either of two effects when used in combination with a retinoid agonist in a model system 16 designed to measure AP-1 activity. First, AGN 193109 could 17 conceivably have antagonized the effect of the agonist, thereby relieving 18 the agonist-dependent inhibition of AP-1 activity. Alternatively, AGN
19 193109 could have potentiated the agonist's activity, thereby exaggerating the agonist-dependent inhibition of AP-1 activity.
21 Example 13 describes the methods used to demonstrate that 22 AGN 193109 potentiated the anti-AP-1 activity of a retinoid agonist.
23 As disclosed below, the AGN 191183 retinoid agonist weakly inhibited 24 AP-1 dependent gene expression. The combination of AGN 193109 and the retinoid agonist strongly inr ~ted AP-1 dependent gene 26 expression. By itself, AGN 193109 h.. j substantially no anti-AP-1 27 activity.

1 Example 13 2 AGN 193109 Potentiates the Anti-AP-1 Activity of a Retinoid Agonist 4 HeLa cells were transfected with 1 ,ug of the Str-AP1-CAT
reporter gene construct and 0.2 ~cg of plasmid pRS-hRARa, described 6 by Giguere et al. in Nature 33:624 (1987), using LIPOFECTAMINE
7 (Life Technologies, Inc.). Str-AP1-CAT was prepared by cloning a 8 DNA fragment corresponding to positions -84 to + 1 of the rat 9 stromelysin-1 promoter (Matrisian et al., Mol. Cell. BioL 6:1679 (1986)) between the HindIII-BamHI sites of pBLCAT3 (Luckow et al., Nucl.
11 Acids Res. 15:5490 (1987)). This sequence of the stromelysin-1 12 promoter contains an AP1 motif as its sole enhancer element 13 (Nicholson et al., EMBO J. 9:4443 (1990). The promoter sequence was 14 prepared by annealing two synthetic oligonucleotides having sequences:
5'-18 GGACCTCGAGGATCCAG-3' 19 (SEQ ID N0:2), and 5'-2o CTGGATCCTCGAGGTCCACCTTTCTGAGCCCAACTI"pT'ATAGA

22 CTTCCATAAGCTTCT-3' (SEQ ID N0:3). Procedures involving 23 transfection, treatment with appropriate compounds and measurement 24 of CAT activity were carried out as described by Nagpal et al. in J. Biol.
Chem. 270:923 (1995), the disclosure of which is hereby incorporated by 26 reference.
27 The results of these procedures indicated that AGN 193109 28 potentiated the anti-AP-1 activity of the retinoid agonist, AGN 191183.
29 More specifically, in the concentration range of from 10-12 to 10-~o M, AGN 191183 did not inhibit the TPA-induced Str-AP1-CAT expression.
..-~w r.a~.....~~ ,... ..~...... . .......~ .W... ..

1 Treatment with AGN 193109 in the concentration range of from 10-'° to 2 10-8 M did not substantially inhibit AP-1 mediated reporter activity.
3 However, the results presented in Figure 10 indicated that stimulation 4 of the transfectants with the combination of AGN 193109 (10-8 M) and AGN 191183 in the concentration range of from 10-'2 to 10-'° M
6 substantially inhibited TPA-induced Str-AP1-CAT expression by an 7 amount of from 12% to 21%. Therefore, AGN 193109 potentiated the 8 anti-AP-1 activity of AGN 191183 under conditions where this retinoid 9 agonist ordinarily did not inhibit AP-1 activity.
l0 We reasoned that AGN 193109 potentiated the agonist-mediated 11 repression of AP-1 activity by a mechanism that likely involved AGN
12 193109-dependent sequestration of NCPs onto RARs. RARs belong to 13 a superfamily of nuclear receptors that also includes receptors for 1,25-14 dihydroxyvitamin D3, glucocorticoid, thyroid hormone, estrogen and progesterone. It was a reasonable assumption that the ability to bind 16 NCPs may be shared among different members of the nuclear receptor 17 superfamily. This led us to speculate that AGN 193109 could potentiate 18 the anti-AP-1 activity of one or more of the ligands that interact with 19 this superfamily of nuclear receptors.
The results presented in the preceding Example clearly indicated 21 that AGN 193109 potentiated the anti-AP-1 activity of a retinoid 22 agonist. More specifically, AGN 193109 lowered the threshold dose at 23 which the anti-AP-1 activity of AGN 191183 could be detected. Since 24 AGN 193109 has substantially no anti-AP-1 activity by itself, its effect on nuclear receptor agonists was s~.-~ergistic. We also foun,l that the 26 AGN 193109 negative hormone p~ :,ntia~ed thc;. anti-AP-1 activity of 27 1,25-dihydroxyvitamin D3, the natural ligand for the vitamin D3 receptor.
28 The observed synergy between AGN 193109 and AGN 191183 in 29 the preceding Example necessarily implied that the anti-AP-1 activity of the retinoid agonist and the AGN 193109-mediated potentiation of that WO 98/58922 PCTlUS98/13065 1 activity must result from different mechanisms. If the mechanisms of 2 action of the two agents were identical, then it follows that the 3 effectiveness of the combination of AGN 193109 and the agonist would 4 have been additive. Instead, the combination was shown to be more effective than either agent alone, an effect that could not have been 6 predicted in advance of this finding.
7 Significantly, the AGN 193109-mediated potentiation of the RAR
8 agonist was performed using an approximately 100-fold molar excess of 9 AGN 193109 over that of the retinoid agonist. Accordingly, the to majority of RARs should have been bound by AGN 193109 leaving very 11 few RARs available for agonist binding. In spite of this fact, the 12 population of RARs that were not bound by AGN 193109 were able to 13 bind retinoid agonist and vigorously stimulate an agonist-dependent 14 response measurable as an inhibition of reporter gene expression. Thus, i 5 our data suggested possible heterogeneity of RARs that are induced by 16 AGN 193109.
17 The negative hormone activity of AGN 193109, attributed to its 18 ability to promote the interaction of RARs and NCPs, provided a basis 19 for understanding the synergy between AGN 193109 and retinoid 20 agonists. Our results were fully consistent with a model in which AGN
21 193109 treatment of cells promoted binding of RARs and NCPs, 22 thereby reducing the number of free NCP and free RAR within the cell.
23 This results in the generation of two populations of RARs that are 24 functionally distinct. The first population is represented by RARs 25 associated with NCPs. Such AGN 193109/RAR/NCP complexes cannot 26 be activated by retinoid agonists. The second population consists of 27 RARs that are not bound by NCP, and that remain available for 28 interaction with agonists. This latter population is designated "RAR*"
29 to indicate free RARs in an environment substantially depleted of NCP.
30 The RAR*s have decreased probabilities of association with NCP

1 through equilibrium binding and have an increased sensitivity to retinoid 2 agonists measurable, for example, as anti-AP-1 activity. This is so 3 because, while the intracellular reservoir of NCP is depleted by virtue of 4 AGN 193109 administration, the reservoir of PCP has not been depleted. Accordingly, free RAR*s can bind a retinoid agonist and 6 interact with PCP factors in an environment substantially depleted of 7 NCP. The ability of AGN 193109 to increase the sensitivity of other 8 nuclear receptors to their respective agonists can be attributed to the 9 ability of these different nuclear receptors to interact with the same NCPs that interact with AGN 193109/RAR complexes. This model of 11 AGN 193109-mediated modulation of NCP availability for nuclear 12 receptor family members is schematically represented in Figure 11.
13 This mechanistic model led us to predict that AGN 193109 could 14 modulate the activities of nuclear receptor ligands other than retinoid agonists. As illustrated in the following Example, we confirmed that 16 AGN 193109 potentiated the activity of 1,25-dihydroxyvitamin D3 in an 17 in vitro transactivation assay.
18 Example 14 describes the methods used to demonstrate that 19 AGN 193109 enhanced the activity of 1,25-dihydroxyvitamin D3 in a transactivation assay.
2 ~ Example 14 22 AGN 193109 Potentiates 1,25-Dih.~xyvitamin Dz Activity 23 Hela cells were transfected using the cationic liposome-mediated 24 transfection procedure described by Felgner et al. in Proc. Natl. Acad.
Sci. USA 84:7413 (1987). 5 X 104 cells were plated in 12-well multiwell 26 plates and grown in DMEM supplemented with 10% FB ~. Cells were 27 cotransfected in serum-free medium using 2 ~,g/well of 28 LIPOFECTAMINE reagent (Life Technologies, Inc.) with 0.7 ~cg of the 29 reporter plasmid MTV-VDRE-Luc, containing two copies of the 1,25-dihydroxyvitamin D3 response element 1 5'-GTACAAGGTTCACGAGGTTCACGTCTTA-3' (SEQ ID N0:4) 2 from the mouse osteopontin gene (Ferrara et al. J. Biol. Chem. 269:2971 3 (1994)) ligated into the reporter plasmid MTV-Luc (Heyman et al. in 4 Cell 68:397 (1992)), and 0.3 ,ug of the plasmid pGEM3Z (Pharmacia, Inc.) as carrier DNA to bring the final concentration of DNA to 1.0 ~cg 6 per well. After six hours of transfection, cells were fed with growth 7 medium containing charcoal extracted FBS at a final concentration of 8 10%. Eighteen hours after transfection cells were treated with vehicle 9 alone (ethanol) or AGN 193109 in ethanol at a final concentration of either 10-$ or 10-~ M. Six hours later 1,25-dihydroxyvitamin D3 was 11 added in ethanol to a final concentration of from 10-I~ to 10-~ M. Cells 12 were lysed and harvested eighteen hours following 1,25-dihydroxyvitamin 13 D3 treatment. Luciferase activity was measured as described above.
14 This experimental system allowed a convenient method of monitoring and quantitating 1,25-dihydroxyvitamin D3-dependent gene expression.
16 The results presented in Figure 12 indicated that, when compared 17 with the result obtained using 1,25-dihydroxyvitamin D3 alone, AGN
18 193109 coadministered with 1,25-dihydroxyvitamin D3 shifted the dose 19 response curve to the left. This confirmed that AGN 193109 potentiated the effectiveness of 1,25-dihydroxyvitamin D3 in the in vitro 21 transactivation assay. More specifically, Figure 12 graphically illustrates 22 that an AGN 193109 concentration as low as 10 - 100 nM rendered the 23 1,25-dihydroxyvitamin D3 approximately 10 fold more active. While a 24 1,25-dihydroxyvitamin D3 concentration of 10-g M was required to produce a luciferase expression of approximately 2,000 rlu, only one-26 tenth as much 1,25-dihydroxyvitamin D3 was required to produce the 27 same luciferase output when the vitamin was coadministered with AGN
28 193109 at a concentration of 10-8 - 10-~ M. Although not shown on the 29 graph in Figure 12, substantially identical results were obtained using AGN 193109 concentrations of 10-9 M and 10-$ M. Thus, .. . - . _ _._. .~.-.. _..._...w.~....--_-.-...W......~..-.._ . .....~~,-.~.~..,-_.. _ ..._~-.......,_- _.. _ ..... -..

1 coadministration with AGN 193109 substantially reduced the amount of 2 1,25-dihydroxyvitamin D3 that was required to produce a similar effect 3 in the absence of the negative hormone.
4 Interestingly, when the above procedure was repeated with cotransfection of a vitamin D receptor (VDR) expression plasmid, there 6 was a coincident decrease in the ability of AGN 193109 to potentiate 7 the activity of 1,25-dihydroxyvitamin D3. We interpreted this result as 8 indicating that over-expression of VDRs could affect the ability of AGN
9 193109 to potentiate the activity of 1,25-dihydroxyvitamin D3. Thus, the 1 o intracellular concentration of a ligand receptor, which may differ in a 11 tissue-specific fashion, can influence the ability of AGN 193109 to 12 potentiate the activity of a ligand that binds the receptor. This was 13 again consistent with a model in which titratable NCPs contributed to 14 the regulation of the Vitamin D3 response, and supported the model set forth above.
16 As illustrated in the following Example, we also confirmed that 17 AGN 193109 potentiated the anti-AP-1 activity of 1,25-dihydroxyvitamin 18 D3. Our model for the activity of AGN 193109 action explains this 19 observation by invoking that NCPs avidly associate with RARs in the presence of this drug. Endogenous vitamin D receptors present in 21 HeLa cells likely were rendered more sensitive to the 1,25-22 dihydroxyvitamin D3 ligand, with the consequence of exaggerating the 23 ability of this ligand to inhibit expression from the Str-AP1-CAT
24 reporter.
Example 15 descrines the methods used to demonstrate that 26 AGN 193109 potentiate~~i the anti-AP-1 activity of 2,25-dihydroxyvitamin 27 D3.

1 Example 15 2 AGN 193109 Potentiates the Anti-AP-1 Activity 3 of 1,25-Dihydrox~Nitamin D3 4 HeLa cells were transfected with 1 ~cg of Str-AP1-CAT using LIPOFECTAMINE according to the method described by Nagpal et al.
6 in J. Biol. Chem. 270:923 (1995). Transfected cells were treated with 7 AGN 193109 alone (10-9 to 10-' M), 1,25-dihydroxyvitamin D3 alone (10-g lz to 10-' M) or 1,25-dihydroxyvitamin D3 (10-'z to 10-' M) in the 9 presence of 10-8 M AGN 193109.
l0 The results of these procedures indicated that AGN 193109 11 potentiated the ability of 1,25-dihydroxyvitamin D3 to inhibit TPA-12 induced AP-1 activity. When used alone in the concentration range of 13 from 10-9 to 10-' M, AGN 193109 had no detectable anti-AP-1 activity.
14 The results presented in Figure 13 indicated that 1,25-dihydroxyvitamin D3 repressed TPA-stimulated activity only in the 10-8 and 10-' M
16 concentration range. Analysis of 1,25-dihydroxyvitamin D3 mediated 17 repression of TPA stimulated CAT activity in the presence of 10-8 M
18 AGN 193109 indicated that anti-AP-1 activity was detectable at 10-» and 19 10-9 M 1,25-dihydroxyvitamin D3 and an increase in activity at 10-$ and 10-' M doses compared to 1,25-dihydroxyvitamin D3 treatment alone.
21 This AGN 193109 dependent modulation of 1,25-dihydroxyvitamin D3 22 mediated anti-AP-1 activity was consistent with our model in which 23 NCP sequestration to RARs made the NCP unavailable for interaction 24 with other nuclear receptor family members. Accordingly, the receptors were rendered more sensitive to the 1,25-dihydroxyvitamin D3 26 treatment.
27 The mechanisms underlying RAR mediated transactivation and 28 anti-AP-1 activity are likely different. This conclusion was based on our 29 observation that high doses of AGN 193109 completely inhibited transactivation without substantially inhibiting anti-AP1 activity. We . .. . ............- . ..... .._...,..~,..~..-"._.~.~.,."~.,.~.~.r.-, .,-_.~..-.. . . . . . ... _.T_.._.,",_.. ..>.",-........__-.._.--_",~.~..-...._ 1 therefore wished to gain additional evidence to support our model for 2 RAR* formation mediated by AGN 193109 treatment. To accomplish 3 this, we investigated whether AGN 193109 could potentiate the activity 4 of the RAR specific agonist AGN 191183 in an in vitro transactivation assay.
6 Example 16 describes the methods used to demonstrate that 7 AGN 193109 potentiated the activity of the RAR specific agonist, AGN
8 191183. The results of this procedure indicated that, under particular 9 circumstances, AGN 193109 enhanced the potency of the RAR specific 1 o retinoid, and provided strong evidence that AGN 193109 promoted 11 RAR* formation.
12 Example 16 13 Potentiation of Retinoid Effectiveness by 14 AGN 193109 Coadministration Hela cells were transfected using the cationic liposome-mediated 16 transfection procedure described by Felgner et al. in Proc. Natl. Acad.
17 Sci. I~SA 84:7413 (1987). 5 X 104 cells were plated in 12 well multiwell 18 plates and grown in DMEM supplemented with 10% FBS. Cells were 19 cotransfected in serum free medium using LIPOFECTAMINE reagent (2 ug/well, Life Technologies, Inc.) with 0.7 ~,g of the reporter plasmid 21 MTV-TREp-Luc, containing two copies of the TREpaI response 22 element 5'-TCAGGTCATGACCTGA-3' (SEQ ID NO:S) inserted into 23 the reporter plasmid MTV-Luc (Heyman et al. in Cell 68:397 (1992)), 24 and 0.1 ~cg of the RAR-y expression plasmid pRShRAR-y (Ishikawa et al. Mol. Endocrinol. 4:837 (1990)). After six hours of lansfection, cells 26 were fed with growth medium ct. utaining charcoal extracted FBS at a 27 final concentration of 10%. Eighteen hours after transfection, cells 28 were treated with vehicle alone (ethanol) or AGN 193109 in ethanol at 29 a final concentration of from 10-11 to 10-8 M. Six hours later, AGN
191183 was added in ethanol to a final concentration of either 0, 10-1 or 1 10-9 M. Cells were harvested after eighteen hours of AGN 191183 2 treatment and luciferase activity was measured as described above.
3 Preliminary experiments indicated that 10-9 M AGN 193109 was 4 relatively ineffective at inhibiting the response to of 10-9 M AGN 191183 in HeLa cells. This contrasted with the ability of 10-9 M AGN 193109 6 to inhibit 10-$ M ATRA in CV-1 cells (Figure 2).
7 The results presented in Figure 14 supported the prediction that 8 AGN 193109 stimulated the formation of RAR*. Consistent with our 9 characterization of the antagonist and negative hormone activities of AGN 193109, treatment with AGN 193109 resulted in a biphasic dose 11 response curve. The lowest doses of AGN 193109 (10-11 and 10-1 M) 12 resulted in a stimulation of luciferase activity over that of AGN 191183 13 alone. This effect suggests that RAR*s are generated by AGN 193109.
14 Curiously, this was also seen for AGN 193109 treatment alone, suggesting that RAR*'s can respond to an endogenous ligand. AGN
16 191183 is a synthetic retinoid agonist and, like ATRA, activates 17 transcription through the RARs. Substitution of AGN 1911$3 for 18 ATRA in Example 7 would give qualitatively similar results (i.e., AGN
19 193109 would antagonize the effect of 10 nM AGN 191183). Example 16 illustrates that, while AGN 193109 can function as an antagonist of 21 RAR agonists, dosing conditions could easily be identified wherein 22 AGN 193109 coadministration potentiated activation mediated by an 23 RAR agonist. It is important to note that the doses of the compounds 24 used in Example 16 are substantially lower than the doses employed in the procedure described under Example 7. We proposed that AGN
26 193109 treatment could lead to RAR heterogeneity RARs versus 27 RAR*s. The apparent heterogeneity (i.e., ability to potentiate) appears 28 to have different windows in transactivation versus AP-1 repression.
29 The reason that the curves are biphasic is because, with increasing amounts of AGN 193109, there is proportionately less RAR available to .. . _...w~...~,..a.~~-.~.~..~,~--..... ~.~..... .n.... -~. .~w,...~.~.~
......_ .~. ..~.~_..._. ..

1 bind the agonist. This doesn't appear to be the case for AP-1 2 repression and we are left to speculate that this difference must reflect 3 two distinct mechanisms for transactivation and AP-1 repression by the 4 same receptor species.
Clinical results have confirmed that some retinoids are useful for 6 inhibiting the growth of premalignant and malignant cervical lesions.
7 Exemplary studies supporting this conclusion have been published by 8 Graham et al. in West. J. Med. 145: 192 (1986), by Lippman et al. in J.
9 Natl. Cancer Inst. 84:241 ( 1992), and by Weiner et al. in Invest. New l0 Drugs 4:241 (1986)).
11 Similar conclusions are supported by the results of in vitro studies 12 that used cultured cells to quantitate the antiproliferative effects of 13 various retinoids. More specifically, Agarwal et al. in Cancer Res.
14 51:3982 (1991) employed the ECE16-1 cell line to model the early stages of cervical dysplasia and demonstrated that retinoic acid could 16 inhibit epidermal growth factor (EGF) dependent cellular proliferation.
17 Example 17 describes the methods used to demonstrate that 18 AGN 193109 can antagonize the activity of the AGN 191183 retinoid 19 agonist which inhibited proliferation of the ECE16-1 cell line.
Example 17 21 AGN 193109 Antagonizes the Antiproliferative Effect 22 of Retinoids in ECE16-1 Cells 23 ECE16-1 cells were seeded at a density of 1 x 104 cells per cm2 24 in complete medium containing DMEM:F12 (3:1), nonessential amino acids, 5% FBS, 5 ~.g/ml transferrin, 2 nM of 3,3',5 triiodothyronine 26 (thyroid hormone or "T3"), 0.1 nM cholera toxin, 2 mM L-glutamine, 1.8 27 x 10-4 M adenine and 10 ng/mI EGF. Cells were allowed to attach to 28 plates overnight and then shifted to defined medium containing 29 DMEM:F12 (3:1), 2 mM L-glutamine, nonessential amino acids, 0.1%
bovine serum albumin, 1.8 x 10-4 M adenine, 5 ~,g/ml transferrin, 2 nM

1 T3, 50 ,ug/ml ascorbic acid, 100 ug/ml streptomycin, 100 units/ml 2 penicillin and 50 ~cg/ml gentamicin. Defined medium (DM) was 3 supplemented with 10 ng/ml EGF. EGF treated cells received 10 nM of 4 the AGN 191183 retinoid agonist in combination with either 0, 0.1, 1.0, 10, 100 or 1000 nM AGN 193109 or 1000 nM AGN 193109 alone.
6 After three days of treatment, cells were harvested as described by 7 Hembree et al. in Cancer Res. 54:3160 (1994) and cell numbers 8 determined using a COULTER counter.
9 The results presented in Figure 15 demonstrated that ECE16-1 cells proliferated in response to EGF but not in defined medium alone.
11 This confirmed the findings published by Andreatta-van Leyen et al. in 12 J. Cell. Physio. 160:265 ( 1994), and by Hembree et al. in Cancer Res.
13 54:3160 (1994). Addition of 10 nM AGN 191183 and 0 nM AGN
14 193109 completely inhibited EGF mediated proliferation. Thus, AGN
191183 was a potent antiproliferative retinoid. Increasing the AGN
16 193109 concentration from 0 nM to 10 nM antagonized the AGN
17 191183 mediated growth inhibition by approximately 50%. A ten-fold 18 molar excess of AGN 193109 completely reversed the antiproliferative 19 effect of AGN 191183. Treatment of cells with 1000 nM AGN 193109 alone had no effect on the EGF mediated proliferation increase. These 21 results proved that AGN 193109 antagonized the antiproliferative effect 22 of a retinoid but had substantially no antiproliferative activity of its own 23 when used to treat cells representing cervical epithelium that is sensitive 24 to growth inhibition by retinoids such as AGN 191183. Notably, there was no evidence that AGN 193109 potentiated the antiproliferative 26 activity of the AGN 191183 agonist using the ECE16-1 model system.
27 In contrast to the model system represented by the ECE16-1 cell 28 Line, there are other examples where cellular proliferation associated 29 with cervical dysplasia cannot be inhibited by retinoid agonists. For example, Agarwal et al. in Cancer Res. 54:2108 (1994) described the use . . ... _. . .. ._._.~.~ _. ... ~w_ ....._ , _... _... r__.... . ...._ ._ . ..
_._. _ 1 of CaSki cells as a model for cervical tumors that are unresponsive to 2 retinoid therapy. As disclosed below, rather than inhibiting cell 3 proliferation, retinoid treatment had substantially no effect on the 4 growth rate of CaSki cells. The following Example addressed the effect of the AGN 193109 negative hormone on the proliferation rates of this 6 cell line. The results unexpectedly proved that AGN 193109 can inhibit 7 the proliferation of cervical tumor cells that are unresponsive to the 8 antiproliferative effects of retinoid agonists.
9 Example 18 describes the methods used to demonstrate that AGN 193109 inhibited the growth of a cervical tumor cell line that did 11 not respond to the antiproliferative effects of other retinoids such as 12 AGN 191183. Significantly, AGN 193109 displayed antiproliferative 13 activity in the absence of added retinoid 14 Example 18 AGN 193109 Inhibits the Proliferation Rate of CaSki 16 Cervical Carcinoma-Derived Cell Line 17 We tested the effect of EGF on CaSki cell proliferation, either 18 alone or in combination with the AGN 191183 retinoid agonist and/or 19 the AGN 193109 negative hormone at a concentration of 10-6 M. Cell proliferation assays were performed as described above for studies 21 involving ECE16-1 cells. EGF was added to the retinoid treated 22 cultures to give a final concentration of 20 ng/ml. Cells were treated 23 with AGN 191183 ( 10-1 to 10-6 M) in the presence or absence of 10-6 M
24 AGN 193109 for a total of three days. The media was replaced with fresh media and each of the two retinoid compounds, as appropriate, 26 every day. Cell numbers were determined using a COULTER counter 27 as described above.
28 The results presented in Figure 16 indicated that CaSki cells were 29 substantially refractory to the effects of a retinoid agonist and that AGN
193109 exhibited antiproliferative activity in the absence of added 1 retinoid. The presence of EGF in the culture media stimulated CaSki 2 cell growth. This conclusion was based on comparison of the stripped 3 bar representing no AGN 191183 and the open bar representing defined 4 growth media ("DM") alone. AGN 191183 treatment had no antiproliferative activity on the CaSki tumor cell line. We discounted 6 any slight increase in the cellular proliferation rate associated with the 7 retinoid agonist, because a ten thousand fold increase in the retinoid 8 agonist concentration was associated with only roughly a 20% increase 9 in the proliferation rate. Thus, the AGN 191183 agonist had substantially no effect on the proliferation rate of CaSki cells.
11 The results presented in Figure 16 also indicated that AGN
12 193109 inhibited proliferation of the CaSki cervical epithelial cell line.
13 This conclusion was based on comparison of the measurements 14 appearing as the "0" AGN 191183 black bar and the "0" AGN 191183 stripped bar. Thus, AGN 193109 was capable of stimulating a biological 16 response in the absence of added retinoid agonist when used to treat 17 cervical tumor cells that were not growth inhibited by retinoid agonists 18 such as AGN 191183.
19 Our discovery that the AGN 193109 negative hormone could inhibit cellular proliferation was consistent with a model in which 21 unliganded RAR mediated the expression of genes that were required 22 for proliferation. While an RAR agonist such as AGN 191183 had 23 substantially no effect, or perhaps promoted cellular proliferation 24 slightly, AGN 193109 had an antiproliferative effect. The AGN 193109 negative hormone likely bound RARs thereby promoting NCP
26 association and causing the RARs to adopt an inactive conformation.
27 According to our model, this repressed gene activity that was positively 28 regulated by unliganded RARs. This ability of AGN 193109 to down-29 regulate the activity of unliganded RARs likely resulted from its ability to promote the association of RARs and NCPs.
.._._. . . ,.........u , .W. .. ..... ...~..Y...~-...w...._.. .. ..

1 Those having ordinary skill in the art will appreciate that some 2 retinoid agonists are useful for controlling the undesirable consequences 3 of cell growth that follows retinal detachment. After retinal detachment 4 the retinal pigment epithelium (RPE) dedifferentiates, proliferates and migrates into the subretinal space. This process can negatively impact 6 the success of surgical procedures aimed at retinal reattachment.
7 Campochiaro et al. in Invest. Opthal & Vis. Sci. 32:65 ( 1991 ) have 8 demonstrated that RAR agonists such as ATRA exhibited an 9 antiproliferative effect on the growth of primary human RPE cultures.
Retinoid agonists have also been shown to decrease the incidence of 11 retinal detachment following retinal reattachment surgery (Fekrat et al.
12 Opthamology 102:412 (1994)). As disclosed in the following Example, 13 we analyzed the ability of the AGN 193109 negative hormone to 14 suppress growth in primary human RPE cultures.
Example 19 describes the methods used to demonstrate that 16 AGN 193109 potentiated the antiproliferative effect of a retinoid 17 antagonist in a primary culture of human retinal pigment epithelium.
18 Example 19 19 AGN 193109 Potentiates the Anti~roliferative Activity of ATRA
Primary cultures of human retinal pigment epithelium (RPE) 21 were established according to the method described by Campochiaro et 22 al. in Invest. Opthal & Yis. Sci. 32:65 (1991). 5 x 104 cells were plated 23 in 16-mm wells of 24-well multiwell plates in DMEM (Gibco) 24 containing 5% FBS. Cells were mock treated with ethanol vehicle alone, ATRA (10-1~ to 10-6 M) °~ ethanol, AGN ~ 93109 (10v~ to 10-6 M) 26 in ethanol, or ATRA (10-lo to w6 M) and 10-6 1 .~.GN 193109. Cells 27 were fed with fresh media containing the appropriate concentrations of 28 these compounds every two days for a total of five days of treatment.
29 Cells were removed from the plates by gentle digestion with trypsin and the number of cells was counted with an electronic cell counter.

1 The results presented in Figure 17 indicated that AGN 193109 2 dramatically potentiated the antiproliferative activity of ATRA on RPE
3 cells. Treatment of primary RPE cells with ATRA led to a dose 4 dependent decrease in RPE cell proliferation with an approximately 40% decrease at 10-6 M ATRA relative to control cultures. AGN
6 193109 treatment did not substantially alter the growth rate of the RPE
7 cells at any concentration tested in the procedure. Unexpectedly, the 8 combination of ATRA (10-11 to 10-6 M) and 10-6 M AGN 193109 had a 9 stronger antiproliferative activity than ATRA alone. Thus, AGN 193109 cotreatment potentiated the antiproliferative effect of ATRA. More 11 specifically, the results shown in the Figure indicated that the 12 antiproliferative effect of 10-8 M ATRA was obtainable using only 10-10 13 M ATRA in combination with 10-~ M AGN 193109. Thus, the AGN
14 193109 negative hormone advantageously enhanced the antiproliferative activity of ATRA by approximately 100 fold.
16 In an independent experiment, comparison of the antiproliferative 17 effect of ATRA (10-1' to 10-6 M) with that of ATRA and 10-6 M AGN
18 193109 again demonstrated the apparent increase in sensitivity of 19 primary RPE cells to ATRA in the presence of AGN 193109. In this system, AGN 193109 neither functioned as a retinoid antagonist nor 21 exhibited an antiproliferative effect when used alone. However, AGN
22 193109 coadministration potentiated the antiproliferative activity of the 23 retinoid agonist.
24 AGN 193109 was tested for its ability to potentiate the anti-proliferative effect of 13-cis retinoic acid (13-cis RA) in primary RPE
26 cultures using conditions and techniques to measure RPE cell 27 proliferation described above. Notably, 13-cis RA is clinically 28 significant. More particularly, 13-cis RA is useful in the treatment of 29 several disease states, including acne (Peck et al. N. Engl. J. Med.
300:329 (1977); Jones et al. Br. J. Dermatol. 108:333 (1980)), and .... _ . .. _.. .._... _ . ..._~..~~ .-H...._~ w.,-...-. ..~w....__.-....-..
~..~ ...~...... w~..~~... . ... ..,.--..~-w...-. .

1 squamous cell carcinoma of the skin and cervix in combination 2 treatment with interferon 2a (Lippman et al. J. Natl. Cancer Inst. 84:241 3 ( 1992); Moore et al. Seminars in Hematology 31:31 { 1994)).
4 The results presented in Figure 18 indicated that both 13-cis RA
(10-12 to i0-6M) and ATRA (10-12 to 10-6M) effectively inhibited RPE
6 cell growth. Notably, the 13-cis isomer was approximately two orders of 7 magnitude less effective in this assay when compared with ATRA.
8 Similar to the results obtained using coadministration of AGN 193109 9 and ATRA (above), coadministration of AGN 193109 (either 10-g or 10-6M) with 13-cis RA (10-12 to 10-6M) dramatically increased the potency 11 of 13-cis RA in mediating repression of RPE cell proliferation. In 12 contrast to treatment with 13-cis RA alone, coadministration of AGN
13 193109 enhanced the potency of 13-cis RA. Thus, AGN 193109 14 potentiated the antiproliferative activity of 13-cis RA.
We next tested the ability of AGN 193109 to potentiate the 16 activities of other nuclear receptor hormones in primary RPE cell 17 cultures. Dexamethasone, a synthetic glucocorticoid receptor agonist, is 18 one member of a class of compounds that have been used clinically for 19 their potent anti-inflammatory and immunosuppressive properties.
Thyroid hormone (T3; 3,3',5'-Triiodothyronine) is a natural thyroid 21 hormone receptor agonist used primarily for hormone replacement 22 therapy in the treatment of hypothyroidism. Methods used in these 23 experiments were identical to those described above for procedures 24 employing ATRA and 13-cis RA.
The results of these procedures indicated that coadministration of 26 AGN 193109 and the nuclear receptor agonists potentiaued the 27 antiproliferative activities of the nuclear receptor agonists. More 28 specifically, the results presented in Figure 19 showed that single-agent 29 treatment of RPE cells with either dexamethasone (10-11 to 10-6M) or ATRA ( 10-12 to 10-6M) was substantially unable to inhibit RPE cell 1 proliferation. However, treatment of RPE cells with dexamethasone 2 (10-11 to 10-6M) and either 10-8 or 10-6M AGN 193109 repressed RPE
3 cell proliferation to an extent that approximated the inhibition caused 4 by treatment with ATRA. Similarly, the results presented in Figure 20 indicated that AGN 193109 potentiated the antiproliferative activity of 6 thyroid hormone. Similar to the results obtained using dexamethasone, 7 the proliferation of RPE cells was refractory to single-agent treatment 8 with thyroid hormone (10-~~ to 10-6M). However, co-treatment of RPE
9 cells with thyroid hormone ( 10-' 1 to 10-6M) and AGN 193109 (either 10-8 l0 or 10-6M) inhibited RPE cell proliferation in a thyroid hormone 11 dependent manner. We concluded that AGN 193109 rendered primary 12 RPE cultures sensitive to the anti-proliferative effects of these nuclear 13 receptor agonists. The mechanism by which AGN 193109 mediated 14 these effects likely involved modulation of NCP/RAR interactions.
We additionally examined the effect of AGN 193109 on the 16 expression of marker genes in other experimental systems that were 17 sensitive to retinoid agonists. Both the MRP8 and stromelysin genes 18 are known to be inhibited by retinoid agonists in a variety of biological 19 systems. For example, Wilkinson et al. in J. Cell Sci. 91:221 (1988) and Madsen et al. in J. Invest. Dermatol. 99:299 (1992) have disclosed that 21 MRP8 gene expression was elevated in psoriasis. Conversely, MRP8 22 gene expression was repressed by the retinoid agonist AGN 190168 in 23 human psoriatic skin (Nagpal et al., submitted 1995), in human 24 keratinocyte raft cultures (Chandraratna et al. J. Invest. Dermatol.
102:625 (1994)) and in cultured human newborn foreskin keratinocytes 26 (Thacher et al. J. Invest. Dermatol. 104:594 (1995)). Nagpal et al. in J.
27 Biol. Chem. 270:923 (1995) have disclosed that stromelysin mRNA levels 28 were repressed by retinoid agonists such as AGN 190168 in cultured 29 human newborn foreskin keratinocytes. We analyzed the regulated expression of these genes following treatment of cultured human .. .. . _.......w~ .w..w~~~.._.. ..w.... ...~.e~._...._..... _.., _.......~..M~~.....w 1 newborn foreskin keratinocytes with either the AGN 191183 retinoid 2 agonist or AGN 193109.
3 Example 20 describes the methods used to demonstrate that 4 AGN 193109 inhibited MRP-8 expression in cultured keratinocytes.
Example 20 6 AGN 193109 Inhibits MRP-8 Expression 7 in Keratinoc. es 8 Primary foreskin keratinocytes were isolated according to the 9 procedure described by Nagpal et al. in J. Biol. Chem. 270:923 (1995) and cultured in keratinocyte growth medium {KGM) that was purchased l1 from Clonetics. After 3 days of treatment with AGN 191183 (10-~ M) 12 or AGN 193109 (10-6 M), total cellular RNA was isolated from treated 13 and control keratinocytes according to standard methods. The mRNA
14 was reverse transcribed into cDNA which then served as the template in a PCR amplification protocol using primers specific for either the 16 glyceraldehyde phosphate dehydrogenase (GAPDH) housekeeping gene 17 or MRP-8. The GAPDH primers had the sequences 5'-18 CCACCCATGGCAAATTCCATGGCA-3' (SEQ ID N0:6) and 5'-19 TCTAGACGGCAGGTCAGGTCCACC-3' (SEQ ID N0:7). The MRP-8 primers had the sequences 5'-21 ACGCGTCCGGAAGACCTGGT-3' (SEQ ID N0:8) and 5'-22 ATTCTGCAGGTACATGTCCA-3' (SEQ ID N0:9). An aliquot from 23 the MRP-8 amplification reaction (10 ~,1) was removed after every cycle 24 of PCR amplification starting from 12 cycles and ending at 21 cycles.
Similarly, an alic~~: ~t of the GAPDH amplification reaction was removed 26 after every PCR :~~,cle starting at .. ~- cycles and ending at 24 cycles.
The 27 samples were electrophoresed on 2% agarose gels and the separated 28 amplification products detected by ethidium bromide staining. The 29 staining intensity of the amplification products served as a quantitative measure of the amount of starting mRNA specific for the given primer 1 set.
2 The results of this procedure indicated that both AGN 191183 3 and AGN 193109 independently inhibited MRP-8 expression in 4 keratinocytes. The intensity of the stained GAPDH amplification product was substantially equivalent in the lanes of the gel representing 6 starting material isolated from control, AGN 191183, and AGN 193109 7 treated keratinocytes. Weak bands representing the GAPDH
8 amplification product were first detectable in lanes corresponding to 9 samples removed after 18 cycles of PCR amplification. The equivalent l0 staining intensities among the various lanes of the gel indicated that 11 equivalent masses of starting material were used for all samples.
12 Accordingly, differences in the intensities of stained bands representing 13 MRP-8 amplification products were indicative of differences in MRP-8 14 mRNA expression among the various starting samples. As expected, the MRP-8 amplified signal was inhibited in AGN 191183 (10-~ M) treated 16 cultures relative to an untreated control. AGN 193109 ( 10-6 M) 17 treatment of cultured keratinocytes also repressed MRP8 expression as 18 judged by lower intensity of stained amplification product.
19 As illustrated in the following Example, AGN 193109 also inhibited expression of a second marker gene in keratinocytes. Nagpal 21 et al. in J. Biol. Chem. 270:923 (1995) disclosed that stromelysin mRNA
22 expression was down-regulated by RAR specific agonists in cultured 23 newborn human foreskin keratinocytes. Nicholson et al. (EMBO J.
24 9:4443 (1990)) disclosed that an AP-1 promoter element played a role in the retinoid-dependent negative regulation of the stromelysin-1 gene.
26 Thus, it was of interest to determine whether AGN 193109 could alter 27 the expression of this gene.
28 Example 21 describes the methods used to demonstrate that 29 AGN 193109 inhibited stromelysin-1 gene expression in the absence of an exogenously added retinoid agonist.
._ _, . ......d..~...-.__.-~..~ww~..,~-.......__._.W.-~..~ .._..w_,~ --~.....~.......~..~ .._-..

1 Example 21 2 AGN 193109 Inhibits Stromelysin-1 Expression 3 in Cultured Keratinoc_ytes 4 Primary foreskin keratinocytes were either mock treated or treated for 24 hours with the RAR agonist AGN 191183 (10-' M), or 6 AGN 193109 (10-6 M). Total RNA prepared from mock-treated and 7 retinoid-treated keratinocytes was reverse transcribed and the resulting 8 cDNA was PCR amplified using ~3-actin or stromelysin-1 oligo primers 9 exactly as described by Nagpal et al. in J. Biol. Chem. 270:923 (1995)), the disclosure of which has been incorporated by reference. A sample 11 (10 ~cl) from the PCR amplification reaction was removed after every 12 three cycles starting from 18 cycles of PCR amplification. The sample 13 was electrophoresed on a 2% agarose gel and detected after ethidium 14 bromide staining.
Results of these procedures indicated that AGN 193109 inhibited 16 stromelysin-1 gene expression in the absence of an exogenously added 17 retinoid agonist. More specifically, ethidium-stained bands representing 18 ~-actin amplification products were easily detectable the agarose gels 19 after 18 cycles of PCR. While all band intensities increased with additional cycles of the amplification reaction, stained bands were 21 somewhat less intense in samples representing AGN 191183 treated 22 cells. This indicated that a slightly lesser amount of RNA must have 23 been present in the starting samples corresponding to cells treated with 24 AGN 191183. The results also indicated that stromelysin-1 mRNA was detectable in mock-treated keratinocytes starting at 33 cycles of PCR
26 amplification. As expected, ~~rom~lysin-1 mRNA expression was 27 inhibited after AGN 191183 (10-~ M) treatment as judged by the weaker 28 band intensity on when compared with samples derived from mock-29 treated samples. When normalized to the intensities of the /3-actin amplification products, and consistent with the results obtained in 1 measurements of MRP-8 expression, AGN 193109 ( 10-6 M) treatment of 2 keratinocytes resulted in down-regulation of stromelysin-1 mRNA levels.
3 Indeed, the down-regulation stimulated by AGN 193109 treatment was 4 indistinguishable from the down-regulation caused by treatment of keratinocytes with the RAR agonist AGN 191183.
6 As disclosed herein, AGN 193109 can have any of three possible 7 effects with respect to modulating the activity of a coadministered 8 steroid superfamily agonist. First, AGN 193109 may have no effect.
9 Second, AGN 193109 may antagonize the effect of the agonist, thereby l0 leading to a decrease in the activity of the agonist. Finally, AGN
11 193109 may potentiate the activity of the agonist, thereby leading to a 12 stimulation of the measured effect produced by the agonist.
13 Compounds having activities that can be modulated by AGN
14 193109 include retinoid receptor agonists and agonists which bind to other members of the steroid receptor superfamily. This latter category 16 of agonists includes vitamin D receptor agonists, glucocorticoid receptor 17 agonists and thyroid hormone receptor agonists. Peroxisome 18 proliferator-activated receptors, estrogen receptor and orphan receptors 19 having presently unknown ligands may also be potentiated by AGN
193109. In the case where the steroid superfamily agonist is an RAR
21 agonist, AGN 193109 may either antagonize or potentiate the activity of 22 that agonist. In the case where the agonist used in combination with 23 AGN 193109 is a compound that can bind to a nuclear receptor other 24 than an RAR, coadministration of AGN 193109 will either have no effect or will sensitize of the system to the agonist so that the activity of 26 the agonist is potentiated.
27 A generalized exemplary procedure for determining which of the 28 three possible activities AGN 193109 will have in a particular system 29 follows. This description illustrates each of the possible outcomes for AGN 193109 coadministration with a steroid receptor superfamily _.. .... . , .. ~ ~...w . w_.. .....,...,...-.~-.~*.~...,----.*...~"~*.*._.~._ . .. . T ...w.___ ..~_....

1 agonist. Biological systems useful for assessing the ability of AGN
2 193109 to modulate the activity of a nuclear receptor agonist include 3 but are not limited to: established tissue culture cell lines, virally 4 transformed cell lines, ex-vivo primary culture cells and in vivo studies utilizing living organisms. Measurement of the biological effect of AGN
6 193109 in such systems could include determination of any of a variety 7 of biological endpoints. These endpoints include: analysis of cellular 8 proliferation, analysis of programmed cell death (apoptosis), analysis of 9 the differentiation state of cells via gene expression assays, analysis of the ability of cells to form tumors in nude mice and analysis of gene 11 expression after transient or stable introduction of reporter gene 12 constructs.
13 For illustrative purposes, an mRNA species designated as mRNA
14 "X" is expressed from gene "X" in primary cultured "Y" cells isolated from the organ "Z." Under standard culture conditions, where several 16 "Y" cell genetic markers are maintained, including expression of gene 17 "X", addition of a retinoid agonist leads to a decrease in the abundance 18 of "X" mRNA. Analysis of gene X expression can be assessed via 19 isolation of cellular mRNA and measurement of the abundance of X
mRNA levels via polymerase chain reaction, ribonuclease protection or 21 RNA blotting procedures such as Northern analyses. After isolation 22 from organ Z, primary Y cells are cultured in an appropriate growth 23 medium. The primary cultures are then plated into tissue culture plates 24 for expansion of the cell population. This step facilitates separation of the cells into four sample groups so that various doses of the retinoid 26 a gist anc' s~GN 193109 can be delivered. The first group will be a 27 control, receiving vehicle only. The second group will receive the RAR
28 agonist, retinoic acid, delivered in ethanol, in amounts sufficient to 29 provide final concentrations in the range of from 10-11 to 10-6 M. The lowest dose may need to be empirically determined depending on the 1 sensitivity of the system. Such determinations fall within the scope of 2 routine experimentation for one having ordinary skill in the art. The 3 third group will receive both the nuclear receptor agonist at the same 4 doses used for treating the cells of group 2, and a constant dose of AGN 193109. The dose of AGN 193109 used for treating the cells of 6 group 3 will also need to be determined empirically, but should 7 approximate the affinity constant (Kd) of AGN 193109 for the RAR
8 subtypes (i.e., at least 10-8 M). The fourth group will receive AGN
9 193109 at doses minimally including that used for agonist l0 coadministration in group 3. An alternative to this dosing regimen 11 would substitute AGN 193109 for the retinoid agonist described in the t 2 foregoing example, as specified in group 2, and a constant dose of 13 retinoid agonist in place of AGN 193109, as specified in groups 3 and 4 14 After a suitable incubation period, cells should be harvested in a manner suitable for determination of the biological endpoint being 16 measured as an indicator of agonist activity.
17 For example, analysis of the effect of AGN 193109 on retinoic 18 acid dependent regulation of gene expression would involve comparison 19 of the abundance of the mRNA species X in the mRNA pool harvested from cells treated according to each of the four protocols described 21 above. RNA derived from control cells will serve to determine the 22 baseline expression of X mRNA and will represent a condition 23 corresponding to no repression. Comparison of this level with that 24 measured in the mRNA pool derived from cells treated with retinoic acid will allow for determination of the effect of this agonist on gene 26 expression. Quantitated levels of the repression of specific mRNAs 27 resulting from retinoic acid treatment can then be compared with 28 mRNA abundances from cells treated in parallel with either AGN
29 193109 alone or AGN 193109 in combination with retinoic acid. While this generalized example illustrates an analysis of the effect of .. ... .._ .._. ~.-~.,....~...~. _ _ ...~.~m_.._ _. .. _.. _.

1 coadministered AGN 193109 on the expression of a gene repressed by a 2 retinoid agonist, the example could alternatively have described analysis 3 of the effect of coadministered AGN 193109 on a gene that was induced 4 by a retinoid agonist. The critical feature for determining whether AGN 193109 will behave as an agonist, as a negative hormone or have 6 no effect in a particular system will involve quantitative comparison of 7 the magnitude of the effect in the presence and absence of AGN
s 193109.
9 An example in which AGN 193109 potentiated the activity of a coadministered agonist would be a case in which AGN 193109 11 cotreatment with retinoic acid resulted in a level of X mRNA
12 expression that is further repressed relative to the level measured in 13 cells treated with retinoic acid alone. More specifically, comparison of 14 the dose response curve of the biological effect (i.e., repression of X
mRNA abundance) plotted on the Y-axis versus the dose of the agonist 16 (logarithmic scale) on the X-axis would allow comparison of agonist-17 mediated repression of X mRNA abundance in the presence and 18 absence of AGN 193109 cotreatment. The ability of AGN 193109 to 19 sensitize the biological response to the agonist, thereby potentiating the activity of the agonist, will be indicated by a leftward shift in the dose 21 response curve. More specifically, in the presence of AGN 193109 less 22 agonist would be required to obtain the same biological effect 23 obtainable using the agonist alone.
24 An example of AGN 193109 mediating antagonism of a coadministered agonis uld be a cap a in which AGN 193109 26 cotreatme~ ~ -~rith retinc: acid resulted in a level of X mRNA
27 expression that is less repressed compared to that measured in cells 28 treated with retinoic acid alone. Comparison of dose response curves of 29 X mRNA repression versus log dose of agonist in the presence and absence of AGN 193109 will demonstrate a shift to the right in the dose 1 response curve. More specifically, in the presence of AGN 193109, 2 more agonist will be necessary to obtain the same biological effect 3 obtainable with single agent treatment with the agonist alone.
4 The above examples wherein AGN 193109 mediates either antagonism or potentiation describe experimental outcomes for 6 coadministration of AGN 193109 with a retinoid agonist. If, however, 7 the agonist coadministered with AGN 193109 is an agonist capable of 8 binding and activating a member of the steroid receptor superfamily 9 other than an RAR, then instead of antagonizing the agonist, it becomes possible that AGN 193109 would have no effect on the activity 11 of the agonist. If AGN 193109 cotreatment with such an agonist results 12 in a level of mRNA expression which is equal to that measured in cells 13 treated with agonist alone, then AGN 193109's ability to affect the 14 availability of NCPs via promotion of RAR:NCP associations will be silent in this system. This would be an example wherein AGN 193109 16 has no effect on a coadministered agonist.
17 Example of Antagonism 18 The method disclosed in the above generalized example for 19 determining the effect of AGN 193109 coadministered with a retinoid agonist is exemplified by the procedure described under Example 7.
21 CV-1 cells cotransfected with one of the three retinoic acid receptors 22 and the retinoid agonist inducible MTV-TREp-Luc reporter construct 23 were dosed with either ethanol (control, group 1), AGN 193109 at final 24 concentrations of from 10-9 to 10-6 M (group 2), AGN 193109 at final concentrations of from 10-9 to 10-6 M coadministered with retinoic acid 26 at 10-8 M (group 3), or retinoic acid (10-8 M, group 4). Comparison of 27 the luciferase activity of group 1 with that of group 4 allowed 28 determination of the level of retinoid agonist induced expression of the 29 luciferase reporter gene in the absence of added AGN 193109.
Comparison of luciferase reporter gene expression in cells of group 3 .~......,~......_..-_._.~..~~d~~--..-~- . .~...~...-...~.~w..~.-,...N ~...._ w~....m....

1 with that measured in cells of group 4 indicated that AGN 193109 2 behaved as an antagonist of the retinoid agonist in this system.
3 Example of Anta o~ n, ism 4 The method disclosed in the generalized example for determining the effect of AGN 193109 coadministered with a retinoid agonist was 6 similarly used to determine in Example 17 that AGN 193109 functioned 7 as an antagonist of a retinoid agonist-mediated repression of EGF-8 stimulated cellular proliferation in ECE-16-1 transformed cervical 9 epithelial cells. In this procedure, treatments of ECE-16-1 cells l0 included a control sample treated with EGF alone (group 1), a sample 11 treated with the combination of EGF and AGN 193109 at a final 12 concentration of 10-6 M (group 2), a sample treated with the 13 combination of EGF and AGN 193109 at final concentrations of from 14 10-~~ to 10-6 M coadministered with a single dose of the retinoid agonist AGN 191183 at 10-$ M (group 3), and a sample treated with the 16 combination of EGF and AGN 191183 at 10-8 M (group 4). After three 17 days of treatment, cellular proliferation rates were determined.
18 Determination that the cells had been stimulated to proliferate by EGF
19 was possible because an additional control treatment was included wherein cells were exposed to defined medium that did not contain 21 EGF. Comparison of the number of cells in group 1 with the number 22 of cells in group 4 allowed for determination that RAR agonist AGN
23 191183 repressed the EGF-stimulated proliferation of ECE-16-1 cells.
24 Comparison of group 3 with group 4 indicated that AGN 193109 antagonized the activity of the RAR agonist in this system.
26 Example of Potentiation 27 The method disclosed in the generalized example for determining 28 the effect of AGN 193109 coadministered with a retinoid agonist was 29 also used in Example 14 to determine that AGN 193109 potentiated the activity of a nuclear receptor agonist in HeLa cells transfected with the 1 1,25-dihydroxyvitamin D3 inducible MTV-VDRE-Luc reporter gene.
2 Treatments of transfected cells included vehicle alone (control, group 3 1 ), 1,25-dihydroxyvitamin D3 at final concentrations of from 10-lfl to 10-' 4 M (group 2), 1,25-dihydroxyvitamin D3 at final concentrations of from 10-1 to 10-~ M coadministered with AGN 193109 at a final concentration 6 of either 10-8 or 10-~ M (group 3), and AGN 193109 as a single agent 7 treatment at a final concentration of either 10-8 or 10-~ M {group 4).
8 Comparison of the luciferase activity measured in group 1 (control) cells 9 with that of group 2 cells allowed for determination that 1,25-dihydroxyvitamin D3 stimulated luciferase activity was dose-dependent.
11 Comparison of luciferase activity measured in cells of group 4 (AGN
12 193109 single agent treatment) with that measured in cells of group 3 13 (AGN 193109 coadministration) similarly allowed for determination of 14 dose-dependent 1,25-dihydroxyvitamin D3 stimulated luciferase activity in the presence of a given concentration of AGN 193109. In this 16 instance, the zero value represented the luciferase activity in cells 17 treated with AGN 193109 alone (group 4). Such a dosing regimen 18 allowed for comparison of three 1,25-dihydroxyvitamin D3 dose response 19 curves. Comparison of the dose response curve of 1,25-dihydroxyvitamin D3 in the absence of AGN 193109 with the curve 21 representing coadministration of AGN 193109 (either 10-8 or 10-~ M) 22 demonstrated potentiation of the agonist activity as evidenced by a 23 leftward shift in the half-maximal response.
24 Example of Potentiation The method disclosed in the generalized example for determining 26 the effect of AGN 193109 coadministered with a retinoid agonist was 27 further used to determine in Example 19 that AGN 193109 potentiated 28 the antiproliferative activity of an RAR agonist in primary cultures of 29 human retinal pigment epithelium cells. Treatments of cells included:
ethanol vehicle alone (group 1), retinoic acid at final concentrations of _ . ....._~"......w.-...,...~. .."..,.u~,...~,x.w_w~.~...~".~.... ., ~
,~.~._.__.w..""...".. ._....._.....W~....~,..~n...~...

1 from 10-'° to 10-6 M (group 2), retinoic acid at final concentrations of 2 from 10-1° to 10-6 M coadministered with IO-6 M AGN 193109 (group 3), 3 and AGN 193109 alone at final concentrations of from 10-x° to 10-6 M
4 (group 4). Comparison of assay results obtained using cells of groups 1 and 2 allowed for determination of the dose dependent inhibition of 6 proliferation of these cells by retinoic acid. Similarly, comparison of 7 results obtained using cells of group 3 with those of group 1 allowed for 8 determination of the dose dependent inhibition of proliferation of these 9 cells by retinoic acid in the presence of coadministered AGN 193109.
Group 4 demonstrated the inability of AGN 193109 to substantially 11 alter the proliferation rate of these cells when used as a single treatment 12 agent. Comparison of the dose response curves of retinoic acid 13 mediated repression of cellular proliferation generated in groups 2 and 14 3 provided the basis for the conclusion that AGN 193109 sensitized primary RPE cells to the antiproliferative effects of the RAR agonist, 16 thereby potentiating the activity of the RAR agonist.
17 As indicated above, Agarwal et al., in Cancer Res. 54:2108 18 (1994)), showed that CaSki cell growth, unlike the growth of HPV
19 immortalized ECE-16-1 cells, was not inhibited by treatment with retinoid agonists. As disclosed herein, we unexpectedly found that 21 CaSki cell growth was inhibited by AGN 193109 in the absence of a 22 retinoid agonist. The following Example illustrates how AGN 193109 23 can be used to inhibit the growth of CaSki cell tumors in vivo.
24 Example 22 Inhibition of raSki CeII Tumor Gro =.~th in Nude Mice 26 Follom.:g Administration of AGN 193109 27 1 X 106 CaSki cells are injected into each of a panel of nude 28 mice. Tumor formation is assessed using techniques that will be 29 familiar to one having ordinary skill in the art. After injection, mice are randomly divided into control and test groups. The control group 1 receives a placebo. The test group is administered with AGN 193109.
2 Animals administered with the placebo receive intragastric intubation of 3 corn oil. The test animals receive 20 ~,Mol/kg AGN 193109 in corn oil 4 daily for the period of the treatment. Tumor volume is measured in cubic milliliters using graduated calipers. Tumor volume is plotted as 6 function of time. Mice receiving AGN 193109 exhibit tumors which are 7 significantly reduced in their growth rate as compared to tumors in 8 control mice as judged by tumor size and number over the period of the 9 study. This result provides an in vivo demonstration that AGN 193109 inhibits the growth of an advanced cervical carcinoma that is resistant to 11 therapy comprising administration of a retinoid agonist.
12 As indicated above, CaSki cells are a model of cervical tumors 13 that are not responsive to retinoid agonist therapy. However, herein we 14 have disclosed that CaSki cell growth was inhibited by AGN 193109 in the absence of treatment with a retinoid agonist. The ability of AGN
16 193109 to inhibit the proliferation of CaSki cells suggested that AGN
17 193109 could be used to therapeutically treat cervical carcinomas that 18 are insensitive to retinoid agonist therapy. The following Example 19 illustrates one method that can be used to assess the therapeutic potential of AGN 193109 in the treatment of a cervical carcinoma.
21 Example 23 22 Assessing the Therapeutic Potential of AGN 193109 23 in Patients Having Cervical Carcinoma 24 A patient presenting with an advanced cervical carcinoma is first identified. A cervical biopsy is obtained according to methods that will 26 be familiar to one having ordinary skill in the art. Cells from the 27 explanted tumor are propagated in tissue culture according to standard 28 techniques to provide cell numbers sufficient to allow division into three 29 sample groups. Culture conditions described by Agarwal et al. in Cancer Res. 54:2108 (1994) are employed for this purpose. The first __ W._~~.._...........,~....~..........~~.._._ ~,._.....~...~...~W...rv~._._._ .. _ _._.~~..~ . ..

1 group is reserved as a control and receives vehicle alone (ethanol). The 2 second group is treated with the RAR agonist retinoic acid at a 3 concentration of from 10-1 to 10-6 M. The third group is treated with 4 AGN 193109 at doses ranging from 10-1 to 10-6 M. Cells are fed with fresh growth medium daily and are provided with the retinoids 6 described above as appropriate for each sample group. Cells are 7 counted after three days using an electric cell counter. Comparison of 8 the number of cells in control cultures with the number of cells in 9 retinoic acid treated cultures indicates the RAR agonist does not substantially inhibit the growth rate of the cultured cervical carcinoma 11 cells. In contrast, cells treated with AGN 193109 exhibit a dose-12 dependent decrease in cell number when compared with cell counts in 13 the control group. This result, wherein AGN 193109 treatment inhibits 14 cultured cervical carcinoma cell proliferation, indicates that AGN
193109 will be a useful therapeutic agent for treating cervical carcinoma 16 patients having metastatic disease.
17 Cervical carcinoma patients having undergone surgery for the 18 removal of primary tumors and who present with metastatic disease are 19 enlisted in a randomized clinical study seeking to demonstrate the therapeutic benefit of AGN 193109 in this indication. Patients are 21 divided into two groups. The first group is a control group while 22 members of the second group are treated with AGN 193109. AGN
23 193109 is combined with a pharmaceutically acceptable excipient to 24 produce a composition suitable for systemic administration, all according to te.:.rl:iques that ~~ill be familiar to one having ordinary skill 26 in the art. The control grouFis administered a placebo formulation and 27 the experimental group is administered with the formulation containing 28 the AGN 193109 negative hormone. Dosing of patients is at the 29 maximum tolerated dose and is performed every other day for a period of from three months to one year. The outcome of the study is 1 quantified via measurement of disease-free survival over time.
2 Individuals receiving AGN 193109 display a significant increase in 3 disease-free survival, including a disproportionate number of patients 4 displaying complete remission of their metastatic disease. This result indicates that AGN 193109 has therapeutic utility for in vivo treatment 6 of cervical carcinomas that are unresponsive to the antiproliferative 7 effects of retinoid agonists, such as retinoic acid.
8 As disclosed above, AGN 193109 potentiated the antiproliferative 9 activity of RAR agonists in primary cultures of human retinal pigment epithelium cells. Accordingly, coadministration of AGN 193109 with an 11 RAR agonist in vivo is reasonably expected to increase the therapeutic 12 index of the agonist because a lesser amount of the RAR agonist will be 13 required to obtain the same therapeutic endpoint. Additionally, AGN
14 193109 has been demonstrated to sensitize primary cultures of human retinal pigment epithelium cells to the antiproliferative effects of 16 glucocorticoid and thyroid hormone receptor agonists. The following 17 rabbit model of PVR will be utilized in two separate studies to 18 demonstrate the increased therapeutic index obtained via 19 coadministration of AGN 193109 with an RAR agonist (13-cis retinoic acid) or a thyroid hormone receptor agonist, respectively. Notably, the 21 rabbit model of retinal redetachment published by Sen et al. in Arch.
22 Opthalmol. 106:1291 (1988), has been used to demonstrate that retinoid 23 agonists which inhibit proliferation of primary RPE cells in vitro also 24 inhibit the frequency of retinal detachment in vivo (Araiz et al. Invest.
Opthalmol. 34:522 (1993)). Thus, with respect to their use as 26 therapeutics in the prevention of retinal detachment, a correlation 27 between the in vitro and in vivo activities of retinoid agonists has already 28 been established. The following Examples illustrate how AGN 193109 29 can be used in therapeutic applications directed at preventing retinal detachment.

1 Example 24 2 Use of AGN 193109 to Increase the Therapeutic Potential of 3 Steroid Superfamily Receptor Agonists in the Treatment 4 of Proliferative Vitreoretinopathy (PVR) In a first study, human RPE cells are injected into the vitreous 6 cavity of rabbit eyes according to the method described by Sen et al. in 7 Arch. Opthalmol. lOG:1291 (1988). After intravitreal injection, the 8 rabbits are divided into five groups. The first group (control) will 9 receive vehicle alone by intravitreal injection. The second group receives retinoic acid as single agent treatment {100 E.cg) by intravitreal 11 injection. The third group receives AGN 193109 as a single agent 12 treatment (100,ug) by intravitreal injection. The fourth group receives 13 by intravitreal injection the RAR agonist (retinoic acid) at a dose one-14 tenth the amount administered to group 2 (l0~cg). The fifth group receives the combination of AGN 193109 (100,ug) and retinoic acid 16 (10~,g) by intravitreal injection. Animals receive a single intravitreal 17 injection of the appropriate treatment one day after intravitreal 18 injection of human RPE cells. Rabbits are examined by indirect 19 ophthalmoscopy on days 7, 14 and 28, and are graded for the frequency and severity of tractional retinal detachment. Rabbits from the group 21 injected with 100 ~,g retinoic acid exhibit a significantly reduced 22 frequency and severity of retinal detachment compared to control 23 rabbits or rabbits receiving either AGN 193109 or retinoic acid (l0~cg) 24 alone. Rabbits in the group administered with the combination of AGN
193109 and retinoic acid ( l0,ug) exhibit significantly reduced frequency 26 and severity of retinal detachment as c~,~npared to those in groups 27 either control, AGN 193109 or retinoic acid (10~,g). This result 28 demonstrates that AGN 193109 improves the therapeutic index of the 29 RAR agonist retinoic acid in an in vivo model of PVR.
In a second study, rabbits are first provided with an injection of 1 human RPE cells into the vitreous cavity of the eye, and then divided 2 into four groups. The first group (control) receives vehicle alone by 3 intravitreal injection. The second group receives thyroid hormone as 4 single agent treatment (100 ~.cg) by intravitreal injection. The third group is administered with AGN 193109 as a single agent treatment 6 (100 ~,g) by intravitreal injection. The fourth group is administered with 7 the combination of AGN 193109 (100,ug) and thyroid hormone (100 8 ~,g). Rabbits are examined by indirect ophthalmoscopy on days 7, 14 9 and 28, and graded for the frequency and severity of tractional retinal l0 detachment. Comparison of the frequency and severity of retinal 11 detachment in the four groups demonstrates that single agent treatment 12 with either AGN 193109 or thyroid hormone does not inhibit retinal 13 detachment when compared with control rabbits. In contrast, the group 14 of rabbits administered with the combination of AGN 193109 and thyroid hormone exhibit significantly reduced incidence and severity of 16 retinal detachment. This result demonstrates that AGN 193109 17 improves the therapeutic index of thyroid hormone in an in vivo model 18 of PVR.
19 The following Example illustrates how AGN 193109 can be used to enhance the therapeutic index of an RAR agonist used to treat 21 human patients following retinal reattachment surgery.
22 Example 25 23 Increasing the Therapeutic Index of RAR Agonist 24 13-cis Retinoic Acid A population of adult volunteers having retinal detachment 26 resulting from PVR is first identified. Individuals undergo surgical 27 repair of the detachments using techniques that are standard in the art.
28 The patients are then divided into five groups. The control group 29 consists of patients who undergo surgical repair of the retinal detachment and do not receive any retinoid compound. The second ... .z._~... ~..w.....~-,~.,.~~....... . ~....~. ~.'.~.~_ W..~. .....
~~.....~... .

1 group receives 40 mg oral 13-cis retinoic acid twice daily for four weeks 2 postoperatively. The third group receives 40 mg oral AGN 103109 3 twice daily for four weeks postoperatively. The fourth group receives 4 4 mg oral 13-cis retinoic acid twice daily for four weeks postoperatively.
The fifth group receives 40 mg oral AGN 193109 in combination with 4 6 mg oral 13-cis retinoic acid twice daily for four weeks postoperatively.
7 The treatment protocol and assessment of drug efficacy is performed 8 essentially as described by Fekrat et al. in Ophthalmology 102:412 9 ( 1995).
The frequency and severity of retinal redetachment in 11 postoperative patients in all five groups is monitored over a period of 12 nine months using ophthalmologic examination techniques that will be 13 familiar to those of ordinary skill in the art. Patients receiving 40 mg 14 oral 13-cis retinoic acid exhibit significantly reduced incidence of retinal redetachment when compared with control patients, patients receiving 4 16 mg oral 13-cis retinoic acid twice daily or patients receiving 40 mg oral 17 AGN 193109 twice daily. Examination of the patient group receiving 18 the combination of 40 mg oral AGN 193109 and 4 mg oral 13-cis 19 retinoic acid twice daily for four weeks postoperatively demonstrates the therapeutic outcome in this patient group is equal to or better than 21 those patients receiving 40 mg oral 13-cis retinoic acid twice daily for 22 four weeks postoperatively. This result demonstrates that the AGN
23 193109 negative hormone improves the therapeutic index of an RAR
24 agonist by virtue of decreasing the frequency and severity of retinal re~'Ptachment in PVR patients.
26 G-_ _y~ralized Assay for Identifying Nuclear Receptor Negative Hormones 27 We have demonstrated above that AGN 193109 can function as a 28 negative hormone capable of repressing the basal transcriptional activity 29 of RAR nuclear receptors. Further, we have described an assay using CV-1 cells co-transfected with the ERE-tk-Luc luciferase reporter 1 plasmid and the ER-RXR-a and RAR-y-VP-16 receptor expression 2 plasmids for distinguishing RAR ligands that are simple antagonists 3 from those having negative hormone activity.
4 We have concluded that RAR negative hormones mediate repression of RAR-mediated transcriptional activity by promoting 6 increased interaction between the RAR and NCPs. Further, we have 7 demonstrated that AGN 193109 can potentiate the effects of agonists of 8 other nuclear receptors in a manner consistent with the mutual sharing 9 of NCPs between members of the steroid superfamily of nuclear l0 receptors. As such, ligands can be designed and screened to identify 11 compounds having negative hormone activity at these non-RAR nuclear 12 receptors.
13 Our method of RAR negative hormone screening based on the 14 use of CV-1 cells co-transfected with the ERE-tk-Luc luciferase reporter plasmid and the ER-RXR-a and RAR-'y-VP-16 receptor 16 expression plasmids can be adapted generally such that the RAR-y 17 moiety of the RAR-y-VP-16 plasmid is converted to that of peroxisome 18 proliferator-activated receptors (PPAR), vitamin D receptor (VDR), 19 thyroid hormone receptor (T3R) or any other steroid superfamily nuclear receptor capable of heterodimerizing with RXR. CV-1 cells co-21 transfected with such plasmids would express high basal levels of 22 iuciferase activity. Ligands capable of binding the ligand binding 23 domain of the receptor substituted for the RAR-'y moiety can be easily 24 screened for negative hormone activity by measuring their ability to repress luciferase activity.
26 For steroid superfamily nuclear receptors that do not 27 heterodimerize with RXR (e.g., glucocorticoid and estrogen receptors) 28 the same end result can be achieved using GR-VP-16 or ER-VP-16 29 receptors and a luciferase reporter plasmid consisting of the appropriate glucocorticoid or estrogen response element fused to a heterologous ~.r..._.~-w..,...m....~._... ,.....~~,..,.m-~.--..~._.~..__.._.-.-.~_M
....w~_ww.-.......~...............___.

1 promoter element and luciferase or other reporter gene. An essential 2 feature of a generalized negative hormone screening assay is the 3 inclusion of at least the iigand binding domain of the particular nuclear 4 receptor for which inverse agonists are to be screened and a method for localizing the nuclear receptor ligand binding domain to the promoter 6 of a reporter gene. This could be achieved using the receptors's natural 7 DNA binding site, or alternatively by construction of a chimeric 8 receptor having a heterologous DNA binding domain and corresponding 9 use of a reporter gene which is under control of a DNA regulatory l0 element which is recognized by the heterologous DNA binding domain.
11 In a preferred embodiment, the plasmid expressing the nuclear receptor 12 for which inverse agonists are to be screened would express this nuclear l3 receptor as a fusion protein containing a constitutive activation domain, 14 such as the HSV VP-16 activation domain, in order to provide allow high basal activity. This high basal activity would effectively increase 16 assay sensitivity, thereby allowing analysis of nuclear receptor ligands 17 which repress basal transcriptional activity in the absence of added 18 nuclear receptor agonist.
19 The following Example illustrates one method that can be used to screen for compounds having negative hormone activity at the thyroid 21 hormone receptor.
22 Example 26 23 Method of Identifying Thyroid Hormone Receptor 24 Negative Hormones CV-1 cells are co-transfected with the luciferase reporter plasmid 26 ERE-tk-Luc and the plasmids ER-RXR-a and T3R-V~ - i6. T3R-VP-16 27 is identical to the plasmid RAR-y-VP-16, except the RAR-y moiety of 28 RAR-y-VP-16 has been substituted by the thyroid hormone receptor 29 cDNA. As such, T3R-VP-16 expresses a fusion protein containing the activation domain of HSV VP-16 in frame with the N-terminus of the 1 thyroid hormone receptor. Standard transfection and cell culture 2 methods are employed for this purpose. After transfection, cells are 3 rinsed and fed with growth medium containing 10% fetal calf serum 4 which has been extracted with activated charcoal. Cells are treated with vehicle alone (ethanol), thyroid hormone (10-9 to 10-~~ M), or compound 6 TR-1 (10-9 to 10-6 M). TR-1 is a synthetic thyroid hormone receptor 7 ligand which exhibits strong affinity for the thyroid hormone receptor in 8 competition binding studies, but which does not activate transfected 9 thyroid hormone receptor in transient cotransfection transactivation to assays using a thyroid hormone responsive reporter gene and a thyroid 11 hormone receptor expression plasmid. Further, TR-1 is capable of 12 antagonizing thyroid hormone mediated transactivation and as such is a 13 thyroid receptor antagonist.
14 Analysis of luciferase activity from CV-1 cell transfected with ERE-tk-Luc, ER-RXRa and T3R-VP-16 demonstrates a high basal 16 level of luciferase reporter activity in vehicle-treated cells. Cells treated 17 with thyroid hormone show a slight increase of luciferase activity in a 18 dose dependent manner. Cells treated with TR-1 exhibit a dose 19 dependent decrease in luciferase activity. This indicates that TR-1 exhibits thyroid receptor inverse agonist activity, presumably due to the 21 increased interaction of a NCP with the thyroid hormone receptor.
22 The proliferation rate of human primary retinal pigment 23 epithelium cells is repressed by treatment with RAR agonists. The 24 therapeutic value of this observation has been demonstrated in post-operative use retinoid therapy after retinal reattachment surgery. We 26 have above demonstrated the AGN 193109 RAR negative hormone can 27 sensitize primary RPE cells to the antiproliferative effect of ATRA and 28 13-cis retinoic acid in coadministration procedures. Further, AGN
29 193109 was also shown to sensitize RPE cells to the antiproliferative effects of other nuclear receptor agonists. More specifically, AGN
..__..._~..~.,."..."~.~..... w~......_....,.~...w....~..~.....~.~..
T..._~~..~._._ .....~._~...-_~..-~~.-..

1 193109 sensitized RPE cells to the antiproliferative effects of the 2 glucocorticoid agonist, dexamethasone, and the thyroid hormone agonist 3 3,3',5-triiodothyronine, T3. This data was consistent with our working 4 model wherein AGN 193109 modulated the availability of NCPs that were shared between the members of the nuclear receptor family.
6 Treatment of RPE cells with the thyroid hormone receptor inverse 7 agonist TR-1 will similarly alter the availability of shared NCPs such 8 that coadministration with a non-thyroid receptor agonist, such as the 9 RAR agonist 13-cis retinoic acid will lead to an increased antiproliferative effect upon the RPE cultures as compared to 13-cis I I retinoic acid as a single agent treatment.
12 The following Example illustrates one method that can be used to 13 render primary RPE cells more sensitive to the antiproliferative activity 14 of an RAR agonist. Notably, this Example further illustrates how the activity of RAR agonists can be potentiated by coadministration with a 16 negative hormone.
I ~ Example 27 18 Sensitizing Primary Retinal Pi mg ent Epithelium Cells I9 to the Antiproliferative Effects of RAR A onists by Coadministration of the TR-1 Thyroid Hormone 21 Inverse A og,~nist 22 Human primary RPE cells are obtained and cultured according to 23 standard methods. The cultured cells are divided into four groups and 24 treated as follows. Group 1 receives vehicle alone (ethanol). Group 2 is treated with 13-cis retinoic acid at concentrations ranging from 10-26 ~ ~ to 10-6 M. Group 3 is treated with the thyroid hormone inverse 27 agonist TR-1 at concentrations ranging from 10-'1 to 10-~ M. Group 4 is 28 co-treated with 13-cis retinoic acid at concentrations ranging from 10-29 1~ to 10-6 M TR-1. Cells are refed with fresh growth medium and re-treated with the appropriate compound every two days for a total of five 1 days of treatment. The proliferation rate over the duration of the 2 experiment is quantitated via measurement of the cell number in the 3 cultures using an electric cell counter.
4 TR-1 treated cells (Group 3) exhibits rates of cellular proliferation which are essentially the same as control (Group 1) cells 6 and there is no effect of this inverse agonist upon the measured growth 7 rate of the cultures. Cells treated with 13-cis retinoic acid {Group 2) 8 exhibit a dose dependent decrease in cell number. Comparison of the 9 dose dependent decrease in cellular proliferation of Group 4 cells l0 (13-cis RA and TR-1 coadministration) with that obtained in Group 3 11 demonstrates the ability of TR-1 thyroid hormone receptor inverse 12 agonist coadministration to sensitize RPE cultures to the 13 antiproliferative effect of 13-cis retinoic acid as measured by the shift in 14 the dose response curve of this RAR agonist to the left in Group 4 as compared to Group 2 cells.
. .m.w,....~._~..~.~.-.~...~...~..~.... ~-..w~.~
....,...~..~~..~..w.~~.~.~..~.--......h.....~...~..~.~."-._.-~.~. .... .. .

SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: Vision Pharmaceuticals L.P., a Texas Limited Partnership doing business as Allergan.
(ii) TITLE OF INVENTION: SYNTHESIS AND USE OF RETINOID
COMPOUNDS HAVING NEGATIVE HORMONE AND/OR ANTAGONIST ACTI
VITIES
(iii) NUMBER OF SEQUENCES: 9 (iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Allergan {B) STREET: 2525 Dupont Drive (C) CITY: Irvine (D) STATE: CA
{E) COUNTRY: USA
(F} ZIP: 92612 (v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Diskette (B) COMPUTER: IBM Compatible (C) OPERATING SYSTEM: Windows 95 (D) SOFTWARE: FastSEQ for Windows Version 2.0 (vi) CURRENT APPLICATION DATA:
{A) APPLICATION NUMBER:
(B) FILING DATE:
(C) CLASSIFICATION:
(vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: 08/613,863 (B) FILING DATE: 11-MAR-1996 (A) APPLICATION NUMBER: 08/522,778 (B) FILING DATE: O1-SEP-1995 (A) APPLICATION NUMBER: 08/522,779 (B) FILING DATE: 01-SEP-1995 (A) APPLICATION NUMBER: 08/542,648 (B) FILING DATE: 13-OCT-1995 (viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Fisher :~arlos A
( B ) REGI STRATIOI '~.J~ER : 3 6 , 510 (C) REFERENCED: .'t' NUMBER: 17171CIP
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: 714-246-4920 (B) TELEFAX: 714-246-4249 (C) TELEX:

(2) INFORMATION FOR SEQ ID NO:1:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 21 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID NO:1:

(2) INFORMA'PION FOR SEQ ID N0:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 101 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID N0:2:

(2) INFORMATION FOR SEQ ID N0:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 101 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID N0:3:

(2) INFORMATION FOR SEQ ID N0:4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 28 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID N0:4:

(2) INFORMATION FOR SEQ ID N0:5:
(i) SEQUENCE CHARACTERISTICS:
SUBSTITUTE SHEET (RULE 26) _. .. .... ~,~~ ~... .~.._...~_.. ~.~~.... _ fi....ri........ ._.....~ww-..r..~:~.w.._~.w~._,~.

(A) LENGTH: 16 base pairs 226 (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID N0:5:

(2) INFORMATION FOR SEQ ID N0:6:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 24 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID N0:6:

(2) INFORMATION FOR SEQ ID N0:7:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 24 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID N0:7:

(2) INFORMATION FOR SEQ ID NO: B:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 20 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID N0:8:

(2) INFORMATION FOR SEQ ID N0:9:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 20 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (xi) SEQUENCE DESCRIPTION: SEQ ID N0:9:

SUBSTITUTE SHEET (RULE 26)

Claims (34)

    CLAIM

    WHAT IS CLAIMED IS:
  1. Claim 1. (CANCEL)
  2. Claim 2. (CANCEL)
  3. Claim 3. (CANCEL)
  4. Claim 4. (CANCEL)
  5. Claim 5. (CANCEL)
  6. Claim 6. (CANCEL)
  7. Claim 7. (AMENDED) A compound of the formula wherein R2 is lower alkyl of 1 to 6 carbons, F, Cl, Br, I, CF3, fluoro substituted alkyl of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 6 carbons;
    m is an integer having the value of 0 - 3;
    R3 is lower alkyl of 1 to 6 carbons or F;
    o is an integer having the value of 0 - 3;
    s is an integer having the value of 1 - 3;
    R8 is an alkyl group of 1 to 10 carbons or trimethylsilylalkyl where the CLAIM

    alkyl group has 1 to 10 carbons, or a cycloalkyl group of 5 to 10 carbons, or is phenyl or lower alkylphenyl;

    R15 is independently H, F, Cl, Br, I, NO2, N(R8)2, COR8, NR8CON(R8)2, CCOR8, OR8, CN, an alkyl group having 1 to 10 carbons, fluoro substituted alkyl group having 1 to 10 carbons, an alkenyl group having 1 to 10 carbons and 1 to 3 double bonds, alkynyl group having 1 to 10 carbons and 1 to 3 triple bonds, or a trialkylsilyl or trialkylsilyloxy group where the alkyl groups independently have 1 to 6 carbons;
    r is an integer having the values of 0 - 5, and the CONH group is in the 6 or 7 position of the benzopyran, or a pharmaceutically acceptable salt of said compound.
  8. 8. A compound in accordance with Claim 7 where r is 1 and R15 is lower alkyl.
  9. 9. A compound in accordance with Claim 8 where R15 is 4-methyl.
  10. 10. A compound in accordance with Claim 7 where R3 is CH3, o is 2 and the CH3 substituents occupy the 2-position of the benzopyran ring.
  11. 11. A compound in accordance with Claim 7 where R2 is Br, and m is 1, and the bromo substituent is in the 8-position of the benzopyran ring.
  12. 12. (AMENDED) A compound of Claim 7 characterized by the formula CLAIM

    where R2 is H or Br;
    s is 1 or 2;
    R3 is H or CH3, and R8 is H, lower alkyl of 1 to 6 carbons, or a pharmaceutically acceptable salt of said compound.
  13. 13. (CANCEL)
  14. 14. (CANCEL)
  15. 15. (CANCEL)
  16. 16. (CANCEL)
  17. 17. (CANCEL)
  18. 18. (CANCEL)
  19. 19.
    A compound in accordance with Claim 12 where R3 is CH3.
  20. 20. A compound in accordance with Claim 19 where R2 is Br.
  21. 21. A compound in accordance with Claim 20 where R2' is H and CLAIM

    R2" is H.
  22. 22. A compound in accordance with Claim 21 where R8 is H or ethyl, or a pharmaceutically acceptable salt of said compound.
  23. 23. A compound in accordance with Claim 20 where R2' is F and R2"
    is H.
  24. 24. A compound in accordance with Claim 23 where R8 is H or ethyl, or a pharmaceutically acceptable salt of said compound.
  25. 25. A compound in accordance with Claim 20 where R2' is F and R2"
    is F.
  26. 26. A compound in accordance with Claim 25 where R8 is H or ethyl, or a pharmaceutically acceptable salt of said compound.
  27. 27. (AMENDED) A method of treating a pathological condition in a mammal, said condition associated with a retinoic acid receptor activity, said method comprising administering to said mammal a retinoid antagonist or negative hormone compound capable of binding to a retinoic acid receptor subtype selected from the group consisting of RAR.alpha., RAR.beta. and RAR.gamma., said antagonist or negative hormone being administered in an amount pharmaceutically effective to provide a therapeutic benefit against said pathological condition in said mammal and wherein said antagonist or negative hormone is a compound in accordance with Claim 7.
  28. 28. The method of Claim 27 wherein the pathological condition is the toxicity or undesired side effects resulting from administration of a retinoid compound to said mammal, and wherein said therapeutic benefit is the prevention or amelioration of said toxicity or undesired side effects.
  29. 29. The method of Claim 27 wherein the retinoid antagonist or negative hormone is administered in order to cure or ameliorate a pre-existing pathological condition caused by intake of a retinoid drug or vitamin A or CLAIM

    vitamin A precursors by the mammal.
  30. 30. The method of Claim 27 wherein the retinoid antagonist or negative hormone is administered topically to block or ameliorate undesired topical side effects of a retinoid drug administered for a therapeutic purpose.
  31. 31. The method of Claim 27 wherein the retinoid antagonist or negative hormone is administered topically to block or ameliorate undesired topical side effects of a retinoid drug administered systemically for a therapeutic purpose.
  32. 32. The method of Claim 27 wherein the retinoid antagonist or negative hormone is administered topically to treat a pre-existing condition or side effect caused by a retinoid drug or vitamin A.
  33. 33. The method of Claim 27 wherein the retinoid antagonist or negative hormone is administered systemically to treat a pre-existing condition or side effect caused by a retinoid drug or vitamin A.
  34. 34. The method of Claim 27 wherein the retinoid antagonist or negative hormone is administered systemically to block or ameliorate bone toxicity caused by coadministration of a retinoid drug or vitamin A.
CA002294601A 1997-06-24 1998-06-24 Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities Abandoned CA2294601A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/880,823 1997-06-24
US08/880,823 US5877207A (en) 1996-03-11 1997-06-24 Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities
PCT/US1998/013065 WO1998058922A1 (en) 1997-06-24 1998-06-24 Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities

Publications (1)

Publication Number Publication Date
CA2294601A1 true CA2294601A1 (en) 1998-12-30

Family

ID=25377185

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002294601A Abandoned CA2294601A1 (en) 1997-06-24 1998-06-24 Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities

Country Status (11)

Country Link
US (1) US5877207A (en)
EP (2) EP1535919A3 (en)
JP (1) JP4295361B2 (en)
KR (1) KR100512058B1 (en)
CN (1) CN1142928C (en)
AT (1) ATE289998T1 (en)
AU (1) AU729140B2 (en)
BR (1) BR9810340A (en)
CA (1) CA2294601A1 (en)
DE (1) DE69829183T2 (en)
WO (1) WO1998058922A1 (en)

Families Citing this family (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6008204A (en) 1995-09-01 1999-12-28 Allergan Sales, Inc. Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities
US5952345A (en) 1995-09-01 1999-09-14 Allergan Sales, Inc. Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities
WO2000018885A1 (en) * 1998-09-29 2000-04-06 Gamida Cell Ltd. Methods of controlling proliferation and differentiation of stem and progenitor cells
CA2346687A1 (en) * 1998-10-08 2000-04-13 Allergan Sales, Inc. Use of rar antagonists or inverse agonists as male anit-fertility agents
US6521641B1 (en) * 1998-10-08 2003-02-18 Allergan, Inc. Male anti-fertility agents
CA2254429A1 (en) * 1998-11-19 2000-05-19 Tully Michael Underhill Compositions for promoting chondrogenesis
WO2000034435A2 (en) * 1998-12-08 2000-06-15 Clontech Laboratories, Inc. Cis-element reporter constructs and uses thereof
WO2000061233A2 (en) * 1999-04-14 2000-10-19 Allergan Sales, Inc. Methods and compositions for the treatment and prevention of lung disease involving a specific rar-gamma agonist
AU4225500A (en) 1999-04-14 2000-11-14 Allergan Sales, Inc. Methods and compositions for the treatment and prevention of lung disease
US6166244A (en) 1999-05-07 2000-12-26 Allergan Sales, Inc. Oxygen, sulfur and nitrogen substituted cyclohexene and cyclohexane derivatives having retinoid-like biological activity
US6043381A (en) * 1999-05-07 2000-03-28 Allergan Sales, Inc. Process for preparing substituted benzo[1,2-g]-chrom-3-ene, benzo[1,2-g]-thiochrom-3-ene and benzo[1,2-g]-1,2-dihydroquinoline derivatives
US6177588B1 (en) 1999-05-07 2001-01-23 Allergan Sales, Inc. 1-alkoxy and 1-acyloxy substituted cyclohex-1-ene compounds and sulfur and 1-alkoxycarbonyl analogs having retinoid-like biological activity
DE60016404T2 (en) 1999-06-11 2005-10-27 Allergan, Inc., Irvine Organosilicate compounds having the ability to modulate cell nucleus-localized hormone receptors
US6906057B1 (en) 1999-06-11 2005-06-14 Allergan, Inc. Methods for modulating FXR receptor activity
US6093838A (en) * 1999-08-16 2000-07-25 Allergan Sales, Inc. Amines substituted with a dihydro-benzofuranyl or with a dihydro-isobenzofuranyl group, an aryl or heteroaryl group and an alkyl group, having retinoid-like biological activity
US6127382A (en) * 1999-08-16 2000-10-03 Allergan Sales, Inc. Amines substituted with a tetrahydroquinolinyl group an aryl or heteroaryl group and an alkyl group, having retinoid-like biological activity
US20030114482A1 (en) * 1999-12-15 2003-06-19 Maurizio Pacifici Use of retinoid receptor antagonists or agonists in the treatment of cartilage and bone pathologies
US6313168B1 (en) * 1999-12-15 2001-11-06 Allergan Sales, Inc. Use of retinoid receptor antagonists in the treatment of cartilage and bone pathologies
US6462075B1 (en) 1999-12-23 2002-10-08 The University Of Georgia Research Foundation, Inc. Chalcone and its analogs as agents for the inhibition of angiogensis and related disease states
US20030125252A1 (en) * 2000-03-14 2003-07-03 Underhill T. Michael Compositions and methods for affecting osteogenesis
US6613917B1 (en) 2000-03-23 2003-09-02 Allergan, Inc. Amines substituted with a dihydronaphthalenyl, chromenyl, or thiochromenyl group, an aryl or heteroaryl group and an alkyl group, having retinoid-like biological activity
AU2001290816A1 (en) 2000-09-13 2002-03-26 Bristol-Myers Squibb Company Retinoic acid receptor antagonists as promoters of angiogenesis
US20020193403A1 (en) * 2001-05-03 2002-12-19 Allergan Sales, Inc. Methods of treating hyperlipidemia
EP1465869B1 (en) * 2001-12-21 2013-05-15 Exelixis Patent Company LLC Modulators of lxr
US7482366B2 (en) 2001-12-21 2009-01-27 X-Ceptor Therapeutics, Inc. Modulators of LXR
IL152904A0 (en) * 2002-01-24 2003-06-24 Gamida Cell Ltd Utilization of retinoid and vitamin d receptor antagonists for expansion of renewable stem cell populations
WO2003062404A1 (en) * 2002-01-25 2003-07-31 Gamida-Cell Ltd. Methods of expanding stem and progenitor cells and expanded cell populations obtained thereby
US6720423B2 (en) 2002-04-30 2004-04-13 Allergan, Inc. Dihydrobenzofuran and dihydrobenzothiophene 2,4-pentadienoic acid derivatives having selective activity for retinoid X (RXR) receptors
US6620963B1 (en) 2002-09-19 2003-09-16 Allergan, Inc. TRICYCLO[6.2.202,7]DODECA-2(7),3,5-TRIEN-4-CARBONYLAMINO-PHENYL AND TRICYCLO[6.2.202,7]DODECA-2(7),3,5-TRIEN-4-CARBONYLAMINO-HETEROARYL AND RELATED COMPOUNDS HAVING RARα RECEPTOR SELECTIVE BIOLOGICAL ACTIVITY
US7105566B2 (en) * 2002-10-22 2006-09-12 Allergan, Inc. Methods of treatment during vascular procedures
US20050026950A1 (en) * 2003-07-30 2005-02-03 Allergan, Inc. Methods of therapeutic treatment using retinoids to achieve consistent bioavailability
US20050026949A1 (en) * 2003-07-30 2005-02-03 Allergan, Inc. Methods of therapeutic treatment using amounts of retinoids without regard to body weight
US20050026958A1 (en) * 2003-07-30 2005-02-03 Allergan, Inc. Methods of therapeutic treatment using amounts of retinoid components
AU2004261301A1 (en) * 2003-07-30 2005-02-10 Allergan, Inc. Methods of therapeutic treatment using amounts of retinoid components
US20060141049A1 (en) * 2003-11-12 2006-06-29 Allergan, Inc. Triamcinolone compositions for intravitreal administration to treat ocular conditions
US20050250737A1 (en) * 2003-11-12 2005-11-10 Allergan, Inc. Therapeutic ophthalmic compositions containing retinal friendly excipients and related methods
US20050101582A1 (en) * 2003-11-12 2005-05-12 Allergan, Inc. Compositions and methods for treating a posterior segment of an eye
US20070224278A1 (en) * 2003-11-12 2007-09-27 Lyons Robert T Low immunogenicity corticosteroid compositions
DE602004019685D1 (en) * 2003-12-26 2009-04-09 Allergan Inc DISUBSTITUTED CHALCOGENOX OXES WITH ANTAGONISTIC EFFECT ON THE RAR (Gamma) RETINOID RECEPTOR
AU2005209201B2 (en) 2004-01-20 2010-06-03 Allergan, Inc. Compositions for localized therapy of the eye, comprising preferably triamcinolone acetonide and hyaluronic acid
US20050244465A1 (en) * 2004-04-30 2005-11-03 Allergan, Inc. Drug delivery systems and methods for treatment of an eye
US20050244466A1 (en) * 2004-04-30 2005-11-03 Allergan, Inc. Photodynamic therapy in conjunction with intraocular implants
US20050244458A1 (en) * 2004-04-30 2005-11-03 Allergan, Inc. Sustained release intraocular implants and methods for treating ocular neuropathies
US8147865B2 (en) * 2004-04-30 2012-04-03 Allergan, Inc. Steroid-containing sustained release intraocular implants and related methods
US8425929B2 (en) * 2004-04-30 2013-04-23 Allergan, Inc. Sustained release intraocular implants and methods for preventing retinal dysfunction
US20050244478A1 (en) * 2004-04-30 2005-11-03 Allergan, Inc. Anti-excititoxic sustained release intraocular implants and related methods
US7799336B2 (en) 2004-04-30 2010-09-21 Allergan, Inc. Hypotensive lipid-containing biodegradable intraocular implants and related methods
BRPI0510485A (en) 2004-04-30 2007-11-13 Allergan Inc biodegradable intravitreal tyrosine kinase inhibitor implants
US20050244461A1 (en) * 2004-04-30 2005-11-03 Allergan, Inc. Controlled release drug delivery systems and methods for treatment of an eye
US8455656B2 (en) 2004-04-30 2013-06-04 Allergan, Inc. Kinase inhibitors
US9498457B2 (en) 2004-04-30 2016-11-22 Allergan, Inc. Hypotensive prostamide-containing biodegradable intraocular implants and related implants
US8722097B2 (en) 2004-04-30 2014-05-13 Allergan, Inc. Oil-in-water method for making polymeric implants containing a hypotensive lipid
US8119154B2 (en) * 2004-04-30 2012-02-21 Allergan, Inc. Sustained release intraocular implants and related methods
US20050244463A1 (en) * 2004-04-30 2005-11-03 Allergan, Inc. Sustained release intraocular implants and methods for treating ocular vasculopathies
US20070059336A1 (en) * 2004-04-30 2007-03-15 Allergan, Inc. Anti-angiogenic sustained release intraocular implants and related methods
US7771742B2 (en) 2004-04-30 2010-08-10 Allergan, Inc. Sustained release intraocular implants containing tyrosine kinase inhibitors and related methods
US20050244462A1 (en) * 2004-04-30 2005-11-03 Allergan, Inc. Devices and methods for treating a mammalian eye
US7993634B2 (en) 2004-04-30 2011-08-09 Allergan, Inc. Oil-in-oil emulsified polymeric implants containing a hypotensive lipid and related methods
US20050244471A1 (en) * 2004-04-30 2005-11-03 Allergan, Inc. Estradiol derivative and estratopone containing sustained release intraocular implants and related methods
US8591885B2 (en) 2004-04-30 2013-11-26 Allergan, Inc. Carbonic anhydrase inhibitor sustained release intraocular drug delivery systems
US20050244472A1 (en) * 2004-04-30 2005-11-03 Allergan, Inc. Intraocular drug delivery systems containing excipients with reduced toxicity and related methods
US8673341B2 (en) * 2004-04-30 2014-03-18 Allergan, Inc. Intraocular pressure reduction with intracameral bimatoprost implants
JP2008505978A (en) * 2004-07-12 2008-02-28 アラーガン、インコーポレイテッド Ophthalmic composition and eye disease treatment method
EP1799812A4 (en) 2004-09-16 2009-09-09 Gamida Cell Ltd Methods of ex vivo progenitor and stem cell expansion by co-culture with mesenchymal cells
BRPI0616454A2 (en) 2005-09-30 2011-06-21 Vitae Pharmaceuticals Inc cancer treatment methods
ES2743202T3 (en) 2005-10-27 2020-02-18 Viacyte Inc Endoderm of the dorsal and ventral proximal intestine expressing PDX1
US8846393B2 (en) 2005-11-29 2014-09-30 Gamida-Cell Ltd. Methods of improving stem cell homing and engraftment
ES2702128T3 (en) * 2006-05-16 2019-02-27 Io Therapeutics Llc Antagonist or reverse RAR agonist for use in the treatment of side effects of chemotherapy and / or radiation therapy
US8969415B2 (en) 2006-12-01 2015-03-03 Allergan, Inc. Intraocular drug delivery systems
US7911053B2 (en) * 2007-04-19 2011-03-22 Marvell World Trade Ltd. Semiconductor packaging with internal wiring bus
US8082730B2 (en) * 2008-05-20 2011-12-27 Caterpillar Inc. Engine system having particulate reduction device and method
BR122017013201B1 (en) 2009-11-09 2018-05-15 Allergan, Inc. COMPOSITION FOR STIMULATION OF HAIR GROWTH
US20130190395A1 (en) 2011-12-13 2013-07-25 Dartmouth College Autoimmune disorder treatment using rxr agonists
BR112014020119A2 (en) 2012-02-13 2020-10-27 Gamida-Cell Ltd culture of mesenchymal stem cells
US9175266B2 (en) 2012-07-23 2015-11-03 Gamida Cell Ltd. Enhancement of natural killer (NK) cell proliferation and activity
US9567569B2 (en) 2012-07-23 2017-02-14 Gamida Cell Ltd. Methods of culturing and expanding mesenchymal stem cells
CN103044477B (en) * 2012-12-07 2015-08-05 中国科学院广州生物医药与健康研究院 Trimethyl silicane replaces benzopyrans compounds and application thereof
CN109602691A (en) 2013-02-15 2019-04-12 阿勒根公司 Sustained drug delivery implantation material
KR102489706B1 (en) 2015-10-31 2023-01-17 아이오 테라퓨틱스, 인크. Treatment of nervous system disorders using combinations of rxr agonists and thyroid hormones
ES2898676T3 (en) 2015-11-25 2022-03-08 Io Therapeutics Inc Selective CYP26-resistant rar-alpha agonists in cancer treatment
JP7169647B2 (en) 2016-03-10 2022-11-11 アイオー セラピューティクス インコーポレイテッド Treatment of muscle disorders with a combination of RXR agonists and thyroid hormones
CA3016876C (en) 2016-03-10 2021-12-28 Io Therapeutics, Inc. Treatment of autoimmune diseases with combinations of rxr agonists and thyroid hormones
CN107176945B (en) * 2016-03-11 2021-06-08 中国科学院上海有机化学研究所 Retinoid compound, preparation method, intermediate and application thereof
US20210322543A1 (en) * 2016-06-02 2021-10-21 Hoyu Co., Ltd. Egg allergy antigen
EP3468600A4 (en) 2016-06-10 2019-11-13 IO Therapeutics, Inc. Receptor selective retinoid and rexinoid compounds and immune modulators for cancer immunotherapy
KR20200029544A (en) 2017-07-13 2020-03-18 아이오 테라퓨틱스, 인크. Immunomodulatory retinoid and rexinoid compounds combined with immunomodulators for cancer immunotherapy
AU2018326617B2 (en) 2017-08-31 2022-12-01 Io Therapeutics, Inc. Rar selective agonists in combination with immune modulators for cancer immunotherapy
AU2018335393A1 (en) 2017-09-20 2020-04-02 Io Therapeutics, Inc. Treatment of disease with esters of selective RXR agonists
CN110090237B (en) * 2019-05-30 2021-05-28 四川农业大学 Medicine for treating liver injury and bone injury caused by vitamin A poisoning and application thereof
US10966950B2 (en) 2019-06-11 2021-04-06 Io Therapeutics, Inc. Use of an RXR agonist in treating HER2+ cancers
EP4077300A1 (en) * 2019-12-19 2022-10-26 Orphagen Pharmaceuticals, Inc. Rar-alpha compounds for inflammatory disease and male contraception
US11896558B2 (en) 2021-12-07 2024-02-13 Io Therapeutics, Inc. Use of an RXR agonist and taxanes in treating Her2+ cancers

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2138000A1 (en) * 1994-01-03 1995-07-04 John E. Starrett, Jr. Retinoid-like compounds
US5776699A (en) * 1995-09-01 1998-07-07 Allergan, Inc. Method of identifying negative hormone and/or antagonist activities
US5675024A (en) * 1995-11-22 1997-10-07 Allergan Aryl or heteroaryl amides of tetrahydronaphthalene, chroman, thiochroman and 1,2,3,4,-tetrahydroquinoline carboxylic acids, having an electron withdrawing substituent in the aromatic or heteroaromatic moiety, having retinoid-like biological activity

Also Published As

Publication number Publication date
ATE289998T1 (en) 2005-03-15
DE69829183T2 (en) 2006-04-06
EP0991636A1 (en) 2000-04-12
JP2002507204A (en) 2002-03-05
AU8261998A (en) 1999-01-04
EP1535919A3 (en) 2005-10-26
CN1142928C (en) 2004-03-24
CN1268130A (en) 2000-09-27
EP0991636B1 (en) 2005-03-02
JP4295361B2 (en) 2009-07-15
DE69829183D1 (en) 2005-04-07
US5877207A (en) 1999-03-02
BR9810340A (en) 2000-09-19
EP1535919A2 (en) 2005-06-01
KR20010020505A (en) 2001-03-15
WO1998058922A1 (en) 1998-12-30
KR100512058B1 (en) 2005-09-02
AU729140B2 (en) 2001-01-25

Similar Documents

Publication Publication Date Title
CA2230672C (en) Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities
CA2294601A1 (en) Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities
US5958954A (en) Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities
US5952345A (en) Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities
US6469028B1 (en) Synthesis and use of retinoid compounds having negative hormone and/or anatgonist activities
US6218128B1 (en) Methods of identifying compounds having nuclear receptor negative hormone and/or antagonist activities
US6942980B1 (en) Methods of identifying compounds having nuclear receptor negative hormone and/or antagonist activities
US20030219832A1 (en) Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities
JP2008280346A (en) Synthesis and use of retinoid compound having negative hormone activity and/or antagonist activity

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued