CA2316351C - Benzopyran and benzothiopyran derivatives having retinoid antagonist-like activity - Google Patents

Benzopyran and benzothiopyran derivatives having retinoid antagonist-like activity Download PDF

Info

Publication number
CA2316351C
CA2316351C CA002316351A CA2316351A CA2316351C CA 2316351 C CA2316351 C CA 2316351C CA 002316351 A CA002316351 A CA 002316351A CA 2316351 A CA2316351 A CA 2316351A CA 2316351 C CA2316351 C CA 2316351C
Authority
CA
Canada
Prior art keywords
compound
formula
accordance
mmol
compounds
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.)
Expired - Lifetime
Application number
CA002316351A
Other languages
French (fr)
Other versions
CA2316351A1 (en
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.)
IO Therapeutics Inc
Original Assignee
Allergan Inc
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 Allergan Inc filed Critical Allergan Inc
Publication of CA2316351A1 publication Critical patent/CA2316351A1/en
Application granted granted Critical
Publication of CA2316351C publication Critical patent/CA2316351C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • 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
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/04Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond

Abstract

2,2-Dialkyl- 4-aryl-substituted benzopyran and benzothiopyran derivatives of formula (I) where the symbols have the meaning described in the specification, 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 or RAR agonists on the bound receptor sites. Specifically, the RAR agonists are administered or coadministered with retinoid drugs to prevent o r ameliorate toxicity or side effects caused by retinoids or vitamin A or vitamin A precursors. The retinoid negative hormones can be use d to potentiate the activities of other retinoids and nuclear receptor agonists.

Description

DEMANDES OU BREVETS VOLUMINEUX

LA PRESENTE PARTIE DE CETTE DBVIANDE OU CE BREVET
COMPREND PLUS D'UN TOME.

CECI EST LE TOME __.IfDE

NOTE: Pour les tomes additionels, veuiiiez contacter le Bureau canadien -des brevets JUMBO APPLICATIONS/PATENTS

THIS SECTION OF THE APPl.ICATIONIPATENT CONTAINS MORE
THAN ONE VOLUME

THiS IS VOLUME `-OF _ - =

NOTE: For additional voiumes=please contact"the Canadian Patent Office ,. .; . .... .. :..
.. : . ..; ., , _ .
... .. ~: .~}
.: ,. . . ;. . . . .. _:_.... . __ ._. ... , WO 99/33821` PCT/US98/27314 BENZOPYRAN AND BENZOTHIOPYRAN DERIVATIVES HAVING RETINOID
ANTAGONIST-LIKE ACTIVITY

Field of the inveDtion The present invention relates to novel compounds having retinoid negative hormone andlor retinoid antagonist-like biological activities.
More specifically, the invention relates to 4-aryl substituted benzopyran, 21 4-aryl substituted benzothiopyran, 4-aryl substituted 1,2-dihydroquinoline 22 and 8-aryl substituted 5,6-dihydronaphthalene derivatives which may 23 also be substituted by a substituted 3-oxo-l-propenyl group. These 24 novel compounds have retinoid antagonist like-activity and are useful 25 for treating or preventing retinoid and vitamin A and vitamin A
26 precursor induced toxicity in mammals and as an adjunct to treatment 27 of mammals with retinoids to prevent or ameliorate unwanted or 28 undesired side effects. The invention further relates to the use of 29 retinoid negative hormones for increasing the biological activities of 1 other retinoids and steroid hormones and inhibiting the basal activity of 2 unliganded retinoic acid receptors.
3 Background of the Invention 4 Compounds which have retinoid-like activity are well known in the art, and are described in numerous United States and other patents 6 and in scientific publications. It is generally known and accepted in the 7 art that retinoid-like activity is useful for treating mammals, including 8 humans, in order to cure or alleviate the symptoms associated with 9 numerous diseases and conditions.
Retinoids (vitamin A and its derivatives) are known to have broad 11 activities, including effects on cell proliferation and differentiation, in a 12 variety of biological systems. This activity has made retinoids useful in 13 the treatment of a variety of diseases, including dermatological disorders 14 and cancers. The prior art has developed a large number of chemical compounds which have retinoid-like biological activity, and voluminous 16 patent and chemical literature exists describing such compounds. The 17 relevant patent literature includes United States Patent Nos. 4,980,369, 18 5,006,550, 5,015,658, 5,045,551, 5,089,509, 5,134,159, 5,162,546, 19 5,234,926, 5,248,777, 5,264,578, 5,272,156, 5,278,318, 5,324,744, 2o 5,346,895, 5,346,915, 5,348,972, 5,348,975, 5,380,877, 5,399,561, 21 5,407,937, (assigned to the same assignee as the present application) and 22 patents and publications cited therein, which particularly describe or 23 relate to chroman, thiochroman and 1,2,3,4-tetrahydroquinoline 24 derivatives which have retinoid-like biological activity. In addition, several applications are pending which are assigned to the assignee of 26 the present application, and which are directed to further compounds 27 having retinoid-like activity.
28 United States Patent Nos. 4,740,519 (Shroot et al.), 4,826,969 29 (Maignan et al.), 4,326,055 (Loeliger et al.), 5,130,335 (Chandraratna et 1 al.), 5,037,825 (Klaus et al.), 5,231,113 (Chandraratna et al.), 5,324,840 2 (Chandraratna), 5,344,959 (Chandraratna), 5,130,335 (Chandraratna et 3 al.), Published European Patent Application Nos. 0 176 034 A (Wuest 4 et al.), 0 350 846 A (Klaus et al.), 0 176 032 A (Frickel et al.), 0 176 033 A (Frickel et al.), 0 253 302 A(Klaus et al.), 0 303 915 A (Bryce et al.), 6 UK Patent Application GB 2190378 A (Klaus et al.), German Patent 7 Application Nos. DE 3715955 Al (Klaus et al.), DE 3602473 Al (Wuest 8 et al., and the articles J. Amer. Acad. Derm. 15: 756 - 764 (1986) (Sporn 9 et al.), Chem. Pharm. Bull. 33: 404-407 (1985) (Shudo et al.), J. Med 1o Chem. 31: 2182 - 2192 (1988) (Kagechika et al.), Chemis rv and Biology 11 of Synthetic Retinoids CRC Press Inc. 1990 pp. 334 - 335, 354 (Dawson 12 et al.), describe or relate to compounds which include a 13 tetrahydronaphthyl moiety and have retinoid-like or related biological 14 activity. United States Patent No. 4,391,731 (Boller et al.) describes tetrahydronaphthalene derivatives which are useful in liquid crystal 16 compositions.
17 An article by Kagechika et al. in J. Med. Chem 32:834 (1989) 18 describe certain 6-(3-oxo-l-propenyl)- 1,2,3,4-tetramethyl- 1,2,3,4-19 tetrahydronaphthalene derivatives and related flavone compounds zo having retinoid-like activity. The articles by Shudo et al. in Chem.
21 Pharnc. BulL 33:404 (1985) and by Jetten et al. in Cancer Research 22 47:3523 (1987) describe or relate to further 3-oxo-l-propenyl derivatives 23 (chalcone compounds) and their retinoid-like or related biological 24 activity.
Unfortunately, compounds having retinoid-like activity (retinoids) 26 also cause a number of undesired side effects at therapeutic dose levels, 27 including headache, teratogenesis, mucocutaneous toxicity, 28 musculoskeletal toxicity, dyslipidemias, skin irritation, headache and 29 hepatotoxicity. These side effects limit the acceptability and utility of 1 retinoids for treating disease.
2 It is now general knowledge in the art that two main types of 3 retinoid receptors exist in mammals (and other organisms). The two 4 main types or families of receptors are respectively designated as the RARs and RXRs. Within each type there are subtypes: in the RAR
6 family the subtypes are designated RAR-a, RA.R-f3 and RAR-y, in 7 RXR the subtypes are: RXR-a, RXB-/3 and RXR-y. Both families of 8 receptors are transcription factors that can be distinguished from each 9 other based on their ligand binding specificities. All-trans-RA (ATRA) 1 o binds and activates a class of retinoic acid receptors (RARs) that 11 includes RAR-a, RAR-0 and RAR-y. A different ligand, 9-cis-RA (9C-12 RA), binds and activates both the RARs and members of the retinoid X
13 receptor (RXR) family.
14 It has also been established in the art that the distribution of the two main retinoid receptor types, and of the several subtypes is not 16 uniform in the various tissues and organs of mammalian organisms.
17 Moreover, it is generally accepted in the art that many unwanted side 18 effects of retinoids are mediated by one or more of the RAR receptor 19 subtypes. Accordingly, among compounds having agonist-like activity at 2o retinoid receptors, specificity or selectivity for one of the main types or 21 families, and even specificity or selectivity for one or more subtypes 22 within a family of receptors, is considered a desirable pharmacological 23 property.
24 Relatively recently compounds have been developed in the art which bind to RAR receptors without triggering the response or 26 responses that are triggered by agonists of the same receptors. The 27 compounds or agents which bind to RAR receptors without triggering a 28 "retinoid" response are thus capable of blocking (to lesser or greater 29 extent) the activity of RAR agonists in biological assays and systems.

1 More particularly, regarding the scientific and patent literature in this 2 field, published PCT Application WO 94/14777 describes certain 3 heterocyclic carboxylic acid derivatives which bind to RAR retinoid 4 receptors and are said in the application to be useful for treatment of 5 certain diseases or conditions, such as acne, psoriasis, rheumatoid 6 arthritis and viral infections. A similar disclosure is made in the article 7 by Yoshimura et al. J Med. Chem. 38: 3163-3173 (1995). Kaneko et al.
8 Med. Chem Res. 1:220-225 (1991); Apfel et al. Proc. Natl. Acad. Sci.
9 USA 89: 7129-7133 Augusty 1992 Cell Biology; Eckhardt et al. Toxicology 1o Letters 70:299-308 (1994); Keidel et al. Molecular and Cellular Biology 11 14:287-298 (1994); and Eyrolles et al. J. Med. Chem. 37: 1508-1517 12 (1994) describe compounds which have antagonist like activity at one or 13 more of the RAR retinoid subtypes.
14 In addition to undesirable side-effects of therapy with retinoid compounds, there occurs occasionally a serious medical condition 16 caused by vitamin A or vitamin A precursor overdose, resulting either 17 from the excessive intake of vitamin supplements or the ingestion of 18 liver of certain fish and animals that contain high levels of the vitamin.
1 s The chronic or acute toxicities observed with hypervitaminosis A
2o syndrome include headache, skin peeling, bone toxicity, dyslipidemias, 21 etc. In recent years, it has become apparent that the toxicities observed 22 with vitamin A analogs, i.e., retinoids, essentially recapitulate those of 23 hypervitaminosis A syndrome, suggesting a common biological cause, 24 i.e., RAR activation. These toxicities are presently treated mainly by supportive measures and by abstaining from further exposure to the 26 causative agent, whether it be liver, vitamin supplements, or retinoids.
27 While some of the toxicities resolve with time, others (e.g., premature 28 epiphyseal plate closure) are permanent.
29 Generally speaking, specific antidotes are the best treatment for = f 1 poisoning by pharmacological agents, but only about two dozen 2 chemicals or classes of chemicals out of thousands in existence have 3 specific known antidotes. A specific antidote would clearly be of value 4 in the treatment of hypervitaminosis A and retinoid toxicity. Indeed, as increasingly potent retinoids are used clinically, a specific antidote for 6 retinoid poisoning could be life saving.

Summary of the Invention The present invention relates to a compound of the formula COzRe Ru Ri Rz where X, is S or O;
X2 is CH or N;

R2 is H, F, CF3 or alkoxy of 1 to 6 carbons;
R2* H, F, or CF3;

R8 is H, or lower alkyl of 1 to 6 carbons;

R14 is unsubstituted phenyl, thienyl or pyridyl, or phenyl, thienyl or pyridyl substituted with one to three Ri5 groups, where Rl5 is lower alkyl of I to 6 carbons, chlorine, CF3, or alkoxy of I to 6 carbons, or a pharmaceutically acceptable salt of said compound.

In a preferred embodiment, X, is S; X2 is CH; R14 is phenyl, 2-thienyl, or 3-pyridyl unsubstituted or substituted with one lower alkyl group having 1 to carbons; R2 is H or F; R2* is H or F; X2 is N.

In another preferred embodiment, X, is 0; X2 is CH; R14 is phenyl, 2-thienyl, or 3-pyridyl unsubstituted or substituted with one lower alkyl group having I
to 6 carbons; R2 is H or F and R2* is H or F.

In another embodiment, the invention relates to a compound of the formula coite Ru R=

wherein X2 is CH or N;

R2 is H, F or OCH3;
R2* is H or F;

R8 is H, or lower alkyl of 1 to 6 carbons, and R14 is selected from the group consisting of phenyl, 4-(lower-alkyl)phenyl, 5-(lower a1kyI)-2-thienyl, and 6-(lower alkyl)-3-pyridyl where lower alkyl has I
to 6 carbons, or a pharmaceutically acceptable salt of said compound.

In a preferred embodiment, X2 is CH; R2 and R2* is H; the R14 group is selected from phenyl, 4-methylphenyl, 4-ethylphenyl, 4-i-propylphenyl, 4-t-butylphenyl, 5-methyl-2-thienyl, 5-ethyl-2-thienyl, 5-t-butyl-2-thienyl and 6-methyl-3-pyridyl; Rg is ethyl or H, or a pharmaceutically acceptable salt of said compound.

In another preferred embodiment, R2 is H. R2* is F, the R14 group is selected from 4-methylphenyl and 4-ethylphenyl; and Rg is ethyl or H or a pharmaceutically acceptable salt of said compound.

- In another preferred embodiment, R2 is F, R2* is H; the R14 group is selected from 4-methylphenyl and 5-methyl-2-thienyl; R8 is ethyl or H or a pharmaceutically acceptable salt of said compound.

In another preferred embodiment, X2 is CH; R2 is OCH3; R2* is H; R14 is 4-methylphenyl; and RS is ethyl or H or a pharmaceutically acceptable salt of said compound.

In another preferred embodiment, X2 is N; R2 is H; R2* is H; R14 is selected from the group consisting of 4-methylphenyl and 4-ethylphenyl; and Rs is ethyl or H
or a pharmaceutically acceptable salt of said compound.

In another preferred embodiment, the invention relates to a compound of the formula Nk CQaR*
po FW
flu where R2* is H or F;

R8 is H, or lower alkyl of 1 to 6 carbons, and R14 is selected from the group consisting of phenyl, and 4-(lower-alkyl)phenyl, where lower alkyl has 1 to 6 carbons, or a pharmaceutically acceptable salt of said compound.

In a preferred embodiment, R2* is H; the R14 group is selected from phenyl, 4-methylphenyl, 4-ethyiphenyl, 4-i-propylphenyl and 4-t-butylphenyl; and R8 is ethyl or H or a pharmaceutically acceptable salt of said compound.

In another preferred embodiment, R2* is F; R14 is selected from the group consisting of 4-methylphenyl and 4-ethylphenyl; and Rg is ethyl or H or a pharmaceutically acceptable salt of said compound.

In another preferred embodiment, the invention relates to a compound of the formula C02%
where R8 is H or lower alkyl of 1 to 6 carbons, or a pharmaceutically acceptable salt of said compound.

In a preferred embodiment, R8 is H or a pharmaceutically acceptable salt of said compound.

In another preferred embodiment, R8 is ethyl.

In another preferred embodiment of the invention, the invention relates to the use of a retinoid antagonist or a negative hormone which binds to a retinoic acid receptor subtype selected from the group consisting of RARa, RARa and RARy to treat in a mammal a pathological condition caused by retinoid or vitamin A or vitamin A precursor induced toxicity or side effects, wherein the antagonist or negative hormone is a compound of the formula coAS
Ru Ri R=

In another preferred embodiment, the invention relates to the use of a retinoid antagonist or a negative hormone which binds to a retinoic acid receptor subtype selected from the group consisting of RARa,,, RARR and RAR., in the manufacture of a medicament to treat in a mammal a pathological condition caused by retinoid or vitamin A or vitamin A precursor induced toxicity or side effects, wherein the antagonist or negative hormone is a compound of the formula ` COOR`
R" Re Rs In a further preferred embodiment, the retinoid antagonist or negative hormone is used in order to cure or ameliorate a pre-existing pathological condition caused by intake of a retinoic drug or vitamin A or vitamin A precursors by the mammal.

In another embodiment, the retinoid antagonist or negative hormone is used topically to block or ameliorate undesired topical side effects of a retinoic drug administered for a therapeutic purpose.

In a further preferred embodiment, the retinoid antagonist or negative hormone is used topically to block or ameliorate undesired topical side effects of a retinoic drug administered systemically for a therapeutic purpose.

In a further prefened embodiment, the retinoid antagonist or negative hormone is used topically to treat a pre-existing condition or side effect caused by a retinoid drug or vitamin A.

In a further preferred embodiment, the retinoid antagonist or negative hormone is used systemically to treat a pre-existing condition or side effect caused by a retinoid drug or vitamin A.

In a further preferred embodiment, the retinoid antagonist or negative hormone is used systemically to block or ameliorate bone toxicity caused by coadministration of a retinoid drug or vitamin A.

The present invention covers compounds of Formula 1 a,.

Formula I
wherein X is S, 0, NR' where R' is H or alkyl of 1 to 6 carbons, or X is (C(R,)J. where R, is independently H or alkyl of 1 to 6 carbons, and n is an integer between 0 and 2;
R2 is hydrogen, lower aAryl of 1 to 6 carbons, F, Cl, Br, I, CF3, fluoro substituted alkyl of 1 to 6 carbons, OH, SH, alkoxy of I to 6 carbons, or alkylthio of 1 to 6 carbons;
R3 is hydrogen, lower alkyl of I to 6 carbons or F;
m is an integer having the value of 0- 3;
o is an integer having the value of 0- 3;
Z is -C=C-, -N-N-, -N=CR; , 1 -(CR, = CR,),,.- where n' is an integer having the value 0 - 5, 2 -CO-NR,-, 3 -CS-NR,-, 4 -NR,-CO, -NR,-CS, 6 -coo-, 7 -OCO-;
8 -CSO-;
9 -OCS-;
Y is a phenyl or naphthyl group, or heteroaryl selected from a 11 group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, 12 pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrrazolyl, said phenyl and 13 heteroaryl groups being optionally substituted with one or two R2 14 groups, or when Z is -(CR,=CR,),,: and n' is 3, 4 or 5 then Y represents a 18 direct valence bond between said (CR2=CR2),,, group and B;
17 A is (CH2)q where q is 0-5, lower branched chain alkyl having 3-6 18 carbons, cycloalkyl having 3-6 carbons, alkenyl having 2-6 carbons and 1 19 or 2 double bonds, alkynyl having 2-6 carbons and 1 or 2 triple bonds;
B is hydrogen, COOH or a pharmaceutically acceptable salt 21 thereof, COORB, CONRgR,o, -CH2OH, CH20R,,, CH2OCOR,,, CHO, 22 CH(OR12)2, CHOR13O, -COR7, CR7(OR,2)2, CR70R130, or tri-lower 23 alkylsilyl, where R7 is an alkyl, cycloalkyl or alkenyl group containing 1 24 to 5 carbons, R8 is an alkyl group of 1 to 10 carbons or trimethylsilylalkyl where the alkyl group has 1 to 10 carbons, or a 26 cycloalkyl group of 5 to 10 carbons, or R8 is phenyl or lower 27 alkylphenyl, R9 and R,o independently are hydrogen, an alkyl group of 1 28 to 10 carbons, or a cycloalkyl group of 5-10 carbons, or phenyl or lower 29 alkylphenyl, Rõ is lower alkyl, phenyl or lower alkylphenyl, R12 is lower 1 alkyl, and R13 is divalent alkyl radical of 2-5 carbons, and 2 R14 is (R,s)r-phenyl, (R,S)r naphthyl, or (Rls)r heteroaryl where 3 the heteroaryl group has 1 to 3 heteroatoms selected from the group 4 consisting of 0, S and N, r is an integer having the values of 0 - 5, and R15 is independently H, F, Cl, Br, I, NO2, N(R8)2, N(R8)COR8, 6 NR8CON(R8)2, OH, OCORB, OR8, CN, an alkyl group having 1 to 10 7 carbons, fluoro substituted alkyl group having 1 to 10 carbons, an 8 alkenyl group having 1 to 10 carbons and 1 to 3 double bonds, alkynyl 9 group having 1 to 10 carbons and 1 to 3 triple bonds, or a trialkylsilyl or 1o trialkylsilyloxy group where the alkyl groups independently have 1 to 6 11 carbons.
12 The present invention further covers compounds of Formula 101 Ru (ROm ~A "8 (O)p~ .Y(R2) 16 (POo R)n 17 Rig O

19 Formula 101 wherein X is S, 0, NR' where R' is H or alkyl of 1 to 6 carbons, 21 or 22 X is [C(Rl)2]õ where R, is independently H or alkyl of 1 to 6 23 carbons, and n is an integer between 0 and 2;
24 R2 is hydrogen, 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 26 carbons, or alkylthio of 1 to 6 carbons;
27 R3 is hydrogen, lower alkyl of 1 to 6 carbons or F;
28 m is an integer having the value of 0 - 3;
29 o is an integer having the value of 0 - 3;

1 Y is a phenyl or naphthyl group, or heteroaryl selected from a 2 group consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, 3 pyrazinyl, thiazolyl, oxazolyl, imidazolyl and pyrrazolyl, said phenyl and 4 heteroaryl groups being optionally substituted with one or two R2 groups;
6 A is (CHZ)q where q is 0-5, lower branched chain alkyl having 3-6 7 carbons, cycloalkyl having 3-6 carbons, alkenyl having 2-6 carbons and 1 8 or 2 double bonds, alkynyl having 2-6 carbons and 1 or 2 triple bonds;
9 B is hydrogen, COOH or a pharmaceutically acceptable salt 1o thereof, COOR8, CONR9Rlo, -CHZOH, CH20R11, CH2OCOR11, CHO, 11 CH(OR12)2, CHOR13O, -COR7, CR7(ORi2)2, CR70R,30, or tri-lower 12 alkylsilyl, where R7 is an alkyl, cycloalkyl or alkenyl group containing 1 13 to 5 carbons, R. is an alkyl group of 1 to 10 carbons or 14 trimethylsilylalkyl where the alkyl group has 1 to 10 carbons, or a cycloalkyl group of 5 to 10 carbons, or R. is phenyl or lower 16 alkylphenyl, R9 and R,o independently are hydrogen, an alkyl group of 1 17 to 10 carbons, or a cycloalkyl group of 5-10 carbons, or phenyl or lower 1s alkylphenyl, Rõ is lower alkyl, phenyl or lower alkylphenyl, R12 is lower 19 alkyl, and R13 is divalent alkyl radical of 2-5 carbons, and R14 is (R,s)r phenyl, (R,s)r-naphthyl, or (R,s)r heteroaryl where 21 the heteroaryl group has 1 to 3 heteroatoms selected from the group 22 consisting of 0, S and N, r is an integer having the values of 0 - 5, and 23 R,s is independently H, F, Cl, Br, I, NO2, N(R8)2, N(R8)COR8, 24 NRSCON(R8)2, OH, OCOR8, ORB, CN, an alkyl group having 1 to 10 carbons, fluoro substituted alkyl group having 1 to 10 carbons, an 26 alkenyl group having 1 to 10 carbons and 1 to 3 double bonds, alkynyl 27 group having 1 to 10 carbons and 1 to 3 triple bonds, or a trialkylsilyl or 28 trialkylsilyloxy group where the alkyl groups independently have 1 to 6 29 carbons;

1 R16 is H, lower alkyl of 1 to 6 carbons;
2 Rõ is H, lower alkyl of 1 to 6 carbons, OH or OCOR,,, and 3 p is zero or 1, with the proviso that when p is 1 then there is no 4 Rõ substituent group, and m is an integer between 0 and 2.
5 The compounds of the present invention are useful for preventing 6 certain undesired side effects of retinoids which are administered for the 7 treatment or prevention of certain diseases or conditions. For this 8 purpose the compounds of the invention may be coadministered with 9 retinoids. The compounds of the present invention are also useful in 1 o the treatment of acute or chronic toxicity resulting from overdose or 11 poisoning by retinoid drugs or Vitamin A.
12 The present invention additionally relates to the use of RAR
13 antagonists for blocking all or some RAR receptor sites in biological 14 systems, including mammals, to prevent or diminish action of RAR
agonists on said receptor sites. More particularly, the present invention 16 relates to the use of RAR antagonists for (a) the prevention and (b) the 17 treatment of retinoid (including vitamin A or vitamin A precursor) 18 chronic or acute toxicity and side effects of retinoid therapy.
19 In one particular aspect of the present invention, there is provided a method of treating a pathological condition in a mammal.
21 The conditions treated are associated with a retinoic acid receptor 22 activity. This method involves administering to the mammal a retinoid 23 antagonist or negative hormone capable of binding to one of the 24 following retinoic acid receptor subtypes: RAR., RAR,, and RAR,,.
The antagonist or negative hormone is administered in an amount 26 pharmaceutically effective to provide a therapeutic benefit against the 27 pathological condition in the mammal.
28 As an antidote to acute or chronic retinoid or vitamin A
29 poisoning the RAR antagonist can be administered to a mammal 1 enterally, i.e., intragastric intubation or food/water admixture, or 2 parenterally, e.g., intraperitoneally, intramuscularly, subcutaneously, 3 topically, etc. The only requirement for the route of administration is 4 that it must allow delivery of the antagonist to the target tissue. The RAR antagonist can be formulated by itself or in combination with 6 excipients. The RAR antagonist need not be in solution in the 7 formulation, e.g., in the case of enteral use.
8 As an adjunct to therapy with retinoids and in order to prevent 9 one or more side effects of the retinoid drug which is administered, the 1 o RAR antagonist can similarly be administered enterally or parenterally.
11 The RAR antagonist and RAR agonist need not be administered by the 12 same route of administration. The key is that sufficient quantities of 13 the RAR antagonist be present continuously in the tissue of interest 14 during exposure to the RAR agonist. For the prevention of retinoid toxicity, it is best that the RAR antagonist be administered concurrently 16 or prior to treatment with the RAR agonist. In many situations the 17 RAR antagonist will be administered by a different route than the 18 agonist. For example undesirable skin effects of an enterally 19 administered retinoid may be prevented or ameliorated by an RAR
2o antagonist which is administered topically.
21 Another aspect of the present invention is a method of identifying 22 retinoid negative hormones. The method includes the following steps:
23 obtaining transfected cells containing a reporter gene transcriptionally 24 responsive to binding of a recombinant retinoid receptor, the recombinant retinoid receptor having at least protein domains located 26 C-terminal to a DNA binding domain of an intact retinoid receptor, 27 measuring a basal level of reporter gene expression in untreated 28 transfected cells, the untreated transfected cells being propagated in the 29 absence of an added retinoid, treating the transfected cells with a 1 retinoid compound to be tested for negative hormone activity, 2 measuring a level of reporter gene expression in treated cells, 3 comparing the levels of reporter gene expression measured in treated 4 cells and untreated cells, and identifying as retinoid negative hormones those retinoid compounds producing a lower level of reporter gene 6 expression in treated cells compared with the basal level of reporter 7 gene expression measured in untreated cells. In certain preferred 8 embodiments of this method the intact receptor is an RAR-a, RAR-f3 9 or RAR-y subtype. In other embodiments, the intact receptor is an lo RXR-a, RXR -,6 or RXR-y subtype. The recombinant receptor can also 11 be either a recombinant RAR or RXR receptor. In some 12 embodiments, the recombinant receptor is a chimeric retinoid receptor 13 having a constitutive transcription activator domain. Such a constitutive 14 transcription activator domain can comprise a plurality of amino acids having a net negative charge or have an amino acid sequence of a viral 16 transcription activator domain, such as the herpes simplex virus VP-16 17 transcription activator domain. In embodiments in which the 18 constitutive transcription activator domain has a net negative charge, the 19 retinoid receptor can be recombinant and have deleted therefrom a 2o DNA binding domain, such as a DNA binding domain specific for a cis-21 regulatory element other than a retinoic acid responsive"element. These 22 elements include an estrogen responsive element. The transfected cell 23 is preferably propagated in a growth medium substantially depleted of 24 endogenous retinoids, such as one that includes activated charcoal-extracted serum. In this method, the reporter gene can be the 26 luciferase gene, in which case, the measuring steps can involve 27 luminometry. The reporter gene can also be the 0-galactosidase gene, 28 in which case the measuring steps would involve a13-galactosidase assay.
29 The transfected cell can be a transfected mammalian cell, such as a 1 Green monkey cell or a human cell.
2 Another aspect of the present invention is a method of 3 potentiating a pharmacologic activity of a steroid superfamily receptor 4 agonist administered to a mammal. This method involves coadministering to the mammal with the steroid superfamily receptor 6 agonist a composition comprising a pharmaceutically effective dose of a 7 retinoid negative hormone to potentiate the pharmacologic activity of 8 the steroid superfamily receptor agonist. The pharmacologic activity is 9 measurable in a reporter gene trans-activation assay in vitro, such as by 1o measuring anti-AP-1 activity. The pharmacologic activity to be 11 potentiated can be an antiproliferative activity, such as activity of the 12 type measurable in retinal pigment epithelium. The steroid superfamily 13 receptor agonist can be any of the following: a retinoid receptor 14 agonist, a vitamin D receptor agonist, a glucocorticoid receptor agonist, a thyroid hormone receptor agonist, a peroxisome proliferator-activated 16 receptor agonist or an estrogen receptor agonist. The retinoid receptor 17 agonist can be an RAR agonist, such as all-trans-retinoic acid or 13-cis 18 retinoic acid. The retinoid receptor agonist can also be an RXR
1 s agonist. A preferred vitamin D receptor agonist is 1,25-2o dihydroxyvitamin D3. A preferred glucocorticoid receptor agonist is 21 dexamethasone. A preferred thyroid hormone receptor agonist is 3,3',5-22 triiodothyronine. The retinoid negative hormone is an RAR-specific 23 retinoid negative hormone, which preferably has a dissociation constant 24 less than or approximately equal to 30 nM. Example of the RAR-specific retinoid negative hormone include AGN 193109, AGN 193385, 26 AGN 193389 and AGN 193871. The composition comprising a 27 pharmaceutically effective dose of a retinoid negative hormone can be 28 coadministered at the same time as the steroid superfamily agonist and 29 be combined prior to coadministration. These can also be 1 coadministered as separate compositions.
2 Brief Description of the Drawings 3 Figure 1 shows the chemical structure of AGN 193109.
4 Figures 2A - 2F are a series of line graphs showing that AGN
193109 inhibited ATRA-dependent transactivation at the RARs.
6 Figures 2A and 2B represent activity at the RAR-a receptor; Figures 2C
7 and 2D represent activity at the RAR-f3 receptor; Figures 2E and 2F
s represent activity at the RAR-y receptor. In Figures 2A, 2C and 2E, 9 open squares represent retinoic acid treatment and filled circles 1 o represent AGN 193109 treatment. In Figures 2B, 2D and 2F the single ii lines represent luciferase activity measured after treatment with 10-8 M
12 ATRA and variable concentrations of AGN 193109.
13 Figures 3A and 3B are line graphs representing luciferase activity 14 detected in CV-1 cells transfected with reporter plasmid ERE-tk-Luc and expression plasmid ER-RAR-a and stimulated with ATRA (Figure 16 3A) or AGN 193109 (Figure 3B) at various concentrations. Data points 17 represent the mean SEM of three independent luciferase 18 determinations. The results of transfections carried out using different 19 amounts of co-transfected ER-RAR-a (0.05, 0.1 and 0.2 g/well) are indicated in each figure.
21 Figures 4A and 4B are line graphs representing luciferase activity 22 in CV-1 cells transfected with reporter plasmid ERE-tk-Luc and 23 expression plasmid ER-RAR-0 and stimulated with ATRA (Figure 4A) 24 or AGN 193109 (Figure 4B) at various concentrations. Data points represent the mean SEM of three independent luciferase 26 determinations. The results of transfections carried out using different 27 amounts of co-transfected ER-RAR-P (0.05, 0.1 and 0.2 g/well) are 28 indicated in each figure.
29 Figures 5A and 5B are line graphs representing luciferase activity i detected in CV-1 cells transfected with reporter plasmid ERE-tk-Luc 2 and expression plasmid ER-RAR-y and stimulated with ATRA (Figure 3 5A) or AGN 193109 (Figure 5B) at various concentrations. Data points 4 represent the mean SEM of three independent luciferase 5 determinations. The results of transfections carried out using different 6 amounts of co-transfected ER-RAR-y (0.05, 0.1 and 0.2 g/well) are 7 indicated in each figure.
8 Figure 6 shows ATRA and AGN 193109 dose responses of CV-1 9 cells cotransfected with the ERE-tk-Luc reporter plasmid and either the 1 o ER-RXR-a chimeric receptor expression plasmid alone, or in 11 combination with the RAR-y-VP-16 expression plasmid. ER-RXR-a 12 cotransfected cells were treated with ATRA (square) and AGN 193109 13 (diamond). Cells cotransfected with the combination of ER-RXR-a and 14 RAR-y-VP-16 were treated with ATRA (circle) or AGN 193109 15 (triangle).
16 Figure 7 shows a line graph representing luciferase activity 17 measurements recorded in lysates of CV-1 cells transfected with the 1 e ERE-tk-Luc reporter and ER-RAR-y expression construct and then 19 treated with ATRA at 10-8 M and the test compounds at the concentrations indicated on the horizontal axis. The test compounds 21 were AGN 193109 (square), AGN 193357 (open diamond), AGN
22 193385 (circle), AGN 193389 (triangle), AGN 193840 (hatched square) 23 and AGN 192870 (filled diamond).
24 Figure 8 shows a line graph representing luciferase activity measurements recorded in_ lysates of CV-1 cells transfected with the 26 ERE-tk-Luc reporter and RAR-y-VP-16 and ER-RXR-a expression 27 constructs and then treated with the test compounds at the 28 concentrations indicated on the horizontal axis. The test compounds 29 were ATRA (open square), AGN 193109 (open circle), AGN 193174 1 (open triangle), AGN .193199 (hatched square), AGN 193385 (hatched 2 circle), AGN 193389 (inverted triangle), AGN 193840 (diagonally filled 3 square) and AGN 193871 (half-filled diamond).
4 Figures 9A, 9B and 9C schematically diagram a mechanism whereby AGN 193109 can modulate the interaction between the RAR
s(shaded box) and negative coactivator proteins (-) illustrated in the 7 context of a transactivation assay. Figure 9A shows that negative 8 coactivator proteins and positive coactivator proteins (+) are in a 9 binding equilibrium with the RAR. In the absence of a ligand, basal io level transcription of the reporter gene results. As illustrated in Figure 11 9B, addition of an RAR agonist promotes the association of positive 12 coactivator proteins with the RAR and results in upregulated reporter 13 gene transcription. As illustrated in Figure 9C, addition of AGN
14 193109 promotes the association of negative coactivator proteins with the RAR and prevents reporter gene transcription.
16 Figure 10 is a bar graph showing the inhibition of TPA-induced 17 Str-AP1-CAT expression as a function of AGN 191183 concentration is (10-10 to 10-12 M) with the AGN 193109 concentration held constant at 19 10-8 M. Results from trials conducted with AGN 191183 alone are shown as hatched bars while stripped bars represent the results from 21 treatment with the combination of AGN 193109 and AGN 191183.
22 Figure 11 schematically diagrams a mechanism whereby AGN
23 193109 can potentiate the activities of RARs and other nuclear receptor 24 family members. As illustrated in the diagram, introduced RARs (open rectangles having AB-C-DEF domains) have increased sensitivity to 26 RAR ligands in the anti-AP1 assay because the negative coactivator 27 protein (ncp), present in limiting supply, is sequestered onto RARs 28 thereby leading to two populations: RAR+ncp and RAR-ncp. RAR-29 ncp has increased sensitivity to ligands. Non-RAR nuclear factors i(shaded rectangles having AB-C-DEF domains) have increased 2 sensitivity to cognate ligands because ncp has been sequestered to the 3 RAR by the activity of AGN 193109. The modular domains of the 4 nuclear receptors are designated using standard nomenclature as "AB"
(ligand independent transactivation domain), "C" (DNA binding 6 domain), and "DEF" (ligand regulated transactivation domain and 7 dimerization domain.
8 Figure 12 is a line graph showing the effect of AGN 193109 on 9 the 1,25-dihydroxyvitamin D3 dose response in CV-1 cells transfected lo with the MTV-DR3-Luc reporter plasmid. Transfectants were treated ii with 1,25-dihydroxyvitamin D3 (filled square), 1,25-dihydroxyvitamin D3 12 and 10-8 M AGN 193109 (filled triangle), and 1,25-dihydroxyvitamin D3 13 and 10-7M AGN 193109 (filled circle).
14 Figure 13 is a bar graph showing the effect of AGN 193109 (10 nM) coadministration on 1,25-dihydroxyvitamin D3-mediated inhibition 16 of TPA induced Str-AP1-CAT activity. Filled bars represent inhibition 17 of CAT activity in transfected cells treated with 1,25-dihydroxyvitamin 18 D3 alone. Open bars represent inhibition of CAT activity in transfected 19 cells treated with the combination of 1,25-dihydroxyvitamin D3 and 2o AGN 193109.
21 Figure 14 is a line graph showing the effect of AGN 193109 alone 22 and in combination with AGN 191183 on HeLa cells cotransfected with 23 RAR-y and the RAR responsive MTV-TREp-Luc reporter construct.
24 Drug treatments illustrated in the graph are: AGN 193109 alone (square), AGN 193109 in combination with AGN 191183 at 10-10 M
26 (diamond) and AGN 193109 in combination with AGN 191183 at 10-9 27 M.
28 Figure 15 is a line graph showing that ECE16-1 cells proliferated 29 in response to EGF (filled square) but not in response to defined 1 medium alone (open circle). Cells treated with AGN 193109 alone are 2 represented by the filled triangle. The filled circles represent results 3. obtained for cells treated with 10 nM AGN 191183 and 0 - 1000 nM
4 AGN 193109.
Figure 16 is a bar graph showing the effect of AGN 193109 on 6 the proliferation of CaSki cells in the presence or absence of the AGN
7 191183 retinoid agonist. All sample groups received 20 ng/ml of 8 epidermal growth factor (EGF) with the exception of the sample 9 propagated in defined medium (DM) alone (open bar). Stripped bars 1 o represent samples propagated in the absence of AGN 193109. Filled 11 bars represent samples propagated in the presence of 1000 nM AGN
12 193109. The concentrations of AGN 191183 used in the procedure are 13 shown on the horizontal axis.
14 Figure 17 is a dose response curve showing that AGN 193109 potentiated the antiproliferative activity of ATRA on retinal pigment 16 epithelium (RPE) cells. Samples treated with ATRA alone are 17 represented by filled squares. Samples treated with the combination of 18 ATRA and AGN 193109 (10-7M) are represented by filled circles. The 19 ATRA concentration used for treating the various samples is given on the horizontal axis.
21 Figure 18 is a dose response curve showing that both 13-cis-RA
22 and ATRA inhibited RPE cell growth, and that AGN 193109 23 potentiated the antiproliferative activity of 13-cis-RA. The various 24 sample treatments shown in the dose response included 13-cis-RA alone (filled square), 13-cis-RA in combination with AGN 193109 (10-6 M) 26 (filled circle), 13-cis-RA in combination with AGN 193109 (10-8 M) 27 (filled triangle), and ATRA (filled diamond). The concentrations of 13-28 cis-RA and ATRA used in the sample treatments are shown on the 29 horizontal axis.

1 Figure 19 is a dose response curve showing that AGN 193109 2 potentiated the antiproliferative activity of dexamethasone in primary 3 RPE cell cultures. The various sample treatments shown in the dose 4 response included ATRA (filled square), dexamethasone alone (filled circle), dexamethasone in combination with AGN 193109 (10-8 M) (filled 6 triangle), and dexamethasone in combination with AGN 193109 (10-6 M) 7 (filled diamond). The concentrations of dexamethasone and ATRA
8 used in the sample treatments are shown on the horizontal axis.
9 Figure 20 is a dose response curve showing that AGN 193109 1 o potentiated the antiproliferative activity of thyroid hormone (T3) in 11 primary RPE cell cultures. The various sample treatments shown in the 12 dose response included ATRA (filled square), T3 alone (filled circle), 13 T3 in combination with AGN 193109 (10-8 M) (filled triangle), T3 in 14 combination with AGN 193109 (10-6 M) (filled diamond). The concentrations of T3 and ATRA used in the sample treatments are 16 shown on the horizontal axis.
17 Detailed Description of the Invention 18 Definitions 19 For the purposes of the present invention, an RAR antagonist is 2o defined as a chemical that binds to one or more of the RAR subtypes 21 with a Kdof less than 1 micromolar (Kd < 1 M) but which does not 22 cause significant transcriptional activation of that RAR subtypes-23 regulated genes in a receptor co-transfection assay. Conventionally, 24 antagonists are chemical agents that inhibit the activities of agonists.
Thus, the activity of a receptor antagonist is conventionally measured by 26 virtue of its ability to inhibit the activity of an agonist.
27 An RAR agonist is defined as a chemical that binds to one or 28 more RAR receptor subtype with I{d of less than 1 micromol (K.d < 1 29 M) and causes transcriptional activation of that RAR-subtype-1 regulated genes in a receptor co-transfection assay. The term "RAR
2 agonist" includes chemicals that may bind and/or activate other 3 receptors in addition to RARs, e.g., RXR receptors.
4 As used herein, a negative hormone or inverse agonist is a ligand 5 for a receptor which causes the receptor to adopt an inactive state 6 relative to a basal state occurring in the absence of any ligand. Thus, 7 while an antagonist can inhibit the activity of an agonist, a negative 8 hormone is a ligand that can alter the conformation of the receptor in 9 the absence of an agonist. The concept of a negative hormone or 1 o inverse agonist has been explored by Bond et al. in Nature 374:272 11 (1995). More specifically, Bond et al. have proposed that unliganded 12 f32-adrenoceptor exists in an equilibrium between an inactive 13 conformation and a spontaneously active conformation. Agonists are 14 proposed to stabilize the receptor in an active conformation.
15 Conversely, inverse agonists are believed to stabilize an inactive receptor 16 conformation. Thus, while an antagonist manifests its activity by virtue 17 of inhibiting an agonist, a negative hormone can additionally manifest 18 its activity in the absence of an agonist by inhibiting the spontaneous 19 conversion of an unliganded receptor to an active conformation. Only a 20 subset of antagonists will act as negative hormones. As disclosed 21 herein, AGN 193109 is both an antagonist and a negative hormone. To 22 date, no other retinoids have been shown to have negative hormone 23 activity.
24 As used herein, coadministration of two pharmacologically active compounds refers to the delivery of two separate chemical entities, 26 whether in vitro or in vivo. Coadministration refers to the simultaneous 27 delivery of separate agents; to the simultaneous delivery of a mixture of 28 agents; as well as to the delivery of one agent followed by delivery of 29 the second agent. In all cases, agents that are coadministered are 1 intended to work in conjunction with each other.
2 The term alkyl refers to and covers any and all groups which are 3 known as normal alkyl, branched-chain alkyl and cycloalkyl. The term 4 alkenyl refers to and covers normal alkenyl, branch chain alkenyl and cycloalkenyl groups having one or more sites of unsaturation. Similarly, 6 the term alkynyl refers to and covers normal alkynyl, and branch chain 7 alkynyl groups having one or more triple bonds.
8 Lower alkyl means the above-defined broad definition of alkyl s groups having 1 to 6 carbons in case of normal lower alkyl, and as 1 o applicable 3 to 6 carbons for lower branch chained and cycloalkyl 11 groups. Lower alkenyl is defined similarly having 2 to 6 carbons for 12 normal lower alkenyl groups, and 3 to 6 carbons for branch chained and 13 cyclo- lower alkenyl groups. Lower alkynyl is also defined similarly, 14 having 2 to 6 carbons for normal lower alkynyl groups, and 4 to 6 carbons for branch chained lower alkynyl groups.
16 The term "ester" as used here refers to and covers any compound 17 falling within the definition of that term as classically used in organic 18 chemistry. It includes organic and inorganic esters. Where B (of 19 Formula 1 or Formula 101) is -COOH, this term covers the products derived from treatment of this function with alcohols or thiols 21 preferably with aliphatic alcohols having 1-6 carbons. Where the ester 22 is derived from compounds where B is -CHZOH, this term covers 23 compounds derived from organic acids capable of forming esters 24 including phosphorous based and sulfur based acids, or compounds of the formula -CH2OCORõ where R11 is any substituted or unsubstituted 26 aliphatic, aromatic, heteroaromatic or aliphatic aromatic group, 27 preferably with 1-6 carbons in the aliphatic portions.
28 Unless stated otherwise in this application, preferred esters are 29 derived from the saturated aliphatic alcohols or acids of ten or fewer Wo 99/33821 PCT/US98/27314 1 carbon atoms or the cyclic or saturated aliphatic cyclic alcohols and 2 acids of 5 to 10 carbon atoms. Particularly preferred aliphatic esters are 3 those derived from lower alkyl acids and alcohols. Also preferred are 4 the phenyl or lower alkyl phenyl esters.
Amides has the meaning classically accorded that term in organic 6 chemistry. In this instance it includes the unsubstituted amides and all 7 aliphatic and aromatic mono- and di- substituted amides. Unless stated 8 otherwise in this application, preferred amides are the mono- and di-s substituted amides derived from the saturated aliphatic radicals of ten 1 o or fewer carbon atoms or the cyclic or saturated aliphatic-cyclic radicals 11 of 5 to 10 carbon atoms. Particularly preferred amides are those 12 derived from substituted and unsubstituted lower alkyl amines. Also 13 preferred are mono- and disubstituted amides derived from the 14 substituted and unsubstituted phenyl or lower alkylphenyl amines.
Unsubstituted amides are also preferred.
16 Acetals and ketals include the radicals of the formula-CK where 17 K is (-OR)2. Here, R is lower alkyl. Also, K may be -OR70- where R7 18 is lower alkyl of 2-5 carbon atoms, straight chain or branched.
19 A pharmaceutically acceptable salt may be prepared for any compounds in this invention having a functionality capable of forming a 21 salt, for example an acid functionality. A pharmaceutically acceptable 22 salt is any salt which retains the activity of the parent compound and 23 does not impart any deleterious or untoward effect on the subject to 24 which it is administered and in the context in which it is administered.
Pharmaceutically acceptable salts may be derived from organic or 26 inorganic bases. The salt may be a mono or polyvalent ion. Of 27 particular interest are the inorganic ions, sodium, potassium, calcium, 28 and magnesium. Organic salts may be made with amines, particularly 29 ammonium salts such as mono-, di- and trialkyl amines or ethanol 1 amines. Salts may also be formed with caffeine, tromethamine and 2 similar molecules. Where there is a nitrogen sufficiently basic as to be 3 capable of forming acid addition salts, such may be formed with any 4 inorganic or organic acids or alkylating agent such as methyl iodide.
Preferred salts are those formed with inorganic acids such as 6 hydrochloric acid, sulfuric acid or phosphoric acid. Any of a number of 7 simple organic acids such as mono-, di- or tri- acid may also be used.
8 Some of the compounds of the present invention may have trans 9 and cis (E and Z) isomers. In addition, the compounds of the present 1 o invention may contain one or more chiral centers and therefore may 11 exist in enantiomeric and diastereomeric forms. The scope of the 12 present invention is intended to cover all such isomers per se, as well as 13 mixtures of cis and trans isomers, mixtures of diastereomers and racemic 14 mixtures of enantiomers (optical isomers) as well. In the present application when no specific mention is made of the configuration (cis, 16 trans or R or S) of a compound (or of an asymmetric carbon) then a 17 mixture of such isomers, or either one of the isomers is intended.
18 Aryl Substituted Benzoavran. Benzothiopyran. 1.2-Dihydroquinoline and 19 5.6-Dihydronaphthalene Derivatives Having Retinoid Antagonist Like Biological Activitv 21 With reference to the symbol Y in Formula 1, the preferred 22 compounds of the invention are those where Y is phenyl, pyridyl, thienyl 23 or furyl. Even more preferred are compounds where Y is phenyl or 24 pyridyl, and still more preferred where Y is phenyl. As far as substitutions on the Y (phenyl) and Y (pyridyl) groups are concerned, 26 compounds are preferred where the phenyl group is 1,4 (para) 27 substituted by the Z and A-B groups, and where the pyridine ring is 2,5 28 substituted by the Z and A-B groups. (Substitution in the 2,5 positions 29 in the "pyridine" nomenclature corresponds to substitution in the 6-1 position in the "nicotinic acid" nomenclature.) In the preferred 2 compounds of the invention either there is no optional R2 substituent 3 on the Y group, or the optional R2 substituent is fluoro (F).
4 The A-B group of the preferred compounds is (CH2)n COOH or 5(CH2)n COOR8, where n and R8 are defined as above. Even more 6 preferably n is zero and R8 is lower alkyl, or n is zero and B is COOH
7 or a pharmaceutically acceptable salt thereof.
8 In many of the presently preferred examples of compounds of the 9 invention X is [C(R,)2]õ where n is 1. Compounds where n is zero lo (indene derivatives) and where X is S or O(benzothiopyran and 11 benzopyran derivatives) are also preferred. When X is [C(R,)2]õ and n 12 is 1, then R, preferably is alkyl of 1 to 6 carbons, even more preferably 13 methyl.
14 The R2 group attached to the aromatic portion of the 15 tetrahydronaphthalene, benzopyran, benzothiopyran or dihydroquinoline 16 moiety of the compounds of Formula 1 is preferably H, F, CF3. or 17 alkoxy such as OCH3. R3 is preferably hydrogen or methyl, even more 18 preferably hydrogen.
19 The R2 group attached to the Y group, when there is such R2 20 substituent, is preferably, F or CF3.
21 Referring now to the group Z in the compounds of the invention 22 and shown in Formula 1, in a plurality of preferred examples Z
23 represents an acetylenic linkage (Z =-C=C-). However, the "linker 24 group" Z is also preferred as a diazo group (Z = -N=N-), as an olefinic 25 or polyolefinic group (Z =-(CR,=CR1)Q,-), as an ester (Z = -COO-), 26 amide (Z = -CO-NR2-) or thioamide (Z = -CS-NR2-) linkage.
27 Referring now to the R14 group, compounds are preferred where 28 R14 is phenyl, 2-pyridyl, 3-pyridyl, 2- thienyl, and 2-thiazolyl. The R,s 29 group (substituent of the R14 group) is preferably H, lower alkyl, 1 trifluoromethyl, chlorine, lower alkoxy or hydroxy.
2 The presently most preferred compounds of the invention 3 according to Formula 1 are shown in Table 1 with reference to Formula 4 2, Formula 3, Formula 4, Formula 5, and Formula 5a.

S ~.
7 R,:
OW !~.
8 ~ ~ ~ ~ =
=, ,,, i 9 cH, cK, 11 Formula 2 Formula 3, RU= sRa R,..
16 / ~ = / / I \-~ \ \ sR**
17 \ I \
18 CH' Formula 4 Fosmula 5 24 R+: ~' ~=

0s CHs 29 Formula Sa 2 Compound Formula R14 Z RZ' RS
3 #

1 2 4-methylphenyl -C=-C- H Et 6 la 2 phenyl -C=-C- H Et 7 2 2 3-methylphenyl -C=-C- H Et 8 3 2 2-methylphenyl -C=-C- H Et 9 4 2 3,5-d'unethylphenyl -C=-C- H Et 5 2 4-ethylphenyl -C=-C- H Et 11 6 2 4-t-butylphenyl -C=-C- H Et 12 7 2 4-chlorophenyl -C=-C- H Et 13 8 2 4-methoxyphenyl -C=-C- H Et 14 9 2 4-trifluoromethylphenyl -C=-C- H Et 10 2 2-pyridyl -C=-C- H Et 16 11 2 3-pyridyl -C= C- H Et 17 12 2 2-methyl-5-pyridyl -C=-C- H Et 18 13 2 3-hydroxyphenyl -C= C- H Et 19 14 2 4-hydroxy phenyl -C=-C- H Et 15 2 5-methyl-2-thiazolyl -C=-C- H Et 21 15a 2 2-thiazolyl -C=-C- H Et 22 16 2 4-methyl-2-thiazolyl -C=-C- H Et 23 17 2 4,5-dimethyl-2-thiazoly -C=-C- H Et 24 18 2 2-methyl-5-pyridyl -C=-C- H H
19 2 2-pyridyl -C=-C- H H
26 20 2 3-methylphenyl -C_C- H H
27 21 2 4-ethylphenyl -C=-C- H H
28 22 2 4-methoxyphenyl -C=-C- H H
29 23 2 4-trifluoromethylphenyl -C=-C- H H
24 2 3,5-dimethylphenyl -C=-C- H H
31 25 2 4-chlorophenyl -C=-C- H H
32 26 2 3-pyridyl -C=-C- H H
33 27 2 2-methylphenyl -C=-C- H H
34 28 2 3-hydroxyphenyl -C=-C- H H
29 2 4-hydroxyphenyl -C=-C- H H
36 30 2 5-methyl-2-thiazolyl -C=-C- H H
37 30a 2 2-thiazolyl -C=-C- H H
38 31 2 4-methyl-2-thiazolyl -C=-C- H H
39 32 2 4,5-dimethyl-2-thiazolyl -C=-C- H H
33 2 5-methyl-2-thienyl -C=-C- H Et 41 33a 2 2-thienyl -C=-C- H Et 42 34 2 5-methyl-2-thienyl -C=-C- H H
43 34a 2 2-thienyl -C=-C- H H

1 35 2 4-methylphenyl -CONH- H Et 2 36 2 4-methylphenyl -CONH- H H
3 37 2 4-methylphenyl -COO- H Et 4 38 2 4-methyiphenyl -COO- H (CH2)2Si(CH3) 39 2 4-methylphenyl -COO- H H
6 40 2 4-methylphenyl -CONH- F Et 7 41 2 4-methylphenyl -CONH F H
8 42 2 4-methylphenyl -CSNH- H Et 9 43 2 4-methylphenyl -CSNH- H H
44 2 4-methyiphenyl -CH = CH- H Et 11 45 2 4-methylphenyl -CH=CH- H H
12 46a 2 4-methylphenyl -N=N- H Et 13 46b 2 4-methylphenyl -N=N- H H
14 47 3 4-methylphenyl -C= C- H Et 48 3 4-methylphenyl -C- C- H H
16 49 4 4-methylphenyl -C=-C- H Et 17 50 4 4-methylphenyl -C=-C- H H
18 51 5 4-methylphenyl - - Et 19 52 5 4-methylphenyl - - H
60 2 4-methylphenyl -C=C- H H
21 60a 2 phenyl -C=-C- H H
22 61 2 4-t-butylphenyl -C=C- H H
23 62 2 4-methylphenyl -CSNH F Et 24 63 2 4-methylphenyl -CSNH F H
64 5a 4-methylphenyl ---- - Et 26 65 5a 4-methylphenyl ---- - H
27 66 2 2-furyl -C=-C- H Et 28 67 2 2-furyl -C=-C- H H

Still further most preferred compounds of the invention are in 31 accordance with Formula 5b which are further identified in Table la and in 32 the ensuing experimental section.

34 co1H
Ru 36 Rs 39 Formula 5b 1 Table la 2 Compound # R14 X, X2 R2 R2 3 Page #
4 242 phenyl S CH H H
243 4-methylphenyl S CH H H
6 244 4-ethylphenyl S CH H H
7 245 4-i-propylphenyl S CH H H
8 246 4-t-butylphenyl S CH H H
9 247 5-methyl-2-thienyl S CH H H
248 5-ethyl-2-thienyl S CH H H
11 249 5-t-butyl-2-thienyl S CH H H
12 250 6-methyl-3-pyridyl S CH H H
13 251 4-methylphenyl S CH H F
14 252 4-ethylphenyl S CH H F
253 4-methylphenyl S N H H
16 254 4-ethylphenyl S N H H
17 255 4-methylphenyl S CH F H
18 256 4-methylphenyl S CH OCH3 H
19 257 5-methyl-2-thienyl S CH F H
274 phenyl 0 CH H H
21 275 4-methylphenyl 0 CH H H
22 276 4-ethylphenyl 0 CH H H
23 277 4-isopropylphenyl 0 CH H H
24 278 4-t-butylphenyl 0 CH H H
279 4-methylphenyl 0 CH H F
26 280 4-ethylphenyl 0 CH H F

1 Aryl and (3-Oxo-l-Propenvl)-Substituted Benzopyran. Benzothiopyran.
2 Dihkdroquinoline and 5 6-Dihydronaphthalene Derivatives Having Retinoid 3 Antagonist-Like Biolocaical Activitv 4 With reference to the symbol Y in Formula 101, the preferred compounds of the invention are those where Y is phenyl, pyridyl, thienyl or 6 furyl. Even more preferred are compounds where Y is phenyl or pyridyl, and 7 still more preferred where Y is phenyl. As far as substitutions on the Y
s(phenyl) and Y (pyridyl) groups are concerned, compounds are preferred 9 where the phenyl group is 1,4 (para) substituted by the -CR16=CR17- and A-B
1o groups, and where the pyridine ring is 2,5 substituted by the -CR16=CRõ
11 and A-B groups. (Substitution in the 2,5 positions in the "pyridine"
12 nomenclature corresponds to substitution in the 6- position in the "nicotinic 13 acid" nomenclature.) In the preferred compounds of the invention there is 14 no optional R2 substituent on the Y group.
The A-B group of the preferred compounds is (CH2)n-COOH or 16 (CH2)õCOOR8, where n and R. are defined as above. Even more preferably 17 n is zero and R. is lower alkyl, or n is zero and B is COOH or a 18 pharmaceutically acceptable salt thereof.
19 In the presently preferred examples of compounds of the invention X
is [C(Rl)2]o where n is 1. Nevertheless, compounds where X is S or 0 21 (benzothiopyran and benzopyran derivatives) are also preferred. When X is 22 [C(R,)2]a and n is 1, then R, preferably is alkyl of 1 to 6 carbons, even more 23 preferably methyl.
24 The R2 group attached to the aromatic portion of the tetrahydronaphthalene, benzopyran, benzothiopyran or dihydroquinoline 26 moiety of the compounds of Formula 101 is preferably H, F or CF3. R3 is 27 preferably hydrogen or methyl, even more preferably hydrogen.
28 Referring now to the R14 group, compounds are preferred where R14 is 29 phenyl, 2-pyridyl, 3-pyridyl, 2- thienyl, and 2-thiazolyl. The R,s group 1(substituent of the R14 group) is preferably H, lower alkyl, trifluoromethyl, 2 chlorine, lower alkoxy or hydroxy.
3 Preferred compounds of the invention according to Formula 101 are 4 shown in Table 2 with reference to Formula 102.

8 zuijocw 13 Formula 102 15 Compound Rts R8 17 102 CH3 Et 1 s 103 H H
19 104 H Et 21 Bioiooigai Activity. Modes of Administration 22 As noted above, the compounds of the present invention are 23 antagonists of one or more RAR receptor subtypes. This means that the 24 compounds of the invention bind to one or more RAR receptor subtypes, but do not trigger the response which is triggered by agonists of the same 26 receptors. Some of the compounds of the present invention are antagonists 27 of all three RAR receptor subtypes (RAR-a, RAR -P and RAR-y), and these 28 are termed "RAR pan antagonists". Some others are antagonists of only one 29 or two of RAR receptor subtypes. Some compounds within the scope of the Wo 99/33821YC'T/US98727314 1 present invention are partial agonists of one or two RAR receptor subtypes 2 and antagonists of the remaining subtypes. The compounds of the invention 3 do not bind to RXR receptors, therefore they are neither agonists nor 4 antagonists of RXR.
Depending on the site and nature of undesirable side effects which are 6 desired to be suppressed or ameliorated, compounds used in accordance with . ,.
7 the invention may be antagonists of only one or two of RAR receptor s subtypes. Some compounds used in accordance with the invention may be 9 partial agonists of one or two RAR receptor subtypes and antagonists of the remaining subtypes. Such compounds are, generally speaking, usable in 11 accordance with the invention if the antagonist effect is on that RAR
12 receptor subtype (or subtypes) which is (are) predominantly responsible for 13 the overdose poisoning or for the undesired side effect or side effects. In this 14 connection it is noted that, generally speaking, a compound is considered an 15antagonist of a given receptor subtype if in the below described co-tranfection 16 assays the compound does not cause significant transcriptional activation of 17 the receptor regulated reporter gene, but nevertheless binds to the receptor ts with a Kdvalue of less than appr-oximately.l M.
19 '. Whether a compound is an RAR antagonist and therefore can be utilized in accordance with the present invention, can be tested in the 21 following assays.
22 : A chimeric receptor transactivation assay which tests for agonist-like 23 activity in the RAR-a, RAR=P, RAR-Y, RXR-a receptor, =stabtypes, and which 24 is based on work published by Feigner P. L. and Hoim M. Focus Vol 11, No.
2(1989) is described in detail in published PCT Application No.
26 W:094/17796, published on August 18, 1994. The latter publication is the 27 PCT counterpart of U. S. application serial no. 08/016,404, filed on February 28 11, 1993, which issued as U.S. Patent No. 5,455,265.

I A compound should not cause 2 significant activation of a reporter gene through a given receptor subtype 3 (RAR-a, RAR-P or RAR-y) in this assay, in order to qualify as an RAR
4 antagonist with utility in the present invention.
A holoreceptor transactivation assay and a ligand binding assay which 6 measure the antagonist/agonist like activity of the compounds of the 7 invention, or their ability to bind to the several retinoid receptor subtypes, 8 respectively, are described in published PCT Application No. W093/11755 9(particularly on pages 30 - 33 and 37 - 41) published on June 24, 19931.
A description 11 of the holoreceptor transactivation assay is also provided below.
12 Holoreceptor Transactivation AssaX
13 CV1 cells (5,000 cells/well) were transfected with an RAR reporter 14 plasmid IvITV-TREp-LUC (50 ng).along with one of the RAR expression vectors (10 ng) in an automated 96-well format by the calcium phosphate 16 procedure of Heymanet al. Cell 68: 397 - 406. For RXR-a and RXR-y 17 transactivation assays, an RXR-responsive reporter plasmid CRBPII-tk-LUC
18 (50 ng) along with the appropriate RXR expression vectors (10 ng) was used 19 substantially as described by Heyman et al. above, and Allegretto et al. J.
Biol. Chem. 268: 26625 - 26633. For RXR-,8 transactivation assays, an RXR-21 responsive reporter plasmid CPRE-tk-LUC (50 mg) along with RXR -P
22 expression vector (10 mg) was used as described in above. These reporters 23 contain DRI elements from hulrianCRBPII and certain DRI elements from 24 promotor, respectiv.ely (see Mangelsdorf et al. The Retinoids: Biologv.
Chemistrk and Medicine, pp. 319 - 349, Raven Press Ltd., New York and 26 Heyman et al., cited above). Afl-galactosidase (50 ng) expression vector was 27 used as an internal control in the transfections to normalize for variations in 28 transfection eff'iciency. The cells were transfected in triplicate for 6 hours, 29 followed by incubation with retinoids for 36 hours, and the extracts were WO "/33821 PCTNS98J27314 i assayed for luciferase andfl-galactosidase activities. The detailed 2 experimental procedure for holoreceptor transactivations has been described 3 in Heyman et al. above, and Allegretto et al. cited above. The results a obtained in this assay are expressed in EC~O numbers, as,they are also in the s chimeric receptor transactivation assay.

s The results s of ligand binding assay are expressed in K,d numbers. .(See Cheng et al.
Biocliemical Phannacology 22: 3099-3108.) 12 Still another transactiivation assay, the "PGR assay" is described in the 13 publication Klein et al, J. Biol. Chem. 271, 22692-22696 (1996), The results of the PGR assay are a-Iso expressed in EC~O
.16 numbers (nanomolar concentration).
17 RARMPGR Holoreceptor, Transactivattion Assay 18 CV-1 cells (4 x 105 cells/well) were transiently transfected with the .19 luciferase reporter plaspnid..:1VITV-4(RSG)-Luc (0.7 ug/well) containing four copies- of the R5G retinoid DNA response element along with the RXRa 21. . expression plasMid pRS-hRXRa (0.1 ug/well) and one of the RAR P-GR
22 expression plasmids (0.05 ug/well) in 12 well plates via calcium phosphate 23 precipitation Chen et al. (1987) Mol. Cell. Biol. 7, 2745-2752 as described by 24 Klein et aL in J. Biol. -Chem. 271, 22692, referenced above. The three . different RAR P-GR 'expression plasmids, pRS-RARa-P-GR, pcDNA3-26 RAR,8-P-GR and pcDI~A3-RARy-P-GR, express RARa, RAR.8 and RARy 27 receptors, respectively, which contain modified DNA binding domains such 28 that their "P-boxes" have been altered to that of the gluoocorticoid receptor.
2a. These. RAR-P-GR.receptprs bind to DNA as heterodimeric complexes with 1 RXR. Specifically, the RAR-P-GR receptors bind retinoic acid response 2 elements designated R5G, comprised of two RAR half sites (nucleotide 3 sequence 5'-GGTTCA-3') separated by 5 base pairs in which the 3'-half site 4 has been modified to that of a glucocorticoid receptor half site, 5'-AGAACA-3'. To allow for various in transfection efficiency a0-galactosidase expression 6 plasmid (0.01 ug/well) was used as an internal control. Alternatively, the 7 assay was performed in a 96-well microtiter plate format (5000 cells/well) in a 8 manner which was identical to that described above except 115 of the amount 9 of the DNA-calcium phosphate precipitant (20 l instead of 100 l) was lo applied to each well. Eighteen hours after introduction of the DNA
11 precipitants, cells were rinsed with phosphate buffered saline (PBS) and fed 12 with D-MEM (Gibco-BRL) containing 10% activated charcoal extracted fetal 13 bovine serum (Gemini Bio-Products). Cells were treated for 18 hours with 14 the compounds indicated in the figures. After rinsing with PBS cells were lysed with luciferase activity was measured as previously described in de Wet 16 (1987) Mol. Cell. Biol. 7, 725-737. Luciferase values represent the 17 mean SEM of triplicate determinations normalized to,8-galactosidase i8 activity.
19 A compound should not cause significant activation of a reporter gene through a given receptor subtype (RAR-a, RAR -0 or RAR-y ) in the 21 transactivation assays, in order to qualify as an RAR antagonist with utility in 22 the present invention. Last, but not least, a compound should bind to at least 23 one of the RAR receptor subtypes in the ligand binding assay with a Kd of 24 less than approximately 1 micromolar (Kd< 1 M) in order to be capable of functioning as an antagonist of the bound receptor subtype, provided the 26 same receptor subtype is not significantly activated by the compound.
27 Table 3 below shows the results of the holoreceptor transactivation 28 assay for certain compounds of Table 1. Table 3a shows the results of the 29 RAR-PGR moloreceptor transactivation assay for the preferred compounds 1 of Table la. Tables 4 and 4a disclose the efficacy (in percentage) in these 2 assays of the test compounds relative to all trans retinoic acid, for certain 3 exemplary compounds of the invention. Tables 5 and 5a show the results of 4 the ligand binding assay for certain exemplary compounds of the invention.

6 Holoreceptor Transactivation Assav 7 Compound # EC,, (nanomolar) 8 RARcr RARB RARy RXRcr RXRB RXRy 1 o 18 0.00 0.00 0.00 0.00 0.00 0.00 11 19 0.00 0.00 0.00 0.00 0.00 0.00 12 20 0.00 0.00 0.00 0.00 0.00 0.00 13 21 0.00 0.00 0.00 0.00 0.00 0.00 14 22 0.00 0.00 0.00 0.00 0.00 0.00 15 23 0.00 0.00 0.00 0.00 0.00 0.00 16 24 0.00 0.00 0.00 0.00 0.00 0.00 17 .25 0.00 0.00 0.00 0.00 0.00 0.00 18 26 0.00 0.00 Ø00 0.00 0.00 0.00 19 27 0.00 0.00 0.00 0.00 0.00 0.00 2o 28 0.00 0.00 0.00 0.00 0.00 0.00 21 29 0.00 0.00 0.00 0.00 0.00 0.00 22 30 0.00 0.00 0.00 0.00 0.00 0.00 23 31 0.00 0.00 0.00 0.00 0.00 0.00 24 32 0.00 0.00 0.00 0.00 0.00 0.00 25 34 0.00 0.00 0.00 0.00 0.00 0.00 2e 36 0.00 0.00 0.00 0.00 0.00 0.00 27 39 0.00 0.00 0.00 0.00 0.00 0.00 28 41 0.00 0.00 0.00 0.00 0.00 0.00 29 45 0.00 0.00 0.00 0.00 0.00 0.00 3o 46b 0.00 0.00 0.00 0.00 0.00 0.00 31 52 0.00 0.00 0.00 0.00 0.00 0.00 32 60 0.00 0.00 0.00 0.00 0.00 0.00 33 61 0.00 0.00 0.00 0.00 0.00 0.00 34 63 0.00 0.00 0.00 0.00 0.00 0.00 35 101 0.00 0.00 0.00 0.00 0.00 0.00 36 103 0.00 0.00 0.00 0.00 0.00 0.00 37 0.0 in Tables 3 and 3a indicates that the compound is less than 20 % as 38 active (efficacious) in this assay than all trans retinoic acid.

1 Table 3a 2 RAR-PGR Holoreceptor Transactivation Assay 3 Compound No. EC50 (nanomolar) 4 RARa RAR,8 RARy 242 0.00 0.00 0.00 6 243 0.00 0.00 0.00 7 244 0.00 0.00 0.00 8 245 0.00 0.00 0.00 9 246 0.00 0.00 0.00 247 0.00 0.00 0.00 11 248 0.00 0.00 0.00 12 249 0.00 0.00 0.00 13 250 0.00 0.00 0.00 14 251 0.00 0.00 0.00 252 0.00 0.00 0.00 16 253 0.00 0.00 0.00 17 254 0.00 0.00 0.00 18 255 0.00 0.00 0.00 19 256 0.00 0.00 0.00 257 0.00 0.00 0.00 21 274 0.00 0.00 0.00 22 275 0.00 0.00 0.00 23 276 0.00 0.00 0.00 24 277 0.00 0.00 0.00 278 0.00 0.00 0.00 28 279 0.00 0.00 0.00 27 280 0.00 0.00 0.00 2 Transactivation Assay Efficacx (% of RA activitv) 3 Compound RARa RARl3 RARy RXRa RXRB RXRy 4 No.
18 4.00 1.00 0.00 2.00 10.00 1.0 s 19 0.00 5.0 3.0 0.0 9.0 4.0 7 20 3.0 4.0 0.00 4.00 0.00 3.0 8 21 2.00 2.00 2.00 3.00 0.00 3.00 9 22 0.00 0.00 2.00 1.00 0.00 2.00 1 o 23 0.00 6.00 3.00 1.00 0.00 4.00 11 24 3.00 7.00 4.00 1.00 0.00 3.00 12 25 2.00 3.00 3.00 5.00 0.00 3.00 13 26 1.00 6.00 0.00 2.00 0.00 3.00 14 27 9.00 14.00 6.00 2.00 0.00 4.00 28 2.00 10.00 2.00 2.00 0.00 3.00 16 29 0.00 6.00 11.00 0.00 6.00 2.00 17 30 3.00 5.00 1.00 0.00 9.00 3.00 18 31 4.00 14.00 2.00 1.00 8.00 6.00 19 32 0.00 2.00 2.00 1.00 0.00 2.00 2o 34 3.00 5.00 2.00 1.00 0.00 3.00 21 36 1.00 5.00 0.00 1.00 7.00 2.00 22 39 1.00 7.00 9.00 2.00 0.00 1.00 23 41 3.00 5.00 6.00 1.00 0.00 3.00 24 45 2.00 0.00 7.00 3.00 8.00 0.00 46b 4.00 5.00 3.00 2.00 0.00 4.00 26 52 0.00 15.00 3.00 0.00 0.00 10.00 27 60 0.00 1.00 4.00 3.00 0.00 3.00 28 61 2.00 2.00 0.00 1.00 0.00 3.00 29 63 2.00 2.00 7.00 1.00 0.00 1.00 101 0.00 4.00 2.00 1.00 0.00 3.0 31 103 4.00 12.0 7.0 0.00 0.0 2.0 1 Table 4a 2 Compound No. Transactivation Assay Efficacy (% of RA Activity) 3 RARa RARP RARy 4 242 0.00 0.00 0.00 243 0.00 0.00 0.00 6 244 0.00 0.00 0.00 7 245 0.00 0.00 0.00 8 246 0.00 0.00 0.00 9 247 0.00 0.00 0.00 248 0.00 0.00 0.00 11 249 0.00 0.00 0.00 12 250 0.00 0.00 0.00 13 251 0.00 0.00 0.00 14 252 0.00 0.00 0.00 253 0.00 0.00 0.00 16 254 0.00 0.00 0.00 17 255 0.00 0.00 0.00 18 256 0.00 0.00 0.00 19 257 0.00 0.00 0.00 274 0.00 0.00 0.00 21 275 0.00 0.00 0.00 22 276 0.00 0.00 0.00 23 277 0.00 0.00 0.00 24 278 0.00 0.00 0.00 279 0.00 0.00 0.00 26 280 0.00 0.00 0.00 2 Ligand Binding Assay 3 Compound # Kd (nanomolar) 4 RARcr RAn RARy RXRa RXItB 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 1 o 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 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 2o 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 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 so 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 1 Table 5a 3 Ligand Binding Assay Kd (nanomolar) 4 Compound No. RARa RAW RARy 1s 255 3 3 3 19 256 > 103 393 203 29 0.0 in Tables 5 and 5a indicates a value greater than 1000nM.

1 As it can be seen from the test results summarized in Tables 3, 3a, 4, 2 4a, 5 and 5a, 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 the remaining RAR
6 subtypes.) Due to this property, the compounds of the invention can be used 7 to block the activity of RAR agonists in biological assays. In mammals, 8 including humans, the compounds of the invention can be coadministered 9 with RAR agonists and, by means of pharmacological selectivity or site-lo specific delivery, preferentially prevent the undesired effects of RAR
agonists.
11 The compounds of the invention can also be used to treat Vitamin A
12 overdose, acute or chronic, resulting either from the excessive intake of 13 vitamin A supplements or from the ingestion of liver of certain fish and 14 animals that contain high level of Vitamin A. Still further, the compounds of the invention can also be used to treat acute or chronic toxicity caused by 16 retinoid drugs. It has been known in the art that the toxicities observed with 17 hypervitaminosis A syndrome (headache, skin peeling, bone toxicity, 18 dyslipidemias) are similar or identical with toxicities observed with other 19 retinoids, suggesting a common biological cause, that is RAR activation.
2o Because the compounds of the present invention block RAR activation, they 21 are suitable for treating the foregoing toxicities.
22 The compounds of the invention are able to substantially prevent skin 23 irritation induced by RAR agonist retinoids, when the compound of the 24 invention is topically coadministered to the skin. Similarly, compounds of the invention can be administered topically to the skin, to block skin irritation, in 26 patients or animals who are administered RAR agonist compounds 27 systemically. The compounds of the invention can accelerate recovery from 28 pre-existing retinoid toxicity, can block hypertriglyceridemia caused by co-29 administered retinoids, and can block bone toxicity induced by an RAR

1 agonist (retinoid).

2 Generally speaking, for therapeutic applications in mammals in 3 accordance with the present invention, the antagonist compounds can be 4 admistered enterally or topically as an antidote to vitamin A, vitamin A
precursors, or antidote to retinoid toxicity resulting from overdose or 6 prolonged exposure, after intake of the causative factor (vitamin A
precursor 7 or other retinoid) has been discontinued. Alternatively, the antagonist 8 compounds are coadministered with retinoid drugs in accordance with the 9 invention, in situations where the retinoid provides a therapeutic benefit, and 1o where the coadministered antagonist alleviates or eliminates one or more 11 undesired side effects of the retinoid. For this type of application the 12 antagonist may be administered in a site-specific manner, for example as a 13 topically applied cream or lotion while the coadministered retinoid may be 14 given enterally.
For therapeutic applications in accordance with the present invention 16 the antagonist compounds are incorporated into pharmaceutical 17 compositions, such as tablets, pills, capsules, solutions, suspensions, creams, 18 ointments, gels, salves, lotions and the like, using such pharmaceutically 19 acceptable excipients and vehicles which per se are well known in the art.
For example preparation of topical formulations are well described in 21 Remington's Pharmaceutical Science, Edition 17, Mack Publishing Company, 22 Easton, Pennsylvania. For topical application, the antagonist compounds 23 could also be administered as a powder or spray, particularly in aerosol form.
24 If the drug is to be administered systemically, it may be confected as a powder, pill, tablet or the like or as a syrup or elixir suitable for oral 26 administration. For intravenous or intraperitoneal administration, the 27 antagonist compound will be prepared as a solution or suspension capable of 28 being administered by injection. In certain cases, it may be useful to 29 formulate the antagonist compounds by injection. In certain cases, it may be 1 useful to formulate the antagonist compounds in suppository form or as 2 extended release formulation for deposit under the skin or intramuscular 3 injection.
4 The antagonist compounds will be administered in a therapeutically effective dose in accordance with the invention. A therapeutic concentration 6 will be that concentration which effects reduction of the particular condition 7 (such as toxicity due to retinoid or vitamin A exposure, or side effect of 8 retinoid drug) or retards its expansion. It should be understood that when 9 coadministering the antagonist compounds to block retinoid-induced toxicity 1o or side effects in accordance with the invention, the antagonist compounds 11 are used in a prophylactic manner to prevent onset of a particular condition, 12 such as skin irritation.
13 A useful therapeutic or prophylactic concentration will vary from 14 condition to condition and in certain instances may vary with the severity of the condition being treated and the patient's susceptibility to treatment.
16 Accordingly, no single concentration will be uniformly useful, but will require 17 modification depending on the particularities of the chronic or acute retinoid 18 toxicity or related condition being treated. Such concentrations can be 1a arrived at through routine experimentation. However, it is anticipated that a 2o formulation containing between 0.01 and 1.0 milligrams of antagonist 21 compound per mililiter of formulation will constitute a therapeutically 22 effective concentration for topical application. If administered systemically, 23 an amount between 0.01 and 5 mg per kg per day of body weight would be 24 expected to effect a therapeutic result.
The basis of the utility of RAR antagonists for the prevention or 26 treatment of RAR agonist-induced toxicity is competitive inhibition of the 27 activation of RAR receptors by RAR agonists. The main distinction between 28 these two applications of RAR antagonists is the presence or absence of 29 preexisting retinoid toxicity. Most of the examples immediately described 1 below relate to the use of retinoids to prevent retinoid toxicity, but the 2 general methods described herein are applicable to the treatment of 3 preexisting retinoid toxicity as well.
4 Description of experiments demonstrating the use of RAR antagonists to prevent or treat retinoid toxicity and/or side effects of retinoid drugs 6 Example 1: skin iritation induced by topically applied agonist is treated with 7 topicall -pa nlied antagonist 8 The compound 4-[(E)-2-(5,6,7,8-tetrahydro-5,5,8,8-9 tetramethylnaphthalen-2-yl)propen-1-yl]benzoic acid, designated AGN
1 o 191183, is known in the prior art as a potent RAR agonist (see for example 11 the descriptive portion and figure 2b of United States Patent No.
5,324,840).
12 (The "AGN" number is an arbitrarily designated reference number utilized by 13 the corporate assignee of the present invention for identification of 14 compounds.) 4-[(5,6-dihydro-5,5-dimethyl-8-(phenyl)-2- naphthalenyl)ethynyl]benzoic 16 acid (AGN 192869, also designated Compound 60a) is a compound the 17 preparation of which is described below. This compound is an RAR
18 antagonist.
19 Skin irritation induced by an RAR agonist, AGN 191183, administered topically, can be blocked by an RAR antagonist, AGN 192869, also 21 administered topically in hairless mice.
22 More particularly skin irritation was measured on a semiquantitative 23 scale by the daily subjective evaluation of skin flaking and abrasions. A
24 single number, the topical irritation score, summarizes the skin irritation induced in an animal during the course of an experiment. The topical 26 irritation score is calculated as follows. The topical irritation score is the 27 algebraic sum of a composite flaking score and a composite abrasion score.
28 The composite scores range from 0-9 and 0-8 for flaking and abrasions, 29 respectively, and take into account the maximum severity, the time of onset, 1 and the average severity of the flaking and abrasions observed.
2 The severity of flaking is scored on a 5-point scale and the severity of 3 abrasions is scored on a 4- point scale, with higher scores reflecting greater 4 severity. The maximum severity component of the composite scores would be 5 the highest daily severity score assigned to a given animal during the course 6 of observation.
7 For the time of onset component of the composite score, a score 8 ranging from 0 to 4 is assigned as follows:

10 Time to An12earance of Flaking or Abrasions 11 of Severity 2 or Greater 13 da s Time of Onset Score 21 The average severity component of the composite score is the sum of 22 the daily flaking or abrasion scores divided by the number of observation 23 days. The first day of treatment is not counted, since the drug compound has 24 not had an opportunity to take effect at the time of first treatment.
To calculate the composite flaking and abrasion scores, the average 26 severity and time of onset scores are summed and divided by 2. The result is 27 added to the maximal severity score. The composite flaking and abrasion 28 scores are then summed to give the overall topical irritation score. Each 29 animal receives a topical irritation score, and the values are expressed as the 3o mean SD of the individual scores of a group of animals. Values are 31 rounded to the nearest integer.
32 Female hairless mice [Crl:SKH1-hrBR] (8-12 weeks old, n=6) were 33 treated topically for 5 consecutive days with acetone, AGN 191183, AGN

1 192869, or some combination of AGN 192869 and 191183. Doses of the 2 respective compounds are given in Table 7. Treatments are applied to the 3 dorsal skin in a total volume of 4 ml/kg (- 0.1 ml). Mice were observed daily 4 and scored for flaking and abrasions up to and including 3 days after the last treatment, i.e., day 8.

7 Experimental Design and Results. Exam lp e 1 e Dose Dose Molar Ratio Topical 9 AGN 191183 AGN 192869 (192869: Irritation 1o Group m c d m c d (191183 Sco e 13 B 0.025 0 -- 8 2 14 C 0.025 0.06 2:1 5 2 D 0.025 0.30 10:1 2 1 16 E 0.025 1.5 50:1 1 0 17 F 0 1.5 -- 0 0 19 The topical irritation scores for Example 1 are given in Table 7.
Neither acetone (vehicle) nor AGN 192869 (antagonist) at a dose of 1.5 21 mg/kg/d (group F) caused observable topical irritation. AGN 191183, the 22 RAR agonist, caused modest topical irritation at a dose of 0.025 mg/kg/d.
23 However, AGN 191183-induced topical irritation was inhibited in a dose-24 dependent fashion by AGN 192869, with nearly complete abrogation of irritation in the presence of a 50-fold molar excess of AGN 192869. This 26 demonstrates that a topical RAR antagonist blocks skin irritation caused by a 27 topical RAR agonist. Complete blockade of RAR agonist-induced skin 28 irritation can be achieved with lower molar ratios of antagonist to agonist 29 when the RAR antagonists is more potent, such as the compound 4-[(5,6-3o dihydro-5,5- dimethyl-8-(4-methylphenyl)-2- naphthalenyl)ethynyl]benzoic acid 31 (AGN 193109, also designated in this application as Compound 60.) 32 Example 2: skin iritation induced bv orally applied agonist is blocked with 33 topically applied antagonist ~

I The potent RAR agonist AGN 191183 (4-[(E)-2- (5,6,7,8-2 tetrahydro-5,5,8,8-tetramethylnaphthalen-2- yl)propen-l-yl]benzoic acid) and 3 the potent RAR antagonist 4-[(5,6-dihydro-5,5-dimethyl-8-(4-4 methylphenyl)-2-naphthalenyl)ethynyl]benzoic acid (AGN 193109, Compound 60) were used in this example and body weight of the experimental animals s(mice) was used as a marker of systemic RAR agonist exposure.
7 Groups of female hairless mice (8-12 weeks old, n=6) were treated by 8 intragastric intubation with corn oil or AGN 191183 (0.26 mg/kg) suspended 9 in corn oil (5 ml/kg). Mice were simultaneously treated topically on the lo dorsal skin with vehicle (97.6% acetone/2.4% dimethylsulfoxide) or solutions ii of AGN 193109 in vehicle (6 ml/kg). Specific doses for the different 12 treatment groups are give in Table 8. Treatments were administered daily for 13 4 consecutive days. Mice were weighed and graded for topical irritation daily 14 as described in Example 1 up to and including 1 day after the last treatment.
Percent body weight change is calculated by subtracting final body weight 16 (day 5) from initial body weight (day 1), dividing by initial body weight, and 17 multiplying by 100%. Topical irritation scores are calculated as described in is Example 1.
19 Topical irritation scores and weight loss for the different groups are given in Table 8. Combined treatment with the topical and oral vehicles, i.e., 21 acetone and corn oil, respectively, caused no topical irritation or weight loss.
22 Similarly, combined treatment with the oral vehicle and the topical antagonist 23 AGN 193109 resulted in no topical irritation or weight loss. Oral AGN
24 191183 by itself induced substantial weight loss and skin irritation. AGN
191183-induced skin irritation was substantially reduced when combined with 26 the lower dose of AGN 193109 and completely blocked at the higher dose of 27 AGN 193109. AGN 191183-induced weight loss was also blocked in a dose-28 related fashion by topical AGN 193109, but the blockade was not complete.
29 Thus, topical AGN 193109 preferentially blocked the dermal toxicity of AGN

1 191183. Presumably, low levels of AGN 193109 were absorbed systemically 2 and thus partially blocked the weight loss induced by AGN 191183. However, 3 such absorption would likely be even less in a species with less permeable 4 skin, such as humans. Alternatively, the weight loss inhibition by AGN

could be due to amelioration of the AGN 191183 induced skin irritation.

7 Experimental Design and Results. Examlp e 2 8 Dose of Topical Dose of Oral % Weight Topical 9 AGN 193109 AGN 191183 Gain or Irritation 1 o Group ~ (m PJkg(d) (Loss) Score) 13 B 0 0.26 (21 6) 8 1 14 C 0.12 0.26 (9 5) 1 1 D 0.47 0.26 (3 5) 0 1 16 E 0.47 0 3 3 0 0 18 Thus, Example 2 demonstrates that RAR antagonists administered 19 topically can be used to block preferentially the skin irritation induced by an 2o RAR agonist administered orally.
21 Example 3: topically applied antagonist accelarates recovery from prexisting 22 retinoid toxicitv 23 In this example, weight loss is induced by topical treatment with the 24 RAR agonist AGN 191183 and then the test animals are topically treated with either vehicle or the RAR antagonist AGN 193109.
26 Female hairless mice (8-12 weeks old, n=5) were treated topically with 27 AGN 191183 (0.13 mg/kg/d) in vehicle (97.6% acetone/2.4% DMSO, 4 ml/kg) 28 daily for 2 days. Groups of these same mice (n=5) were then treated 29 topically either with vehicle or AGN 193109 in vehicle (4 ml/kg) daily for consecutive days beginning on day 3. Mice were weighed on days 1-5 and on 31 day 8. Body weights are expressed as the mean SD. Means were 32 compared statistically using an unpaired, two-tailed t-test. Differences were 33 considered significant at P < 0.05.

~

2 Results. Example 3 3 Treatment Body Weight (g) 4 (days 3-5) 7 vehicle 24.6t1.5 23.9t1.2 21.4t1.2 20.3 1.7 21.0 1.4 24.7 1.0 8 AGN 193109 23.9f1.0 233 t1.2 21.4t0.6 22.2t0.7 22.8 0.8 25.0 1.1 The time course of body weights in Example 3 are given in Table 9.
11 Body weights of both groups of mice were lowered in parallel on days 2 and 12 3 as a result of AGN 191183 treatment on days 1 and 2. Body weights in the 13 two groups were not significantly different on days 1, 2, or 3. However, AGN
14 193109 treatment significantly increased body weight relative to vehicle treatment on days 4 and 5. These data indicated that recovery from AGN
16 191183-induced body weight loss was accelerated by subsequent treatment 17 with AGN 193109. Body weights were not significantly different between the 1s two groups of mice on day 8, indicating that full recovery was achievable in 19 both groups given sufficient time. Thus, RAR antagonists are effective in 2o alleviating RAR agonist-induced toxicity even if RAR agonist-induced toxicity 21 precedes RAR antagonist treatment, i.e., in the RAR agonist poisoning 22 scenario.
23 Example 4: orally applied antagonist blocks hypertriglyceridemia incuded by 24 orally cogdministered retinoid agonist 5-[(E)-2-(5,6,7,8-tetrahydro-3,5,5,8,8-pentamethylnaphthalen-2-26 yl)propen-1-yl]-2- thiophencarboxylic acid, is a known RAR/RXR pan-27 agonist (see United States Patent No. 5,324,840 column 32) and is designated 28 AGN 191659. This compound was used orally to induce acute 29 hypertriglyceridemia in rats, and AGN 193109 Compound 60 was coadministered orally to block the AGN 191659-induced hypertriglyceridemia.
31 Male Fischer rats (6-7 weeks old, n=5) were treated by intragastric 32 intubation with corn oil (vehicle), AGN 191659, AGN 193109 or a 1 combination of AGN 191659 and AGN 193109. AGN 191659 and AGN
2 193109 were given as fine suspensions in corn oil. The experimental design, 3 including doses, is given in Table 10.
4 Blood was withdrawn from the inferior vena cava under carbon dioxide 5 narcosis. Serum was separated from blood by low speed centrifugation.
6 Total serum triglycerides (triglycerides plus glycerol) were measured with a 7 standard spectrophotometric endpoint assay available commercially as a kit 8 and adapted to a 96-well plate format. Serum triglyceride levels are 9 expressed as the mean SD. Means were compared statistically by one-way 1 o analysis of variance followed by Dunnett's test if significant differences were 11 found. Differences were considered significant at P < 0.05.
12 As shown in Table 10, AGN 191659 by itself caused significant 13 elevation of serum triglycerides relative to vehicle treatment. AGN 193109 14 by itself did not significantly increase serum triglycerides. Importantly, the 15 combination of AGN 193109 and AGN 191659 at molar ratios of 1:1 and 5:1 1 e reduced serum triglycerides to levels that were not signficantly different from 17 control.
1 s TABLE 10 19 erimental Desig;n and Results. Example 4 2o Gron Treatment f dosel Serum Trig.lxgerides (mai) 22 A vehicle 55.0 3.1 23 B AGN 193109 (19.6 mg/kg) 52.4 6.3 24 C AGN 191659 (3.7 mg/kg) 122.5 27.6 25 D AGN 193109 (3.9 mg/kg) 55.7 14.7 26 + AGN 191659 (3.7 mg/kg) 27 E AGN 193109 (19.6 mg/kg) 72.7 8.9 28 + AGN 191659 (3.7 mg/kg) 30 Example 4 demonstrates that an RAR antagonist can be used to block 31 hypertriglyceridemia induced by a coadministered retinoid.
32 Example 5: parenterally aRplied antag,onist blocks bone toxicitX incuded by 33 parenterally_coadministered retinoid agonist A

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

12 R+4 13 (Rj =
14 R~

17 Formula 1 18 In Formula 1, X is S, 0, NR' where R' is H or alkyl of 1 to 6 carbons, 19 or X is [C(R)z]A where R1 is H or alkyl of 1 to 6 carbons, and n is an 21 integer between 0 or 1;
22 R2 is hydrogen, lower alkyl of 1 to 6 carbons, F, CF3, fluor substituted 23 alkyl of 1 to 6 carbons, OH, SH, alkoxy of 1 to 6 carbons, or alkylthio of 1 to 24 6 carbons;
R3 is hydrogen, lower alkyl of 1 to 6 carbons or F;
26 m is an integer having the value of 0 - 3;
27 o is an integer having the value of 0 - 3;
28 Z is -C=C-, 29 -N=N-, A

1 -N=CR,-, 2 -CR, =N, 3 -(CR,=CR,),,.- where n' is an integer having the value 0 - 5, 4 -CO-NR,-, -CS-NR,-, 6 -NR,-CO, 7 -NR,-CS, 8 -COO-, 9 -OCO-;
-CSO-;
11 -OCS-;
12 Y is a phenyl or naphthyl group, or heteroaryl selected from a group 13 consisting of pyridyl, thienyl, furyl, pyridazinyl, pyrimidinyl, pyrazinyl, 14 thiazolyl, oxazolyl, imidazolyl and pyrrazolyl, said phenyl and heteroaryl groups being optionally substituted with one or two R2 groups, or 16 when Z is -(CR,=CR,),,.- and n' is 3, 4 or 5 then Y represents a direct 17 valence bond between said (CR2=CR2)n. group and B;
18 A is (CH2)q where q is 0-5, lower branched chain alkyl having 3-6 19 carbons, cycloalkyl having 3-6 carbons, alkenyl having 2-6 carbons and 1 or 2o double bonds, alkynyl having 2-6 carbons and 1 or 2 triple bonds;
21 B is hydrogen, COOH or a pharmaceutically acceptable salt thereof, 22 COORB, CONR9R,o, -CH2OH, CH20R11, CHZOCOR,,, CHO, CH(OR12)2, 23 CHOR13O, -COR7, CR7(OR1z)2, CR70R130, or tri-lower alkylsilyl, where R7 24 is an alkyl, cycloalkyl or alkenyl group containing 1 to 5 carbons, R. is an alkyl group of 1 to 10 carbons or trimethylsilylalkyl where the alkyl group has 26 1 to 10 carbons, or a cycloalkyl group of 5 to 10 carbons, or R$ is phenyl or 27 lower alkylphenyl, R9 and R,o independently are hydrogen, an alkyl group of 28 1 to 10 carbons, or a cycloalkyl group of 5-10 carbons, or phenyl or lower 29 alkylphenyl, Rõ is lower alkyl, phenyl or lower alkylphenyl, R12 is lower alkyl, 1 and R,3 is divalent alkyl radical of 2-5 carbons, and 2 Rõ is (R15)r phenyl, (Rls),-naphthyl, or (R15)r heteroaryl where the 3 heteroaryl group has 1 to 3 heteroatoms selected from the group consisting of 4 0, S and N, r is an integer having the values of 0 - 5, and R,s is independently H, F, Cl, Br, I, NO2, N(R8)2, N(R8)COR8,.
6 NR8CON(R8)2, OH, OCORe, OR8, CN, an alkyl group having 1 to 10 7 carbons, fluoro substituted alkyl group having 1 to 10 carbons, an alkenyl 8 group having 1 to 10 carbons and 1 to 3 double bonds, alkynyl group having 1 9 to 10 carbons and 1 to 3 triple bonds, or a trialkylsilyl or trialkylsilyloxy group 1o where the alkyl groups independently have 1 to 6 carbons.
11 Sknthetic Methods - A.r,yl Substituted Com ounds 12 The exemplary RAR antagonist compounds of Formula 1 can be made 13 by the synthetic chemical pathways illustrated here. The synthetic chemist 14 will readily appreciate that the conditions set out here are specific embodiments which can be generalized to any and all of the compounds 1 s represented by Formula 1.

O
5 r (RO AICb / grz ~ (Ra) 7 R, R, R, 8 Fonnula 6 Fomula7 R1 ~ MQBr 10 E120 Pda2(PPii3)2 Et3N / Cul/ 70=C

12 HO ,=
r 13 (Ra3o (F6 14 rmr (F6m 15 Fortnula 15 Fomwla 8 17 PT~sOH L2003 18 j,M.oH
19 Ru 20 r ~
21 ~
R, Foar~ula 16 23 FomwM g 29 Reaction Scheme 1 O
4 Y( PdC~(PPh3)2 (R~o Cul/Et3N/70=C
(Ra) 6 (~)m R~ R (Ri)m 7 Formula 10 i Fomwla9 Fomula 11 9 NaN(SiNe3)2 TNF/ 78'C
Homologs and 11 DerivaGves 12 ONa Y( (R3) 14 (~)m R, Fomwla 12 CI /

18 I~)s 19 g YN OTf Y(Rz) 21 (R3)0R14ZnCl (Ro Owm 22 Pd(PPh3)4 3 R' R' (%)m TNF/SO=C , , 24 Fortrwta 14 Fomwla 13 Homologs and 26 Derivatives 29 Reaction Scheme 1 (continued) 1 Reaction Scheme 1 illustrates the synthesis of compounds of Formula 1 2 where the Z group is an ethynyl function (-C=C-) and X is [C(R,)2]. where n 3 is 1. In other words, Reaction Scheme 1 illustrates the synthesis of ethynyl 4 substituted dihydronaphthalene derivatives of the present invention. In accordance with this scheme, a tetrahydronaphtalene-1-one compound of 6 Formula 6 is brominated to provide the bromo derivative of Formula 7. The 7 compounds of Formula 6 already carry the desired R,, R2 and R3 8 substituents, as these are defined above in connection with Formula 1. A
9 preferred example of a compound of Formula 6 is 3,4-dihydro-4,4-1o dimethyl-1(2H)- naphthalenone, which is described in the chemical literature ii (Arnold et al. J. Am. Chem. Soc. 69: 2322 - 2325 (1947)). A presently 12 preferred route for the synthesis of this compound from 1-bromo-3-13 phenylpropane is also described in the experimental section of the present 14 application.
The compounds of Formula 7 are then reacted with 16 (trimethylsilyl)acetylene to provide the (trimethylsilyl)ethynyl-substituted 3,4-17 dihydro- naphthalen-1(2H)-one compounds of Formula 8. The reaction with 18 (trimethylsilyl)acetylene is typically conducted under heat (approximately 1 s 100 C) in the presence of cuprous iodide, a suitable catalyst, typically having 2o the formula Pd(PPh3)2C12, an acid acceptor (such as triethylamine) under an 21 inert gas (argon) atmosphere. Typical reaction time is approxilnately 24 22 hours. The (trimethylsilyl)ethynyl- substituted 3,4-dihydro-naphthalen-1(2H)-23 one compounds of Formula 8 are then reacted with base (potassium 24 hydroxide or potassium carbonate) in an alcoholic solvent, such as methanol, to provide the ethynyl substituted 3,4-dihydro-l-naphthalen-1(2H)ones of 26 Formula 9. Compounds of Formula 9 are then coupled with the aromatic or 27 heteroaromatic reagent X,-Y(RZ)-A-B' (Formula 10) in the presence of 28 cuprous iodide, a suitable catalyst, typically Pd(PPh3)ZC12, an acid acceptor, 29 such as triethylamine, under inert gas (argon) atmosphere. Alternatively, a i zinc salt (or other suitable metal salt) of the compounds of Formula 9 can be 2 coupled with the* reagents of Formula 10 in the presence of Pd(PPh3)4 or 3~ similar complex. Typically, the coupling reaction with the reagent X,-Y(Rz)-4 A-B' (Formula 10) is conducted at room or moderately elevated temperature.
Generally speaking, coupling between an ethynylaryl derivative :or its zinc salt 6 and a halogen substituted aryl or heteroaryl compound, such as the reagent 7 of Formula 10, is described in United States Patent No. 5,264,450.
8 The 9 compounds of Formula 11 are precursors to exemplary compounds of the io present invention, or derivatives thereof protected in the B' group, from 1 t which the protecting group can be readily removed by reactions well known 12 -in the art. The com'pounds of Formula 11 can also be converted into further 13 precursors to the exemplary compounds by such reactions and 14 transformations which are well known in the art. Such reactions are i s indicated in Reaction Scheme 1 by conversion into "homologs and 16 derivatives". One such conversion employed for the synthesis of several 17 exemplai'y compounds- is saponification of an ester group (when B or B' is an ia ester): to provide the free carboxylic acid or its salt.
ts The halogen substituted aryl or heteroaryl compounds of Formula 10 20 can, generally speaking, be obtained by reactions well known in the art. An 21 example of such compound-is ethyl 4-iodobenzoate which is obtainable, for 22 example, by esterifieation of 4-iodobenzoic.acid. Another example is ethyl 23 iodonicotinoate which' can be'obtained by conducting a halogen exchange 24reaction on 6-chloronicotinic acid, followed by esterification. Even more 25 generally speaking, regarding derivatization of compounds of Formula 11 26 and/or the synthesis of aryl and heteroaryl compounds of Formula 10 which 21 can thereafter be reacted with compounds of Formula 9, the following well 2a known and published general principles and synthetic methodology can be 29 employed.

1 Carboxylic acids are typically esterified by refluxing the acid in a 2 solution of the appropriate alcohol in the presence of an acid catalyst such as 3 hydrogen chloride or thionyl chloride. Alternatively, the carboxylic acid can 4 be condensed with the appropriate alcohol in the presence of dicyclohexylcarbodiimide and dimethylaminopyridine. The ester is recovered 6 and purified by conventional means. Acetals and ketals are readily made by 7 the method described in March, Advanced Organic Chemistry, 2nd Edition, 8 McGraw-Hill Book Company, p. 810). Alcohols, aldehydes and ketones all 9 may be protected by forming respectively, ethers and esters, acetals or ketals 1o by known methods such as those described in McOmie, Plenum Publishing 11 Press, 1973 and Protecting Groups, Ed. Greene, John Wiley & Sons, 1981.
12 To increase the value of n in the compounds of Formula 10 before 13 affecting the coupling reaction of Reaction Scheme 1(where such compounds 14 corresponding to Formula 10 are not available from a commercial source) aromatic or heteroaromatic carboxylic acids are subjected to homologation by 16 successive treatment under Arndt-Eistert conditions or other homologation 17 procedures. Alternatively, derivatives which are not carboxylic acids may also 18 be homologated by appropriate procedures. The homologated acids can then 19 be esterified by the general procedure outlined in the preceding paragraph.
Compounds of Formula 10, (or other intermediates or exemplary 21 compounds) where A is an alkenyl group having one or more double bonds 22 can be made for example, by synthetic schemes well known to the practicing 23 organic chemist; for example by Wittig and like reactions, or by introduction 24 of a double bond by elimination of halogen from an alpha-halo-arylalkyl-carboxylic acid, ester or like carboxaldehyde. Compounds of Formula 10 (or 26 other intermediates or exemplary compounds) where the A group has a triple 27 (acetylenic) bond can be made by reaction of a corresponding aromatic 28 methyl ketone with strong base, such as lithium diisopropylamide, reaction 29 with diethyl chlorophosphate and subsequent addition of lithium 1 diisopropylamide.
2 The acids and salts derived from compounds of Formula 11 (or other 3 intermediates or exemplary compounds) are readily obtainable from the 4 corresponding esters. Basic saponification with an alkali metal base will 5 provide the acid. For example, an ester of Formula 11 (or other 6 intermediates or exemplary compounds) may be dissolved in a polar solvent 7 such as an alkanol, preferably under an inert atmosphere at room 8 temperature, with about a three molar excess of base, for example, lithium 9 hydroxide or potassium hydroxide. The solution is stirred for an extended 1 o period of time, between 15 and 20 hours, cooled, acidified and the 11 hydrolysate recovered by conventional means.
12 The amide may be formed by any appropriate amidation means known 13 in the art from the corresponding esters or carboxylic acids. One way to 14 prepare such compounds is to convert an acid to an acid chloride and then 15 treat that compound with ammonium hydroxide or an appropriate amine.
16 Alcohols are made by converting the corresponding acids to the acid 17 chloride with thionyl chloride or other means (J. March, Advanced Organic 18 Chemistrv, 2nd Edition, McGraw-Hill Book Company), then reducing the 1s acid chloride with sodium borohydride (March, Ibid, p. 1124), which gives the 2o corresponding alcohols. Alternatively, esters may be reduced with lithium 21 aluminum hydride at reduced temperatures. Alkylating these alcohols with 22 appropriate alky halides under Williamson reaction conditions (March, Ibid, 23 p. 357) gives the corresponding ethers. These alcohols can be converted to 24 esters by reacting them with appropriate acids in the presence of acid 25 catalysts or dicyclohexylcarbodiimide and dimethylaminopyridine.
26 Aldehydes can be prepared from the corresponding primary alcohols 27 using mild oxidizing agents such as pyridinium dichromate in methylene 28 chloride (Corey, E. J., Schmidt, G., Tet. Lett. 399, 1979), or dimethyl 29 sulfoxide/oxalyl chloride in methylene chloride (Omura, K., Swern, D., 1 Tetrahedron 34: 1651 (1978)).
2 Ketones can be prepared from an appropriate aldehyde by treating the 3 aldehyde with an alkyl Grignard reagent or similar reagent followed by 4 oxidation.
Acetals or ketals can be prepared from the corresponding aldehyde or 6 ketone by the method described in March, Ibid, p. 810.
7 Compounds of Formula 10 (or other intermediates, or exemplary 8 compounds) where B is H can be prepared from the corresponding 9 halogenated aromatic or hetero aromatic compounds, preferably where the lo halogen is I.
11 Referring back again to Reaction Scheme 1, the compounds of 12 Formula 11 are reacted with sodium bis(trimethylsilyl)amide and 2-(N,N-13 bis(trifluoromethylsulfonyl)amino)-5-chloropyridine in an inert ether type 14 solvent, such as tetrahydrofuran, at low temperatures (-78 C and 0 C). This is shown in Reaction Scheme 1 where the usually unisolated sodium salt 16 intermediate is shown in brackets as Formula 12. The reaction results in the 17 trifluoromethylsulfonyloxy derivatives represented in Formula 13. (Tf =
18 SOZCF3). The compounds of Formula 13 are then converted to the ia exemplary compounds of the invention, shown in Formula 14, by reaction with an organometal derivative derived from the aryl or heteroaryl compound 21 R14H, such that the formula of the organometal derivative is R14Met (Met 22 stands for monovalent metal), preferably R44Li. (R,4 is defined as in 23 connection with Formula 1.) The reaction with the organometal derivative, 24 preferably lithium derivative of the formula R14Li is usually conducted in an inert ether type solvent (such as tetrabydrofuran) in the presence of zinc 26 chloride (ZnC12) and tetrakis(triphenylphosphine)-palladium(0) (Pd(PPh3)4).
27 The organolithium reagent R14Li, if not commercially available, can be 28 prepared from the compound R14H (or its halogen derivative R14 X, where 29 X, is halogen) in an ether type solvent in accordance with known practice in 1 the art. The temperature range for the reaction between the reagent R14Li 2 and the compounds of Formula 13 is, generally speaking in the range of 3 approximately -78 C to 50 C. The compounds of Formula 14 can be 4 converted into further homologs and derivatives in accordance with the reactions discussed above.
6 The intermediate 7-bromo-tetrahydronaphthalene-l- one compounds of 7 Formula 7 shown in Reaction Scheme 1 can also be converted with a 8 Grignard reagent of the formula R14MgBr (R,a is defined as in connection 9 with Formula 1) to yield the tertiary alcohol of Formula 15. The tertiary lo alcohol is dehydrated by treatment with acid to provide the 3,4-dihydro-7-11 bromonaphthalene derivatives of Formula 16, which serve as intermediates 12 for the synthesis of additional compounds of the present invention (see 13 Reaction Schemes 6, 7, and 8).

2 . / r HO=C~ r ( 8rC(R3)2C(Fl3)'2~2H ~(R,)2 /
3 HX ~ ~)m Foenuia 18 (R~:~X
4 ga" %hn Fomvula 17 FomwV 19 H+/e Meo 0 / H ~r,=Me3 r ~o ~ (Ra1 %)m Pd02(PPh3)2 11 Formuta 21 Cu I/ E13 N/ 70= C (~)m Fomw1a20 13 MeOH

14 ~B
Y~A
Pdp2(PPh3)2 16 (Ra) Cul/Et3N/70=C (R3) 17 (%~ X, Y(Fis)=A--B' (~kn Fomula22 Fomwla 10 Fortaula 23 19 Homologs and 1. NaN(SSMe3~
Qorivadves THF / 78' C

2= c`ro wm .IB
22 R14 Y(<j , Tf Y(R=) R 14ZnCl 23 Pd(PPh3)4 / /
~ ~ tR,) Foffnub25 \- Foemula24 26 iHomolops and arivatlv.s 29 Reaction Scheme 2 WO 99/33821 PCTlUS98/27314 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, 0 or NR' 3 and the Z group is an ethynyl function (-C-C-). Starting material for this 4 sequence of the reaction is a bromophenol, bromothiophenol or bromoaniline of the structure shown in Formula 17. For the sake of simplifying the 6 present specification, in the ensuing description X can be considered 7 primarily sulfur as for the preparation of benzothiopyran derivatives. It 8 should be kept in mind, however, that the herein described scheme is also 9 suitable, with such modifications which will be readily apparent to those 1 o skilled in the art, for the preparation of benzopyran (X = 0) and 11 dihydroquinoline (X = NR') compounds of the present invention. Thus, the 12 compound of Formula 17, preferably para bromothiophenol, para 13 bromophenol or para bromoaniline is reacted under basic condition with a 3-14 bromo carboxylic acid of the Formula 18. In this reaction scheme the symbols have the meaning described in connection with Formula 1. An le example for the reagent of Formula 18 where R3 is hydrogen, is 3-17 bromopropionic acid. The reaction with the 3-bromocarboxylic acid of 18 Formula 18 results in the compound of Formula 19. The latter is cyclized by 19 treatment with acid to yield the 6-bromothiochroman-4-one derivative (when X is S) or 6- bromochroman derivative (when X is 0) of Formula 20. The 21 bromo compounds of Formula 20 are then subjected to substantially the same 22 sequence of reactions under analogous conditions, which are described in 23 Reaction Scheme 1 for the conversion of the bromo compounds of Formula 24 7 to the compounds of the invention. Thus, briefly summarized here, the 2s bromo compounds of Formula 20 are reacted with (trimethylsilyl)acetylene to 26 provide the 6- (trimethylsilyl)ethynyl- substituted thiochroman-4-one or 27 chroman-4-one compounds of Formula 21. The 6- (trimethylsilyl)ethynyl-28 substituted thiochroman-4-one compounds of Formula 21 are then reacted 29 with base (potassium hydroxide or potassium carbonate) to provide the 1 ethynyl substituted 6-ethynyl substituted thiochroman-4-ones of Formula 22.
2 Compounds of Formula 22 are then coupled with the aromatic or 3 heteroaromatic reagent X,-Y(R2)-A-B' (Formula 10) under conditions 4 analogous to those described for the analogous reactions of Reaction Scheme 5 1, to yield the compounds of Formula 23.
6 The compounds of Formula 23 are then reacted still under conditions 7 analogous to the similar reactions described in Reaction Scheme 1 with 8 sodium bis(trimethylsilyl)amide and 2-[N,N-9 bis(trifluoromethylsulfonyl)amino]-5-chloropyridine to yield the 4-1 o trifluoromethylsulfonyloxy benzothiopyran or benzopyran derivatives 11 represented in Formula 24. The compounds of Formula 24 are then 12 converted to compounds shown in Formula 25, by reaction with an 13 organometal derivative derived from the aryl or heteroaryl compound R14H, 14 as described in connection with Reaction Scheme 1.
15 Similarly to the use of the intermediate 7-bromo-16 tetrahydronaphthalene-l-one compounds of Formula 7 of Reaction Scheme 17 1, the intermediate 6-bromothiochroman- 4-one compounds of Formula 20 18 can also be used for the preparation of further compounds within the scope 19 of the present invention, as described below, in Reaction Schemes 6, 7 and 8.
2o The compounds of Formula 25, can also be converted into further homologs 21 and derivatives, in reactions analogous to those described in connection with 22 Reaction Scheme 1.

r H-EMMe3 6 (Rs)o ~ PdpZ(PPh3y2 (R,)o Cul/ E18N/70=C (Rz)m 7 Fomtila26 Fomula27 MeOH

O Y(%) 12 Pdp2(PPh3)2 O
bo Cul/E1SN/70=C
13 (Ra)c (~)m )' Y~)-A~' (F6)o 14 Fomwla29 Fomiuta 10 (R2)M
Fomula28 J 1. NaN(SiMe3~
16 THF /78=C
17 2' c'`~
18 %
19 .18 OT( Y(r4) Ru Yc~ 4~
R1~CI
21 Pd(PPh3)4 b --- \
22 THF / 50' C o (Rehn 23 Fomwla 30 Fomula 31 Homolops and Ded,ratiws 26 Reaction Bcheme 3 29 Reaction Scheme 3 1 Reaction Scheme 3 discloses a synthetic route to compounds where, 2 with reference to Formula 1, X is [C(R,)2],,, n is 0 and the Z group is an 3 ethynyl function (-C=_C-). In accordance with this scheme, a 6- bromo-2,3-4 dihydro-1H-inden-1-one derivative of Formula 26 is reacted in a sequence of reactions (starting with reaction with trimethylsilylacetylene) which are s analogous to the reactions described above in connection with Reaction 7 Schemes 1 and 2, to provide, through intermediates of the formulas 27 - 30, 8 the indene derivatives of Formula 31. In a preferred embodiment within the 9 scope of Reaction Scheme 3, the starting material is 6-bromo-2,3-dihydro-3,3-io dimethyl-lH-inden-l-one that is available in accordance with the chemical 11 literature (See Smith et al. Org. Prep. Proced. Int. 1978 10, 123-131).
12 Compounds of Formula 26, such as 6-bromo-2,3-dihydro-3,3-dimethyl-l.H-13 inden-l-one, can also be used for the synthesis of still further exemplary 14 compounds for use in the present invention, as described below.

2 t. NC13 / R100C{ R, + other isomer 3 2. CrO3 (F66 HOAc / Ac20 ~ R~ (~~x)m Fo mula 32 Fomuta 33 6 HOCH2CH2ON ~
pTsOH

8r (R~ R, R14Mg 9 R0.'(%), 11 m E120 R R (~kn R, , , 12 Fomw{a 35 Formula34 14 pTsOH

16 ~ = t~~o~~ yCN
N. 0 ql~ R,. R, Forrnuta 37 CHO
/ / R
18 ~ \ ~ R~ NaH/THF ` ~
R ( 2 D18AL-H N (Rt)m 19 . Rskn , Fomwta36 Formula38 22 (Eqspj".
Fomwla 38 23 !DA/THF

R'd R R' Homologs and E~ i / , \ \
26 Derfvatives N

28 Fomti440 29 Reaction Scheme 4 1 Referring now to Reaction Scheme 4 a synthetic route to exemplary 2 compounds is disclosed where, with reference to Formula 1, Z is -s(CR,=CR,),,: , n' is 3, 4 or 5 and Y represents a direct valence bond between 4 the (CR,=CR,)n. group and B. This synthetic route is described for examples where the X group is [C(R,)2]õ and n is 1 (dihydronaphthalene derivatives).
6 Nevertheless, it should be understood that the reactions and synthetic 7 methodology described in Reaction Scheme 4 and further ensuing schemes, is 8 also applicable, with such modifications which will be readily apparent to 9 those skilled in the art, to derivatives where X is is S, 0, NR' to (benzothiopyran, benzopyran or dihydroquinoline derivatives) or [C(Rl)2]n 11 and n is 0 (indene derivatives).
12 In accordance with Reaction Scheme 4, a 1,2,3,4-13 tetrahydronaphthalene derivative of Formula 32 is reacted with an acid 14 chloride (R,COCI) under Friedel Crafts conditions, and the resulting acetylated product is oxidized, for example in a Jones oxidation reaction, to 16 yield a mixture of isomeric 6- and 7- acetyl-1(2H)- naphthalenone derivatives 17 of Formula 33. In a specific preferred example of this reaction, the starting 18 compound of Formula 32 is 1,2,3,4-tetrahydro-1,1- dimethylnaphthalene (a 19 known compound) which can be prepared in accordance with a process described in the experimental section of the present application. The 7-21 acetyl-1(2H)-naphthalenone derivative of Formula 33 is reacted with ethylene 22 glycol in the presence of acid to protect the oxo function of the exocyclic 23 ketone moiety, as a ketal derivative of Formula 34. The ketal of Formula 34 24 is thereafter reacted with a Grignard reagent of the formula R14MgBr (the symbols are defined as in connection with Formula 1), to yield the tertiary 26 alcohol of Formula 35. Thereafter the dioxolane protective group is removed 27 and the tertiary alcohol is dehydrated by treatment with acid to provide the 28 3,4- dihydro-7-acetylnaphthalene derivatives of Formula 36. The ketone 29 function of the compounds of Formula 36 is subjeceted to a Homer Emmons 1(or analogous) reaction under strongly alkaline conditions with a phosphonate 2 reagent of Formula 37, to yield, after reduction, the aldehyde compounds of 3 Formula 38. Still another Horner Emmons (or analogous) reaction under 4 strongly alkaline conditions with a reagent of Formula 39 provides 5 compounds of Formula 40. The latter can be converted into further 6 homologs and derivatives in accordance with the reactions described above.
7 A specific example of the Horner Emmons reagent of Formula 37 which is 8 used for the preparation of a preferred compound is 9 diethylcyanomethylphosphonate; an example of the Horner Emmons reagent 1o of Formula 39 is diethyl-(E)-3- ethoxycarbonyl-2-methylallylphosphonate.

12 p ~
N^ H2SO4 14 Fomuk6 Fomtuh 41 15 IH2IFd EtOAe o 17 ~
HOAC
18 0N--YPY-A-e Fom,ul. sa (~kn ~
19 R+ R+
20 FonmL sa Fonnut. 42 21 1. wN(Siw1.3?2 tNF1=rr c 2.c 24 7f A A
~ a14ZnCl ~
25 Pd(PPI-1)4 b 26 A, htFisac 27 Fomul*45 Foenih48 28 Hmdogs wid Drm-aUws 29 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 Reaction 3 Scheme 4 this process is described for examples where the X group is 4[C(R,)2]õ and n is 1(dihydronaphthalene derivatives). Nevertheless, it should be understood that the synthetic methodology described is also applicable, 6 with such modifications which will be readily apparent to those skilled in the 7 art, to all azo compounds for use in the invention, namely to derivatives 8 where X is is S, 0, NR' (benzothiopyran, benzopyran or dihydroquinoline 9 derivatives) or [C(R,)Z]n and n is 0 (indene derivatives). Thus, a nitro group 1 o is introduced into the starting compound of Formula 6 under substantially ii standard conditions of nitration, to yield the 3,4-dihydro-7-nitro-1(2H)-12 naphthalenone derivative of Formula 41. The latter compound is reduced to 13 the 3,4-dihydro-7-amino-1(2H)-naphthalenone derivative of Formula 42 and 14 is thereafter reacted with a nitroso compound of the formula ON-Y(R2)-A-B
(Formula 43) under conditions normally employed (glacial acetic acid) for 16 preparing azo compounds. The nitroso compound of Formula 43 can be 17 obtained in accordance with reactions known in the art. A specific example 18 for such compound, which is used for the synthesis of a preferred compound 1s is ethyl 4-nitrosobenzoate. The azo compound of Formula 44 is thereafter reacted with sodium bis(trimethylsilyl)amide and 2-[N,N-21 bis(trifluoromethylsulfonyl)amino]-5-chloropyridine to yield the 4-22 trifluoromethylsulfonyloxy derivatives represented in Formula 45. The 23 compounds of Formula 45 are then converted to the azo compounds shown 24 in Formula 46, by reaction with an organometalic derivative derived from the aryl or heteroaryl compound R14H. These latter two reactions, namely the 26 conversion to the 4-trifluoromethylsulfonyloxy derivatives and subsequent 27 reaction with the organometal derivative, have been described above in 28 connection with Reaction Schemes 1, 2 and 3, and are employed in several 29 presently preferred synthetic processes leading to exemplary RAR antagonist 1 compounds.

R,. R,.
4 r I. t-Bul.i CO=H
K)O, THF a80 C (R3) Rs)m 2. C02 (~)m 6 R, R, 7 Fomwla t 6 Fomwla 47 HX2--Y(Rz}--A--8 9 Foffnula 48 (X2=NRiorO) 12 R,. O A.A
13 Homologs and (R3) Y(6 Dedvagves 14 R R, Nm Fomwla49 18 Reaction Scheme 6 19 Reaction Scheme 6 discloses a presently preferred synthetic process for the preparation of compounds where, with reference to Formula 1, the Z
21 group is COO- or CONR, (R, is preferably H). These ester and amide 22 derivatives are prepared from the 3,4-dihydro-7- bromonaphthalene 23 derivatives of Formula 16, which can be obtained as described in Reaction 24 Scheme 1. Thus, the compounds of Formula 16 are reacted with strong base, such as t-butyllithium, in an inert ether type solvent, such as tetrahydrofuran, 26 at cold temperature, and carbon dioxide (C02) is added to provide the 5,6-27 dihydro-2-naphthalenecarboxylic acid derivatives of Formula 47. Compounds 28 of Formula 47 are then reacted with compounds of the formula X2-Y(R2)-A-29 B (Formula 48) where X2 reperesent an OH or an NR, group, the R, 1 preferably being hydrogen. Those skilled in the art will recognize that the 2 compounds of Formula 48 are aryl or heteroaryl hydroxy or amino derivatives 3 which can be obtained in accordance with the state-of-the- art. The reaction 4 between the compounds of Formula 47 and Formula 48 can be conducted under various known ester or amide forming conditions, such as coupling of 6 the two in the presence of 1-(3-dimethylaminopropyl)-3- ethylcarbodiimide 7 hydrochloride and 4- dimethylaminopyridine. Alternatively, the compounds 8 of Formula 47 can be converted into the corresponding acid chloride for s coupling with the compounds of Formula 48 in the presence of base. The lo amide or ester compounds of Formula 49 can be converted into further 11 homologs and derivatives, as described above. Although Reaction Scheme 6 12 is described and shown for the example where the X group of Formula 1 is 13 [C(R,)2)õ and n is 1(dihydronaphthalene derivatives), the herein described 14 process can be adapted for the preparation of benzopyran, benzothiopyran, dihydroquinoline and indene derivatives as well.
16 Compounds of the present invention where with reference to Formula 17 1, Z is -OCO-, NR,CO, as well as the corresponding thioester and thioamide 18 analogs, can be prepared from the intermediates derived from the 19 compounds of Formula 16 where the bromo function is replaced with an amino or hydroxyl group and in accordance with the teachings of United 21 States Patent Nos. 5,324,744.

23 Ru 1. iAg/THF R14 ~q'S
V'-(R2)- 2. ZnC 12 / /. Y(RZ) 24 3. Ni(O) ! THF tRa~\ i n X,--Y(FY~-A--9 t~)n 26 R' a' Formula 10 Ri N
27 Fomwlal6 Fortnula50 28 Homologs and Derivatives 29 Reaction Scheme 7 1 Reaction Scheme 7 discloses a presently preferred synthetic process for 2 the preparation of compounds where with reference to Formula 1, Z is -3(CR,=CR,),,: and n' is 0. These compounds of Formula 50 can be obtained 4 in a coupling reaction between compounds of Formula 16 and a Grignard reagent derived from the halo compounds of Formula 10. The coupling 6 reaction is typically conducted in the presence of a zinc salt and a nickel 7(Ni(0)) catalyst in inert ether type solvent, such as tetrahydrofuran. The 8 compounds of Formula 50 can be converted into further homologs and 9 derivatives, as described above.

O ~g 12 Pd2(F'PA3)2(OAe)2 O
r Et3N / P(o-MePh)3 y(e) 13 (3FI3) (Ra) Nm CHz=CFi-=Y(R=)-A-=-B \ ( 14 R, Fortnula 51 ~
R, R, Fortnula 7 Formula 52 1. NaN(SiMe3)2 17 THF / 78= C
2. ch~`N

R14 A Tt A
(i~) 21 (~ I Y(~) R1~~ Y
Pd(~
23 q~ R~ 3)4 (~)o /
22 ~
(R~)m THF / SC C R
, Forrrwfa 54 Fomwla53 26 Homolofls and DerivaUves 29 Reaction Scheme 8 1 Referring now to Reaction Scheme 8 a presently preferred synthetic 2 process is disclosed for the preparation of compounds where Z is -s(CR, =CR,),,: and n' is 1. More particularly, Reaction Scheme 8 discloses 4 the presently preferred process for preparing those compounds which are 5 dihydronaphtalene derivatives and where the Z group represents a vinyl (-6 CH=CH-) function. However, the generic methodology disclosed herein can 7 be extended, with such modifications which will be apparent to those skilled 8 in the art, to the analogous benzopyran, benzothiopyran, dihydroquinoline 9 compounds, and to compounds where the vinyl group is substituted. Thus, in 1o accordance with Reaction Scheme 8 the 7-bromo-1(2H)-naphthalenone 11 derivative of Formula 7 is reacted with a vinyl derivative of the structure -12 CH2=CH-Y(R2)-A-B (Formula 51) in the presence of a suitable catalyst, 13 typically having the formula Pd(PPh3), an acid acceptor (such as 14 triethylamine) under an inert gas (argon) atmosphere. The conditions of this 15 reaction are analogous to the coupling of the acetylene derivatives of 16 Formula 9 with the reagent of Formula 10 (see for example Reaction Scheme 17 1), and this type of reaction is generally known in the art as a Heck reaction.
18 The vinyl derivative of Formula 51 can be obtained in accordance with the 19 state of the art, an example for such a reagent used for the synthesis of a 20 preferred compound to be used in the invention is ethyl 4-vinylbenzoate.
21 The product of the Heck coupling reaction is an ethenyl derivative of 22 Formula 52, which is thereafter converted into compounds used in the 23 present invention by treatment with sodium bis(trimethylsilyl)amide and 2-24 [N,N-bis(trifluoromethylsulfonyl)amino]-5-chloropyridine to yield the 4-25 trifluoromethylsulfonyloxy derivatives of Formula 53, and subsequent reaction 26 with an organometal derivative derived from the aryl or heteroaryl compound 27 R14H, as described above. The resulting compounds of Formula 54 can be 28 converted into further homologs and derivatives.
29 The compounds of Formula 54 can also be obtained through synthetic i schemes which employ a Wittig or Horner Emmons reaction. For example, 2 the intermediate of Formula 33 (see Reaction Scheme 4) can be reacted with 3 a triphenylphosphonium bromide (Wittig) reagent or more preferably with a 4 diethylphosphonate (Homer Emmons) reagent of the structure (EtO)2P0-6 CH27-Y(Rz)-A-B, as described for analogous Homer Emmons reactions in 6 United States Patent No. 5,324,840.
7 The just mentioned Homer Emmons reaction provides a intermediate compounds analogous in structure to Formula 52, and can be 9 converted into compounds of Formula 54 by the sequence of reactions lo described in Reaction Scheme 8 for the compounds of Formula 52.

R
4 i ~ C01H \re~ ~ OMe ~ I R3 R' HS +
~ ~ '_'~ Hp2C~
(R?)m R~ R3 105'C (R?)m 6 Formula 55 Formula 56 Formuls 57 7 1. (COCt)2 2. SnC14 bz l rt CH2CI2 8 0'C/2h 9 O~y 0 ~ ~ Olak Q Ti0 i OH BBr3 R
R (R pyr RR (R CH2CI2 RR (Rz 11 -23' C
Formula 60 Formula 59 Formula 58 1_TMSCCH
13 Pd(m I Cu(I) Et3N /90'C
14 2. KZC03 / MeOH
B' 0 0 1F(R21 A' / Pd(II)/Cup) 16 i Et3N / 90'C i R ~I
17 R (R2)m Xry(RZ}A-B, R (R26 18 Formula 61 Fortnula 62 1. NaN(TMS)2 / THF
\~- ArNRn2 B, ~B, Rx (R~ A' OTf A Y(R~
R 14ZnCi 21 i\~ Pd( o)/rHF ~

22 R (R~m R \ (R~m 23 Formula 64 Formuls 63 Homolops and Darivattvas 29 Reaction Scheme 9 1 A number of particularly preferred compounds of the invention, for 2 example those shown in Formula 5b and Table la are prepared in accordance 3 with Reaction Scheme 9. An important feature of the compounds prepared 4 in accordance with Reaction Scheme 9 is that the group X with reference to Formula 1 is S and the 2 position of the benzothiopyrane (thiochromene) 6 ring of the compounds is substituted with alkyl groups.
7 Referring now to Reaction Scheme 9, a 4-methoxythiophenol of 8 Formula 55 serves as a starting material. The 4-methoxythiophenol of 9 Formula 55 may be substitituted with one or more R2 groups as defined in 1 o connection with Formula 1. In the synthesis of the preferred compounds in 11 accordance Reaction Scheme 9 the R2 group of Formula 55 is either H, 12 halogen such as F, or an alkoxy group, preferably metboxy. Examples for 13 compounds of Formula 55 which are used for the synthesis of specific 14 preferred compounds in accordance with the invention are 4-methoxythiophenol and 3-fluoro-4-methoxythiophenol. The thiophenol of 16 Formula 55 is reacted with an alkenoic acid of Formula 56 by heating in 17 piperidine. This reaction results in addition of the thiophenol to the double 18 bond of the alkenoic acid (Michael addition) to give rise to compounds of 19 Formula 57. The alkenoic acid of Formula 56 includes the R3 substituents which for the herein described preferred embodiments are alkyl, most 21 preferably methyl, groups. An example for the alkenoic acid of Formula 56 22 is 3-methyl-2-butenoic acid. Reaction of thiophenols of Formula 55 with 2-23 alkenoic acids to synthesize 2,2-dimethyl-thiochroman-4-one derivatives is 24 generally described by Ferreira et al. Synthesis 1987 p 149.
The aromatic acid of Formula 57 is converted to the corresponding 26 acid chloride, for example by reaction with oxalyl chloride, and the resulting 27 acid chloride is thereafter cyclized by heating in an inert solvent such as 28 CH2C12 in the presence of stannic tetrachloride (SnC14), to give rise to 2,2-29 disubstituted thiochroman-4-one compounds of Formula 58. The 2,2-1 disubstituted thiochroman-4-one of Formula 58 is reacted with 2 borontribromide to remove the methyl group from the methoxy function and 3 to yield the 2,2-disubstituted 6-hydroxy-thiochroman-4-one of Formula 59.
4 The hydroxy compound of Formula 59 is thereafter reacted with trifluoromethanesulfonic anhydride in anhydrous pyridine to give rise to the 6 6- trifluoromethanesulfonyloxy derivative of Formula 60.
7 The 2,2-disubstituted 6-trifluoromethanesulfonyloxy-thiochroman-4-one 8 of Formula 60, shown in Reaction Scheme 9, is reacted with s(trimethylsilyl)acetylene to provide 2,2-disubstituted 6-(trimethylsilyl)ethynyl-to thiochroman-4-one compounds which are subsequently reacted with base 11 (KZC03) in an alcoholic solvent, such as methanol, to yield the 2,2-12 disubstituted 6-ethynyl-thiochroman-4-one compounds of Formula 61. The 13 reaction with (trimethylsilyl)acetylene is typically conducted under heat 14 (approximately 95 C) in the presence of a suitable catalyst, typically having the formula Pd(PPh3)2C12, and an acid acceptor (such as triethylamine) under 16 an inert gas (argon) atmosphere. This reaction is analogous to the reactions 17 of the 6-bromo-thiochroman-4-one compounds of Formula 20 with 18 (trimethylsilyl)acetylene, shown in Reaction Scheme 2. Thereafter, the 2,2-1s disubstituted 6-ethynyl-thiochroman-4-one compound of Formula 61 is coupled with the aromatic or heteroaromatic reagent X,-Y(R2)-A-B' 21 (Formula 10, defined above) under conditions similar to those described for 22 the analogous reactions of Reaction Scheme 1 and Reaction Scheme 2, to 23 yield the compounds of Formula 62. In the synthesis of the herein described 24 specific preferred examples of the compounds of Formula 62 ethyl 4-iodobenzoate, ethyl 2-fluoro-4-iodobenzoate and ethyl 6-iodonicotinate serve 26 as examples for the reagent X,-Y(R2)-A-B' (Formula 10).
27 The compounds of Formula 62 are then reacted, still under conditions 28 similar to the analogous reactions described in Reaction Scheme 1 and 29 Reaction Scheme 2, with sodium bis(trimethylsilyl)amide and 2-[NN-WO 99/33821 PCT/US98l27314 1 bis(trifluoromethylsulfonyl)amino]-5-chloropyridine to yield the 4-2 trifluoromethylsulfonyloxy benzothiopyran (thiochromene) derivatives 3 represented by Formula 63. The compounds of Formula 63 are then 4 converted to compounds shown in Formula 64, by reaction with an 5 organometal derivative derived from the aryl or heteroaryl compound R,4H, 6 R14Br or R141 as described in connection with Reaction Scheme 1 and 7 Reaction Scheme 2. Examples of reagents and conditions utilized in 8 accordance with Reaction Scheme 9 to convert the 4-9 trifluoromethanesulfonyloxy compounds of Formula 63 into preferred 1o compounds of the invention in accordance with Formula 64 are:
11 4-methylbromobenzene, tetrahydrofuran (THF), t-butyllithium and zinc 12 chloride (ZnC12) to make the organometal reagent, followed by reaction with 13 the 4-trifluoromethylsulfonyloxy (triflate) derivative in the presence of 14 tetrakis(triphenylphosphine)palladium(O) catalyst;
15 2-methylthiophene, THF, n-butyllithium and ZnCIZ followed by the 16 reaction with the triflate in the presence of 17 tetrakis(triphenylphosphine)palladium(0);
18 bromobenzene, THF, t-butyllithium and ZnCIZ followed by the reaction 1s with the triflate in the presence of tetrakis(triphenylphosphine)palladium(0);
20 4-ethylbromobenzene, THF, t-butyllithium and ZnC12 followed by the 21 reaction with the triflate in the presence of 22 tetrakis(triphenylphosphine)palladium(O);
23 4-iso-propylbromobenzene, THF, t-butyllithium and ZnC12 followed by 24 the reaction with the triflate in the presence of 25 tetrakis(triphenylphosphine)palladium(0);
26 4-t-butylbromobenzene, THF, t-butyllithium and ZnC12 followed by the 27 reaction with the triflate in the presence of 28 tetrakis(triphenylphosphine)palladium(0);
29 2-ethylthiophene, THF, n-butyllithium and ZnC12 followed by the 1 reaction with the triflate in the presence of 2 tetrakis(triphenylphosphine)palladium(O);
3 2-t-butylthiophene, THF, n-butyllithium and ZnC12 followed by the 4 reaction with the triflate in the presence of tetrakis(triphenylphosphine)palladium(0), and 6 4-bromo-2-methylpyridine, THF, t-butyllithium and ZnC12 followed by 7 the reaction with the triflate in the presence of 8 tetrakis(triphenylphosphine)palladium(O).
9 The 4-aryl or 4-heteroaryl benzothiopyran (thiochromene) derivatives lo represented by Formula 64 are preferred compounds within the scope of the 11 invention. Most preferably the R3 group is methyl in these compounds. The 12 compounds of Formula 64 can be converted to further homologs and 13 derivatives as described above in connection with the preceding reaction 14 schemes. A frequently utilized reaction in this regard, which gives rise to a number of specific examplary compounds of the invention is saponification of 1s an alkyl ester (B or B' represents an esterified carboxylic acid group) to yield 17 a free carboxylic acid or its pharmaceutically acceptable salt.

i Br E AcCi p i & ~ q(Rzl.
7 H ~ I CiC~ A~ 155 `C -H (R~(Fries 8 rearrarpemsrrt) Formula 65 Fomnula 66 Formula 67 ^

O ~ J
~
RAaa toUOWed by a ~ ~N Mid~eal addqion) bz 190 'C
12 q-B.
O (R~ p H 1.TMSCCH p 13 =~ F'd(II) / Cu(n Pd(In / Cum Bt iI Et3N / 90'C EbN / 70 'C
=t- ~
14 R - R 2. K2CO3 R
R (R?)T XrY(RZ)-/1-B' R (R~ MsOH R (R$,n Formula 70 Foffnuy 69 Formuls 68 16 I 1. NaN(TMS)2 THF / -78'C
17 2. 18 ~8' 8' OTi (R~ q ~~~~ Rw ~~ q~
THF / 30 "C
21 ~ ~ ----~ ~
R R14ZnC4 R
22 R (R2)m R (Ra)in 23 Fomwla 71 Formula 72 24 Homolops and Darivatlvss 29 Reaction Scheme 10 1 A number of particularly preferred compounds of the invention, shown 2 in Formula 5b and Table la, where with reference to Formula 1 X is 0 and 3 the 2-position of the benzopyrane (chromene) ring of the compounds is 4 substituted with alkyl groups, are prepared in accordance with Reaction Scheme 10. A 4-bromophenol compound of Formula 65 serves as a starting 6 material and R2 is defined as in connection with Formula 1, although 7 preferably the 4 bromophenol is unsubstituted, or is alkyl, halogen or alkoxy a substituted (the R2 group symbolizes alkyl, halogen or alkoxy). The 4-9 bromophenol of Formula 65 is acetylated to provide the acetyloxy-4-1o bromobenzene derivative of Formula 66. The acetyloxy-4-bromobenzene 11 derivative of Formula 66 is then subjected to a rearrangement reaction (Fries 12 rearrangement) by heating with aluminum chloride (A1C13) to provide the 3-13 bromo-6-hydroxyacetophenone derivatives of Formula 67. The latter 14 compounds are heated in piperidine in the presence of piperidine trifluoroacetate with a ketone of the formula R3-CO-R3, where the R3 group 1 e is lower alkyl, causing the 3-bromo-6-hydroxyacetophenone derivatives of 17 Formula 67 to undergo a Knoevenagel condensation followed by a Michael 18 addition, and thereby ring close and provide the 2,2-dialkyl-6-bromo-19 chroman-4-one derivatives of Formula 68.
The 2,2-dialkyl-6-bromo-chroman-4-one derivatives of Formula 68 are 21 then reacted with (trimethylsilyl)acetylene under conditions similar to those 22 described for the reaction with (trimethylsilyl) acetylene of the bromo 23 compounds of Formula 15 and 20 in Reaction Schemes 1 and 2, respectively, 24 and for the 6-trifluorosulfonyloxythiochroman-4-one compounds of Formula 60 in Reaction Scheme 9. The resulting 2,2-disubstituted 6-28 (trimethylsilyl)ethynyl-chroman-4-one compounds (not shown in Reaction 27 Scheme 10) are then treated with base (as in the previously described 28 analogous reactions). to provide 2,2-disubstituted 6-ethynyl-chroman-4-one 29 compounds of Formula 69. The compounds of Formula 69 are then coupled 1 with the aromatic or heteroaromatic reagent X,-Y(R2)-A-B' (Formula 10, 2 defined above) under conditions similar to those described for the analogous 3 reactions of Reaction Scheme 1, Reaction Scheme 2 and Reaction Scheme 9 4 to yield the compounds of Formula 70. Examples for the reagent X,-Y(R2)-A-B' (Formula 10) used in the synthesis of the herein described specific 6 preferred embodiments of the compounds of Formula 70 are ethyl 4-7 iodobenzoate and ethyl 2-fluoro-4-iodobenzoate.
8 Referring still to Reaction Scheme 10, the 6-(aryl) or s 6(heteroaryl)ethynyl 2,2-disubstituted chroman-4-one compounds of Formula 1o 70 are converted into the 6-(aryl) or 6-(heteroaryl)ethynyl 2,2-disubstituted 4-11 trifluoromethylsulfonyloxy benzopyran (chromene) derivatives of Formula 71 12 by treatment with sodium bis(trimethylsilyl)amide and 2-[N,N-13 bis(trifluoromethylsulfonyl)amino]-5-chloropyridine. This is in analogy to the 14 conversion of the corresponding thiochroman-4-one compounds (Formula 62) 1 s into the corresponding 4-trifluoromethylsulfonyloxy benzothiopyran 16 (thiochromene) derivatives of Formula 63. The 6-(aryl) or 6-17 (heteroaryl)ethynyl 2,2-disubstituted 4-trifluoromethylsulfonyloxy benzopyran 18 (chromene) derivatives of Formula 71 are then reacted with an organometal is derivative derived from the aryl or heteroaryl compound R1,,H, R14Br, or 2o as described above in connection with Reaction Scheme 1, Reaction Scheme 21 2 and Reaction Scheme 9, to yield preferred compounds of the invention 22 shown in Formula 72. Examples of reagents and conditions utilized in 23 accordance with Reaction Scheme 10 to to convert the 4-24 trifluoromethanesulfonyloxy compounds of Formula 71 into preferred 25 compounds of the invention in accordance with Formula 72 are:
26 bromobenzene, THF, t-butyllithium and ZnC12 followed by the reaction 27 with the triflate in the presence of tetrakis(triphenylphosphine)palladium(0);
28 4-methylbromobenzene, THF, t-butyllithium and ZnC12 followed by the 29 reaction with the triflate in the presence of 1 tetrakis(triphenylphosphine)palladium(O);
2 4-ethylbromobenzene, THF, t-butyllithium and ZnC12 followed by the 3 reaction with the triflate in the presence of 4 tetrakis(triphenylphosphine)palladium(0);
5 4-iso-propylbromobenzene, THF, t-butyllithium and ZnC12 followed by 6 the reaction with the triflate in the presence of 7 tetrakis(triphenylphosphine)palladium(O), and 8 4-t-butylbromobenzene, THF, t-butyllithium and ZnC12 followed by the 9 reaction with the triflate in the presence of 1o tetrakis(triphenylphosphine)palladium(0).
11 The preferred 2,2-disubstituted benzopyran (chromene) compounds of 12 the invention, represented by Formula 72, can be converted to further 13 homologs and derivatives, as described above. Saponification of the alkyl 14 esters represented by Formula 72 when B' represents an esterified carboxyl 1 s group, yields the specially preferred carboxylic acid (or pharamaceutically 16 acceptable salt) compounds of the invention.
17 Synthetic Methods - Aryl and S3-Oxy4-Propenvll-Substitutgd Com ounds 18 The exemplary RAR antagonist compounds of Formula 101 can be 19 made by the synthetic chemical pathways illustrated here. The synthetic 20 chemist will readily appreciate that the conditions set out here are specific 21 embodiments which can be generalized to any and all of the compounds 22 represented by Formula 101.

~)m 1. Alp3 0 4 ~ RtsC't2~
R,$
~ (Rs)o isomeeic.
2 C103 diketone R, R, HOAdAr20 R, 0 p=TsOH

11 R14 OH (R*)m O (Roo)m 12 Rte (FR14MpBr R
s~o I ~E----- (Rjo "
13 THF or E120 O
R, R, O~ ! R, R, O ` O
14 ~J V, 16 1. i) NaN(SiMe3)2/THFh78'C

N (TT~
18 H* 2.11 X/a-u~
1) 2~G=~
19 i) Pd(0) / THF / 5o'C

R,d 21 (%)^ R,~ Y(R=)=A-8 Rõ
(~)m A
R+=
22 (Rs)o FORMULA 108 (POo R17 Y(R=) 23 R, R, NaOH I EtOH
O R,e R, R1 0 Homologs and 26 Dotivatlvos, 29 Reaction Scheme 101 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 Rõ 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 compound of Formula 6 103 which is appropriately substituted with the R3 and R2 groups (as these 7 are defined in connection with Formula 101) serves as the starting material.
e A preferred example of a compound of Formula 103 is 1,3,3,4-9 tetrahydro-1,1-dimethyl-naphthalene, which is described in the chemical 1 o literature (Mathur et al. Tetrahedron, 1985, 41:1509. A presently preferred 11 route for the synthesis of this compound from 1-bromo-3-phenylpropane is 12 also described in the experimental section of the present application.
13 The compound of Formula 103 is reacted in a Friedel Crafts type 14 reaction with an acid chloride having the structure R16CH2COC1 (R16 is defined as in connection with Formula 101) and is thereafter oxidized with 16 chromium trioxide in acetic acid to provide the isomeric 6 and 7 acyl-3,4-17 dihydro-1(2H)-naphthalenone derivatives. Only the 6-acyl derivative which is 18 of interest from the standpoint of the present invention, is shown by 19 structural formula (Formula 104) in Reaction Scheme 101. In the preparation of the presently preferred compounds of this invention the R, 21 groups represent methyl, R2, R3 and R,b are H, and therefore the preferred 22 intermediate corresponding to Formula 104 is 3,4-dihydro-4,4-dimethyl-6-23 acetyl-1(2H)- naphthalenone.
24 The exocyclic ketone function of the compound of Formula 104 is thereafter protected as a ketal, for example by treatment with ethylene glycol 26 in acid, to provide the 1,3-dioxolanyl derivative of Formula 105. The 27 compound of Formula 105 is then reacted with a Grignard reagent of the 28 formula R14MgBr (R14 is defined as in connection with Formula 101) to give 29 the 1,2,3,4- tetrahydro-1-hydroxy-naphthalene derivative of Formula 106.
The 1 exocyclic ketone function of the compound of Formula 106 is then 2 deprotected by treatment with acid and dehydrated to give the compound of 3 Formula 107.
4 An alternate method for obtaining the compounds of Formula 107 from the compounds of Formula 105 is by reacting the compounds of 6 Formula 105 with sodium bis(trimethylsilyl)amide and 2-[N,N-7 bis(trifluoromethylsulfonyl)amino]-5-chloropyridine (Tf = SOZCF3) in an inert 8 ether type solvent, such as tetrahydrofuran, at low temperatures (-78 C and a 0 C). This reaction proceeds through a sodium salt intermediate which is lo usually not isolated and is not shown in Reaction Scheme 101. The overall 11 reaction results in a trifluoromethylsulfonyloxy derivative, which is thereafter 12 reacted with an organometal derivative derived from the aryl or heteroaryl 13 compound R14H, such that the formula of the organometal derivative is 14 R14Met (Met stands for monovalent metal), preferably R14Li. (R14 is defined as in connection with Formula 101.) The reaction with the organometal is derivative, preferably lithium derivative of the formula R14Li is usually 17 conducted in an inert ether type solvent (such as tetrahydrofuran) in the is presence of zinc chloride (ZnC12) and tetrakis(triphenylphosphine)-ie palladium(0) (Pd(PPh3)4). The organolithium reagent R14,Li, if not commercially available, can be prepared from the compound R14H (or its 21 halogen derivative R14 X, where X, is halogen) in an ether type solvent in 22 accordance with known practice in the art. The temperature range for the 23 reaction between the reagent R14Li and the trifluoromethylsulfonyloxy 24 derivative is, generally speaking, in the range of approximately -78 C to 50 C.
The compounds of the invention are formed as a result of a 26 condensation between the ketone compound of Formula 107 and an 27 aldehyde or ketone of Formula 108. In the preparation of the preferred 28 exemplary compounds of the invention the reagent of Formula 108 is 4-29 carboxybenzaldehyde (Rl,-H). Examples of other reagents within the scope 1 of Formula 108 and suitable for the condensation reaction and for the 2 synthesis of compounds within the scope of the present invention (Formula 3 101) are: 5-carboxy-pyridine-2-aldehyde, 4- carboxy-pyridine-2-aldehyde, 4-4 carboxy-thiophene-2- aldehyde, 5-carboxy-thiophene-2-aldehyde, 4-carboxy-furan-2-aldehyde, 5-carboxy-furan-2-aldehyde, 4- carboxyacetophenone, 2-6 acetyl-pyridine-5-carboxylic acid, 2-acetyl-pyridine-4-carboxylic acid, 2-acetyl-7 thiophene-4-carboxylic acid, 2-acetyl-thiophene-5- carboxylic acid, 2-acetyl-8 furan-4-carboxylic acid, and 2-acetyl-furan-5-carboxylic acid. The latter 9 compounds are available in accordance with the chemical literature; see for 1o example Decroix et al., J. Chem. Res.(S), 4: 134 (1978); Dawson et al., J.
Med.
11 Chem. 29:1282 (1983); and Queguiner et al., Bull Soc. Chimique de France 12 No. 10, pp. 3678 - 3683 (1969). The condensation reaction between the 13 compounds of Formula 107 and Formula 108 is conducted in the presence of 14 base in an alcoholic solvent. Preferably, the reaction is conducted in ethanol in the presence of sodium hydroxide. Those skilled in the art will recognize 16 this condensation reaction as an aldol condensation, and in case of the herein 17 described preferred examples (condensing a ketone of Formula 107 with an 18 aldehyde of Formula 108) as a Claisen-Schmidt reaction. (See March:
1s Advanced Organic Chemistry: Reactions. Mechan'>smg, and Structure, pp. 694 - 695 McGraw Hill (1968). The compounds of Formula 109 are within the 21 scope of the present invention, and can also be subjected to further 22 transformations resulting in additional compounds of the invention.
23 Alternatively, the A-B group of Formula 108 may be a group which is within 24 the scope of the invention, as defined in Formula 101, only after one or more synthetic transformations of such a nature which is well known and within the 26 skill of the practicing organic chemist. For example, the reaction performed 27 on the A-B group may be a deprotection step, homologation, esterification, 28 saponification, amide formation or the like.
29 Generally speaking, regarding derivatization of compounds of Formula 1 109 and/or the synthesis of aryl and heteroaryl compounds of Formula 108 2 which can thereafter be reacted with compounds of Formula 107, well known 3 and published general principles and synthetic methodology can be employed 4 which is also described above under the subtitle "Synthetic Methods - Aryl 5 Substituted Compounds".

8 a OCH3 R1RtZ^ OCH3 9 (R~o Mp c~o CHZa2 ~
10 O R' 12 ICrO3 HOAc 14 R, OH 0 15 / OCH3 R14M9Br OCH3 I E---- a 16 (~o ~ T}iF or E120 ( '~o 17 R+ R, R, F~

19 H"

21 Ri= OCH 1.88r3 Ri= O C R
i i ~ CH2az i i u Hx ,.
22 (Ralo I (Ralo 'I
~ 2. R16CH20Op O
23 R. ~ DY^k* R, R, 29 Reaction Scheme 102 R,- ~ R, ~

OyCH2R,s i~r =C / /
(~o ~~ (~)o 6 O (Fries CH2R,=
rourangement) R
7 R, N R, O

CI (%)M-6 pyridne 12 iA .18 13 R" R" OyY(F6) OH AA KOH ay O
14 ( Rs)o Y(~ yYnd`ne (R0)O CIt2R,s R, R, O OH R1 R16 18 Hy804 HOAc R, AA
O Y(y Homologs and 21 (P.0I ' Derivativos o 22 R,g 23 R, R, 29 Reaction Scheme 102 (continued) 1 Referring now to Reaction Scheme 102, a synthetic route to those 2 compounds of the invention is described in which, with reference to Formula 3 101 p is zero, R2 in the aromatic portion of the condensed ring structure is 4 OH and Rõ is OH. Those skilled in the art will readily recognize that these compounds are P-diketones in the enol form. Reaction Scheme 102 also 6 describes a synthetic route to those compounds of the invention where p is 1.
7 Those skilled in the art will readily recognize that the latter compounds are 8 flavones. Thus, in accordance with this scheme a 1,2,3,4- tetrahydro-6-9 methoxynaphthalene-l-one derivative of Formula 110 is reacted with dialkyl 1o zinc (R1Zn) in the presence of titanium tetrachloride in a suitable solvent ii such as CH2C12 to replace the oxo function with the geminal dialkyl group 12 R,R,, to yield a compound of Formula 111, where R, is lower alkyl. In 13 preferred embodiments of the compounds of the invention which are made in 14 accordance with Reaction Scheme 102 the R3 group is hydrogen and R, are methyl. Accordingly, the dialkyl zinc reagent is dimethyl zinc, and the 16 preferred starting material of Formula 110 is 1,2,3,4- tetrahydro-6-17 methoxynaphthalene-1-one. The latter compound is commercially available, 18 for example from Aldrich Chemical Company. The 1,2,3,4-tetrahydro-1,2-1 s dialkyl-6-methoxy naphthalene derivative of Formula 111 is thereafter oxidized with chromium trioxide in acetic acid and acetic anhydride to give a 21 1,2,3,4-tetrahydro- 3,4-dialkyl-7-methoxy naphthalen-l-one derivative of 22 Formula 112. The ketone compound of Formula 112 is reacted with a 23 Grignard reagent (R14MgBr, R14 is defined as in connection with Formula 24 101) to yield a 1- hydroxy-l-aryl-3,4-dihydro-3,4-dialkyl-7-methoxy naphthalene derivative of Formula 113. The hydroxy compound of Formula 26 113 is subjected to elimination by heating, preferably in acid, to yield the 27 dihydronaphthalene compound of Formula 114. The methyl group is 2e removed from the phenolic methyl ether function of the compound of 29 Formula 114 by treatment with boron tribromide in a suitable solvent, such as 1 CHZC12, and therafter the phenolic OH is acylated with an acylating agent 2 that introduces the R,6CH2CO group, to give a compound of Formula 115.
3 In the preferred embodiment R16 is H, and therefore the acylating agent is 4 acetyl chloride or acetic anhydride. The acetylation reaction is conducted in a basic solvent, such as pyridine. The acylated phenol compound of Formula 6 115 is reacted with aluminum chloride at elevated temperature, causing it to 7 undergo a Fries rearrangement and yield the 1-aryl-3,4-dialkyl-3,4-dihydro-6-8 acyl-7-hydroxy-naphthalene compound of Formula 116. The phenolic 9 hydroxyl group of the compound of Formula 116 is acylated with an 1o acylating agent (such as an acid chloride) that introduces the CO-Y(R2)-A-B
11 group to yield a compound of Formula 117. In the acid chloride reagent CI-12 CO-Y(R2)-A-B (or like acylating reagent) the symbols Y, R2 and A-B have 13 the meaning defined in connection with Formula 101. In the preparation of 14 a preferred compound of the invention in accordance with this scheme this reagent is C1COC6H4COOEt (the half ethyl ester half acid chloride of 16 terephthalic acid).
17 The compound of Formula 117 is reacted with strong base, such as 18 potassium hydroxyde in pyridine, to yield, as a result of an intramolecular 19 Claisen condensation reaction, a compound of Formula 118. The compounds of Formula 118 are within the scope of the invention and of Formula 101, 21 where there is an OH for the R2 substituent in the aromatic portion of the 22 condensed ring moiety and Rõ is OH. In connection with the foregoing 23 reaction (intramolecular Claisen condensation) and the previously mentioned 24 Fries rearrangement it is noted that these probable reaction mechanisms are mentioned in this description for the purpose of fully explaining the herein 26 described reactions, and for facilitating the work of a person of ordinary skill 27 in the art to perform the herein described reactions and prepare the 26 compounds of the invention. Nevertheless, the present inventors do not wish 29 to be bound by reaction mechanisms and theories, and the herein claimed 1 invention should not be limited thereby.
2 The compounds of Formula 118 are reacted with strong acid, such as 3 sulfuric acid, in a suitable protonic solvent, such as acetic acid, to yield the 4 flavone compounds of Formula 119. The compounds of Formula 119 are also compounds of the invention, within the scope of Formula 101 where p is 6 1. Both the compounds of Formula 118 and Formula 119 can be subjected 7 to further reactions and transformations to provide further homologs and 8 derivatives, as described above in connection with Reaction Scheme 101.
9 This is indicated in Reaction Scheme 102 as conversion to homologs and 1 o derivatives.

11 N6 8rC(R3)2CH(R3)CO2H HO=C%CH~ N6 12 base (FP6,)=C~ N.

17 0 (Rp)M pd(n)02 0 18 (Rjo 02 / Cua'2 (F6)0 ZL4.-.RI. 18 Wseker O oxidation p TsOH

0 A)m R,. OH VQ%)m 26 (R,)o \ ' R,= R ~B~ 10 (PS)o ~ R,=
THF or E120 27 0~..~

29 Reaction Scheme 103 7 R,. OH Nm a,. (P2)m 8 +
H /
(Ftz)o pis i ZR~o R,=

O

13 a,Ay(p,)-A-e NaOH / EtOH

17 R,. (~lm / A=e 18 Homologs and Rõ Y(R2) Derivatives No I

R,e 29 Reaction Scheme 103 (continued) 1 Referring now to Reaction Scheme 103 a synthetic route is shown 2 leading to those compounds of the invention where, with reference to 3 Formula 101 X is S, 0 or NR', p is zero and Rl, is H or lower alkyl.
4 However, by applying the generic principles of synthesis shown in Reaction Scheme 102 the presently shown synthetic process can be modified or 6 adapted by those of ordinary skill in the art to also obtain compounds of the 7 invention where X is S, 0 or NR' and p is 1, or where X is S, 0 or NR' and 8 p is zero, the R2 group in the aromatic portion of the condensed ring moiety 9 is OH and R17 is OH.
The starting compound of Reaction Scheme 103 is a phenol, 11 thiophenol or aniline derivative of Formula 120. In the presently preferred 12 compounds of the invention the R2 and R16 groups are both hydrogen, and 13 the preferred starting compounds of Formula 120 are 3- ethenyl-thiophenol 14 or 3-ethenyl-phenol which are known in the chemical literature (Nuyken, et al. Polym. Bull (Berlin) 11:165 (1984). For the sake of simplifying the present 16 specification, in the ensuing description X can be considered primarily sulfur 17 as for the preparation of benzothiopyran derivatives of the present invention.
18 It should be kept in mind, however, that the herein described scheme is also 19 suitable, with such modifications which will be readily apparent to those skilled in the art, for the preparation of benzopyran (X = 0) and 21 dihydroquinoline (X = NR') compounds within the scope of the present 22 invention. Thus, the compound of Formula 120 is reacted under basic 23 condition with a 3-bromo carboxylic acid of the Formula 121. In this reaction 24 scheme the symbols have the meaning described in connection with Formula 101. An example for the reagent of Formula 121 where R3 is hydrogen, is 3-26 bromopropionic acid. The reaction with the 3-bromocarboxylic acid of 27 Formula 121 results in the compound of Formula 122. The latter is cyclized 28 by treatment with acid to yield the 7-ethenyl-thiochroman-4-one derivative 29 (when X is S) or 7-ethenyl-chroman derivative (when X is 0) of Formula 123.

1 The 7-ethenyl- thiochroman-4-one or 7-ethenyl-chroman-4-one derivative of 2 Formula 123 is oxidized in the presence of Pd(II)CIZ and CuC12 catalysts to 3 provide the corresponding 7-acyl (ketone) compound of Formula 124. Those 4 skilled in the art will recognize the latter reaction as a Wacker oxidation.
The exocyclic ketone group of the compound of Formula 124 is protected in 6 the form of a ketal, for example by treatment with ethylene glycol in acid, to 7 provide the 1,3-dioxolanyl derivative of Formula 125. Thereafter the 8 compound of Formula 125 is subjected to a sequence of reactions analogous 9 to those described for the compounds of Formula 105 in Reaction Scheme io 101. Thus, the 1,3- dioxolanyl derivative of Formula 125 is reacted with a 11 Grignard reagent of the formula R14MgBr to give the tertiary alcohol of 12 Formula 126, which is thereafter dehydrated in acid to provide the 13 benzothiopyran (X is S), benzopyran (X is 0) or dihydroquinoline (X is NR') 14 derivative of Formula 127. The ketone compound of Formula 127 is then reacted in the presence of base with the reagent of Formula 108 in an aldol 16 condensation (Claisen-Schmidt) reaction to provide compounds of the 17 invention of Formula 128. The compounds of Formula 128 can be converted ls into further homologs and derivatives, as described above in connection with is Reaction Schemes 101 and 102.
Specific Examples 21 Z-hydroxy-2-methyl-5-phenvlpentane 22 To a mixture of magnesium turnings 13.16 g (0.541 mol) in 200 ml of 23 anhydrous EtzO was added 100.0 g (0.492 mol) of 1-bromo-3-phenyl propane 24 as a solution in 100 ml of Et20. After of 5-10 ml of the solution had been added, the addition was stopped until the formation of the Grignard reagent 26 was in progress. The remaining bromide was then added over 1 hour. The 27 Grignard reagent was stirred for 20 minutes at 35 C and then 31.64 g (0.541 28 mol) of acetone was added over a 45 minute period. The reaction was 29 stirred overnight at room temperature before being cooled to 0 C and i acidified by the careful addition of 20% HCI. The aqueous layer was 2 extracted with Et20 (3 x 200 ml) and the combined organic layers washed 3 with water, and saturated aqueous NaCI before being dried over MgSO4.
4 Removal of the solvent under reduced pressure and distillation of the residue afforded 63.Og (72%) of the product as a pale-yellow oil, bp 99-102 C / 0.5 6 mm Hg. 1H NMR (CDC13): 8 7.28-7.18 (5H, m), 2.63 (2H, t, J = 7.5 Hz), 7 1.68 (2H, m), 1.52 (2H, m), 1.20 (6H, s).
8 1.2.3.4-tetrahkdro-1.1-dimethXlnaphthalene 9 A mixture of P205 (55.3 g, 0.390 mol) in 400 ml of inethanesulfonic lo acid was heated to 105 C under argon until all of the solid had dissolved.
ti The resulting solution was cooled to room temperature and 2-hydroxy-2-12 methyl-5-phenylpentane (63.0 g, 0.354 mol) added slowly with stirring.
After 13 4 hours the reaction was quenched by carefully pouring the solution onto I
L
14 of ice. The resulting mixture was extracted with Et20 (4 x 125 ml)and the t s combined organic layers washed with water, saturated aqueous NaHCO31 16 water, and saturated aqueous NaCl before being dried over MgSO4.
17 Concentration of the solution under reduced pressure, followed by distillation 18 afforded 51.0 g (90%) of the product as a clear colorless oil, bp. 65-67 C
/ 1.1 19 mmHg. 1H NMR (CDC13): 8 7.32 (1H, d, J = 7.4 Hz), 7.16-7.05 (3H, m), 2o 2.77 (2H, t, J = 6.3 Hz), 1.80 (2H, m), 1.66 (2H, m), 1.28 (6H, s).
21 3.4-dihydro-4.4-dimethvl-1(2H1-naphthalenone (Compound A) 22 A solution of 350 ml of glacial acetic acid and 170 ml of acetic 23 anhydride was cooled to 0 C and Cr031 25.0 g (0.25 mol) carefully added in 24 small portions. The resulting mixture was stirred for 30 minutes before 120 25 ml of benzene was added. 1,2,3,4-tetrahydro-1,1- dimethylnaphthalene was 2s added slowly as a solution in 30 ml of benzene. Upon completing the 27 addition the reaction was stirred for 4 hours at 0 C. The solution was diluted 28 with H20 (200 ml) and extracted with Et20 (5 x 50 ml). The combined 29 organic layers were washed with water, saturated aqueous NaCO3, and 1 saturated aqueous NaCI, before being dried over MgSO4. Removal of the 2 solvents under reduced pressure, and distillation afforded 16.0 g (74%) of the 3 product as a pale-yellow oil, bp 93-96 C / 0.3 mm Hg 1H NMR (CDC13): S
4 8.02 (1H, dd, J = 1.3, 7.8 Hz), 7.53 (1H, m ), 7.42 (1H, d, J = 7.9 Hz), 7.29 5(1H, m), 2.74 (2H, t, J = 6.8 Hz), 2.02 (2H, t, J = 6.8 Hz), 1.40 (6H, s).
6 3.4-dihvdro-4.4-dimethyl-7-bromo-1(2H)-naphthalenone l,Compounm 7 A 100 ml three-necked flask, fitted with an efficient reflux condenser 8 and drying tube, and addition funnel, was charged with a mixture of A1C13 9 9.5g (71.4 mmol) and 3 ml of CH2C12. The 3,4-dihydro- 4,4-dimethyl-1(2H)-1 o naphthalenone (5.0g, 28.7 mmol), was added dropwise with stirring (Caution:
11 Exothermic Reaction!) to the mixture at room temperature. Bromine, 5.5 g 12 (34.5 mmol), was then added very slowly, and the resulting mixture stirred for 13 2 hours at room temperature. (Note: if stirring stops, the mixture can be 14 warmed to 70 C until stirring resumes.) The reaction was then quenched by 15 the slow addition of ice-cold 6M HCI. The mixture was extracted with Et20 16 and the combined organic layers washed with water, saturated aqueous 17 NaHCO3, and saturated NaCI, before being dried over MgSO4. Removal of 18 the solvent under reduced pressure, and distillation of the residue afforded 19 5.8 g (80%) of the product as a pale-yellow oil which solidified on standing, 2o bp: 140 C / 0.4 mm Hg. 1H NMR (CDC13): S 8.11 (1H, d, J = 3.0 Hz), 7.61 21 (1H,dd,J=3.0,9.0Hz),7.31(1H,d,J=9.0Hz),2.72(2H,t,J=6.0Hz), 22 2.01 (2H, t, J = 6.0 Hz), 1.28 (6H, s).
23 1.2.3.4-tetrahvdro-l-h,ydroU-1-(4-met!Wphenyl)-4.4-dimethXl-7-24 bromonaphthalene (.,Compound C) 25 To a mixture of magnesium turnings (648.0 mg, 27.0 mmol) in 25 ml of 26 THF was added a solution of 4- bromotoluene (5.40 g, 31.8 mmol) in 10 ml 27 of THF in two portions. The reaction was initiated by the addition of 2 ml of 28 the solution, followed by the slow addition of the remaining solution via an 29 addition funnel. The mixture was stirred at room temperature for 1 hour, 1 and then the solution was transferred to a second flask using a canula. To 2 the resulting Grignard reagent was added 4.0 g (15.9 nlmol) of 3,4-dihydro-3 4,4-dimethyl-7- bromo-1(2H)-naphthalenone (Compound B) as a solution in 4 15 ml of THF. The resulting solution was heated to reflux overnight, cooled to room temperature, and the reaction quenched by the careful addition of s ice-cold 10% HCI. Extraction with Et20 was followed by washing of the 7 combined organic layers with H20 and saturated aqueous NaCl, then drying 8 over MgSO4. Removal of the solvent under reduced pressure provided an oil 9 which afforded the product as a colorless solid after column chromatography to (hexanes / EtOAc , 96 : 4). 1H NMR (CDC13): S 7.36 (1H, dd, J = 2.1, 7.6 11 Hz), 7.26 (3H, m), 7.12 (3H, s), 2.34 (3H, s), 2.24-2.04 (2H, m), 1.81 (1H, m), 12 1.55 (1H, m), 1.35 (3H, s), 1.30 (3H, s).
13 3.4-dih, dro-1- (4-methylphenyl)-4.4-dimethyl-7- bromonaphthalene 14 (_Compound D) A flask equipped with a Dean-Stark trap was charged with 3.4 g of is (9.85 mmol) of 1,2,3,4-tetrahydro-l-hydroxy-l-(4-methylphenyl)-4,4-17 dimethyl-7-bromonaphthalene (Compound C) and 40 ml of benzene. A
18 catalytic amount of p-toluenesulfonic acid monohydrate was added and the ia resulting solution heated to reflux for 2 hours. Upon cooling to room temperature, Et20 was added and the solution washed with H20, saturated 21 aqueous NaHCO3, and saturated aqueous NaCI then dried over MgSO4.
22 Removal of the solvents under reduced pressure, and column 23 chromatography (100% hexane / silica gel) afforded the title compound as a 24 colorless solid. 1H NMR (CDC13): 8 7.32 (1H, dd, J = 2.1, 8.2 Hz), 7.21 (5H, m), 7.15 (1H, d, J = 2.1 Hz), 5.98 (1H, t, J = 4.7 Hz), 2.40 (3H, s), 2.32 26 (2H, d, J = 4.7 Hz), 1.30 (6H, s).
27 7-Ethwvl-3.4-dihydro-4.4-dimethylnaphthalen-1(2H)-one ,Com op und E~
28 To a solution (flushed for 15 minutes with a stream of argon) of 7 g 29 (27.6 mmol) of 3,4-dihydro-4,4- dimethyl-7-bromo-1(2H)-naphthalenone 1(Compound B) in 150 ml of triethylamine was added 0.97 g (1.3 mmol) of 2 bis(triphenylphosphine)palladium(II) chloride and 0.26 g (1.3 mmol) of 3 cuprous iodide. The solution mixture was flushed with argon for 5 minutes 4 and then 39 ml (36.6 mmol) of (trimethylsilyl)acetylene was added. The reaction mixture was sealed in a pressure tube and placed in a preheated oil 6 bath (100 C) for 24 hours. The reaction mixture was then filtered through 7 Celite, washed with Et20 and the filtrate concentrated in vacuo to give crude 8 7-(trimethylsilyl)ethynyl-3,4-dihydro- 4,4-dimethyl.naphthalen-1(2H)-one. To a 9 solution of this crude TMS-acetylenic compound in 50 ml of methanol was 1 o added 0.6 g (4.3 mmol) of K2C03. The mixture was stirred for 8 hours at 11 ambient temperature and then filtered. The filtrate was concentrated in 12 vacuo, diluted with Et20, washed with water, 10% HC1 and brine, dried over 13 MgSO4 and concentrated in vacuo. Purification by column chromatography 14 (silica, 10% EtOAc-hexane) yielded the title compound as a white solid.
PMR (CDC13) : 8 1.39 (6H, s), 2.02 (2H, t, J = 7.0 Hz), 2.73 (2H, t, J = 7.0 16 Hz), 3.08 (1H, s), 7.39 (1H, d, J = 8.2 Hz), 7.61 (1H, dd, J = 1.8 , 8.2 Hz), 17 8.14(1H,d,J = 9 1.8 Hz).
18 Ethvl-4-iodobenzoate 19 To a suspension of 10 g (40.32 mmol) of 4-iodobenzoic acid in 100 ml 2o absolute ethanol was added 2 ml thionyl chloride and the mixture was then 21 heated at reflux for 3 hours. Solvent was removed in vacuo and the residue 22 was dissolved in 100 ml ether. The ether solution was washed with saturated 23 NaHCO3 and saturated NaC1 solutions and dried (MgSO4). Solvent was then 24 removed in vacuo and the residue Kugelrohr distilled (100 C; 0.55 mm) to give the title compound as a colorless oil, PMR (CDC13): S 1.42 (3H, t, J- 7 2e Hz), 4,4 (2H, q, J- 7 Hz), 7.8 (4H).
27 6-iodonicotinic acid 28 Sodium iodide (20.59 g, 137.40 mmol) was cooled to -78 C under argon 29 and then hydriodic acid (97.13 g, 759.34 mmol) was added. The cooling bath 1 was removed and the suspension was stirred for 5 minutes. To this mixture 2 was added 6-chloronicotinic acid (22.09 g, 140.20 mmol) and the resulting 3 mixture was slowly warmed to ambient temperature with stirring. The 4 mixture was heated to reflux at 125 C for 24 hours, cooled to ambient temperature and poured into acetone (500 ml) at 0 C. The yellow solid was 6 collected by filtration and washed with 200 ml of 1N aqueous NaHSO3 7 solution. Recrystallization from methanol (crystals were washed with ethyl 8 ether) afforded the title compound as white crystals: mp 177-179 C [lit. mp 9 187-192, Newkome et al. "Reductive Dehalogenation of Electron-Poor 1o Heterocycles: Nicotinic Acid Derivatives" J. Org Chem. 51: 953-954 (1986).
11 1H NMR (DMSO-d6): 8 8.81 (1H, dd, J= 0.8, 2.4 Hz), 8.01 (1H, dd, J
12 0.8, 8.2 Hz), 7.91 (1H, dd, J = 2.4, 8.2 Hz).
13 EthX16-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-(3-1e dimethylaminopropyl)-3- ethylcarbodiimide hydrochloride (19.86 g, 103.6 17 mmol) in dichloromethane (250 ml). 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 (550 21 ml). The combined organic phases were washed with saturated aqueous 22 NaCI, dried (MgSO4) and concentrated to a yellow solid. Purification by 23 flash chromatography (silica, 10% EtOAc-hexane) afforded the title 24 compound as white needles: mp 48-49 C; 1H NMR (CDC13): S 8.94 (1H, d, J = 2.1 Hz), 7.91 (1H, dd, J = 2.1, 8.2 Hz), 7.85 (1H, d, J = 8.2 Hz), 4.41 26 (2H, q, J = 7.1 Hz), 1.41 (3H, t, J = 7.1 Hz).
27 Ethv14-j(5.6,7.8-tetrahydro-5.5-dimethyl-8-oxo-2-2s naphthalenyl)ethynvllbenzoate (Compound Fl 29 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 with a 2 stream of argon, and 6 g (21.7 mmol) of ethyl 4-iodobenzoate in 100 ml of 3 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 then 6 stirred at ambient temperature for 18 hours. The reaction mixture was 7 filtered through Celite and the filtrate was concentrated in vacuo.
Purification 8 by flash chromatography (silica, 10 % EtOAc-hexane) yielded the title s compound as a white solid. PMR (CDC13) : 8 1.41 (3H, t, J = 7.2 Hz), 1.41 io (6H, s), 2.04 (2H, t, J 6.5 Hz), 2.76 (2H, t, J = 6.5 Hz), 4.40 (2H, q, J =
11 7.2 Hz), 7.44 (1H, d, J 8.2 Hz), 7.59 (2H, d, J = 8.4 Hz), 7.68 (1H, dd, J
12 1.8, 8.2 Hz), 8.04 (2H, d, J = 8.4Hz),8.15(1H,d,J = 1.8 Hz).
13 Ethy14-[(5.6-dihydro-5,5-dimethyl-8-(trifluoromethxlsulfonyl)oxy-2-i 4 naphthalenyl)ethvnyl]benzoate ,Compound.,Gl.
To a cold solution (-78 C) of 291.6 mg (1.59 mmol) of sodium 16 bis(trimethylsily)amide in 5.6 ml of THF was added a solution of 500.0 mg 17 (1.44 mmol) of ethyl 4- [(5,6,7,8-tetrahydro-5,5-dimethyl-8-oxo-2-18 naphthalenyl)ethynyl]benzoate (Compound F) in 4.0 ml of THF. The 19 reaction mixture was stirred at -78 C for 35 minutes and then a solution of 2o 601.2 mg (1.59 mmol) of 5-chloro(2-bis-triflouromethylsulfonyl)imide in 4.0 21 ml of THF was added. After stirring at -78 C for 1 hour, the solution was 22 warmed to 0 C and stirred for 2 hours. The reaction was quenched by the 23 addition of saturated aqueous NH4C1. The mixture was extracted with 24 EtOAc (50 ml) and the combined organic layers were washed with 5%
aqueous NaOH, water, and brine. The organic phase was dried over Na2SO4 26 and then concentrated in vacuo to a yellow oil. Purification by column 27 chromatography (silica, 7% EtOAc-hexanes) yielded the title compound as a 28 colorless solid. 1H NMR (CDC13): 8 8.04 (2H, dd, J 1.8, 8.4 Hz), 7.60 29 (2H, dd, J = 1.8, 8.4 Hz), 7.51 (2H, m), 7.32 (1H, d, J 8.0 Hz), 4.40 (2H, t q, J 7.1 Hz), 6.02 (1H, t, J = 5.0 Hz), 2.44 (2H, d, J = 5.0 Hz), 1.43 (3H, 2 t, J 7.1 Hz), 1.33 (6H, s).
3 Ethy14-[(5.6-dihYdro-5.5-dimeth,Y1-8-(4-methylphenyl)-2-4 naphthalenvl)ethvn,-1]benzoate (Compound 1) A solution of 4-lithiotoluene was prepared by the addition of 189.9 mg 6 (1.74 ml, 2.96 mmol) of t-butyl lithium (1.7M solution in hexanes) to a cold 7 solution (-78 C) of 253.6 mg (1.482 mmol) of 4-bromotoluene in 2.0 ml of 8 THF. After stirring for 30 minutes a solution of 269.4 mg (1.977 mmol) of s zinc chloridb in 3.0 ml of THF was added. The resulting solution was warmed to room temperature, stirred for 30 minutes, and added via cannula 11 to a solution of 472.9 mg (0.988 mmol) of ethyl 4-[(5,6-dihydro-5,5-12 dimethyl-8-(trifluoromethylsulfonyl)oxy-2-naphthalenyl)ethynyl]benzoate 13 (Compound G) and 50 mg (0.04 mmol) of 14 tetrakis(triphenylphosphine)palladium(0) in 4.0 ml of THF. The resulting solution was heated at 50 C for 45 minutes, cooled to room temperature and 16 diluted with sat. aqueous NH4C1. The mixture was extracted with EtOAc (40 17 ml) and the combined organic layers were washed with water and brine.
18 The organic phase was dried over Na2SO4 and concentrated in vacuo to a 19 yellow oil. Purification by column chromatography (silica, 5% EtOAc-2o hexanes) yielded the title compound as a colorless solid. 1H NMR (d6-21 acetone): 8 1.35 (6H, s), 1.40 (3H, t, J = 7.1 Hz), 2.36 (2H, d, J = 4.7 Hz), 22 2.42 (3H,s), 4.38 (2H, q, J = 7.1 Hz), 5.99 (1H, t, J = 4.7 Hz), 7.25 (5H, m), 23 7.35 (2H, m), 7.52 (2H, d, J = 8.5 Hz), 7.98 (2H, d, J = 8.5 Hz).
24 Ethv14-[(5.6-dihydro-5.5-dimethyl-8-phen l-y 2-na htp halenvl)ethvn,~~l]benzoate (Compound Ia) 26 Employing the same general procedure as for the preparation of ethyl 27 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-28 naphthalenyl)ethynyl]benzoate (Compound 1), 203.8 mg (0.43 mmol) of ethyl 29 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)o)W-2-1 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title 2 compound (colorless solid) using 58.2 mg (0.36 ml, 0.69 mmol) of 3 phenyllithium (1.8M solution in cyclohexane/Et2O), 116.1 mg (0.85 mmol) of 4 zinc chloride and 13.8 mg (0.01 mmol) of tetrakis(triphenylphosphine)palladium(0). PMR (CDC13): 8 1.36 (6H, s), 6 1.40 (3H, t, J = 7.1Hz), 2.37 (2H, d, J 4.7 Hz), 4.38 (2H, q, J = 7.1 Hz), 7 6.02(1H,t,J=4.7Hz),7.20(1H,d,J= 1.5Hz),7.27(1H,m),7.39(6H, s m), 7.52 (2H, d, J = 8.2 Hz), 7.98 (2H, d, J = 8.2 Hz).
9 Ethv14-[,(5.6-dihydro-5.5-dimethyl-8-(3-methylphenyl)-lo naphthalenyl, ethWvllbenzQate (Compound 2) 11 Employing the same general procedure as for the preparation of ethyl 12 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-13 naphthalenyl)ethynyl]benzoate (Compound 1), 250.0 mg (0.522 mmol) of 14 ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title 16 compound (colorless solid) using 284.8 mg (2.090 mmol) of zinc chloride, 24 17 mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) in 2.0 ml of ts THF, and 3-methylphenyl lithium (prepared by adding 201.2 mg (1.86 ml, 19 3.14 mmol) of tert-butyllithium (1.7M solution in pentane) to a cold solution 2o (- 78 C) of 274.0 mg (1.568 mmol) of 3-methylbromobenzene in 2.0 ml of 21 THF). 1H NMR (CDC13): 8 7.99 (2H, d, J = 8.4 Hz), 7.51 (2H, d, J = 8.4 22 Hz), 7.39-7.14 (7H, m), 5.99 (1H, t, J = 4.7 Hz), 4.37 (2H, q, J = 7.1 Hz), 23 2.60 (3H, s), 2.35 (2H, d, J = 4.7 Hz), 1.39 (3H, t, J = 7.1 Hz), 1.34 (6H, s).
24 Ethyl 4-[(5.6-dihydro-5.5-dimeth 1-8- (2-methylphenvl)-2-naphthalenyl)ethynyl]benzoate (Compound 3) 26 Employing the same general procedure as for the preparation of ethyl 27 4-[(5,6-dihydro-5,5-dimethyl-8-(4-methylphenyl)-2-28 naphthalenyl)ethynyl]benzoate (Compound 1), 200.0 mg (0.418 mmol) of 29 ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-i naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title 2 compound (colorless solid) using 199.4 mg (1.463 mmol) of zinc chloride, 24 3 mg (0.02 mmol) of tetrakis(triphenyiphosphine)palladium(0) in 4.0 ml of 4 THF, and 2-methylphenyl lithium (prepared by adding 133.9 mg (1.23 ml, 2.09 mmol) of tert-butyllithium (1.7M solution in pentane) to a cold solution s(- 78 C) of 178.7 mg (1.045 mmol) of 2-methylbromobenzene in 2.0 ml of 7 THF). 1H NMR (CDC13): S 7.97 (2H, d, J = 8.4 Hz), 7.50 (2H, d, J = 8.4 a Hz), 7.49-7.19 (6H, m), 6.81 (1H, d, J = 1.6 Hz), 5.89 (1H, t, J = 4.5 Hz), 9 4.36 (2H, q, J = 7.1 Hz), 2.43-2.14 (2H, dq, J = 3.7, 5.4 Hz), 2.15 (3H, s), to 1.39-1.34 (9H, m).
Eth y1 4[(5.6-dihvdro-5.5-dimeth -1-8-(3.5-dimethylphen,-1)-2-12 naphthalenyl)ethmvl enzoate lCompound 4) 13 Employing the same general procedure as for the preparation of ethyl 14 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-naphthalenyl)ethynyl]benzoate (Compound 1), 250.0 mg (0.522 mmol) of 16 ethyl4[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-17 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title 18 compound (colorless solid) using 249.0 mg (1.827 mmol) of zinc chloride, 24 19 mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) in 2.0 ml of 2o THF, and 3,5-dimethylphenyl lithium (prepared by adding 167.7 mg (1.54 ml, 21 2.62 mmol) of tert- butyllithium (1.7M solution in pentane) to a cold solution 22 (-78 C) of 249.0 mg (1.305 mmol) of 3,5- dimethylbromobenzene in 2.0 ml of 23 THF). 1H NMR (CDC13): 8 7.98 (2H, d, J = 8.4 Hz), 7.52 (2H, d, J = 8.4 24 Hz), 7.40-7.33 (2H, m), 7.20 (1H, d, J = 1.6 Hz), 7.00 (IH, s), 6.97 (2H, s), 5.97(1H,t,J=4.8Hz),4.37(2H,q,J=7.1Hz),2.36(6H,s),2.34(2H,d,J
28 = 4.8 Hz), 1.39 ( 3H, t, J = 7.1 Hz), 1.37 (6H, s).
27 Ethy14-[,(5.6-dihvdro-5.5-dimethvl-8-(4-ethylphenyl)-2.-_ 28 naphthalenvl, ethyDyl]benzoate (Compound 5) 29 Employing the same general procedure as for the preparation of ethyl 1 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-2 naphthalenyl)ethynyl]benzoate (Compound 1), 250.0 mg (0.522 mmol) of 3 ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-4 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title s compound (colorless solid) using 249.0 mg (1.827 mmol) of zinc chloride, 24 6 mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) in 2.0 ml of 7 THF, and 4-ethylphenyl lithium (prepared by adding 167.7 mg (1.54 ml, 2.62 8 mmol) of tert-butyllithium (1.7M solution in pentane) to a cold solution 9 (-78 C) of 244.0 mg (1.305 mmol) of 4-ethylbromobenzene in 2.0 ml of THF).
lo 1H NMR (CDC13): 8 7.99 (2H, d, J = 8.4 Hz), 7.51 (2H, d, J = 8.4 Hz), 11 7.42 - 7.24 (7H, m), 5.99 (1H, t, J = 4.7 Hz), 4.37 (2H, q, J = 7.1 Hz), 2.71 12 (2H, q, J = 7.6 Hz), 2.35 (2H, d, J = 4.7 Hz), 1.39 ( 3H, t, J = 7.1 Hz), 1.34 13 (6H, s).
14 Ethyl 4-[(5.6-dihxdro-5.5-dimethyl-8-(,4-(1.1-dimethvlethyl)phenyl)-2-t 5 naphthalenti, et ynyl]benzoate_ (Compound 61 16 Employing the same general procedure as for the preparation of ethyl 17 4-[(5,6-dihydro-5,5-dimethyl-8-(2- thiazolyl)-2-naphthalenyl)ethynyl]benzoate 18 (Compound 1), 250.0 mg (0.52 mmol) of ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-is (trifluoromethylsulfonyl)oxy-2-naphthalenyl)ethynyl]benzoate (Compound G) 20 was converted into the title compound (colorless solid) using 142.4 mg (1.045 21 mmol) of zinc chloride and 4-tert- butylphenyl lithium (prepared by adding 22 100.6 mg (0.97 ml, 1.57 mmol) of tert-butyllithium (1.5M solution in pentane) 23 to a cold solution (-78 C) of 167.0 mg (0.78 mmol) of 4-tert-24 butylbromobenzene in 1.0 ml of THF) . 1H NMR (CDC13): 8 7.99 (2H, d, J
25 = 8.4 Hz), 7.55 (2H, d, J = 8.4 Hz), 7.28-7.45 (7H, m), 6.02 (1H, t, J =
4.9 2e Hz), 4.38 (2H, q, J = 7.2 Hz), 2.36 (2H, d, J = 4.9 Hz), 1.59 (3H, s), 1.40 27 (3H, t, J = 7.2 Hz), 1.39 (9H, s), 1.35 (6H, s).
28 Ethyl 4-[(5 6-dihydro-5 5-dimethy-1-8-(4-chlorophenyl)-2-2s naphthalenyllTy.pyl]benzoate (Compound 7) 1 Employing the same general procedure as for the preparation of ethyl 2 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-3 naphthalenyl)ethynyl]benzoate (Compound 1), 250.0 mg (0.522 mmol) of 4 ethyl 4-[(5,6-dihydro-5,5- dilnethyl-8-(trifluoromethylsulfonyl)oxy-2-naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title 6 compound (colorless solid) using 249.0 mg (1.827 mmol) of zinc chloride, 24 7 mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) in 2.0 ml of 8'THF, and 4-chlorophenyl lithium (prepared by adding 167.7 mg (1.54 ml, 2.62 9 mmol) of tert-butyllithium (1.7M solution in pentane) to a cold solution (-1o 78 C) of 252.4 mg (1.305 mmol) of 4-chloro-l- bromobenzene in 2.0 ml of 11 THF). 1H NMR (CDC13): 8 7.98 (2H, d, J = 8.4 Hz), 7.53 (2H, d, J = 8.4 12 Hz), 7.40-7.27 (6H, m), 7.12 (1H, d, J = 1.6 Hz), 6.00 (1H, t, J = 4.8 Hz), 13 4.37 (2H, q, J = 7.1 Hz), 2.35 (2H, d, J = 4.8 Hz), 1.40 (2H, t, J = 7.1 Hz), 14 1.34 (6H, s).
Ethv14-[(5.6-dih,ydro-5.5-dimethvl-8-(4-methoxyphenA)-2-18 na hthaleny_)ethvn,ti~l]benzoate (Compound 8) 17 Employing the same general procedure as for the preparation of ethyl 18 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-19 naphthalenyl)ethynyl]benzoate (Compound 1), 250.0 mg (0.522 mmol) of 2o ethyl4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-21 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title 22 compound (colorless solid) using 249.0 mg (1.827 mmol) of zinc chloride, 24 23 mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) in 2.0 ml of 24 THF, and 4-methoxyphenyl lithium (prepared by adding 167.7 mg (1.54 ml, 2s 2.62 mmol) of tert-butyllithium (1.7M solution in pentane) to a cold solution 26 (-78 C) of 244.1 mg (1.305 mmol) of 4-methoxy-l-bromobenzene in 2.0 ml of 27 'I'HF). 1H NMR (CDC13): S 7.98 (2H, d, J = 8.5 Hz), 7.52 (2H, d, J = 8.6 28 Hz), 7.40-7.21 (5H, m), 6.95 (2H, d, J = 8.7 Hz), 5.97 (1H, t, J = 4.7 Hz), 29 4.37 (2H, q, J = 7.1 Hz), 4.34 (3H, s), 2.34 (2H, d, J = 4.7 Hz), 1.39 (3H, t, J

1 = 7.1 Hz), 1.34 (6H, s).
2 Ethy14-j(5.6-dihydro-5.5-dimethxl-8:(4-trifluoromethXuhe vl)2-s naphthalenyl)th,,myl]benzoate (Compound 9) 4 Employing the same general procedure as for the preparation of ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-6 naphthalenyl)ethynyl]benzoate (Compound 1), 250.0 mg (0.522 mmol) of 7 ethyl4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-8 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title 9 compound (colorless solid) using 249.0 mg (1.827 mmol) of zinc chloride, 24 io mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) in 2.0 ml of ii THF, and 4-trifluoromethylphenyl lithium (prepared by adding 167.7 mg 12 (1.54 ml, 2.62 mmol) of tert-butyllithium (1.7M solution in pentane) to a cold 13 solution (-78 C) of 296.6 mg (1.305 mmol) of 4-14 trifluoromethylbromobenzene in 2.0 ml of THF). 1H NMR (CDC13): 8 7.98 (2H, d, J = 8.5 Hz), 7.67 (2H, d, J = 8.3 Hz), 7.54 - 7.36 (6H, m), 7.10 (1H, ~s d, J = 1.6Hz),6.06(1H,t,J=4.8Hz),4.37(2H,q,J=7.1Hz),2.38(2H, 17 d, J = 4.8 Hz), 1.39 (3H, t, J = 7.1 Hz), 1.35 (6H, s).
18 Ethy14-[(5.6-dihydro-5.5-dimethy_1-8-(2-pyridyl)-2-ia naphthalenyl)eth3myllbenzoate (Compound 10) Employing the same general procedure as for the preparation of ethyl 21 4-[(5,6-dihydro-5,5-dimethyl-8-(4-methylphenyl)-2-22 naphthalenyl)ethynyl]benzoate (Compound 1), 250.0 mg (0.52 mmol) of ethyl 23 4-[(5,6-dihydro-5,5-d'unethyl-8-(trifluoromethylsulfonyl)oxy-2-24 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title compound (colorless solid) using 142.4 mg (1.045 mmol) of zinc chloride and 26 2-lithiopyridine (prepared by the addition of 100.6 mg (0.97 ml, 1.57 mmol) 27 of tert-butyllithium (1.5M solution in pentane) to a cold solution (-78 C) of 28 123.8 mg (0.784 mmol) of 2-bromopyridine in 1.0 ml of THF). 1H NMR
29 (d6-acetone): 8 8.64 (1H, m), 7.99 (2H, d, J = 8.5 Hz), 7.85 ( 1H, ddd, J

1 1.8, 7.7, 9.5 Hz), 7.58 (2H, d, J = 8.4 Hz), 7.50 (1H, d, J = 7.7 Hz), 7.47 (2 2 H, d, J = 1.1Hz),7.35(2H,m),6.32(1H,t,J=4.8Hz),4.34(2H,q,J=
3 7.2 Hz), 2.42 (2H, d, J = 7.4 Hz), 1.35 (3H, t, J = 7.0 hz), 1.35 (6H, s).
4 Ethyl 4-[(5.6-dih d ro-S,S-dimet ~-8-(3-pyr'dv1~2-s naphthalenvl)eth -~n,yl]benzoate (Compound 11) s Employing the same general procedure as for the preparation of ethyl 7 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-8 naphthalenyl)ethynyl]benzoate (Compound 1), 170.0 mg (0.35 mmol) of ethyl 9 4-[(5,6-dihydro-5,5-dimethyl-8-(trifluoromethylsulfonyl)oxy-2-1o naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title 11 compound (colorless solid) using 142.4 mg (1.045 mmol) of zinc chloride and 12 3-lithiopyridine (prepared by the addition of 100.2 mg (0.92 ml, 1.56 mmol) 13 of tert-butyllithium (1.5M solution in pentane) to a cold solution (-78 C) of 14 123.8 mg (0.784 mmol) of 3-bromopyridine in 1.0 ml of THF). 1H NMR
15 (CDC13): S 8.63-8.61 (2H, dd, J = 1.7 Hz), 7.99 2H, d, J = 8.4 Hz), 7.67 1s (1H, dt, J = 7.9 Hz), 7.52 (2H, d, J = 8.4 Hz), 7.43-7.34 (3H, m), 7.10 (1H, 17 d, J = 1.6Hz),6.07(1H,t,J=4.7Hz),4.37(2H,q,J=7.1Hz),2.40(2H, 18 d, J = 4.7 Hz), 1.390 (3H, t, J = 7.1 Hz), 1.36 (6H, s).
19 Ethyl 4-[15,6-dihydro-5.5-dimeth,yl-8-l,2-methyl-5-pyridyl, -2-2o naphthalenyllethynyl]benzoate(Compound 12) 21 Employing the same general procedure as for the preparation of ethyl 22 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenlyl)-2-23 naphthalenyi)ethynyl]benzoate (Compound 1), 250.0 mg (0.522 mmol) of 24 ethyl4-[(5,6- dihydro-5,5-d'unethyl-8-(trifluoromethylsulfonyl)oxy-2-25 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title 26 compound (colorless solid) using 142.4 mg (1.045 mmol) of zinc chloride and 27 2-methyl-5- lithiopyridine (prepared by the addition of 100.5 mg (0.92 ml, 28 1.57 mmol) of tert-butyllithium (1.7 M solution in pentane) to a cold solution 29 (-78 C) of 134.8 mg (0.784 mmol) of 2-methyl-5-bromopyridine in 1.0 ml of 1 THF). 1H NMR (CDC13): S 8.50 (1H, d, J = 2.2 Hz), 7.99 (2H, d, J = 8.3 2 Hz), 7.56 (1H, dd, J = 2.3, 8.0 Hz), 7.53 (2H, d, J = 8.4 Hz), 7.43 (1H, dd, J
3 2.3, 8.0 Hz), 7.37 (2H, d, J = 8.0 Hz), 7.21 (1H, d, J 8.1 Hz), 7.11 (1H, 4 d, J = 1.5Hz),6.04(1H,t,J=4.7Hz),4.38(2H,q,J=7.2Hz),2.63(3H, s), 2.38 (2H, d, J = 4.6 Hz), 1.40 (3H, t, J = 7.1 Hz), 1.35 (6H, s).
e EthY14-[ j5.6-dihvdro-5.5-dimethyl-8-((j2.2- dimethvlet, hvll-7 dimethvls, iloxy)phenyl)-2- naphthalenvl, ethvnyl]benzoate lCompound Hl 8 Employing the same general procedure as for the preparation of ethyl 9 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-1 o naphthalenyl)ethynyl]benzoate (Compound G), 150.0 mg (0.314 mmol) of ii ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-12 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title 13 compound (colorless solid) using 150.0 mg (1.10 mmol) of zinc chloride, 24 14 mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) in 2.0 ml of THF, and 3-((2,2-dimethylethyl)dimethylsiloxy)phenyl lithium (prepared by 1 s adding 100.2 mg (0.92 ml, 1.564 mmol) of tert-butyllithium (1.7M solution in 17 pentane) to a cold solution (-78 C) of 226.0 mg (0.787 mmol) of 3-((2,2-18 dimethylethyl)dimethylsiloxy)bromobenzene in 2.0 ml of THF). 1H NMR
19 (CDC13): 8 7.98 (2H, d, J = 8.4 Hz), 7.51 (2H, d, J = 8.4 Hz), 7.40-7.22 (4H, 2o m), 6.95 (1H, d, J = 7.6 Hz), 6.84-6.82 (2H, m), 6.00 (1H, t, J = 4.7 Hz), 21 4.37(2H,q,J=7.1Hz),2.35(2H,d,J=4.7Hz),1.39(3H,t,J=7.1Hz), 22 1.34 (3H, s), 0.99 (9H, s), 0.23 (6H, s).
23 Ethyl 4-[(5.6-dihydro-5.5-dimethyl-8-(,4((2.2-dimethvlethyl)-24 dimethvlsiloxvlphenvl); 2- naphthalenvllethpvl]benzoate (Compound Il Employing the same general procedure as for the preparation of ethyl 26 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-27 naphthalenyl)ethynyl]benzoate (Compound 1), 210.0 mg (0.439 mmol) of 28 ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-29 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title 1 compound (colorless solid) using 209.0 mg (1.53 mmol) of zinc chloride, 24 2 mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) in 2.0 ml of 3 THF, and 4-((2,2-dimethylethyl)dimethylsiloxy)phenyl lithium (prepared by 4 adding 140.3 mg (1.30 ml, 2.19 mmol) of tert-butyllithium (1.7M solution in pentane) to a cold solution (-78 C) of 315.0 mg (1.09 mmol) of 4-((2,2-6 dimethylethyl)dimethylsiloxy)bromobenzene in 2.0 ml of THF). 1H NMR
7 (CDC13): S 7.98 (2H, d, J = 8.4 Hz), 7.51 (2H, d, J = 8.4 Hz), 7.39-7.25 (3H, 8 m), 7.21 (2H, d, J 8.5 Hz), 5.87 (2H, d, J 8.5 Hz), 5.96 (1H, t, J 4.7 9 Hz),4.37(2H,q,J=7.1Hz),2.33(2H,d,J=4.7Hz),1.39(3H,t,J=7.1 io Hz), 1.33 (6H, s), 1.01 (9H, s), 0.25 (6H, s).
ii Ethy14-[(5.6-dihydro-5.5-dimeth,Yl-8-(3 hydrox,yphenyl)-2-12 naphthalenyl,le hynyl]benzoate (Compound 13) 13 To a solution of ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(3-((2,2-14 dimethylethyl)-d`unethylsiloxy)- phenyl)-2-naphthalenyl)ethynyl]benzoate (Compound H) 60.0 mg (0.114 mmol) in 1.0 ml of THF at room temperature 16 was added 91.5 mg (0.35 ml, 0.35 mmol) of tetrabutylamonium flouride (1 M
17 solution in THF). After stirring overnight, the solution was diluted with is EtOAc and washed with HZO and saturated aqueous NaCI, before being 19 dried over MgSO4. Removal of the solvents under reduced pressure, 2o followed by column chromatography (4:1, Hexanes:EtOAc) afforded the title 21 compound as a colorless solid. 1H NMR (CDC13): S 7.98 (2H, d, J = 7.8 22 Hz), 7.52 (2H, d, J = 8.3 Hz), 7.39 - 7.21 (4H, m), 6.93 (1H, d, J = 7.5 Hz), 23 6.84(1H,d,7.1Hz),6.83 (1H,s),6.01 1H,t,J = 4.7 Hz), 4.91 (1H,s),4.39 24 (2H, q, J = 7.1 Hz), 2.35 (2H, d, J = 4.7 Hz), 1.39 (3H, t, J = 7.1 Hz), 1.34 (6H, s).
2e Ethyl 4-1(5.6-dihydro-5.5-dimet]W-8-(4-h dry o=henYl)-2-27 naghthalenvllethynvllbenzoate (Compound 14) 28 To a solution of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(4-((2,2-29 dimethylethyl)- dimethylsiloxy)phenyl)-2-naphthalenyl)ethynyl]benzoate 1 (Compound I) 50.0 mg (0.095 mmol) in 1.0 ml of THF at room temperature .2 was added 73.2 mg (0.29 ml, 0.29 mmol) of tetrabutylamonium fluoride (1 M
3 solution in THF). After stirring overnight, the solution was diluted with 4 EtOAc and washed with H20 and saturated aqueous NaCI, before being dried over MgSO4. Removal of the solvents under reduced pressure, e followed by column chromatography (4:1, Hexanes:EtOAc) afforded the title 7 compound as a colorless solid. 1H NMR (CDC13): S 7.98 (2H, d, J = 8.2 8 Hz), 7.52 (2H, d, J= 8.3 Hz), 7.41 - 7.20 (5H, m), 6.88 (2H, d, J = 8.4 Hz), 9 5.96(1H,t,J=4.5Hz),4.37(2H,q,J=7.1Hz),2.34(2H,d,J=4.5Hz), 1o 1.39 (3H, t, J = 7.1 Hz), 1.34 (6H, s).
11 Ethy14-[,(5.6-dihydro-5.5-dimethyl-8:(5-methAthiazol-2-yl)-2-12 naphthalenvl)ethvnvl] benzoate(Compound 15) 13 Employing the same general procedure as for the preparation of ethyl 14 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-naphthalenyl)ethynyl]benzoate (Compound 1), 264.0 mg (0.552 mmol) of 16 ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-1 7 naphthalenyl)ethynyljbenzoate (Compound G) was converted into the title i s compound (colorless solid) using 150.0 mg (1.10 mmol) of zinc chloride, 14 19 mg (0.012 mmol) of tetrakis(triphenylphosphine)palladium(0) in 4.0 ml of 2o THF, and 5-methylthiazol-2-yl lithium (prepared by adding 53.2 mg (0.53 ml, 21 0.83 mmol) of n- butyllithium (1.55 M solution in hexanes) to a cold solution 22 (-78 C) of 82.0 mg (0.83 mmol) of 5-methylthiazole in 5.0 ml of THF). 1H
23 NMR (CDC13): 8 7.99 (2H, d, J = 7.8 Hz), 7.88 (1H, d, J = 1.5 Hz), 7.55 24 (2H, d, J = 7.8 Hz), 7.54 (1H, s), 7.45 (1H, dd, J = 1.5, 8.0 Hz), 7.35 (1H, d, J=7.9Hz),6.48(1H,t,J=4.8Hz),4.38(2H,q,J=7.1Hz),2.51(3H,s), 2e 2.38 (2H, d, J = 4.8 Hz), 1.40 (3H, s), 1.32 (6H, s).
27 EthY14-[15.6-dihydro- .5-dimethyl-8-(2-thiazolyl)-28 naphthalenyl, ethygvllbenzoate (Compound 15a1 29 A solution of 2-lithiothiazole was prepared by the addition of 41.2 mg 1 (0.42 ml, 0.63 mmol) of n-butyl- lithium (1.5M solution in hexanes) to a cold 2 solution (-78 C) of 53.4 mg (0.63 mmol) of thiazole in 1.0 ml of THF. The 3 solution was stirred at for 30 minutes and then a solution of 113.9 mg (0.84 4 mmol) of zinc chloride in 1.5 ml of THF was added. The resulting solution was warmed to room temperature, stirred for 30 minutes and then the s organozinc was added via cannula to a solution of 200.0 mg (0.42 mmol) of 7 ethyl 4-[(5,6- dihydro-5,5-dimethyl-8-(trifluoromethylsulfonyl)oxy-2-8 naphthalenyl)ethynyl]benzoate (Compound G) and 12.4 mg (0.01 mmol) of 9 tetrakis(triphenylphosphine)palladium(0) in 1.5 ml of THF. The resulting 1 o solution was heated at 50 C for 45 minutes, cooled to room temperature and 11 diluted with sat. aqueous NH4C1. The mixture was extracted with EtOAc (40 12 ml) and the combined organic layers were washed with water and .brine. The 13 organic phase was dried over Na2SO4 and concentrated in vacuo to a yellow 14 oil. Purification by column chromatography (silica, 20% EtOAc-hexanes) yielded the title compound as a colorless oil. PMR (CDC13): S 1.35 (6H, s), 1s 1.40(3H,t,J=7.1Hz),2.42(2H,d,J=4.8Hz),4.38(2H,q,J=7.1Hz), 17 6.57(1H,t,J=4.8Hz),7.33(1H,d,J=3.3Hz),7.36(1H,d,J=8.0Hz), 18 7.46 (1H, dd, J = 1.7, 8.1 Hz), 7.55 (2H, d, J = 8.4 Hz), 7.87 (1H, d, J =
1.7 19 Hz),7.92(1H,d,J=3.3Hz),8.00(2H,d,J=8.4Hz).
2o Ethv14-[(5.6-dihydro-5.5-dimethyl-8-(4-methylthiazol-2-v112-21 naphthalei~,vl, )ethMvll benzoate fCompound 16) 22 Employing the same general procedure as for the preparation of ethyl ra 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-24 naphthalenyl)ethynyl)benzoate (Compound 1), 295.0 mg (0.617 mmol) of ethyl 4-[(5,6-dihydro-5,5- d'unethyl-8-(trifluoromethylsulfonyl)oxy-2-26 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title 27 compound (colorless solid) using 168.0 mg (1.23 mmol) of zinc chloride, 16 28 mg (0.014 mmol) of tetrakis(triphenylphosphine)palladium(0) in 6.0 ml of 29 THF, and 4-methylthiazol-2-yl lithium (prepared by adding 59.6 mg (0.60 ml, 1 0.93 mmol) of n- butyllithium (1.55 M solution in hexanes) to a cold solution 2(-78 C) of 92.0 mg (0.93 mmol) of 4-methylthiazole in 6.0 ml of THF). 1H
3 NMR (CDC13): 8 8.00 (2H, d, J = 8.4 Hz), 7.80 (1H, d, J= 1.7 Hz), 7.55 4 (2H, d, J = 8.4 Hz), 7.45 (1H, dd, J = 1.7, 8.0 Hz), 7.35 (1H, 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), e 2.39 (2H, d, J = 4.7 Hz), 1.40 (3H, t, J = 7.2 Hz), 1.33 (3H, s).
7 Ethy14-((5.6-dih,ydro-5.5-dimethyl-8-(4.5-dimethylthiazol-2-vl)-2-na htg halenyl) 8 ethyn,yl] benzoate (Comnound 171 9 Employing the same general procedure as for the preparation of ethyl lo 4-[(5,6-dihydro-5,5-dimethyl-8-(4-methylphenyl)-2-ti naphthalenyl)ethynyl]benzoate (Compound 1), 200.0 mg (0.418 mmol) of 12 ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-13 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title 14 compound (colorless solid) using 110.0 mg (0.84 mmol) of zinc chloride, 12 mg (0.011 mmol) of tetrakis(triphenylphosphine)palladium(0) in 2.0 ml of 16 THF, and 4,5-dimethylthiazol-2-yl lithium (prepared by adding 40.2 mg (0.39 17 ml, 0.63 mmol) of n- butyllithium (1.55 M solution in hexanes) to a cold ts solution (-78 C) of 71.0 mg (0.63 mmol) of 4,5-dimethylthiazole in 2.0 ml of 19 THF). 1H NMR (CDC13): 8 8.00 (2H, d, J = 8.4 Hz), 7.82 (1H, d, J = 1.7 2o Hz), 7.54 (2H, d, J = 8.4 Hz ), 7.43 (1H, dd, J = 1.7, 8.0 Hz), 7.33 91H, d, J
21 = 8.0 Hz), 6.45 (1H, t, J = 4.9 Hz), 4.38 (2H, q, J= 7.1 Hz), 2.41 (3H, s), 22 2.40 (3H, s), 2.37 (2H, d, J = 4.9 Hz), 1.40 (3H, t, J = 7.1 Hz), 1.32 (6H, s).
23 4-(j5.6-Dihvdro-5.5-dimethrl-S-j2-methyl_5_p -ridy112-.
2a naphthalenyl)gthvnyl]benzoic acid (Compound 18) A solution of 81.7 mg (0.194 mmol) of ethyl 4- [(5,6-dihydro-5,5-2s dimethyl-8-(2-methyl-5-pyridyl)-2- naphthalenyl)ethynyl]benzoate (Compound 27 12) and 40.7 mg (0.969 mmol) of LiOH-H20 in 3 ml of THF/water (3:1, v/v), 28 was stirred overnight at room temperature. The reaction was quenched by 29 the addition of saturated aqueous NH4C1 and extracted with EtOAc. The 1 combined organic layers were washed with water and brine, dried over 2 NaZS04 and concentrated in vacuo to give the title compound as a colorless 3 solid. 1H NMR (d6-DMSO): 8 8.41 (1H, d, J = 1.9 Hz), 7.90 (2H, d, J =
4 8.3 Hz), 7.63 (1H, dd, J = 2.3, 7.9 Hz), 7.59 (2H, d, J = 8.3 Hz), 7.49 (2H, m),7.33(1H,d,J=7.8Hz),6.95(1H,s),6.11(1H,t,J=4.5Hz),2.52(3H, 6 s), 2.37 (2H, d, J = 4.6 Hz), 1.31 (6H, s).
7 4-[(5.6-Dihvdro-5.5-dimethXl-2-pyr~ idyl, -2- naphthalenyl)ethyl~yl]benzoic 8 acid (Compound 19) 9 A solution of 80.0 mg (0.196 mmol) of ethyl 4- [(5,6-dihydro-5,5-t o dimethyl-8-(2-pyridyl)-2- naphthalenyl)ethynyl]benzoate (Compound 10) and 11 20.6 mg (0.491 mmol) of LiOH-H20 in 3 ml of THF/water (3:1, v/v), was 12 stirred overnight at room temperature. The reaction was quenched by the 13 addition of saturated aqueous NH4C1 and extracted with EtOAc. The 14 combined organic layers were washed with water and brine, dried over Na2SO4 and concentrated in vacuo to give the title compound as a colorless is solid. 1H NMR (d6-DMSO): 8 8.64 (1H, m), 7.94 (2H, d, J = 8.3 Hz), 7.87 17 (1H,dt,J=1.7,7.8Hz),7.58(2H,d,J=8.3Hz),7.50(1H,d,J=8.2Hz), is 7.47 (2H, s), 7.37 ( 1H, m), 7.25 (1H, s), 6.30 (1H, t, J = 4.6 Hz), 2.39 (2H, 19 d, J = 4.6 Hz), 1.31 (6H, s).
2o 4-[(5.6-Dihydro-5.5-dimethyl-8-(3-methylphenyl)-2-21 pa htp halen,yl)ethy,nvllbenzoic acid (.ompound 201 22 To a solution of ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(3-23 methylphenyl)-2-naphthalenyl)ethynyl]benzoate (Compound 2) 30.0mg (0.071 24 mmol) in 3 ml 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 was heated to 50 C for 2s 2 hours, cooled to room temperature, and acidified with 10% HCI.
27 Extraction with EtOAc, followed by drying over Na2SO4, and removal of the 28 solvents under reduced pressure afforded the title compound as a colorless 29 solid. 1H NMR (DMSO): 8 7.90 (2H, d, J = 8.5 Hz), 7.59 (2H, d, J = 8.5 1 Hz), 7.46 (2H, s), 7.32-7.13 (4H, m), 7.10 (1H, s), 6.98 (1H, t, J = 4.5 Hz), 2 2.34 (3H, s), 2.31 (2H, d, J = 4.5 Hz), 1.30 (6H, s).
3 4-[(5.6-Dihvdro-5 5-dimethvl-8-(4-ethylphenyll-, 2naphthalen lv )ethywl]benzoic 4 acid (Compound 21) To a solution of ethyl4-[(5,6-dihydro-5,5-dimethyl-8-(4-ethylphenyl)-2-s naphthalenyl)ethynyl]benzoate (Compound 5) 47.0mg (0.108 mmol) in 3 ml 7 of EtOH and 2 ml of THF was added 28.0 mg (0.70 mmol, 0.7 ml) of NaOH
8 (1.0 M aqueous solution). The solution was heated to 50 C for 2 hours, 9 cooled to room temperature, and acidified with 10% HCI. Extraction with EtOAc, followed by drying over Na2SO4, and removal of the solvents under 11 reduced pressure afforded the title compound as a colorless solid. 1H NMR
12 (DMSO): 8 7.90 (2H, d, J= 8.3 Hz), 7.59 (2H, d, J = 8.3 Hz), 7.46 (2H, s), 13 7.29-7.21 (4H, m), 7.02 (1H, s), 6.01 (1H, t, J = 4.5 Hz), 2.64 (2H, q, J =
7.5 14 Hz), 2.33 (2H, d, J = 4.5 Hz), 1.29 (6H, s), 1.22 (3H, t, J = 7.5 Hz).
4-[(5.6-Dihydro-5 5-dimethyl-8-(4-methoxyphenyl)-2-le naphthalen,-1)eth3Myl]benzoic acid (Compound 22) 17 To a solution of ethyl4-[(5,6-dihydro-5,5-d'unethyl-8-(4-le methoxyphenyl)-2-naphthalenyl)ethynyl]benzoate (Compound 8) 80.0 mg 19 (0.183 mmol) in 3 ml of EtOH and 2 ml of THF was added 40.0 mg (1.00 mmol, 1.0 ml) of NaOH (1.0 M aqueous solution). The solution was heated 21 to 50 C for 2 hours, cooled to room temperature, and acidified with 10%
22 HCI. Extraction with EtOAc, followed by drying over Na2SO4, and removal 23 of the solvents under reduced pressure afforded the title compound as a 24 colorless solid. 1H NMR (DMSO): 8 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 26 (1H, t, J = 4.4 Hz), 3.79 (3H, s), 2.31 (2H, d, J = 4.7 Hz), 1.29 (6H, s).
27 4-[,(5.6-Dihvdro-5.5-d'unethyl-8-(4-trifluoromethylphenvl)-2-2e na hthalenylle hynyl]benzoic acid (Compound 23) 29 To a solution of ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(4-1 trifluoromethylphenyl)- -2-naphthalenyl)ethynyl]benzoate (Compound 9) 70.0 2 mg (0.148 mmol) in 3 ml of EtOH and 2 ml of THF was added 60.0 mg (1.50 3 mmol, 1.50 ml) of NaOH (1.0 M aqueous solution). The solution was heated 4 to 50 C for 2 hours, cooled to room temperature, and acidified with 10%
HCI. Extraction with EtOAc, followed by drying over Na2SO4, and removal e of the solvents under reduced pressure afforded the title compound as a 7 colorless solid. iH NMR (DMSO): 8 7.90 (2H, d, J = 8.3 Hz), 7.80 (2H, d, 8 J = 8.1 Hz), 7.61-7.47 (6H, m), 6.97 (2H, s), 6.16 (1H, t, J = 4.5 Hz), 2.37 9 (2H, d, J = 4.6 Hz), 1.30 (6H, s).
t o 4-[(5.6-Dih,ydro-5.5-dimeth,l-8- 3 5-dimethXlpheny~ 2-11 na , htllalenyl)ethvnvllbenzoic acid (Compound 24) 12 To a solution of ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(3,5-13 dimethylphenyl)-2-naphthalenyl)ethynyl]-benzoate (Compound 4) 90.0 mg 14 (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 was heated te to 50 C for 2 hours, cooled to room temperature, and acidified with 10%
17 HCI. Extraction with EtOAc, followed by drying over Na2SO4, and removal 1 e of the solvents under reduced pressure afforded the title compound as a 19 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 (1H, s), 6.97 (1H, s), 5.97 ( 1H, t, J = 4.5 Hz), 21 2.31 (2H, d, J = 4.5 Hz), 2.30 (6H, s), 1.29 (6H, s).
22 4-[,(5.6-Dihvdro-5.5-dimethyl-8-(4-chlorophenyl)-2-23 naphthalenLI)ethynyllbenzoic acid (Compound 25) 24 To a solution of ethyl 4-[(5,6-dihydro-5,5-dimethyl-8-(4-chlorophenyl)-2-naphthalenyl)ethynyl]benzoate (Compound 7) 80.0 mg (0.181 26 mmol) in 3 ml of EtOH and 2 ml of THF was added 48.0 mg (1.20 mmol, 27 1.20 ml) of NaOH (1.0 M aqueous solution). The solution was heated to 28 50 C for 2 hours, cooled to room temperature, and acidified with 10% HCI.
29 Extraction with EtOAc, followed by drying over Na2SO4, and removal of the 1 solvents under reduced pressure afforded the title compound as a colorless 2 solid. 1H NMR (DMSO): S 7.90 (2H, d, J = 8.3 Hz), 7.60 (2H, d, J = 8.3 3 Hz),7.51-7.48(4H,m),7.34(2H,d,J = 8.4Hz),6.97(1H,s),6.07(1H,t,J
4 = 4.5 Hz), 2.34 (2H, d, J = 4.6 Hz), 1.29 (6H, s).
4-[(5.6-Dihvdro-5.5-dimethy-1-8-(3-Midyl, -12-naphthalenyl)eth3wl]benzoic acid s (Compound 20 7 To a solution of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(3-pyridyl)-2-8 naphthalenyl)ethynyl]benzoate (Compound 11) 45.0 mg (0.110 mmol) in 3 ml 9 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 was heated to 50 C for 2 11 hours, cooled to room temperature, and acidified with 10% HCI. Extraction 12 with EtOAc, followed by drying over Na2SO4, and removal of the solvents 13 under reduced pressure afforded the title compound as a colorless solid. 1H
14 NMR (DMSO): 8 8.60 (1H, d, J = 4.6 Hz), 8.55 (1H, s), 7.90 (2H, d, J
8.3 Hz), 7.76 (1H, d, J = 7.5 Hz), 7.60 (2H, d, J = 8.3 Hz), 7.51-7.46 (3H, 16 m),6.94(1H,s),6.14(1H,t,J=4.5Hz),2.37(2H,d,J=4.5Hz),1.31(6H, 17 s).
18 4-[(5.6-Dihvdro-5.5-dimetlW-8-(2-methylphenyl)-2-19 naphthalenyll_,ethytlyl]benzoic acid (ComQ,ound 27) To a solution of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(2-21 methylphenyl)-2-naphthalenyl)ethynyl]- benzoate (Compound 3) 80.0 mg 22 (0.190 mmol) in 3 ml of EtOH and 2 ml of THF was added 60.0 mg (1.50 23 mmol, 1.50 ml) of NaOH (1.0 M aqueous solution). The solution was heated 24 to 50 C for 2 hours, cooled to room temperature, and acidified with 10%
HCI. Extraction with EtOAc, followed by drying over Na2SO4, and removal 26 of the solvents under reduced pressure afforded the title compound as a 27 colorless solid. 1H NMR (DMSO): 8 7.89(2H, d, J = 8.4 Hz), 7.57 (2H, d, J
28 = 8.4 Hz), 7.46 (2H, s), 7.29-7.14 (4H, m), 6.59 (1H, s), 5.90 (1H, t, J =
4.7 29 Hz), 2.39 (2H, m), 2.60 (3H, s), 1.39 (3H, s), 1.29 (3H, s).

1 4-[(5.6-Dihydro-5.5-dimethyl-3-hydro henyl)-2-2 naphthalenvl, ethvn,ti~l]benzoic acid (Compound 28) 3 To a solution of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(3-((2,2-4 dimethylethyl)- dimethylsiloxy)phenyl)-2-naphthalenyl)ethynyl]benzoate s(Compound H) 40.0 mg (0.076 mmol) in 3 ml of EtOH and 2 ml of THF was 6 added 40.0 mg (1.00 mmol, 1.00 ml) of NaOH (1.0 M aqueous solution).
7 The solution was heated to 50 C for 2 hours, cooled to room temperature, 8 and acidified with 10% HCI. Extraction with EtOAc, followed by drying over 9 NaZS04, and removal of the solvents under reduced pressure afforded the 1o title compound as a colorless solid. 1H NMR (d6-acetone): 8 7.90 (2H, d, J
11 = 8.3 Hz), 7.49 (2H, d, J = 8.4 Hz), 7.35 (2H, s), 7.15-7.07 (2H, m), 6.77-6.69 12 (3H, m), 5.92 (1H, t, J = 4.7 Hz), 2.25 (2H, d, J = 4.7 Hz), 1.23 (6H, s).
13 4-[(5.6-Dihydro-5.5-dimethyl-8-(4-hydroxypheMl)-2-14 naphthalenyl)eth3MLI]benzoic acid (Compound 29) ts To a solution of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(4-((2,2-16 dimethylethyl)- dirnethylsiloxy)phenyl)-2-naphthalenyl)ethynyl]benzoate 17 (Compound I) 75.0 mg (0.143 mmol) in 3 ml of EtOH and 2 ml of THF was 18 added 60.0 mg (1.50 mmol, 1.50 ml) of NaOH (1.0 M aqueous solution).
1s The solution was heated to 50 C for 2 hours, cooled to room temperature, 2o and acidified with 10% HCI. Extraction with EtOAc, followed by drying over 21 NaZSO4, and removal of the solvents under reduced pressure afforded the 22 title compound as a colorless solid. 1H NMR (d6-acetone): S 8.01 (2H, d, J
23 = 8.3 Hz), 7.59 (2H, d, J = 8.4 Hz), 7.45 (2H, s), 7.20-7.17 (3H, m), 6.92-6.89 24 (2H, m), 5.97 (1H, t, J = 4.7 Hz), 2.35 (2H, d, J = 4.7 Hz), 1.34 (6H, s).
25 4-[(5.6-Dihvdro-5.5-dimethyl-8-(5-methvlthiazol-2-yl1-2 _na htp halenyl)ethyl~yl]
26 benzoic acid (Compound 30) 27 To a solution of ethyl 4-[5,6-dihydro-5,5-dimethyl-8-(5-methylthiazol-2-28 yl)-2-naphthalenyl]ethynylbenzoate (Compound 15) (100 mg, 0.23 mmol) and 29 4 ml of EtOH at room temperature was added aqueous NaOH (1 ml, 1 M, 1 1 mmol). The resulting solution was warmed to 50 C for 1 hour and 2 concentrated in vacuo. The residue was suspended in a solution of CHzCIz 3 and ether (5:1) and acidified to pH 5 with 1M aqueous HCI. The layers were 4 separated and the organic layer was washed with brine, dried (Na2SO4), filtered and the solvents removed under reduced pressure to give the title 6 compound as a white solid. 1H NMR (d6-DMSO): 8 7.96 (1H, d, J = 1.7 7 Hz), 7.95 (2H, d, J = 8.0 Hz), 7.65 (2H, d, J = 8.0 Hz), 7.64 (1H, s), 7.53 8 (1H,dd,J=1.7,8.0Hz),7.46(1H,d,J=8.0Hz),6.59(1H,t,J=4.5Hz), 9 2.50 (3H, s), 2.39 (2H, d, J = 4.5 Hz), 1.27 (6H, s).
to 4-[(5.6-dihydro -5.5-dimethXl-8 f 2-thiazol}-l,1-2-nanhthaleMI)ethyr~yl~benzoic 11 acid (Compound 30a~
12 A solution of 33.9 mg (0.08 mmol) of ethyl 4-[(5,6-dihydro-5,5-13 dimethyl-8-(2-thiazolyl)-2- naphthalenyl)ethynyl]benzoate (Compound 15a) 14 and 8.5 mg (0.20 mmol) of LiOH-HZO in 3 ml of THF/water (3:1, v/v), was stirred overnight at room temperature. The reaction was quenched by the 16 addition of sat. aqueous NH4C1 and extracted with EtOAc. The combined 17 organic layers were washed with water and brine, dried over Na2SO4 and 18 concentrated in vacuo to give the title compound as a colorless solid. PMR
1s (d6-DMSO): S 1.29 (6H, s), 2.42 (2H, d, J = 4.6 Hz), 6.68 (1H, t, J= 4.6 2o Hz), 7.51 (2H, m), 7.62 (2H, d, J = 8.2 Hz), 7.77 (1H, d, J = 3.3 Hz), 7.93 21 (2H, d, J = 8.2 Hz), 7.98 (1H, d, J = 3.3 Hz).
22 4-((5.6-Dihydro-5.5-dimethyl-8-(4-methylthiazol-2 vl)-2-naphthalenyl)eth3mvl]
23 benzoic acid(Comoound 31) 24 To a solution of ethyl 4-[5,6-dihydro-5,5-dimethyl-8-(4-methylthiazol-2-yl)-2-naphthalenyl]ethynylbenzoate (Compound 16) (145.0 mg, 0.34 mmol) 26 and 4 ml of EtOH at room temperature was added aqueous NaOH (1 ml, 1 27 M, 1 mmol). The resulting solution was warmed to 50 C for 1 hour and 28 concentrated in vacuo. The residue was suspended in a solution of CH2C12 29 and ether (5:1) and acidified to pH 5 with 1M aqueous HCI. The layers were 1 separated and the organic layer was washed with brine, dried (Na2SO4), 2 filtered and the solvents removed under reduced pressure to give the title 3 compound as a white solid. 1H NMR (d6-DMSO): 8 7.94 (2H, d, J = 8.1 4 Hz), 7.87 (1H, d, J = 1.6Hz),7.63(2H,d,J=8.3Hz),7.50(1H,dd,J=
1.6, 8.1 Hz), 7.45 (1H, d, J = 8.1 Hz), 7.27 (1H, s), 6.58 (1H, t, J = 4.8 Hz), 6 2.43 (3H, s), 2.37 (2H, d, J = 4.8 Hz), 1.26 (6H, s).
7 4-[(5.6-DihXdro-5.5-dimethxl-8 -(4.5-dimethylthiazol-2 ;yl)-2-8 na,phthalenyl)ethvnyl] benzoic acid (Com , ound 32) 9 To a solution of ethyl 4-[5,6-dihydro-5,5-dimethyl-8-(4,5-1o dimethylthiazol-2-yl)-2-naphthalenyl]ethynylbenzoate (Compound 17) (58.0 ii mg, 0.13 mmol) and 4 ml of EtOH at room temperature was added aqueous 12 NaOH (1 ml, 1 M, 1 mmol). The resulting solution was warmed to 50 C for 1 13 hour and concentrated in vacuo. The residue was suspended in a solution of 14 CHZCI2 and ether (5:1) and acidified to pH 5 with 1M aqueous HCI. The layers were separated and the organic layer was washed with brine, dried 16 (Na2SO4), filtered and the solvents removed under reduced pressure to give 17 the title compound as a white solid. 1H NMR (d6-DMSO): 8 7.94 (2H, d, J
18 = 8.4 Hz), 7.86 (1H, d, J = 1.6 Hz), 7.61 (2H, d, J = 8.3 Hz), 7.50 (1H, dd, J
19 = 1.6, 8.0 Hz), 7.45 (1H,d,J = 8.0Hz),6.51 (1H,t,J = 4.9 Hz), 2.37 (3H, s), 2.36 (2H, d, J = 4.6 Hz), 2.32 (3H, s), 1.26 (6H, s).
21 Ethvl 4-[(5.6-dihydro-5.5-dimethv5-methyl-2-thienyl, -) 2-naphthalenYl) 22 ethynyl] benzoate (Compound 331 23 Employing the same general procedure as for the preparation of ethyl 24 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-2s naphthalenyl)ethynyl]benzoate (Compound 1), 170.0 mg (0.366 mmol) of 26 ethyl4-[(5,6-dihydro-5,5-d'unethyl-8-(trifluoromethylsulfonyl)oxy-2-27 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title 28 compound (colorless solid) using 202.0 mg (1.48 mmol) of zinc chloride, 24 29 mg (0.022 mmol) of tetrakis(triphenylphosphine)palladium(0) in 2.0 ml of 1 THF, and 5-methyl-2-lithiothiophene (prepared by adding 58.6 mg (0.36 ml, 2 0.915 mmol) of n- butyllithium (2.5 M solution in hexanes) to a cold solution 3(-78 C) of 89.8 mg (0.915 mmol) of 2-methylthiophene in 2.0 ml of THF).
4 1H NMR (CDC13): S 8.00 (2H, d, J 8.3 Hz), 7.59 (1H, d, J = 1.7 Hz), 7.55 (2H, d, J = 8.2 Hz), 7.43 (1H, dd, J 1.7, 8.0 Hz), 7.35 (1H, d, J = 8.0 Hz), e 6.87(1H,d,J=3.5Hz),6.74(1H,d,J=2.8Hz),6.15(1H,t,J=4.8Hz), 7 4.38 (2H, q, J = 7.1 Hz), 2.52 (3H, s), 2.32 (2H, d, J = 4.8 Hz). 1.40 (3H, t, 8 7.1 Hz), 1.32 (6H, s).
9 Ethvl4[(5.6-dih,ydro-5.5-dimeth,yl-8-(2-thienyl)-2-1o naphthalen,~-1)eth3m,yl]benz oate (Compound 33a) 11 Employing the same general procedure as for the preparation of ethyl 12 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-13 naphthalenyl)ethynyl]benzoate (Compound 1), 250.0 mg (0.52 mmol) of ethyl 14 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title 1 e compound (colorless solid) using 186.8 mg (1.37 mmol) of zinc chloride 37.1 17 mg (0.03 mmol) of tetrakis(triphenylphosphine)palladium(0) and 2-1 s lithiothiophene (prepared by the addition of 65.9 mg (0.69 ml, 1.03 mmol) of 1s n-butyllithium (1.5M solution in hexane) to a cold solution (-78 oC) of 86.5 2o mg (1.03 mmol) of thiophene in 1.0 ml of THF). PMR (CDC13): 8 1.33 (6H, 21 s), 1.36(3H,t,J = 7.1 Hz), 2.38 (2H, d, J = 4.7 Hz), 4.34 (2H, q, J = 7.2 22 Hz), 6.25 (1H, t, J = 4.7 Hz), 7.13 ( 2H, m), 7.47 (4H, m), 7.62 ( 2H, d, J
23 8.5 Hz), 8.00 (2H, d, J = 8.5 Hz).
24 4-[(5.6-Dihydro-5.5-dimethyLI-8-(5-methyl-2-thienyl)-2-naphthalen,yll_, ethywl benzoic acid (Compound 34) 26 To a solution of ethyl 4-[5,6-dihydro-5,5-dimethyl-8-(5-methyl-2-27 thienyl)-2-naphthalenyl]ethyliylbenzoate (Compound 33) (35.0 mg, 0.082 28 mmol) in 2 ml of EtOH and 1 ml THF at room temperature was added 29 aqueous NaOH (1 ml, 1 M, 1 mmol). The resulting solution was stirred at I room temperature overnight and then acidified with 10% HCI. Extraction 2 with EtOAc, followed by drying over Na2SO4, and removal of the solvents 3 under reduced pressure afforded the title compound as a colorless solid. 1H
4 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 = 4.7 Hz), 2.49 (3H, s), 2.35 e(2H, d, J = 4.7 Hz), 1.32 (6H, s).
7 4-((5.6-dihydro-5.5-dimethyl-8-(2-thienvl)-2-na ht~ halenyl)ethynXllbenzoic acid 8 (Compound 34a) 9 Employing the same general procedure as for the preparation of 4-io [(5,6-dihydro-5,5-dimethyl-8-(2- thiazolyl)-2-naphthalenyl)ethynyl]benzoic acid 11 (Compound 30a), 70.0 mg (0.17 mmol) of ethyl 4-[(5,6- dihydro-5,5-12 dimethyl-8-(2-thienyl)-2- naphthalenyl)ethynyl]benzoate (Compound 33a) was 13 converted into the title compound (colorless solid) using 17.8 mg (0.42 mmol) 14 of LiOH in H20. PMR (d6-DMSO): 8 1.27 (6H, s), 2.33 (2H, d, J = 4.9 Hz), 6.23 (1H, t, J =4.9Hz),7.14(2H,m),7.38-7.56(4H,m),7.61(2H,d,J=
16 8.3 Hz), 7.92 (2H, d, J = 8.3Hz).
17 5.6-Dihydro-5.5-dimethyl-8-(4-methxlphenyl)-2-naphthalenecarboxylic acid 18 (Compound K) 19 A solution of 3,4-dihydro-i-(4-methylphenyl)-4,4- dimethyl-7-2o bromonaphthalene (Compound D) (250.0 mg, 0.764 mmol) in 2.0 ml of THF
21 was cooled to -78 C and 1.0 ml of t-butyllithium (1.68 mmol, 1.7 M solution 22 in pentane) was added slowly. After stirring for 1 hour at -78 C gaseous 23 (generated by evaporation of Dry-Ice, and passed though a drying tube) was 24 bubbled through the reaction for 1 hour. The solution was then allowed to warm to room temperature and the reaction was quenched by the addition of 26 10% HCI. Extraction with EtOAc was followed by washing the combined 27 organic layers with HZO and saturated aqueous NaCl, and drying over 28 MgSO4. Removal of the solvents under reduced pressure and washing of the 29 solid with hexanes afforded the title compound as a colorless solid. 1H NMR

1 (CDC13):S 7.94(1H,dd,J= 1.8, 8.1 Hz), 7.76 (1H, d, J = 1.8 Hz), 7.45 2(1H, d, J = 8.1 Hz), 7.24 (4H, m), 6.01 (1H, t, J = 4.7 Hz), 2.40 (3H, s), 2.36 3 (2H, d, J= 4.7 Hz), 1.35 (6H, s).
4 Ethyl 4-[[j5.6-dihydro-5.5-dimethy1=84-methy,~nhenvl_, 1-2-naQhthalenxl)carbonyl]a_, mino]-benzoate (Compound 35) 6 A solution of 170.0 mg (0.58 mmol) 5,6-dihydro-5,5-dimethyl-8-(4-7 methylphenyl)-2-naphthalenecarboxylic acid (Compound K) 115.0 mg (0.70 8 mmol) of ethyl 4-aminobenzoate, 145.0 mg (0.76 mmol) of 1-(3-9 dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, and 92.4 mg (0.76 1o mmol) of 4-dimethylaminopyridine in 6.0 ml of DMF was stirred overnight at 11 room temperature. Ethyl acetate was added and the resulting solution 12 washed with H20, saturated aqueous NaHCO3, and saturated aqueous NaCl, 13 then dried over MgSO4. After removal of the solvent under reduced 14 pressure, the product was isolated as a colorless solid by column chromatography (10 to 15% EtOAc/hexanes). 1H NMR (CDC13): S 8.02 16 (2H,d,J=8.7Hz),7.72(2H,m),7.65(2H,d,J=8.7Hz),7.52(1H,d,J=
17 1.8Hz),7.48(1H,d,J=8.0Hz),7.25(4H,m),6.15(1H,t,J=4.9Hz),4.36 18 (2H, q, J = 7.1 Hz), 2.40 (3H, s), 2.38 (2H, d, J = 4.9 Hz), 1.39 (3H, t, J
19 7.1 Hz), 1.37 (6H, s).
2o 4-[[ 5.( 6-DihAro -5.5-dimeth,yl-8-(4-methylphenyl)^
21 na hthalenyllca, rbonXl]amino]-benzoic acid (Compound 36) 22 To a solution of 26.5 mg (0.06 mmol) ethyl 4[[(5,6-dihydro-5,5-23 dimethyl-8-(4-methylphenyl)-2- naphthalenyl)carbonyl]amino]-benzoate 24 (Compound 35) in 3.0 ml EtOH and 4.0 ml of THF was added 240.1 mg NaOH (6.00 mmol, 3.0 ml of a 2M aqueous solution). After stirring at room 26 temperature for 72 hours, the reaction was quenched by the addition of 10%
27 HCI. Extraction with EtOAc, and drying of the organic layers over MgSO4, 28 provided a solid after removal of the solvent under reduced pressure.
29 Crystallization from CH3CN afforded the title compound as a colorless solid.

1 1H NMR (d6-DMSO): 8 10.4 (1H, s), 7.91-7.81 (5H, m), 7.54 (1H, d, J
2 8.1 Hz), 7.45 (1H, d, J = 1.7 Hz), 7.23 (4H, s), 6.04 (1H, t, J = 4.7 Hz), 2.35 3 (5H, s), 1.33 (6H, s).
4 Ethvl 4-([(5.6-dihvdro-5 5-dimethyl-8-(4-methyl-phenvl)-2-s naphthalenyllcarboW oxy]-benzoate (Compound 37) 6 A solution of 25.0 mg (0.086 mmol) 5,6-dihydro-5,5-dimethyl-8-(4-7 methylphenyl)-2-naphthalenecarboxylic acid (Compound K) 17.5 mg (0.103 8 mmol) of ethyl 4-hydroxybenzoate, 21.4 mg (0.112 mmol) of 1-(3-9 dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, and 12.6 mg (0.103 io mmol) of 4-dimethylaminopyridine'in 2.0 ml of DMF was stirred overnight at 11 room temperature. Ethyl acetate was added and the resulting solution 12 washed with H20, saturated aqueous NaHCO3, and saturated aqueous NaC1, 13 before being dried over MgSO4. After removal of the solvent under reduced 14 pressure, the product was isolated by column chromatography as a pale-15 yellow solid (10% EtOAc/hexanes). 1H NMR (CDC13): 8 8.08 (2H, d, J
16 8.1 Hz), 8.05 (1H, dd, J = 1.8, 8.1 Hz), 7.89 (1H, d, J = 1.8 Hz), 7.50 (2H, d, 17 J=8.1Hz),7.22(5H,m),6.05(1H,t,J=4.7Hz),4.37(2H,q,J=7.1Hz), ie 2.39 (2H, d, J = 4.7 Hz), 2.38 (3H, s), 1.39 (3H, t, J = 7.1 Hz), 1.37 (6H, s).
19 2-Trimethylsilvlethvl4-[[(5 6-dihydro-5.5-dimethyl-8- 4-methvlphenyl)-2-2o naphthalenti)carbonyl]oxy]-benzoate (Compound 381 21 A solution of 93.5 mg (0.320 mmol) 5,6-dihydro-5,5-dimethyl-8-(4-22 methylphenyl)-2-naphthalenecarboxylic acid (Compound K) 76.0 mg (0.319 23 mmol) of 2-trimethylsilylethyl-4-hydroxybenzoate, 80.0 mg (0.417 mmol) of 1-24 (3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, and 51.0 mg 25 (0.417 mmol) of 4-dimethylaminopyridine in 4.0 ml of DMF was stirred 28 overnight at room temperature. Ethyl acetate was added and the resulting 27 solution washed with H20, saturated aqueous NaHCO3, and saturated 28 aqueous NaCI, before being dried over MgSO4. After removal of the solvent 29 under reduced pressure, the product was isolated as a colorless solid by 1 column chromatography (5% EtOAc/hexanes). 1H NMR (CDC13): 8 8.08 2 (2H, d, J = 8.8 Hz), 8.05 (1H, dd, J = 1.8, 8.1 Hz), 7.50 (1H, d, J = 8.1 Hz), 3 7.26- 7.18 (6H, m), 6.05 (1H, t, J = 4,7 Hz), 4.42 (2H, t, J = 8.4 Hz), 2.40 4 (2H, d, J = 4.7 Hz), 2.39 (3H, s), 1.38 (6H, s), 0.09 (9H, s).
4-([(5.6-Dihvdro-5.5-dimethyl-8-(4-meth,_phen~)-2-e nanh_.a thalen~!1_)carbonylloxy]-benzoic acid(Comvou,) 7 A solution of 110.0 mg (0.213 mmol) 2- tritnethylsilylethyl 4[[(5,6-8 dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-naphthalenyl)carbonyl]oxy)-9 benzoate (Compound 38) and 167.3 mg of tetrabutylammonium flouride 1 o (0.640 mmol, 0.64 ml of a 1M solution in THF) in 2.0 ml THF was stirred at 11 room temperature for 22 hours. Ethyl acetate was added and the resulting 12 solution washed with H20 and saturated aqueous NaCl then dried over 13 MgSO4. Removal of the solvents under reduced pressure and washing of the 14 residual solid with EtOAc and CH3CN afforded the title compound as a colorless solid. 1H NMR (d6-acetone): 8 8.10 (2H, d, J = 8.8 Hz), 8.06 (1H, 1s dd,J=2.0,8.1Hz),7.82(1H,d,J= 1.9Hz),7.64(1H,d,J=8.1Hz),7.35 17 (2H, d, J = 8.6 Hz), 7.25 (4H, m), 6.08 (1H, t, J = 4.7 Hz), 2.42 (2H, d, J
18 4.7 Hz), 2.35 (3H, s), 1.39 (6H, s).
19 thv12-fluoro-4-[[(5.6-dihvdro-5.5-dimethyl-8-(4-metlkylhenyl)-2-2o naphthalenvl)carbonyl]amino]-benzoate (Compound 40), 21 A solution of 115.0 mg (0.41 mmol) 5,6-dihydro-5,5-dimethyl-8-(4-22 methylphenyl)-2-naphthalenecarboxylic acid (Compound K) 89.0 mg (0.49 23 mmol) of ethyl 2-fluoro-4-aminobenzoate, 102.0 mg (0.53 mmol) of 1-(3-24 dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, and 65.0 mg (0.53 mmol) of 4-dimethylaminopyridine in 5.0 ml of DMF was stirred at 50 C for 26 1 hour and then overnight at room temperature. Ethyl acetate was added 27 and the resulting solution washed with H20, saturated aqueous NaHCO3, and 28 saturated aqueous NaCI, before being dried over MgSO4. After removal of 29 the solvent under reduced pressure, the product was isolated as a colorless 1 solid by column chromatography (20% EtOAc/hexanes). 1H NMR (CDC13):
2 8 7.96(1H,s),7.89(1H,t,J=8.4Hz),7.70(2H,m),7.52(1H,d,J= 1.9 3 Hz), 7.45 (1H, d, J = 8.1 Hz), 7.23 (5H, m), 6.04 (1H, t, J = 4.8 Hz), 4.36 4 (2H, q, J = 7.1 Hz), 2.38 (3H, s), 2.35 (2H, d, J = 4.8 Hz), 1.39 (3H, t, J
7.1 Hz), 1.36 (6H, s).
6 2-Fluoro-4-[[(5.6-dihydro-5.5-dimethyl-8-(4-methv_lphenyl)-2-7 naRhthalenyl)cT arbonyl]amino]-benzoic acid (Compound 41) 8 To a solution of 41.6 mg (0.091 mmol) ethyl 2- fluoro-4-[[(5,6-9 dihydro-5,5-dimethyl-8- (4-methylphenyl)-2-naphthalenyl)carbonyl]amino]-1 o benzoate (Compound 40) in 2.0 ml EtOH and 2.0 ml of THF was added 40.0 11 mg NaOH (1.00 mmol, 1.0 ml of a 1 M aqueous solution). After stirring at 12 room temperature for overnight, the reaction was quenched by the addition 13 of 10% HC1. Extraction with EtOAc, and drying of the organic layers over 14 MgSO41 provided a solid after removal of the solvent under reduced pressure.
Crystallization from CH3CN afforded the title compound as a pale-yellow 16 solid. 1H NMR (d6-acetone): 8 9.84 (1H, s), 7.94-7.83 (3H, m), 7.64 (1H, 17 dd, J = 2.0 Hz), 7.53 (2H, d, J = 8.1 Hz), 7.23 (4H, s), 6.04 (1H, t, J =
4.7 18 Hz), 2.38 (2H, d, J = 4.7 Hz), 2.36 (3H, s), 1.35 (6H, s).
19 Ethy14[[(5.6-dihydro-5.5-dimethv4-methylphenyl)- 2-2o naRhthalenyl)thiocarbon, l]amino]-benzoate (Comvou,nd 42) 21 A solution of 110.0 mg (0.25 mmol) ethyl4-[[(5,6-dihydro-5,5-22 dimethyl-8-(4-methylphenyl)- 2-naphthalenyl)carbonyl]amino]-benzoate 23 (Compound 35) and 121.0 mg (0.30 mmol) of [2,4-bis(4-methoxyphenyl)- 1,3-24 dithia-2,4-diphosphetane-2,4-disulfide] (Lawesson's Reagent) in 12.0 ml of benzene was refluxed overnight. Upon cooling to room temperature, the 26 mixture was filtered and the filtrate concentrated under reduced pressure.
27 The title compound was isolated by column chromatography (10 to 25%
2s EtOAc / hexanes) as a yellow solid. 1H NMR (CDC13): 8 8.92 (1H, s), 8.06 29 (2H, t, J = 8.5 Hz), 7.88-7.70 (3H, m), 7.42 (2H, d, J= 8.1 Hz), 7.18 (4H, ~ m),6.03(1H,t,J=4.7Hz),4.37(2H,q,J=7.1Hz),2.38(3H,s),2,36(2H, 2 d, J = 4.7 Hz), 1.56 (3H, t, J = 7.1 Hz), 1.35 (6H, s).
3 4-[[(5.6-Dihydro-5 5-dimethyl-8-(4-methyll2henyl):2-4 nanhthalenyllthiocarbonA]amino]-benzoic acid (Compound 43) To a solution of 84.0 mg (0.184 mmol) ethyl 4[[(5,6-dihydro-5,5-6 dimethyl-8-(4-methylphenyl)-2- naphthalenyl)thiocarbonyl)aminoj-benzoate 7 (Compound 42) in 2.0 ml EtOH and 2.0 ml of THF was added 60.0 mg 8 NaOH (1.50 mmol, 1.5 ml of a 1 M aqueous solution). After stirring at room 9 temperature overnight, the reaction was quenched by the addition of 10%
t o HCI. Extraction with EtOAc, and drying of the organic layers over MgSO4, ii provided a solid after removal of the solvent under reduced pressure.
12 Crystallization from CH3CN afforded the title compound as a yellow solid.
13 1H NMR (d6-acetone): 8 10.96 (1H, s), 8.05 (4H, m), 7.72 (1H, dd, J = 2.0, 14 8.0Hz),7.54(1H,s),7.46(1H,d,J=8.1Hz),7.20(4H,m),6.04(1H,t,J=
4.7 Hz), 2.38 (2H, d, J = 4.7 Hz), 2.33 (3H, s), 1.35 (6H, s).
le 2-acetyl-6-bromonaphtha lene ,l,Com pound L) 17 To a cold (10 C) mixture of 44.0 g (0.212 mol) of 2-bromonaphthalene 18 and 34.0 g (0.255 mol) of aluminum chloride in 400 ml of nitrobenzene was ts added 21.0 g (267 mmol) of acetyl chloride. The mechanically stirred reaction mixture was warmed to room temperature, and heated to 40 C for 21 18 hours. After cooling to 0 C in an ice bath, the reaction was quenched by 22 the addition of 12M HCl (70 ml). The layers were separated and the organic 23 phase was washed with water and dilute aqueous NaZCO3. Kugelrohr 24 distillation, followed by recrystallization from 10% EtOAc-hexane yielded g of the title compound as a tan solid. iH NMR (CDC13): 8 8.44 (1H, br s), 26 8.04-8.10 (2H, m), 7.85 (IH, d, J = 8.5 Hz), 7.82 (1H, d, J = 8.8 Hz), 7.64 27 (1H, d, J = 8.8 Hz), 2.73 (3H, s).
2s 6-bromo-2-naphthalenecarboxylic acid (Compound M~
29 To a solution of sodium hypochlorite (62 ml, 5.25% in water (w/w), 3.6 1 g, 48.18 mmol) and sodium hydroxide (6.4 g, 160.6 mmol) in 50 ml of water 2 was added a solution of 2-acetyl-6-bromonaphthalene (Compound L) 4 g, 3 (16.06 mmol) in 50 ml of 1,4-dioxane. The yellow solution was heated to 4 70 C in an oil bath for 2 hours, cooled to ambient temperature, and extracted with ethyl ether (2 x 50 ml). The aqueous layers were diluted with NaHSO3 6 solution (until KI indicator solution remained colorless) and then acidified 7 (pH <2) with 1N sulfuric acid to give a white precipitate. The mixture was e extracted with ethyl ether, and the combined organic phase washed with 9 saturated aqueous NaCl, dried (MgSOd) and concentrated to give 3.54 g to (88%) of the title compound as a solid. 1H NMR (DMSO-d6): S 8.63 (1H, 11 br s), 8.32 (1H, d, J = 2.0 Hz), 8.10 (1H, d, J = 8.8 Hz), 8.00-8.05 (2H, m), 12 7.74 (1H, dd, J = 2.0, 8.8 Hz).
13 Ethv16-bromo-2-naphthalenecarbo ate (Compound N) 14 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 mmol) was added 18M
is sulfuric acid (2 ml). The solution was refluxed for 30 minutes, cooled to 17 room temperature, and the reaction mixture partitioned between pentane 18 (100 ml) and water (100 ml). The aqueous phase was extracted with pentane is (100 ml) and the combined organic layers washed with saturated aqueous NaCl (100 ml), dried (MgSO4), and concentrated to yield an off-white solid.
21 Purification by flash chromatography (silica, 10% EtOAc-hexane) afforded 22 the title compound as a white solid. 1H NMR (CDC13): 8 8.58 (1H, br s), 23 8. 10 (1H, dd, J = 1.7, 9 Hz), 8.06 (1H, d, J = 2 Hz), 7.83 (1H, d, J = 9 Hz), 24 7.80 (1H, d, J = 9 Hz), 7.62 (1H, dd, J = 2, 9 Hz).
EthyLl (E)-4-[2 (5.6.7.8-tetrahvdro-5.5-dimethvl-8-oxo-2-naphthalenYle~yll-2e benzoate (Compound 0) 27 To a solution of 520.0 mg (2.00 mmol) of 3,4- dihydro-4,4-dimethyl-7-28 bromo-1(2H)-naphthalenone (Compound B) and 510.0 mg (2.90 mmol) of 29 ethyl 4- vinylbenzoate in 4.0 ml of triethylamine (degassed by sparging with 1 argon for 25 minutes), was added 124.0 mg (0.40 mmol) of tris(2-2 methylphenyl) phosphine, followed by 44.0 mg (0.20 mmol) of 3 palladium(II)acetate. The resulting solution was heated to 95 C for 2.5 4 hours, cooled to room temperature, and concentrated under reduced pressure. Purification by column chromatography (10% EtOAc / hexanes) 6 afforded the title compound as a colorless solid. 1H NMR (CDC13): 8 8.19 7 (1H,d,J=2.0Hz),8.03(2H,d,J=8.4Hz),7.69(1H,dd,J=2.0,8.2Hz), 8 7.57 (2H, d, J = 8.4 Hz), 7.45 (1H, d, J = 8.2 Hz), 7.20 (2H, s), 4.39 (2H, q, 9 J=7.1Hz),2.76(2H,t,J=6.5Hz),2.04(2H,t,J=6.5Hz),1.41(3H,t,J
1 o = 7.1 Hz, and 6H, s).
11 Ethy-I (E)-4-[2-(5 6-dihvdro-5 5-d'nnethyl-8-(trifluorometh ls~fon, lv )oxyz2-12 naphthalenvl)ethenxll- benzoate (Compound P) 13 To a cold (-78 C) solution of 440.0 mg (2.40 mmol) of sodium 14 bis(trimethylsilyl)amide in 10.0 ml of THF was added 700.0 mg (2.00 mmol) i s of ethyl (E)-4-[2- (5,6,7,8-tetrahydro-5,5-dimethyl-8-oxo-2-1 s naphthalenyl)ethenyl]-benzoate (Compound 0) as a solution in 25.0 ml of 17 THF. After stirring at -78 C for 1.5 hours, 960.0 mg (2.40 mmol) of 2[N,N-18 bis(trifluoromethylsulfonyl)amino]-5-chloropyridine was added in one portion.
19 After 30 minutes the solution was warmed to 0 C and stirred for 3 hours.
2o The reaction was quenched by the addition of saturated aqueous NH4C1, and 21 extracted with EtOAc. The combined extracts were washed with 5% aqueous 22 NaOH, dried (Na2SO4)1 and the solvents removed under reduced pressure.
23 The title compound was isolated as a colorless solid by column 24 chromatography (7% EtOAc / hexanes). 1H NMR (CDC13): 8 8.04 (1H, d, J
25 =8.4Hz),7.57(2H,d,J=8.4Hz),7.52(1H,s),7.49(1H,d,J=8.0Hz), 26 7.33(1H,d,J=8.0Hz),7.20(1H,d,J= 16.4Hz),7.10(1H,d,J= 16.4 27 Hz),6.00(1H,t,J=4.9Hz),4.39(2H,q,J=7.1Hz),2.43(2H,d,J=4.9 28 Hz), 1.41 (3H, t, J = 7.1 Hz), 1.32 (6H, s).
29 Ethy-I (El-,,4-(2 (5.6-di~dro-5.5-d'unethyl-8-(4- methvlAhenv1)-2-WO 99/33821 PCr/US98/27314 I naphthalentil,, ethenyl]-benzoate (Compound 44) 2 A solution of 4-lithiotoluene was prepared at -78 C by the addition of 3 130.7 mg of t-butyllithium (2.04 mmol; 1.20 ml of a 1.7M solution in pentane) 4 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 mg (2.30 mmol) of ZnCl2 in 2.0 ml of 6 THF was added. The resulting solution was warmed to room temperature, 7 stirred for 1.25 hour and then added via canula to a solution of 285.0 mg 8 (0.590 mmol) of ethyl (E)-4-[2- (5,6-dihydro-5,5-dimethyl-8-9 (trifluoromethylsulfonyl)oxy-2-naphthalenyl)ethenyl]- benzoate (Compound P) io and 29.0 mg (0.025 mmol) of tetrakis(triphenylphosphine)palladium(0) in 2.0 11 ml of THF. The resulting solution was stirred at room temperature for 1 12 hour and then at 55 C for 2 hours. Upon cooling to room temperature the 13 reaction was quenched by the addition of saturated aqueous NH4C1. The 14 mixture was extracted with EtOAc, and the combined extracts were washed with 5% aqueous NaOH, saturated aqueous NaCl, and dried over Na2SO4 ie before being concentrated under reduced pressure. The title compound was 17 isolated by column chromatography (10% EtOAC / hexanes) as a colorless 18 solid. 1H NMR (CDC13): 8 7.96 (2H, d, J = 8.1 Hz), 7.47 (2H, d, J = 8.1 ta Hz), 7.43-7.16 (7H, m), 7.07 (1H, d, J = 16.3 Hz), 6.93 (1H, d, J = 16.3 Hz), 2o 5.97 (1H, t, J = 4.7 Hz), 4.39 (2H, q, J = 7.0 Hz), 2.41 (3H,s),2.33(1H,d,J
21 = 4.7 Hz), 1.38 (3H, t, J= 7.0 Hz), 1.33 (6H, s).
22 (El-,4-j2-(5.6-Dihydro-5.5-dimethy-8-(4-methvlphenyl,)-2-2s naphthalejW)ethenvl 1 benzoic acid Compound 45 24 To a solution of 65.0 mg (0.190 mmol) of ethyl (E)-4-[2-(5,6-dihydro-5,5-dimethyl-8-(4-methylphenyl)- 2-naphthalenyl)ethenyl]-benzoate 26 (Compound 44) in 4.0 ml of THF was added 30.0 mg of LiOH (0.909 mmol, 27 1.0 ml of a 1.1M solution) and 1.0 ml of MeOH. The solution was heated to 2s 55 C for 3 hours, cooled to room temperature, and concentrated under 29 reduced pressure. The residue was dissolved in HZO and extracted with 1 hexanes. The aqueous layer was acidified to pH 1 with 10% HCI, and 2 extracted with Et20. The combined organic layers were washed with 3 saturated aqueous NaCI, diluted with EtOAc to give a clear solution, and 4 dried over Na2SO4. The solvents were removed under reduced pressure to give the title compound as a colorless solid. 1H NMR (d6-DMSO): S 7.86 6 (2H, d, J = 8.4 Hz), 7.66 (2H, d, J = 8.4 Hz), 7.58 (1H, dd, J = 1.7, 8.1 Hz), 7 7.41 (1H, d, J = 8.1 Hz), 7.28 (1H, d, J= 16.5 Hz), 7.23 (4H, s), 7.08 (1H, d, s J = 1.7 Hz), 7.07 (1H, d, J = 16.5 Hz), 5.97 (1H, t, J = 4.6 Hz), 2.35 (3H, s), 9 2.31 (1H, d, J = 4.6 Hz), 1.29 (6H, s).
1o Ethyl 4-[2-,1,1-dimethy-l-3-(4-methylphenyl)-5-indenyl, ethynA]benzoate 11 (Compound 4 12 A solution of 32.0 mg (0.187 mmol) of 4- bromotoluene in 1.0 ml THF
13 was cooled to -78 C and 24.0 mg of t-butyllithium (0.375 mmol, 0.22 ml of a 14 1.7 M solution in pentane) was slowly added. The yellow solution was stirred for 30 minutes at which time 29.8 mg (0.219 mmol) of ZnC12 was added as a 16 solution in 1.0 ml THF. The resulting solution was warmed to room 17 temperature and after 30 minutes added to a second flask containing 29.0 mg 1a (0.062 mmol) of ethyl 4-[2- (1,1-d'unethyl-3-(trifluoromethylsulfonyl)oxy-5-1 s indenyl) ethynyl)benzoate (Compound FF) and 2.9 mg (0.003 mmol) of 2o tetrakis(triphenylphosphine)palladium (0) in 1.0 ml THF. The resulting 21 solution was warmed to 50 C for 1 hour and then stirred at room 22 temperature for 4 hours. The reaction was quenched by the addition of 23 saturated aqueous NH4C1, and then extracted with Et20. The combined 24 organic layers were washed with water, saturated aqueous NaCI, and dried over MgSO4 before being concentrated under reduced pressure. The title 26 compound was isolated as a colorless oil by column chromatography (10%
27 Et20 / hexanes). 1H NMR (300 MHz, CDC13): 8 8.03 (2H, d, J = 8.5 Hz), 28 7.66 (1H, s), 7.58 (2H, d, J = 8.5 Hz), 7.50 (2H, d, J = 8.0 Hz), 7.46 (1H, d, 29 J = 7.9 Hz), 7.38 (1H, d, J= 7.7 Hz), 7.28 (2H, d, J = 9 Hz), 6.43 (1H, s), 1 4.40 (2H, q, J = 7.2 Hz), 2.43 (3H, s), 1.41 (3H, t; + 6H, s).
2 4-[2-(1.1-dimethvl-3-t4-methvl henyl, -5- indenkl)ethynyl]benzoic acid 3 (C_ ompognd 48) 4 To a solution of 10.0 mg (0.025 mmol) of ethyl 4- [2-(1,1-dimethyl-3-(4-methylphenyl)-5- indenyl)ethynyl)benzoate (Compound 47) in 0.5 ml 6 THF/H20 (3:1 v/v) was added 5.2 mg (0:12 mmol) LiOH H20. After stirring 7 at room temperature for 48 hours the solution was extracted with hexanes e and the aqueous layer was acidified with saturated aqueous NH4C1. Solid s NaCI was added and the resulting mixture extracted with EtOAc. The combined organic layers were dried (Na2SO4) and concentrated under 11 reduced pressure to give the title compound as a colorless solid. 1H NMR
12 (300 MHz, d6-DMSO): 8 7.95 (2H, d, J = 8.3 Hz), 7.65 (2H, d, J = 8.3 Hz), 13 7.57 (2H, m), 7.49 (3H, m), 7.30 (2H, d, J = 7.9 Hz), 6.61 (1H, s), 2.36 (3H, 14 s), 1.36 (6H, s).
L{4-bromothiopheno~,cyloropionic acid ts To a solution of 1.44 g (35.7 mmol) of NaOH in 20.0 ml degassed H20 17 (sparged with argon) was added 6.79 g (35.7 mmol) of 4-bromothiophenol.
18 The resulting mixture was stirred at room temperature for 30 minutes. A
19 second flask was charged with 2.26 g (16.3 mmol) of K2C03 and 15 ml of degassed H20. To this solution was added (in portions) 5.00 g (32.7 mmol) of 21 3- bromopropionic acid. The resulting potassium carboxylate solution was 22 added to the sodium thiolate solution, and the resulting mixture stirred at 23 room temperature for 48 hours. The mixture was filtered and the filtrate 24 extracted with benzene, and the combined organic layers were dicarded. The aqueous layer was acidified with 10% HCl and extracted with EtOAc. The 26 combined organic layers were washed with saturated aqueous NaCI, dried 27 over MgSO4, and concentrated under reduced pressure. The resulting solid 2s was recrystallized from EtzO - hexanes to give the title compound as off-29 white crystals. 1H NMR (CDC13): 8 7.43 (2H, d, J= 8.4 Hz), 7.25 (2H, d, J

1 = 8.4Hz),3.15(2H,t,J=7.3Hz),2.68(2H,t,J =7.3Hz).
2 2.3-di ydro-6-bromo -(410=1-benzothiopyran-4-one 3 A solution of 3.63 g (13.9 mmol) of 3-(4- bromothiophenoxy)propionic 4 acid in 60 ml methanesulfonic acid was heated to 75 C for 1.5 hours. After cooling to room temperature the solution was diluted with H20 and extracted a with EtOAc. The combined organic layers were washed with 2N aqueous 7 NaOH, H20, and saturated aqueous NaC3 and then dried over MgSO4.
a Removal of the solvent under reduced pressure afforded a yellow solid from e which the product was isolated by column chromatography (3% EtOAc-io hexanes) as a pale-yellow solid. 1H NMR (CDC1,): S 8.22 (iH, d, J = 2.1 ii Hz), 7.48 1H, dd, J = 2.1,8.3 Hz), 7.17 (1H, d; J = 8.5 Hz), 3.24 (2H, t, J=
12 6.4Hz),2.98(2H,t,J =6.7Hz).
13 2 3-dihvdr2-trimet yjsiWethvnvll-(4H1-1- nzothiopylgil-4-one i4 A solution of 1.00 g (4.11 mmol) 2,3-dihydro-6- bromo-(4H)-1-benzothiopyran-4-one and 78.3 mg (0.41 mmol) CuI in 15.0 ml THF and 6.0 is ml Et2NH was sparged with argon for 5 minutes. To this solution was added 17 2.0 ml (1.39 g,14.2 mmol) of (trimethylsilyl)acetylene foUowed by 288.5 mg is (0.41 mmol) of bis(triphenylphosphine)palladium(II) chloride. The resulting ts dark solution was stirred at. room temperature for 3 days and then filtered 2o through a pad of Celite, which was washed with EtOAc. The filtrate was 2i washed with H20 and saturated aqueous NaCi before being dried over 22 MgSO4. The title compound was isolated as an orange oil by column 2s chromatography (4% EtOAc - hezanes). 1H NMR (CDC13): S 8.13 (114, d, J
24 = 1.9Hz),7.36(1H,dd,J=2.1,8.2Hz),7.14(1H,d,J = 8.2 Hz), 3.19 (2H, 26 d, J = 63 Hz), 2.91 (2H, d, J= 6.3 Hz), 0.21(9H, s).
za v A solution containing 600.0 mg (2.25 mmol) of 2,3- dihydro-6-(2-2a trimethylsilylethynyl)-(4H)-1- benzothiopyran-4-one and 100.0 mg (0.72 29 mmol) IG2C703 in 15 ml MeOH was stirred at room temperature for 20 hours.
* Trade-mark 1 The solution was diluted with H20 and extracted with Et20. The combined 2 organic layers were washed with H20 and saturated aqueous NaCI before 3 being dried over MgSO4. Removal of the solvents under reduced preesure 4 afforded the title compound as an orange solid. 1H NMR (CDC13): 8 8.17 s (1H,d,J=1.8Hz),7.40(1H,dd,J=1.8,8.2Hz),7.19(1H,d,J=8.2Hz), 6 3.22 (2H, t, J = 6.3Hz),3.08(1H,s)2.94(2H,t,J = 6.3 Hz).
7 E1hY14-[L-(6(2.3-dihydro-(4H)-1-benzothioRyran-4-onvl), ethnti-l]benzoate 8 A solution of 405.0 mg (2.15 mmol) 2,3-dihydro-6- ethynyl-(4H)-1-9 benzothiopyran-4-one and 594.0 mg (2.15 mmol) of ethyl 4-iodobenzoate in 15 ml Et3N and 3 mt THF was sparged with argon for 15 minutes. To this 11 solution was added 503.0 mg (0.72 mmol) of 12 bis(triphenylphosphine)palladium(II) chloride and 137.0 mg (0.72 mmol) CuI.
13 This solution was stirred for 20 hours at room temperature and then filtered 14 through a pad of Celite; which was washed with EtOAc. Removal of the t s solvents under reduced pressure afforded a brown solid. Column 16 chromatography (3% EtOAc-hexanes) afforded the title compound as an 17 orange solid. 1H NMR (deacetone): 8 8.15 (1H, d, J= 2.0 Hz), 8.02 (2H, ie d,J=8.5Hz),7.69(2H,d,J=8.5Hz),7.61(1H,dd,J=2.1,8.3Hz),7.40 ia (1H,d,J=8.2Hz),4.35(2H,q,J=7.1Hz),3.40(2H,t,J=6.3Hz),2.96 (2H, t, J 6.3 Hz), 1.37 (3H, t, J 7.1 Hz).
21 Eft4-f2-(,6-(4-(trifluoromethvlsulfonyl)oy-(2L-1-benzothiopyr~
22 nyl), eMA]benzoate 23 To a solution of 221.9 mg (1.21 mmol) of sodium 24 bis(trimethylsilyl)amide in 3.0 ml THF cooled to -78 C was added 370.0 mg (1.10 mmol) of ethyl4[2-(6-(2,3- dihydro-(4H)-1-benzothiopyran-4 2s onyl))ethynyljbenzoate in 4.0 ml THF. After 30 minutes, a solution of 2-27 [N,N- bis(trifiuoromethylsulfonyl)amino]-5-chloropyridine in 4.0 m11'HF was 2a slowly added. The reaction was slowly warmed to room temperature and 29 after 5 hours quenched by the addition of saturated aqueous NH4C1. The * Trade-mark 1 mixture was extracted with EtOAc, and the combined organic layers were 2 washed with 5% aqueous NaOH, HZO, and saturated aqueous NaCl before 3 being dried over MgSO4. Removal of the solvents under reduced pressure, 4 followed by column chromatography (4% EtOAc-hexanes) afforded the title compound as a pale-yellow solid. 1H NMR (d6-acetone): S 8.12 (2H, d, J
6 8.5Hz),7.66(2H,d,J = 8.5Hz),7.56(1H,d,J = 1.7Hz),7.49(1H,dd,J =
7 1.7, 8,1 Hz), 7.40 (1H, d, J = 8.1 Hz), 6.33 (1H, t, J = 5.7 Hz), 4.35 (2H, q, J
8= 7.1 Hz), 3.82 (2H, d, J = 5.7 Hz), 1.37 (3H, t, J= 7.1 Hz).
9 Ethyl 4-[2-(6 (4(4-methylphel ,yll f2H,L1-benzothiop,y nn,til), ethywl]benzoate lo (Compound 49) 11 To a solution of 120.8 mg (0.70 mmol) of 4- bromotoluene in 2.0 ml 12 THF at -78 C was added 88.4 mg (1.38 mmol, 0.81 ml of a 1.7 M solution in 13 pentane) of t-butyllithium. After 30 minutes a solution of 131.6 mg (0.97 14 mmol) ZnC12 in 2.0 ml THF was added and the reulting pale-yellow solution is warmed to room temperature. Stirring for 40 minutes-was followed by 16 addition of this solution to a second flask containing 129.2 mg (0.28 mmol) of 17 ethyl 4-[2-(6-(4- (trifluoromethylsulfonyl)oxy-(2H)-1-18 benzothiopyranyl))ethynyl]benzoate, 14.0 mg (0.012 mmol) 19 tetrakis(triphenylphosphine)palladium (0), and 2.0 ml THF. The resulting 20 solution was heated to 50 C for 5 hours, cooled to room temperature, and 21 quenched by the addition of saturated aqueous NH4C1. The mixture was 22 extracted with EtOAc, and the combined organic layers were washed with 23 H20 and saturated aqueous NaCI, then dried (MgSO4) and concentrated to 24 an orange oil. The title compound was isolated as a colorless solid by 25 column chromatography (3 to 5% EtOAc-hexanes). 1H NMR (d6-acetone): S
26 7.98 (2H, d, J = 8.3 Hz), 7.58 (2H, d, J = 8.2 Hz), 7.44-7.38 (2H, m), 27 7.26-7.15 (5H, m), 6.14 (1H, t, J = 5.8 Hz), 4.34 (2H, q, J = 7.1 Hz), 3.53 28 (2H, d, J = 5.8 Hz), 2.37 (2H, s), 1.35 (3H, t, J = 7.1 Hz).
29 4-[2-(6 (4-4(4-met lohenvl)-(2H.)-1-benzothioQvranyl))ethynA]-benzoic acid WO 99/33821 PC`r/US98/27314 1 jComRound 5Q) 2 To a solution of 29.0 mg (0.07 mmol) ethyl 4-[2-(6-(4-(4-3 methylphenyl)-(2H)-1- benzothiopyranyl))ethynyl ]benzoate (Compound 49) 4 in 2.0 ml THF and 2.0 ml EtOH was added 160.0 mg (4.00 mmol, 2.0 ml of a s 2 M aqueous solution). The resulting solution was stirred at 35 C for 2 6 hours, and then cooled to room temperature and stirred an additional 2 7 hours. The reaction was quenched by the addition of 10% aqueous HCl and e extracted with EtOAc. The combined organic layers were washed with H20 9 and saturated aqueous NaCI, and dried over Na2SO4. Removal of the io solvents under reduced pressure afforded a solid which was washed with 11 CH3CN and dried under high vacuum to give the title compound as a pale-12 yellow solid. 1H NMR (d6-DMSO): 8 7.90 (2H, d, J = 8.4 Hz), 7.59 (2H, d, 13 J = 8.4 Hz), 7.40 (4H, m), 7.25-7.13 (4H, m), 7.02 (1H, d, J = 1.7 Hz), 6.11 14 (1H, t, J = 5.7 Hz), 3.54 (2H, d, J = 5.7 Hz), 2.34 (3H, s).
15 3.4-Dihydro-4.4-dimethyl-7-acetv,l_1f 2,-naphthalenone (Compound R): and is 3.4-dihvdro-4.4-dimethyl-6-acetyl- 1(,2W-naphthalenone (Compound S) 17 To a cold (0 C) mixture of aluminum chloride (26.3 g, 199.0 mmols) in 18 dichloromethane (55 ml) was added acetylchloride (15 g, 192 mmols) and 19 1,2,3,4-tetrahydro-1,1-dimethylnaphthalene (24.4g, 152 mmols) in 2o dichloromethane (20 ml) over 20 minutes. The reaction mixture was warmed 21 to ambient temparature and stirred for 4 hours. Ice (200 g) was added to the 22 reaction flask and the mixture diluted with ether (400 ml). The layers were 23 separated and the organic phase washed with 10% HCl (50 ml), water (50 24 ml), 10% aqueous sodium bicarbonate, and saturated aqueous NaCl (50 ml) 25 before being dried over MgSO4. Ths solvent was removed by distillation to 26 afford a yellow oil which was dissolved in benzene (50 ml).
27 To a cold (0 C) solution of acetic acid (240 ml) and acetic anhydride 28 (120 ml) was added chromiumtrioxide (50 g, 503 mmols) in small portions 29 over 20 minutes under argon. The mixture was stirred for 30 mins at 0 C and 1 diluted with benzene (120 mi). The benzene solution prepared above was 2 added with stirring via an addition funnel over 20 minutes. After 8 hours, the a reaction was quenched by careful addition of isopropanol (50 ml) at 0 C, 4 followed by water (100 ml). After 15 minutes, the reaction mixture was s diiuted with ether (1100 ml) and water (200 ml), and then neutralized with e solid sodium bicarbonate (200 g). The ether layer was washed with water (100 7 m1), and saturated aqueous NaC1(2 x 100 ml), and dried over MgSO{.
8 Removal of the solvent under reduced pressure afforded a mixture of the e isomeric diketones which were separated by chromatography ( 5% EtOAc /
hexanes). (Compound R): 1H NMR (CDCI,) : 8 8.55 (iH, d, J= 2.0 Hz), tt 8.13(1H,dd,J=2.0,8.3Hz),7.53(1H,d,J=8.3Hz),2.77(?,H,t,J=6.6 12 Hz), 2.62 (3H, s), 2.05 (?H, t, J= 6.6 Hz), 1.41(6H, s). (Compound S): IH
ts NMR (CDC13) : 8 8.10 (lli, d, J= 8.1 Hz), 8.02 (1H, d, J= 1.6 Hz), 7.82 14 (1H, dd, J= 1.6, 8.1 Hz), 2.77 (2H, t, J= 7.1 Hz), 2.64 (3H, s), 2.05 (2H, t, J
1 6 = 7.1 Hz), 1.44 (6H, s).
16 z 4-Divdro-4 d-dimethvl 7(2 QcAgW-1.3-dioxolartyl)):1(2Ft):Agphthalenone 17 jCompQund ie A mixture of 3,4-dihydro-4,4-dimathyl-7-acetyl- 1(?H)-naphthalenone ,g (Compound R) (140.0 mg, 0.60 mmol), ethylene glyaol (55.0 mg, a90 mmol), 2o p- toluenesulfanic acid monohydrate (4 mg) and benzene (25 mi) was 21 refluxnd using a Dean-Stark apparatus for 12 hours. The reaction was 22 quenched by the addition of 10% aqueous sodium bicarbonate, and extracted 23 with ether (2 x 75 ml). The combined organic layers were washed with water 24 (5 ml), and saturated aqueous NaQ (5 ml), and dried over MgSO4. Removal 25 of the soh-ent under reduced pressure afforded the title compound as an oil.
2s 1HNMR(CDAS):8 8.13(IH,d,J=2.0Hz),7.64(IH,dd,J=2.0,8.2 27 Hz), 7.40 (1H, d, J= &2 Hz), 3.97. 4.10 (2H, m), 3.70-3.83 (2H, in), 2.73 28 (2H, t, J= 6.5 HZ), 201(2H, t, J= 6.5 Hz), 1.64 (3H, S), 1.39 (6H, s).
29 1,23 4-Tetrahvdro-1 +x-1.-(4-methyln^hepml)-a.4-dnMethvl-7-(2:(2-* Trade-mark 1 methyl-1.3-dioxolanyl) naphthalene (Compound Ul 2 To a solution of 195.4 mg (1.00 mmol) p- tolulylmagnesiumbromide 3(1.0 ml; 1M solution in ether) in 2 ml THF was added a solution of 3,4-4 dihydro-4,4- dimethyl-7-(2-(2-methyl-1,3-dioxolanyl))-1(2H)- naphthalenone 5(Compound T) 135.0 mg, 0.52 mmol) in 5 ml THF. The solution was refluxed s for 16 hours, cooled to room temperature, and diluted with ether (50 ml).
7 The solution was washed with water (5 ml), saturated aqueous NH4C1 (5 ml), 8 and dried over MgSO4. Removal of the solvents under reduced pressure and 9 column chromatography (5% EtOAc / hexanes) afforded the title compound 1o as a solid. 1H NMR (CDCl3) : 8 7.37 (2H, d), 7. 21 (1H, s), 7.13 (2H, d, J
11 = 8.5 Hz), 7.08 (2H, d, J = 8.5 Hz), 3.88- 3.99 (2H, m), 3.58-3.75 (2H, m), 12 2.34 (3H, s), 2.12-2.30 (2H, m), 1.79-1.90 (1H, m), 1.57 (3H, s), 1.48-1.58 13 (1H, m), 1.38 (3H, s), 1.31 (3H, s).
14 3.4-Dih, dro-1- 4-methvlphenyl)-4.4-dimethvl-7-acetylnaphthalene (Compound 16 A mixture of 1,2,3,4-tetrahydro-l-hydroxy-l-(4- methylphenyl)-4,4 17 dimethyl-7-(2-(2-methyl-1,3- dioxolanyl))naphthalene (Compound U) 130.0 18 mg (0.38 mmol), p-toluenesulfonic acid monohydrate (4 mg) and benzene (5 19 ml) was -refluxed for 16 hours. Upon cooling to room temperature, the 2o reaction mixture was diluted with ether (100 ml) and washed with 10%
21 aqueous sodium bicarbonate, water, and saturated aqueous NaCI. The 22 organic layer was dried over MgSO4 and the solvents were removed under 23 reduced pressure to give the title compound as a solid. 1H NMR (CDC13) : 8 24 7.83(1H,dd,J= 1.8,8.0Hz),7.66(1H,d,J= 1.8 Hz), 7.45 (1H, d, J = 8.0 25 Hz), 7.25 (2H, d, J = 8.5 Hz), 7.22 (2H, d, J 8.5 Hz), 6.03 (1H, t, J =
2e 6.3Hz), 2.47 (3H, s), 2.41 (3H, s), 2.37 (2H, d, J = 6.3Hz), 1.36 (6H, s).
27 (E)-5.6-dihydro-5.5-dimethvl-8-(4-methyiphenyl)-2-naphthalenyl)-2-28 butenenitrile (Compound Wl 29 To a slurry of NaH (48.0 mg, 2.00 mmol) in THF (6 ml), was added i diethylcyanomethylphosphonate (450.0 mg, 2.50 mmol). After 40 mins, a 2 solution of 3,4-dihydro- 1-(4-methylphenyl)-4,4-dimethyl-7-acetylnaphthalene 3 (Compound V) 95.0 mg, (0.33 mmol) in THF (4 ml) was added. The mixture 4 was stirred for 16 hours, diluted with ether (100 ml), and washed with water, and saturated aqueous NaC1 before being dried over MgSO4. Removal of 6 the solvents under reduced pressure, and column chromatography (3%
7 EtOAc / hexanes) afforded the title compound as a solid. 1H NMR (CDC13):
8 8 7.39 (1H, d, J = 1H), 7.32 (1H, dd, J 2.0, 8.1Hz), 7.20-7.25 (4H, brs), 9 7.15(1H,d,J=2.0Hz),6.03(1H,t,J=6.0Hz),5.44(1H,s),2.42(3H,s), 1 o 2.36 (2H, d, J = 6.0 Hz), 2.35 (3H, s), 1.35 (6H, s).
ii (E, -Z3-(5.6-dihydro-5.5-dimethvl-8-(4-meth, rlphenyl)-2-na hthalenY)-2-butenal 12 (Compound X) 13 To a cold solution (-78 C) of (E)-3-(5,6-dihydro- 5,5-dimethyl-8-(4-14 methylphenyl)-2-naphthalenyl)-2- butenenitrile (Compound W) 84.0 mg, 0.29 mmol) in dichloromethane (4 ml) was added 0.50 ml (0.50 mmol) of 16 diisobutylaluminumhydride (].M solution in dichloromethane). After stirring 17 for 1 hour, the reaction was quenched at -78 C by adding 2-propanol (1 ml) 18 diluted with ether (100 ml). Upon warming to room temperature, the solution 1 s was washed with water, 10% HCI, and saturated aqueous NaCI. The organic layer was dried over MgSO4 and the solvent removed under reduced pressure 21 to give the title compound as an oil. 1H NMR (CDC13) : 8 10.12 (1H, d, J
22 = 7.9 Hz), 7.43 (2H, s), 7.19-7.28 (5H, m), 6.27 (1H, d, J = 7.9 Hz), 6.03 23 (1H, t, J = 4.8 Hz), 2.47 (3H, s), 2.42 (3H, s), 2.37 (2H, d, J = 4.8 Hz), 1.37 24 (6H, s).
EthYl (E.E.E)-3-methyl-7-(5.6-dihydro-5.5-dimethyl-8-(.4-methylphenvl)-2-2s naphthalenxl)-2.4.6-octatrienoate (Compound 51) 27 To a cold (-78 C) solution of diethyl-(E)-3- ethoxycarbonyl-2-28 methylallylphosphonate [prepared in accordance with J. Org. Chem. 39: 821 29 (1974)] 264.0 mg, (1.00 mmol) in THF (2 ml) was added 26.0 mg (0.41 mmol, 1 0.65 ml)of n-butyllithium in hexanes (1.6 M solution) followed immediately 2 by the addition of (E)-3-(5,6- dihydro-5,5-dimethyl-8-(4-methylphenyl)-2-3 naphthalen- yl)-2-butenal (Compound X) 82.0 mg, 0.26 mmol) in THF (3 4 ml). After 1 hour, the reaction mixture was diluted with ether (60 ml), washed with water (5 ml), saturated aqueous NaCI (5 ml) and dried over 6 MgSO4. After removal of the solvents under reduced pressure, the title 7 compound was isolated as an oil by column chromatography (5% EtOAc /
8 hexanes, followed by HPLC using 1% EtOAc / hexanes). 1H NMR (acetone-9 d6) : 8 7.36-7.43 (2H, m), 7.18-7.27 (4H, m), 7.17 (1H, d, J = 1.7 Hz), 7.08 lo (1H, dd, J = 11.2,15.2Hz),6.46(1H,d,J= 11.2 Hz), 6.38 (1H, d, J = 15.2 Hz),5.98(1H,t,J=4.7Hz),5.78(1H,s),4.10(2H,q,J=7.1Hz),2.35 12 (3H, s), 2.33 (3H, s), 2.32 (2H, d, J = 4.7 Hz), 2.12 (3H, s), 1.31 (6H, s), 1.22 13 (3H, t, J= 7.1 Hz).
14 (E.E.E)-3-methyl-7-(5.6-dihydro-5.5-dimethvl-8-(4-methylphenyl)-2-1s ophthalenyl)-2.4.6-octatrienoic acid (Compound 52) t e To a solution of ethyl (E,E,E)-3-methyl-7-(5,6- dihydro-5,5-dimethyl-8-17 (4-methylphenyl)-2- naphthalenyl)-2,4,6-octatrienoate (Compound 51) 85.0 18 mg, 0.20 mmol) in THF (lml) and methanol (lml) was added 12.0 mg (0.50 is mmol) of LiOH (0.5 ml, 1M solution). The mixture was stirred for 6 hours, 2o diluted with ether (60 ml), acidified with 10% HCl (1 ml). The solution was 21 washed with water, and saturated aqueous NaCl, before being dried over 22 MgSO4. Removal of the solvents under reduced pressure afforded the title 23 compound as a solid, which was purified by recrystallization from acetone.
24 1H NMR (acetone-d6) : 8 7.35-7.45 (2H, m), 7.19-7.28 (4H, m), 7.17 (1H, d, 25 J = 1.8Hz), 7.09 (1H, dd, J = 11.5, 15.1 Hz), 6.48 (1H, d, J = 11.5 Hz), 6.42 26 (1H, d, J = 15.lHz),5.99 (1H, t, J = 4.7 Hz),5.82 (1H, s), 2.36 (3H, s), 2.33 27 (2H, d, J = 4.7Hz), 2.32 (3H, s), 2.13 (3H, s), 1.32 (6H, s).
28 3 4-dihydro-4.4-dimethyl-7-nitro-1,2H1,-naphthalenone (,Compound Y) 29 To 1.7 ml (3.0g, 30.6 mmol, 18M) H2S04 at -5 C (ice-NaCl bath) was 1 slowly added 783.0 mg (4.49 mmol) of 3,4-dihydro-4,4-dimethyl-1(2H)-2 naphthalenone. A solution of 426.7 mg (6.88 mmol, 0.43 ml, 16M) HNO3, 3 and 1.31g (0.013 mol, 0.74 ml, 18 M) H2S04 was slowly added. After 20 4 minutes, iee was added and the resulting mixture extracted with EtOAc. The combined extracts were concentrated under reduced pressure to give a e residue from which the tide compound, a pale yellow solid, was isolated by 7 column chromatography (10% EtOAC / hexanes). 1H NMR (CDC'13) : S
s 8.83(1H,d,J=2.6Hz),8.31(1H,dd,J=2.8,8.9Hz),7.62(IH,d,J=8.7 a Hz), 281 (2H, t, J= 6.5 Hz), 2.08 (2H, t, J= 6.5 Hz),1,45 (6H, s).
to 3.4-dihydro-4.4.dimethvl-7-amino-lf=:naohth~ alenonr, (Cmno~und Z) 11 A solution of 230.0 mg (1.05 mmol) 3,4-dihydro-4,4-dimethyl-7-12 nitro-1(2H)-naphthalenone (Compound Y) in 5.0 ml of EtOAc was stirred at 13 room temperature with a catalytic amount of 10% Pd-C under 1 atm of Hz 14 for 24 hours. The catalyst was removed by filtration through a pad of Celite;
and the filtrate concentrated under reduced pressure to give the title ts compound as a dark green oil. 1H NMR (CDCI3) : S 7.30 (1H, d, J = 2.7 17 Hz), 7.22 (1H, d, J= 8.4 Hz), 6.88 (1H, dd, J = 2.7, 8.5 Hz), 2.70 (2H, t, J
ts 6.6 Hz), 1.97 (2H, t, J= 6.6 HZ), 1.34 (6H, s).
ts EUD14-((5.6.7.8-tetrahvdro-5.5-dimethvl-8-oxo-2 naFhthalenvl)zoj-benzoate (Compoun AAl 21 To a solution of 198.? mg (1.05 mmol) 3,4-dihydro-4,44methyl-7-22 amino-1(2H)-naphthalenone (Compound Z) in 5.0 ml glaeial acetic acid was zs added 180.0 mg (1.00 mmol) of ethyl 4nitrosobenzoate. The resulting 24 solution was stirred overnight at room tcmperature, and then concentrated under reduced pressure. The product was isolated from the residual oil as a 29 red solid, by column chromatography (15% EtOAc - hexanes). 1H NMR
27 (CDCI,) : S 8.57 (1H, d, J= 2.0 Hz), 8.19 (2H, d, J= 8.4 Hz), 8.07 (1H, d, 28 J= 8.0 Hz), 7.94 (2H, d, J= 8.4 Hz), 7.58 (1H, d, J 8.6 Hz), 4.41 (2H, q, 2a J= 7.1 Hz), 2.79 (2H, t, J 6.6 Hz), 2.07 (2H, t, J 7.02 Hz), 1.44 (6H, s), * Trade-mark 1 1.42 (3H, t, J = 7.1 Hz).
2 Eth_y-l 4-[f 5.6-dihydro-5 5-dimethy-1-8ltrifluoromethylsulfonyl)ox,y-2-s naphthalenvl)azo]- benzoate (Compound BB) 4 To a solutiori of 90.4 mg sodium bis(trimethylsilyl)amide (0.48 mmol, 0.48 ml of a 1.0 M THF solution) in 2.0 ml THF at -78 C, was added 153.0 s mg (0.437 mmol) of ethyl 4-[(5,6,7,8-tetrahydro-5,5- dimethyl-8-oxo-2-7 naphthalenyl)azo]-benzoate (Compound AA) in 2.0 ml THF. The dark red 8 solution was stirred at -78 C for 30 minutes and then 204.0 mg (0.520 mmol) 9 of 2-[N,N-bis(trifluoromethylsulfonyl)amino]-5-chloropyridine was added as a 1 o solution in 2.0 ml THF. The reaction mixture was allowed to warm to room 11 temperature and after 3 hours it was quenched by the addition of H20. The 12 organic layer was concentrated to a red oil under reduced pressure. The 13 product was isolated by column chromatography (25% EtOAc / hexanes) as a 14 red oil. 1H NMR (CDC13) : 8 8.21 (2H, d, J = 8.6 Hz), 7.96 (2H, d, J = 8.6 Hz), 7.94 (2H, m), 7.49 (1H, d, J = 8.2 Hz), 6.08 (1H, t, J = 2.5 Hz), 4.42 16 (2H, q, J = 7.1 Hz), 2.49 (2H, d, J = 4.8 Hz), 1.44 (3H, t, J = 7.1 Hz), 1.38 17 (6H, s).
18 Ethy14-[(5 6-dihvdro-5 5-dimethyl- 8-(4-methylphenyl -) 2-naphthalenXl)azo-19 benzoat (Compound 46a) A solution of 4-lithiotoluene was prepared by the addition of 62.9 mg 21 (0.58 ml, 0.98 mmol) of t-butyl lithium (1.7 M solution in pentane) to a cold 22 solution (-78 C) of 84.0 mg (0.491 mmol) of 4-bromotoluene in 1.0 ml of 23 THF. After stirring for 30 minutes a solution of 107.0 mg (0.785 mmol) of 24 zinc chloride in 2.0 ml of THF was added. The resulting solution was warmed to room temperature, stirred for 30 minutes, and added via cannula 26 to a solution of 94.7 mg (0.196 mmol) of ethyl4-[(5,6-dihydro-5,5-dimethyl-27 (trifluoromethylsulfonyl)oxy-2-naphthalenyl)azo]- benzoate (Compound BB) 28 and 25 mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) in 2.0 ml 29 of THF. The resulting solution was heated at 50 C for 1.5 hours, cooled to 1 room temperature and diluted with sat. aqueous NH4C1. The mixture was 2 extracted with EtOAc (40 ml) and the combined organic layers were washed 3 with water and brine. The organic phase was dried over Na2SO41 4 concentrated in vacuo, and the title compound isolated as a red solid by column chromatography (25% EtOAc-hexanes) 1H NMR (CDC13) : 8 8.21 6 (2H, d, J = 8.6 Hz), 7.96 (2H, d, J = 8.6 Hz), 7.94 (2H, m), 7.49 (1H, d, J
7 8.2 Hz), 6.08 (1H, t, J = 2.5 Hz), 4.42 (2H, q, J = 7.1 Hz), 2.49 (2H, d, J
8 4.8 Hz), 1.44 (3H, t, J= 7.1 Hz), 1.38 (6H, s).
9 4-j(5.6-dihydro-5.5-dimeth,Y-8-(4-methylphenyl)-2-naphthalenyI)azo]-benzoic 1 o acid (Compound 46b) 11 To a solution of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(4-12 methylphenyl)-2-naphthalenyl)azo]- benzoate (Compound 46a) 16.5 mg, 0.042 13 mmol) in THF (2 ml) and ethanol (iml) was added 80.0 mg (2.00 mmol) of 14 NaOH (2.0 ml, 1M aqueous solution). The mixture was stirred for 12 hours at room temperature, acidified with 10% HCI, and extracted with EtOAc.
16 The combined organic layers were washed with water, and saturated aqueous 17 NaCl, then dried over MgSO4. Removal of the solvents under reduced 18 pressure, and recrystallization of the residue from EtOAC / hexane, afforded 19 the title compound as a red solid. 1H NMR (acetone-d6) : 8 8.19 (2H, d, J
= 8.4 Hz), 7.92 (2H, d, J = 8.5 hz), 7.88 (2H, dd, J = 2.1, 6.1 Hz), 7.66 (1H, 21 s), 7.64 (2H, d, J = 2.3 Hz), 7.28 (4H, d, J = 3.0 Hz), 6.09 (1H, t, J =
2.5 22 Hz), 2.42 (2H, d, J = 4.8 Hz), 2.39 (3H, s), 1.40 (6H, s).
23 6-(2-Trimethylsil,-l)ethynyl-2.3-dihYdro-3.3-dimethyl-lH-inden-l-one 24 (Compound "
To a solution of 815.0 mg (3.41 mmol) 6-bromo-2,3- dihydro-3,3-26 d'unethyl-lH-inden-l-one (See Smith et al. Org. Prep. Proced. Int. 1978 10 27 123-131) in 100 ml of degassed Et3N (sparged with argon for 20 min) was 28 added 259.6 mg (1.363 mmol) of copper(I) iodide, 956.9 mg (1.363 mmol) of 29 bis(triphenylphosphine)palladium(II)chloride, and 3.14 g (34.08 mmol) of WO 99/33821 PCTlUS98127314 i (trimethylss'lyi)acetylene. This mixture was heated at 70 C for 42 hours, 2 cooled to room temperature, and filtered through a pad of silica gel and 3 washed with ether. The 8ltrate was washed with water,l M HCI, water, and 4 finaly with saturated aqueous NaCI before being dried over MgSO4, Concentration of the solution under reduced pressure, followed by column s chromatography (silica gel; 10% Et20 - hexanes) afforded the title compound 7 as a brown oil. 1H NMR (300 MHz, CDCl3): S 7.79(1H, d, J= 1.4 Hz), e 7.69 (1H, dd, J= 1.6, 8.3 Hz), 7.42 (1H, d, J = 8.5 Hz), 2.60 (2H, s), 1.41 s (6H, s), 0.26 (9H, s).
6-Eth3nvl-2.3-dih dry o-3.3-dimetWl-lH-inden-l-one (Qmnoun! d DDl 11 To a solution of 875.0 mg (3.41 mmol) 6-(2- trimethylsilyl)ethynyl-2,3-12 dihydro-3,3-dimethyl-lH-inden-1=one (Compound CC) in 28 ml of MeOH, is was added 197.3 mg (1.43 nnmol) of KzCO, in one portion. After stirring for 14 6 hours at room temperature the mixture was filtered though a pad of Celite*
and the filtrate concentrated under reduced pressure. The residual oil was ts placed on a silica gel column and eluted with 5% EtOAc-hexanes to give the title product as a eolorless oil. 1H NMR (300 MHz, CDC13): 8 7.82 (1H, s), 18 7.72(1H,dd,J= 1.6,7.8Hz),7.47(1H,d,J=8.4Hz),3.11(1H,s),2.61 ts (2H, s), 1.43 (6H, s).
qo Et 4(2-(5-diW-dro-5.5-dimethvl-7-oao-2-indW)ethy,_,_evl nmatP
21 (ComRoand P.FI
22 . A solution of 280.0 mg (l.SaO mmol) 6-ethynyl-2,3- dihydro-3,3-23 dimethyl-lH-inden-l-one (Compound DD) and 419.6 mg (1.520 mmol) ethyl 24 4-iodobenzoate in 5 mi Et,N was sparged with argon for 40 minutes. To this solution was added 271.0 mg (1.033 mmol) of triphenylphosphine, 53.5 mg 2s (0.281 mmol) of eopper(I) iodide, and 53.5 mg (0.076 mmol) of 27 bis(triphenylphosphine)palladium(II) chloride. The resulting mixture was 2a heated to reflux for 2.5 hours, cooled to room temperature, and difuted with 29 EtzO. After 5ltration through a pad of Celite, the filtrate was washed with * Trade-mark I H20, 1 M HCI, H20, and saturated aqueous NaCI, then dried over MgSO4, 2 and concentrated under reduced pressure. The title compound was isolated as 3 a pale-yellow solid by column chromatography (15% EtOAc-hexanes). 1H
4 NMR (300 MHz, d6-acetone): S 8.05 (2H, d, J = 8.6 Hz), 7.87 (1H, dd, J
1.4, 8.1 Hz), 7.75 (2H, m), 7.70 (2H, d, J = 8.5 Hz), 4.36 (2H, q, J = 7.1 Hz), s 2.60 (2H, s), 1.45 (6H, s), 1.37 (3H, t, J = 7.1 Hz).
7 Ethvl 4-[(1.1-dimethvl-3-(trifluoromethyl-sulfonyl)xy-5-8 indenyllethynyljbenzoate (Compound FF) a A solution of 88.0 mg (0.48 mmol) of sodium bis(trimethylsilyl)amide in 0.5 ml THF was cooled to -78 C and 145.0 mg (0.436 mmol) of ethyl 4-[2-11 (5,6-dihydro-5,5-dimethyl-7-oxo-2-indenyl)ethynyl]benzoate (Compound EE) 12 was added as a solution in 1.0 ml THF. After 30 minutes 181.7 mg (0.480 13 mmol) of 2-(N,N- bis(trifluoromethansulfonyl)amino)-5-chloro-pyridine was 14 added as a solution in 1.0 ml THF. The reaction was allowed to slowly warm t s to room temperature and quenched after 5 hours by the addition of saturated 16 aqueous NH4C1. The mixture was extracted with EtOAc, and the combined 17 organic layers washed with 5% aqueous NaOH, H2O, and saturated aqueous 18 NaC1, then dried (MgSOa) and concentrated under reduced pressure. The 19 product was isolated as a colorless solid by column chromatography (10%
Et20-hexanes). 1H NMR (300 MHz, d6-acetone): 8 8.05 (2H, d, J = 8.3 21 Hz), 7.69 (2H, d, J = 8.4 Hz), 7.63 (2H, s), 7.55 (1H, s), 4.36 (2H, q, J =
7.1 22 Hz), 1.44 (6H, s), 1.37 (3H, t, J = 7.1 Hz).
23 4-[(5.6-dih,ydro-5.5-dimethy-8^(4-methylphenyl)-2-24 naphthalenA)ethynti]benzoic acid (Compound 601 A solution of 142.6 mg (0.339 mmol) of ethyl 4- [(5,6-dihydro-5,5-2s dimethyl-8-(4-methylphenyl)-2- naphthalenyl)ethynyl]benzoate (Compound 1) 27 and 35.6 mg (0.848 mmol) of LiOH-H20 in 12 ml of THF/water (4:1, v/v), 28 was stirred overnight at room temperature. The reaction mixture was 29 extracted with hexanes, and the hexane fraction extracted with 5% aqueous t NaOH. The aqueous layers were combined and acidified with 1M HCI, and 2 then extracted with EtOAc and Et20. The combined organic layers were 3 dried over Na2SO4 and concentrated in vacuo to give the title compound as a 4 colorless solid. 1H NMR (d6-DMSO): 8 7.91 (2H, d, J = 8.4 Hz), 7.60 (2H, d, J 8.4 Hz), 7.47 (2H, s), 7.23 (4H, q, J 8.1 Hz), 7.01 (1H, s), 6.01 (1H, 6 t, J 4.6 Hz), 2.35 (3H, s), 2.33 (2H, d, J 4.8 Hz), 1.30 (6H, s).
7 4-[(5.6-dihydro-5.5-dimethxl-8-phen l-y 2-naphthalenyl)ethvnyl]benzoic acid e (Compound 60a) 9 Employing the same general procedure as for the preparation of 4-io [(5,6-dihydro-5,5-dimethyl-8-(2- thiazolyi)-2-naphthalenyl)ethynyl]benzoic acid ii (Compound 30a), 27.0 mg (0.07 mmol) of ethyl 4-[(5,6- dihydro-5,5-12 dimethyl-8-phenyl-2- naphthalenyl)ethynyl]benzoate (Compound la) was 13 converted into the title compound (colorless solid) using 5.9 mg (0.14 mmol) 14 of LiOH in HZO. PMR (d6 DMSO): 8 1.31 (6H, s), 2.35 (2H, d, J = 4.5 Hz), 6.05 (1H, t, J =J =J = 4.5 Hz), 7.00 (1H, s), 7.33 (2H, d, J = 6.2 Hz), 7.44 ie (4H, m), 7.59 (2H, d, J = 8.1 Hz), 7.90 (2H, d, J= 8.1 Hz).
17 4-[(5.6-Dihydro-5.5-dimethyl8-(4-(1.1- dimeth, lY ethyl)phenyl)-2-18 naphthalen3LI)e h,xnvl]benzoic acid (Compound 61) 19 A solution of 80.0 mg (0.173 mmol) of ethyl 4- [(5,6-dihydro-5,5-2o d'nnethyl-8-(4-(1,1- dimethylethyl)phenyl)-2-naphthalenyl)ethynyl]benzoate 21 (Compound 6) and 18.1 mg (0.432 mmol) of LiOH-H20 in 6 ml of 22 THF/water (3:1, v/v), was stirred overnight at room temperature. The 23 reaction mixture was extracted with hexanes, and the remaining aqueous layer 24 acidified with 1M HCI, and then extracted with EtOAc. The combined organic layers were dried over NaZSO4 and concentrated in vacuo to give the 26 title compound as a colorless solid. 1H NMR (d6-DMSO): 8 7.82 (2H, d, J
27 = 8.2 Hz), 7.44 (6H, m), 7.25 (2H, d, J = 8.3 Hz), 7.02 (1H, s), 6.01 (1H, t, J
2s = 4.6 Hz), 2.32 (2H, d, J = 4.7 Hz), 1.32 (9H, s), 1.29 (6H, s).
29 Ethy12-fluoro-4-[[(5.6-dihyJiro-5.5-dimethyLl-8-(4-methythenti)-2-1 naphthalenyl)thiocarbonyl]amino]-benzoate (Compound 62~
2 A solution of 54.4 mg (0.119 mmol) ethyl 2-fluoro-4-[[(5,6-dihydro-5,5-3 dimethyl-8-(4-methylphenyl)-2-naphthalenyl)carbonyl]amino]-benzoate 4 (Compound 40) and 57.7 mg (0.143 mmol) of [2,4-bis(4- methoxyphenyl)-1,3-dithia-2,4-diphosphetane-2,4- disulfide] (Lawesson's Reagent) in 12.0 ml of 6 benzene was refluxed overnight. Upon cooling to room temperature, the 7 mixture was filtered and the filtrate concentrated under reduced pressure.
8 The title compound was isolated by column chromatography (10 to 25%
9 EtOAc / hexanes) as a yellow solid. 1H NMR (CDC13): 8 9.08 (1H, s), 7.92 1o (1H, br s), 7.90 (1H, t, J = 8.2 Hz), 7.66 (1H, dd, J = 2.0, 6.0 Hz), 7.38 (3H, 11 m), 7.18 (4H, m), 6.01 (1H, t, J = 4.7 Hz), 4.35 (2H, q, J = 7.1 Hz), 2.36 12 (3H, s), 2.33 (2H, d, J = 4.7 Hz), 1.38 (3H, t, J = 7.1 Hz), 1.33 (6H, s).
13 2-fluoro-4-[j(5.6-dihxdro-5.5-dimethA-8-(4-methylphenyl)-2-14 naphthalenyl)thiocarbonYl]aminoj-benzoic acid ( ompound 63).
1 s To a solution of 46.5 mg (0.098 mmol) ethyl 2- fluoro-4-[[(5,6-16 dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-naphthafenyl)thiocarbonyl]amino]-1 7 benzoate (Compound 62) in 1.0 ml EtOH and 1.0 ml of THF was added 55 18 mg NaOH (1.4 mmol) and 1.0 ml of HZO. After stirring at room temperature 19 for overnight EtOAc was added, and the reaction quenched by the addition 20 of 10% HCI. Extraction with EtOAc was followed by washing of the 21 combined organic layers with H20, saturated aqueous NaCI, and drying over 22 MgSO4. Removal of the solvent under reduced pressure provided a solid 23 which after crystallization from CH3CN afforded the title compound as a 24 pale-yellow solid. 1H NMR (dd-acetone): 8 11.05 (1H, s), 8.02 (1H, m), 7.99 25 (1H, t, J = 8.3 Hz), 7.75 (1H, m), 7.69 (1H, dd, J = 2.0, 6.1 Hz), 7.52 (1H, 26 s), 7.46 (1H, d, J = 8.1 Hz), 7.21 (4H, m), 6.04 (1H, t, J = 4.8 Hz), 2.37 (2H, 27 d, J = 4.8 Hz), 2.33 (3H, s), 1.36 (6H, s).
28 Ethy15'.6'-dihydro-5',5'-dimethyl-4-methylphenYl)-[2.2'-binaphthalene]-6-2s carboxylate (C'.mpound 64), 1 A solution of 3,4-dihydro-l-(4-methylphenyl)-4,4- dimethyl-7-2 bromonaphthalene Compound D) 0.45 g, 1.40 mmol) and THF (2.1 ml) was 3 added to magnesium turnings (0.044 g, 1.82 mmol) at room temperature 4 under argon. Two drops of ethylene dibromide were added, and the solution, which slowly became cloudy and yellow, was heated to reflux for 1.5 hours.
6 In a second flask was added zinc chloride (0.210 g, 1.54 mmol), which was 7 melted under high vacuum, cooled to room temperature and dissolved in s THF (3 ml). The Grignard reagent was added to the second flask and, after 9 30 minutes at room temperature, a solution of ethyl 6-bromo-2-1o naphthalinecarboxylate (Compound N) 0.293 g, (1.05 mmol) and THF (2 ml) 11 were added. In a third flask was prepared a solution of Ni(PPh3)4 and THF
12 as follows: To a solution of NiCIZ(PPh3)Z (0.82 g, 1.25 mmol) and PPh3 (0.66 13 g, 2.5 mmol) in THF (3.5 ml) was added a 1M solution of 14 diisobutylaluminum hydride and 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 16 temperature for 15 minutes. Three 0.60 ml aliquots of the Ni(PPh3)4 solution 17 were added at 15 minutes intervals to the second flask. The resulting 1 e suspension was stirred at room temperature for 2 hours. The reaction was 19 quenched by the addition of 5 ml 1N aqueous HCl and stirred for 1 hour 2o before extracting the products with ethyl acetate. The organic layers were 21 combined, washed with brine, dried (MgSO4), fil' tered and the solvent 22 removed in-vacuo. The residue was crystalized from hexanes to give 130 mg 23 of pure material. The mother liquor was concentrated under reduced 24 pressure and the residue purified by silica gel chromatography (95:5-hexanes:ethyl acetate) to give an additional 170 mg of the title compound 26 (overall yield = 300 mg, 64 %) as a colorless solid. 1H NMR (CDC13) 8 8.57 27 (s, 1H), 8.05 (dd, 1H, J = 1.7, 8.0 Hz), 7.84-7.95 (overlapping d's, 3H), 7.66 28 (dd, 1H, J = 1.7, 8.5 Hz), 7.58 (dd, 1H, J = 2.0, 8.0 Hz), 7.48 (d, 1H, J =
8.0 29 Hz), 7.43 (d, 1H, J = 2.0 Hz), 7.32 (d, 2H, J = 8.0 Hz), 7.21 (d, 2H, J =
8.0 1 Hz), 6.04 (t, 1H, J = 4.8 Hz), 4.44 (q, 2H, J= 7.1 Hz), 2.40 (s, 3H), 2.39 (d, 2 2H, J = 4.8 Hz), 1.45 (t, 3H, J = 7.1 Hz), 1.39 (s, 6H).
3 5'.6'-Dihydro-5'.5'-d'unetll-8':(4-methlphenxl)-[ .2'-binaphthalene]-6-Comoound 65) 4 carbo lic acid (, .
_.~Y-._.
A solution of ethyl 5',6'-dihydro-5',5'-dimethyl- 8'-(4-methylphenyl)-6 [2,2'-binaphthalene]-6-carboxylate (Compound 64) 0.19 g, 0.43 mmol), EtOH
7 (8 ml) and iN aqueous NaOH (2 ml) was heated to 60 C for 3 hours. The 8 solution was cooled to 0 C and acidified with 1N aqueous HCI. The product s was extracted into ethyl acetate, and the organic layers combined, washed 1 o with water, brine, dried (MgSO4), filtered and the solvent removed in-vacuo.
11 The residue was recrystalized from THF/ethyl acetate at 0 C to give 35 mg of 12 pure material. The mother liquor was concentrated under reduced pressure 13 and the residue purified by silica gel chromatography (100% ethyl acetate) to 14 give an additional 125 mg of the title compound (overall yield = 160 mg, 90 %) as a colorless solid. 1H NMR (DMSO-d6) 8 8.57 (s, 1H), 8.11 (d, 1H, J
16 = 8.7 Hz), 7.96-7.82 (overlapping d's, 3H),7.65 (d, 2H, J= 7.6 Hz), 7.50 (d, 17 1H, J = 7.9 Hz), 7.28 (s, 1H), 7.26 (d, 2H, J = 8.3 Hz), 7.21 (d, 2H, J =
8.3 1s Hz), 6.01 (t, 1H, J = 4.5 Hz), 3.34 (br s, 1H), 2.31 (s, 3H), 2.31 (d, 2H, J
1a 4.5 Hz), 1.31 (s, 6H).
2o Ethy14-((5.6-dihydro-5.5-d'nnethyl $_(2-l, -) 2-naphthalen,ti-1)ethwyl]benzoate 21 (Compound 22 Employing the same general procedure as for the preparation of ethyl 23 4-[(5,6-dihydro-5,5-dimethyl-8-(4- methylphenyl)-2-24 naphthalenyl)ethynyl]benzoate (Compound 1), 250.0 mg (0.52 mmol) of ethyl 4-[(5,6-dihydro-5,5- dimethyl-8-(trifluoromethylsulfonyl)oxy-2-26 naphthalenyl)ethynyl]benzoate (Compound G) was converted into the title 27 compound (colorless solid) using 142.4 mg (1.045 mmol) of zinc chloride, 28 24.1 mg (0.02 mmol) of tetrakis(triphenylphosphine)palladium(0) and 2-29 lithiofuran (prepared by the addition of 53.4 mg (0.52 ml, 0.78 mmol) of n-1 butyllithium (1.5M solution in hexane) to a cold solution (-78 C) of 53.4 mg 2 (0.784 mmol) of furan in 1.0 ml of THF). PMR (CDC13): 8 1.32 (6H, s), 1.41 3 (3H, t, J = 7.1 Hz), 2.35 (2H, d, J = 5.0 Hz), 4.39 (2H, q, J = 7.1 Hz), 6.41 4 (1H,t,J=5.0Hz),6.50(2H,s),7.36(1H,d,J=8.0Hz),7.45(1H,dd,J=
1.7, 8.0 Hz), 7.49 (1H, s), 7.57 (2H, d, J = 8.2 Hz), 7.63 (1H, d, J = 1.7 Hz), 6 8.02 (2H, d, J = 8.2 Hz).
7 4-[(5.6-dihxdro-5.5-dimethvl-8-(2-fuU1, -) 2-naphthalenvl)ethXWj]benzoic acid 8 (Compound 67) 9 Employing the same general procedure as for the preparation of 4-1o [(5,6-dihydro-5,5-dimethyl-8-(2- thiazolyl)-2-naphthalenyl)ethynyl]benzoic acid 11 (Compound 30a), ethyl 4-[(5,6-dihydro-5,5-dimethyl-8- (2-furyl)-2-12 naphthalenyl)ethynyl]benzoate (Compound 66) was converted into the title 13 compound (colorless solid) using 16.0 mg (0.38 mmol) of LiOH in H20.
14 PMR (d6 DMSO): S 1.26 (6H, s), 2.33 ( 211, d, J = 4.9 Hz), 6.41 (1H, t, J
4.9 Hz), 6.60 (2H, m), 7.45-7.53 (3H, m), 7.64 (2H, d, J = 8.3 Hz), 7.75 (1H, 1 s d, J = 1.6 Hz), 7.93 (2H, d, J = 8.3 Hz).
17 3.4-diWro-4,4-dimet , -7-ace ,t l-1(2H)-naphthalenone (Compound 100C) 1 s and 3.4-dihvdro-4.4-dimethvl-6-a g W-1(2H)-naphthalenone (Compound To a cold (0 C) mixture of aluminum chloride (26.3 g, 199.0 mmols) 21 in dichloromethane (55 ml) was added acetylchloride (15 g, 192 mmols) and 22 1,2,3,4- tetrahydro-1,1-dimethylnaphthalene (24.4g, 152mmols) in 23 dichloromethane (20 ml) over 20 minutes. The reaction mixture was warmed 24 to ambient temparature and stirred for 4 hours. Ice (200 g) was added to the reaction flask and the mixture diluted with ether (400 ml). The aqueous and 26 organic layers were separated and the organic phase was washed with 10%
27 HC1 (50 ml), water (50 ml), 10% aqueous sodium bicarbonate, and saturated 28 aqueous NaCI (50 ml) and then dried over MgSO4. The solvent was removed 29 by distillation to afford a yellow oil which was dissolved in benzene (50 ml).

1 To a cold (0 C) solution of acetic acid (240 ml) and acetic anhydride 2 (120 ml) was added chromium trioxide (50 g, 503 mmols) in small portions 3 over 20 minutes under argon. The mixture was stirred for 30 minutes at 0 C
4 and diluted with benzene (120 ml). The benzene solution prepared above was added with stirring via an addition funnel over 20 minutes. After 8 hours, the e reaction was quenched by the careful addition of isopropanol (50 ml) at 0 C, 7 followed by water (100 ml). After 15 minutes, the reaction mixture was 8 diluted with ether (1100 ml) and water (200 ml), and then neutralized with s solid sodium bicarbonate (200 g). The ether layer was washed with water 1o (100 ml),and saturated aqueous NaCI (2 x 100 ml), and dried over MgSO4.
11 Removal of the solvent under reduced pressure afforded a mixture of the 12 isomeric diketones which were separated by chromatography ( 5% EtOAc /
13 hexanes). (Compound 100C): 1H NMR (CDC13) : 8 8.55 (1H, d, J = 2.0 14 Hz),8.13(1H,dd,J=2.0,8.3Hz),7.53(1H,d,J=8.3Hz), 2.77 (2H, t, J
= 6.6 Hz), 2.62 (3H, s), 2.05 (2H, t, J = 6.6 Hz), 1.41 (6H, s). (Compound 16 100D): 1HNMR(CDC13):8 8.10(1H,d,J=8.1Hz),8.02(1H,d,J= 1.6 17 Hz), 7.82 (1H, dd, J = 1.6, 8.1 Hz), 2.77 (2H, t, J = 7.1 Hz), 2.64 (3H, s), 18 2.05 (2H, t, J = 7.1 Hz), 1.44 (6H, s).
19 3.4-dihvdro-4.4dimeth,til-6-(~2-methyl-1.3-dioxolany~))-1(2H~-naphthalenone 2o (Compound 100E) 21 A solution of 1.80 g (8.34 mmol) of a 1:5 mixture of 3,4-dihydro-4,4-22 dimethyl-7-acetyl-1(2H)- naphthalenone (Compound 100C); and 3,4-23 dihydro-4,4- dimethyl-6-acetyl-1(2H)-naphthalenone (Compound 100D) in 50 24 ml benzene was combined with 517.7 mg (8.34 mmol) of ethylene glycol and 20.0 mg (0.11 mmol) of p- toluenesulfonic acid monohydrate. The resulting 26 solution was heated to reflux for 18 hours, cooled to room temperature, and 27 concentrated under reduced pressure. The title compound was isolated by 28 column chromatography (10% EtOAc - hexanes) as a colorless oil. 1H NMR
29 (CDC13) : 8 8.01 (1H, d, J = 8.2 Hz), 7.51 (1H, s), 7.43 (1H, dd, J = 1.7, 6.4 1 Hz), 4.07 (2H, m), 3.79 (2H, m), 2.74 (2H, t, J = 6.5 Hz), 2.04 (2H, t, J =
7.1 2 Hz), 1.67 ( 3H, s), 1.46 (6H, s).
3 1.2.3.4-tetrahydro-l-hydroU-I-(4-methylphenyl)-4.4-dimethLI-6-(2 (2-4 methyl-1.3-dioxolanYl))naphthalene(Compound 100F) To a solution of 496.2 mg (2.54 mmol) p- tolylmagnesiumbromide in s 20 ml THF (2.54 ml; 1M solution in ether) was added a solution of 3,4-7 dihydro-4,4-dimethyl-6-(2-(2-methyl-1,3-dioxolan- yl))-1(2H)-naphthalenone 8 (Compound 100E, 200.0 mg, 0.769 mmol) in THF (5 ml). The solution was 9 refluxed for 16 hours, cooled to room temperature, and washed with water, 1 o saturated aqueous NH4C1, and dried over MgSO4. Removal of the solvents ii under reduced pressure and column chromatography (10% EtOAc / hexanes) 12 afforded the title compound as a colorless solid. 1H NMR (CDC13) : 8 7.49 13 (1H,d,J= 1.7 Hz), 7.19 (2H, m), 7.10 (2H, d, J = 7.9 Hz), 7.04 (1H, d, J =
14 8.2 Hz), 4.05 (2H, m), 3.80 (2H, m), 2.34 (3H, s), 2.21 (1H, m), 2.10 (1H, m), 1.88 (1H, m), 1.65 (3H, s), 1.54 (1H, m), 1.39 (3H, s), 1.33 (3H, s).
18 3.4-dih dro-1- 4-methYlphenyll-4.4-dimethvl-6-acetylnaphthalene (Compound ~7 IOOG) 18 A solution of 1,2,3,4-tetrahydro-l-hydroxy-l-(4- methylphenyl)-4,4-is dimethyl-6-(2-(2-methyl-1,3- dioxolanyl))naphthalene (Compound 100F 160.0 mg, 0.52 mmol), p-toluenesulfonic acid monohydrate (4 mg) and 30 ml 21 benzene was refluxed for 12 hours. After cooling to room temperature, the 22 reaction mixture was diluted with ether (100 ml) and washed with 10%
23 aqueous sodium bicarbonate, water, and saturated aqueous NaCI. The 24 organic layer was dried over MgSO4 and the solvents were removed under 2s reduced pressure to give the title compound, which was isolated by column 28 chromatography (10% EtOAc-hexanes) as a yellow oil . 1H NMR (CDC13) : S
27 7.97(1H,d,J = 1.8Hz),7.67(1H,dd,J = 1.7,6.4Hz),7.22(4H,s),7.13 28 (1H,d,J=8.1Hz),6.10(1H,t,J=4.5Hz),2.59(3H,s),2.40(3H,s),2.38 29 (2H, d, J = 4.7 Hz), 1.38 (6H, s).

1 4-j3-oxo-3-(7.8-dihydro-5- 4-meth,ylphenyl -) 8.8-dimethyl-2-na hthalenxl)-1-2 propenvl]-benzoic acid (Compound 101) 3 To a solution of 78.7 mg (0.272 mmol) 3,4-dihydro- 1-(4-4 methylphenyl)-4,4-dimethyl-6-acetylnaphthalene (Compound 100G) in 4.0 ml of MeOH was added 53.1 mg (0.354 mmol) of 4-carboxy benzaldehyde, and e 80. mg (2.00 mmol; 2.0 ml of 1M aqueous NaOH). The resulting solution 7 was stirred at room temperature for 12 hours, concentrated under reduced 8 pressure, and the residual oil dissolved in EtOAc. The solution was treated 9 with 10% HCI, and the organic layer was washed with H20, and saturated lo aqueous NaCI, then dried over Na2SO4. Removal of the solvents under 11 reduced pressure gave the title compound as a colorless solid which was 12 purified by recrystallization from CH3CN. 1H NMR (acetone-d6) : 8 8.00 13 (7H,m),7.83(1H,d,J= 15.6Hz),7.24(4H,s),7.13(1H,d,J = 8.1 Hz), 14 6.12 (1H, t, J = 4.5 Hz), 2.42 (2H, d, J = 4.8 Hz), 2.38 (3H, s), 1.41 (6H, s).
3.4-dihvdro-l- hen,yl-4.4-dimethyl-6-acetvlnaphthalene (Compound 100w 16 To a solution of 508.0 mg (1.95 mmol) of 3,4- dihydro-4,4-dimethyl-6-17 (2-(2-methyl-1,3-dioxolanyl))- 1(2H)-naphthalenone (Compound 100E) in 10 18 ml of THF was added 496.2 mg (2.54 mmol; 2.54 ml of a 1 M solution in 19 Et20) of phenylmagnesium bromide. The resulting solution was heated to 2o reflux for 8 hours, H20 was added and heating continued for 30 minutes.
21 The THF was removed under reduced pressure and the aqueous residue was 22 extracted with EtOAc. The combined organic layers were dried (MgSO4), 23 concentrated under reduced pressure, and the title compound isolated from 24 the residue by column chromatography (10% EtOAc - hexanes) as a colorless oil. 1H NMR (CDC13) : 8 7.97 (1H, d, J = 1.8 Hz), 7.67 (1H, dd, J = 2.1, 26 8.0 Hz), 7.34 (5H, m), 7.10 (1H, d, J = 8.1 Hz), 6.12 (1H, d, J = 4.6 Hz), 27 2.59 (3H, s), 2.39 (2H, d, J = 4.8 Hz), 1.38 (6H, s).
28 4-[3-ox o=3-(7.8-dih, dro-5-phenyl-8.8-dimethyl-2-naphthalenyl)-l-propenvl]-2s benzoic acid (Compound 1U

1 To a solution of 115.0 mg (0.42 mmol) of 3,4- dihydro-l-phenyl-4,4-2 dimethyl-6-acetylnaphthalene (Compound 100H) and 65.0 mg (0.43 mmol) of 3 4-formyl- benzoic acid in 5.0 ml EtOH and 1.0 ml THF, was added 120.0 mg 4 (3.00 mmol; 3.0 ml of a 1 M aqueous solution) of NaOH. The resulting yellow solution was stirred at room temperature for 12 hours. The solution s was acidified with 6% aqueous HCl and extracted with EtOAc. The 7 combined organic layers were dried (MgSO4), concentrated under reduced 8 pressure, and the title compounds was isolated by column chromatography 9 (50% EtOAc - hexanes) as a pale yellow solid. 1H NMR (CDC13) : 8 8.13 1o (2H, d, J = 7.7 Hz), 8.04 (1H, s), 7.81 (1H, d, J = 15.5 Hz), 7.75 (3H, m), 11 7.60 (iH, d, J = 15.5 Hz), 7.35 (5H, m), 7.14 (1H, d, J = 8.1 Hz), 6.15 (1H, 12 t, J = 4.2 Hz), 2.41 (2H, d, J = 4.2 Hz), 1.41 (6H, s).
13 3-Fluoro-4-methoxythiophenol (Compound 200) 14 A flask containing Mg-turnings (651.9 mg, 26.8 g-atoms) was carefully flame dried under argon. Upon cooling to room temperature the flask was 16 charged with 14.0 mL of THF, the mixture was heated to reflux temperature 17 and 3-fluoro-4-methoxy-l-bromobenzene (5.OOg , 24.4 mmol) was slowly 18 added while the mixture was at reflux. After 6 hours the formation of the 19 Grignard reagent appeared complete. The resulting opaque orange solution was cooled to 0 C and sulfur (781.8 mg, 24.4 g-atoms) was added in three 21 portions over five minutes. After stirring overnight at room temperature the 22 brown mixture was carefully poured into an ice 10% aqueous HCl mixture.
23 Extraction with EtOAc was followed by washing the combined organic layers 24 with H20 and saturated aqueous NaCl and drying over MgSO4. Removal of the solvents under reduced pressure and distillation (bulb-to-bulb) of the 26 residue afforded 2.14 g (55%) of the title compound as a colorless oil, (bp 27 90-100 C / 5mm). IH NMR (300 MHz, CDC13) 8: 7.06 (2H, m), 6.85 (1H, t, 28 J = 8.6 Hz), 3.87 (3H, s), 3.43 (1H, s).
29 3-(3-fluoro-4-methoxv-uhenxlsulfan,ti-l)-3-methvl-buMic acid (C-ompound 201) 1 A heavy-walled screw cap tube was charged with 3-methyl-2-butenoic 2 acid (1.23 g, 12.33 mmol), 3-fluoro-4-methoxy thiophenol (Compound 200, 3 1.95 g, 12.33 mmol), and piperidine (314.8 mg, 3.70 mmol). This mixture was 4 heated to 105 C for 23 hours, cooled to room temperature and dissolved in EtOAc (100mL). The resulting solution was washed with 1M aqueous HCI, 6 H20, and saturated aqueous NaCI before being dried over Na2SO4.
7 Concentration of the dry solution under reduced pressure afforded 3.22 g of 8 a clear yellow oil which was used in the next reaction without further 9 purification. 1H NMR (300 MHz, CDC13) 8: 7.32 (2H, m), 6.94 (1H, t, J
1 o 8.4 Hz), 3.92 (3H, s), 2.54 (2H, s), 1.41 (6H, s).
11 3-(3-fluoro-4-methoxv_-phenylsulfanyl)-T..3-meth 1-b~! utvroyl chloride (Compound 12 2Q~2 13 To a solution of 3-(3-fluoro-4-methoxy-phenylsulfanyl)-3-methyl-14 butyric acid (Compound 201, 3.00 g, 11.6 mmol) in 40 mL of benzene at room temperature was added a solution of oxalyl chloride (2.21 g, 17.4 mmol) 16 in 5 mL of benzene over 30 minutes. After 3 hours, the solution was washed 17 with ice cold 5% aqueous NaOH (CAUTION: a large volume of gas is 18 released during this procedure), followed by ice cold H20, and finally 19 saturated aqueous NaCI. The solution was dried (Na2SO4) and concentrated 2o under reduced pressure to give 2.83 g of a clear yellow oil. This material was 21 used without further purification in the next step. 1H NMR (300 MHz, 22 CDC13) S: 7.27 (2H, m), 6.96 (1H, t, J = 8.4 Hz), 3.93 (3H, s), 3.13 (2H, s), 23 1.42 (6H, s).
24 6-Methoxy-7-fluoro-2.2-dimethXl-thiochroman-4-one (Compound 203) To a solution of the acyl chloride (Compound 202, 2.80 g, 10.1 mmol) 26 in 35 mL of CH2C12 at 0 C was added dropwise a solution of SnCl4 (2.64 g, 27 10.1 mmol) in 5 mL of CHZC12. After 2.5 hours the reaction was quenched 28 by the slow addition of 20 mL H20. The organic layer was washed with 1M
29 aqueous HCI, 5% aqueous NaOH, H20, and finally saturated aqueous NaCI

i before being dried over MgSO4. Concentration under reduced pressure 2 afforded 1.90 g (78%) the title compound as a tan solid. 1H NMR (300 MHz, 3 CDC13) 8: 7.72 (1H, d, J = 8.9 Hz), 6.95 (1H, d, J = 11.0 Hz), 3.91 (3H, s), 4 2.85 (2H, s), 1.47 (6H,s).
6-Hydroxti7-fluoro-2.2-dimethvlthiochroman-4-one (Compound 204) 6 To a solution of 6-methoxy-7-fluoro-2,2-dimethyl-thiochroman-4-one 7 (Compound 203, 1.75 g, 7.29 mmol) in 25 mL CH2C12 cooled to -23 C was 8 added BBr3 (5.46 g, 21.8 mmol; 21.8 mL of a 1M solution in CH2CI2) over a 9 10 minute period. After stirring for 23 hours at -230 C the solution was io warmed to 0 C for 3 hours, and then cooled to -78 C and quenched by the i i slow addition of 25 mL of H20. Upon warming to room temperature the 12 aqueous layer was extracted with CH2C12 and the combined organic layers 13 were washed with saturated aqueous NaHCO3, H20, and saturated aqueous 14 NaCI before being dried over MgSO4. Removal of the solvents under reduced pressure, followed by column chromatography (10 to 20% EtOAc /
16 hexanes) afforded 1.12 g (68%) of the title compound as a colorless solid.

17 NMR (300 MHz, CDC13) 8: 7.82 (1H, d J = 9.3 Hz), 6.97 (1H, d, J = 10.4 ia Hz), 5.54 (1H, s), 2.86 (2H,s), 1.46 (6H, s).
19 2.2-Dimethvl-4-oxo-7-fluoro-thiochroman-6-yd trifluoromethanesulfonate (Compound 205) 21 To a solution of 6-hydroxy-7-fluoro-2,2-dimethylthiochroman-4-one 22 (Compound 204, 1.12 g, 4.94 mmol) in 25.0 mL of anhydrous pyridine at 0 C
23 -was added trifluoromethanesulfonic anhydride (1.61 g, 5.68 mmol). After 18 24 hours at 0 C the solution was concentrated and the residual oil dissolved in Et20, washed with H20 followed by saturated aqueous NaCl, and dried over 26 MgSO4. Removal of the solvents under reduced pressure and column 27 chromatography (5 to 8% EtOAc / hexanes) afforded 1.28 g (72%) of the 28 title compound as a colorless solid. 1H NMR (300 MHz, CDC13) 8: 8.07 (1H, 29 d, J = 7.8 Hz), 7.13 (1H, d, J = 9.7 Hz), 2.89 (2H, s), 1.50 (6H, s).

1 2.2-Dimethyl-6-trimeth ylsilanylethynxl-7-fluoro-thiochroman-4-one 2 (Compound 206) 3 A solution of 2,2-dimethyl-4-oxo-7-fluoro-thiochroman-6-yl 4 trifluoromethanesulfonate (Compound 205, 1.28 g, 3.57 mmol) in 6.0 mL
Et3N and 15.0 mL DMF was sparged with argon for 15 minutes. To this 6 solution was added trimethylsilylacetylene (2.0 g, 20.4 mmol) and 7 bis(triphenylphosphine)-palladium(II) chloride (100.0 mg, 0.14 mmol). The 8 solution was heated to 95 C for 6 hours, cooled to room temperature and 9 stirred overnight. The solution was diluted with H20 and extracted with EtOAc. The combined organic layers were washed with H20, saturated 11 aqueous NaCI and dried over MgSO4. Concentration of the dry solution 12 under reduced pressure and isolation of the product by column 13 chromatography (3 to 5% EtOAc / hexanes) afforded 850.0 mg (78%) of the 14 title compound as an orange solid. 1H NMR (300 MHz, CDC13) 8: 8.22 (1H, d, J = 7.5 Hz), 6.92 (1H, d, J = 9.3 Hz), 2.85 (2H, s), 1.46 (6H, s), 0.25 (9H, 16 S).
17 6-Ethynyl-7-fluoro-2.2-dimethylthiochroman-4-one (A) ...(Compound 207).1 and 18 6-Eth p yl-7-methoxk-2.2-dimethkthiochroman-4-one (B) ompound 208) 19 A solution of 2,2-dimethyl-6-trimethylsilanylethynyl-7-fluror-thiochroman-4-one (Compound 206, 850.0 mg, 2.78mmol) and I{2C03 (180.0 21 mg, 1.30 mmol) in 20.0 mL MeOH was stirred overnight at room 22 temperature. The solution was concentrated to one-third of the original 23 volume, diluted with EtOAc, washed with H20 and saturated aqueous NaCl, 24 and dried over MgSO4. Removal of the solvent under reduced pressure followed by column chromatography (5 to 10% EtOAc / hexanes) of the 26 residual solid afforded 330.0 mg (51%) of Compound 207 as a yellow solid, 27 and 217.0 mg (32%) of Compound 208 as a yellow solid. (Compound 207):
28 1H NMR (300 MHz, CDC13) 8: 8.18 (1H, d, J = 7.5 Hz), 6.90 (1H, d, J = 8.3 29 Hz), 3.28 (1H, s), 2.81 (2H, s), 1.42 (6H, s). (Compound 208); 1H NMR (300 1 MHz, CDC13) 8: 8.19 (1H, s), 6.64 (1H, s), 3.90 (3H, s), 3.26 (1H, s), 2.80 2 (2H, s), 1.44 (6H, s).
3 EthXl 4-[(2.2-dilnethv1-4-oxo-7-fluoro-thiochroman-6 ;A)e hvnvl]-benzoate 4 (Compound 209.) A solution of 6-ethynyl-7-fluoro-2,2-dimethylthiochroman-4-one s(Compound 207, 330.0 mg, 1.41 mmol) and ethyl 4-iodobenzoate (392 mg, 7 1.41 mmol) in 10.0 mL Et3N was purged with argon for 15 minutes. To this 8 solution was added bis(triphenylphosphine)-palladium(II) chloride (247 mg, 9 0.35 mmol) and copper(I) iodide (67.0 mg, 0.35 mmol). After sparging for an 1 o additional 10 minutes with argon, the solution was stirred overnight at room 11 temperature. The reaction mixture was diluted with EtOAc and filtered 12 through a pad of Celite using an EtOAc wash. Concentration of the filtrate 13 under reduced pressure, followed by column chromatography (3 to 5%
14 EtOAc / hexanes) of the residual solid afforded 490.0 mg (91%) of the title 1s compound as a yellow solid. 1H NMR (300 MHz, CDC13) 8: 8.31 (1H, d, J
te 7.6 Hz), 8.04 (2H, d, J = 8.3 Hz), 7.60 (2H, d, J= 8.3.Hz), 7.00 (1H, d, J
17 9.3 Hz), 4.40 (2H, q, J = 7.1 Hz), 2.89 (2H, s), 1.50 (6H, s), 1.41 (3H, t, J
18 7.1 Hz).
19 Ethy14-[j2.2-dimethyl-4-oxo-7-methoxv-thiochroman-6-vl, ethyn,yl]-benzoate 20 (Compound 210) 21 A solution of 6-ethynyl-7-methoxy-2,2-d'unethylthiochroman-4-one 22 (Compound 208, 217.0 mg, 0.88 mmol) and ethyl 4-iodobenzoate (245.0 mg, 23 0.89 mmol) in 10.0 mL Et3N and 2 mL THF was purged with argon for 15 24 minutes. To this solution was added bis(triphenylphosphine)-palladium(II) 25 chloride (154.0 mg, 0.22 mmol) and copper(I) iodide (42.0 mg, 0.22 mmol).
2s After sparging for an additional 10 minutes with argon, the solution was 27 stirred overnight at room temperature. The reaction mixture was diluted 28 with EtOAc and filtered through a pad of Celite using an EtOAc wash.
29 Concentration of the filtrate under reduced pressure, followed by column 1 chromatography (5 to 10% EtOAc / hexanes) of the residual solid afforded 2 320.0 mg (92%) of the title compound as an orange solid. 1H NMR (300 3 MHz,CDC13)8:8.29(1H,s),8.03(2H,d,J=8.4Hz),7.60(2H,d,J=8.4 4 Hz), 6.70 (1H, s), 4.40 (2H, q, J = 7.1 Hz), 3.96 (3H, s), 2.86 (2H, s), 1.50 (6H, s), 1.41 (3H, t, J = 7.1 Hz).
s Ethy14-(2.2-dimethvl-4-trifluoromethanesulfonvloU-7-fluoro-(2H)-7 thiochromen-6-yleth~,n, -~l)-benzoate (Compound 211) 8 A solution of sodium bis(trimethylsilyl)amide (256.7 mg, 1.44 mmol) in 9 3.4 mL of THF was cooled to -78 C and a solution of ethyl 4-(2,2-dimethyl-1o 4-oxo-7-fluoro-thiochroman-6-ylethynyl)-benzoate (Compound 209, 460.0 mg, 11 1.20 mmol) in 3.0 mL was added slowly. After 30 minutes a solution of 2-12 [N,N-bis(trifluoromethanesulfonyl)amino]-5-pyridine (518.0 mg, 1.32 mmol) 13 in 2.0 mL of THF was added. After 5 minutes the solution was warmed to 14 room temperature and stirred for 6 hours. The reaction was quenched by the addition of saturated aqueous NHaCI and extracted with EtOAc. The 16 combined organic layers were washed with 5% aqueous NaOH and HZO
17 before being dried (MgSO4) and concentrated under reduced pressure. The 18 title compound, 549.0 mg (89%), was isolated by column chromatography (3-19 5% EtOAc / hexanes) as a pale-yellow solid. 1H NMR (300 MHz, CDC13) S:
2o 8.04 (2H, d, J = 8.4 Hz), 7.61 (2H, d, J = 8.4 Hz), 7.60 (1H, d, J = 6.7 Hz), 21 7.08 (1H, d, J = 9.0 Hz), 5.88 (1H, s), 4.40 (2H, q, J = 7.1 Hz), 1.53 (6H, s), 22 1.41 (3H, t, J = 7.1 Hz).
2s Eth,k-14-(2.2-dimeth,yl-4-trifluoromethanesulfonyloU-7-methoxy-(2H)-24 thiochromen-6 : l,y, ethynyl)-benzoate(Compound 2121 A solution of sodium bis(trimethylsilyl)amide (155.9 mg, 0.85 mmol) in 28 2.9 mL of THF was cooled to -78 C and a solution of ethyl 4-(2,2-dimethyl-27 4-oxo-7-methoxy-thiochroman-6-ylethynyl)-benzoate (Compound 210, 280.0 28 mg, 0.71 mmol) in 3.0 mL was added slowly. After 30 minutes a solution of 29 2-[N,N-bis(trifluoromethanesulfonyl)amino]-5-pyridine (355.0 mg, 0.85 mmol) 1 in 2.0 mL of THF was added. After 5 minutes the solution was warmed to 2 room temperature and stirred fo 6 hours. The reaction was quenched by the 3 addition of saturated aqueous NH4CI and extracted with EtOAc. The 4 combined organic layers were washed with 5% aqueous NaOH and H20 before being dried (MgSO4) and concentrated under reduced pressure. The e title compound, 300.0 mg (80%), was isolated by column chromatography 7 (15% EtOAc / hexanes) as an orange solid. 1H NMR (300 MHz, CDC13) 8:
8 8.02 (2H, d, J = 8.3 Hz), 7.61 (2H, d, J = 8.3 Hz), 7.57 (1H, s), 6.82 (1H, s), s 5.76 (1H, s), 4.38 (2H, q, J = 7.1 Hz), 3.92 (3H, s), 1.51 (6H, s), 1.40 (3H, t, J=7.1Hz).
ii Ethyl4-((4-(4-methylphenLl)-2.2-dimethyl-7-fluoro-(2H)-thiochromen-6-yl]-t2 ethynyl]-benzoate (Compound 213) 13 A solution of 4-methylbromobenzene (290.0 mg, 1.69 mmol) in 2.0 mL
14 of THF was cooled to -78 C and tert-butyllithium (217.2 mg, 3.39 mmol, 2.0 mL of a 1.7M solution in pentane) was added to give a yellow solution.
16 After 30 minutes a solution of ZnC12 (409.0 mg, 3.00 mmol) in 4.0 mL THF
17 was slowly added via cannula. The resulting solution was warmed to room 18 temperature and transferred via cannula to a solution of ethyl4-(2,2-1s dimethyl-4-trifluoromethanesulfonyloxy-7-fluoro-(2H)-thiochromen-6-2o ylethynyl)-benzoate (Compound 211, 158.0 mg, 0.31 mmol) and 21 tetrakis(triphenylphosphine)palladium(0) (24.0 mg, 0.02 mmol) in 2.0 mL
22 THF. This solution was heated to 50 C for 2 hours, cooled to room 23 temperature, and the reaction quenched by the addition of saturated aqueous 24 NH4C1. This solution was extracted with EtOAc and the combined organic layers washed with HZO and saturated aqueous NaCI before being dried 26 (MgSO4) and concentrated under reduced pressure. The title compound, 27 120.0 mg (86%), was isolated by column chromatography (3% EtOAc /
28 hexanes). 1H NMR (300 MHz, CDC13) 8: 8.00 (2H, d, J = 8.3 Hz), 7.54 (2H, 29 d,J=8.3Hz),7.27-7.13(6H,m),5.79(1H,s),4.38(2H,q,J=7.1Hz),2.42 1 (3H, s), 1.48 (6H, s), 1.40 (3H, t, J = 7.1 Hz).
2 Ethyl 4-[[4 (5-methyl-thiophen-2-,rl)-2.2-dimethyl-7-fluoro-(2H)-thiochromen-3 6-vffl-ethynv_1]-benzoate (Compound 214) 4 A solution of 2-methylthiophene (130.0 mg, 1.30 mmol) in 2.0 mL of THF was cooled to -78 C and n-butyllithium (83.3 mg, 1.30 mmol, 0.84 ml of 6 a 1.6M solution in hexanes) was added and the solution warmed to 0 C
7 during 1.5 hours. A solution of ZnC1Z (341.0 mg, 2.50 mmol) in 3.0 mL THF
s was slowly added via cannula. The resulting solution was warmed to room 9 temperature, stirred for 40 minutes, and transferred via cannula to a solution of ethyl 4-[(2,2-dimethyl-4-trifluoromethanesulfonyloxy-7-fluoro-(2H)-ii thiochromen-6-yl)ethynyl]-benzoate (Compound 211, 158.0 mg, 0.31 12 mmol) and tetrakis(triphenylphosphine) palladium(0) (24.0 mg, 0.02 mmol) in 13 2.0 mL THF. This solution was heated to 50 C for 2 hours, cooled to room 14 temperature, and the reaction quenched by the addition of saturated aqueous NH4C1. Thei solution was extracted with EtOAc and the combined organic 16 layers washed with H20 and saturated aqueous NaCI before being dried 17 (MgSO4) and concentrated under reduced pressure. The title compound, 18 70.0 mg (50%), was isolated by column chromatography (3% EtOAc /
19 hexanes) as a waxy solid. 'H NMR (300 MHz, CDC13) 8:8.01 (2H, d, J = 8.3 2o Hz), 7.61 (1H, d, J = 7.5 Hz), 7.58 (2H, d, J 8.3 Hz), 7.13 (1H, d, J = 7.2 21 Hz), 6.82 (1H, d, J = 4.4 Hz), 6.73 1H, d, J 4.4 Hz), 5.96 (1H, s), 4.39 22 (2H, q, J = 7.1 Hz), 2.52 (3H, s), 1.46 (6H, s), 1.41 (3H, t, J = 7.1 Hz).
23 Eth,y14-[[4-(4-meth,,-lphenyl)-2.2-dimethyl-7-methoxy-(2H)-thiochromen-6-vll 24 ethynyl]-benzoate (Compound 215) A solution of 4-methylbromobenzene (499.0 mg, 2.92 mmol) in 2.0 mL
26 of THF was cooled to -78 C and tert-butyllithium (374.2 mg, 5.84 mmol, 3.4 27 ml of a 1.7M solution in pentane) was added to give a yellow solution.
After 28 30 minutes a solution of ZnC12 (613.0 mg, 4.50 mmol) in 4.0 mL THF was 29 slowly added via cannula. The resulting solution was warmed to room 1 temperature and transferred via cannula to a solution of ethyl 4-(2,2-2 dimethyl-4-trifluoromethanesulfonylo)cy-7-methoxy-(2H)-thiochromen-6-3 ylethynyl)-benzoate (Compound 212, 285.0 mg, 0.54 mmol) and 4 tetrakis(triphenylphosphine)palladium(0) (24.0 mg, 0.02 mmol) in 2.0 mL
THF. This solution was heated to 50 C for 2 hours, cooled to room s temperature, and the reaction quenched by the addition of saturated aqueous 7 NH4Cl. The solution was extracted with EtOAc and the combined, organic 8 layers were washed with H20 and saturated aqueous NaC1 before being dried s(MgSO4) and concentrated under reduced pressure. The title compound, 1 o 200.0 mg (79%), was isolated by column chromatography (2-5% EtOAc /
11 hexanes) as a colorless solid. 1H NMR (300 MHz, CDC13) 8: 7.98 (2H, d, J
12 8.3 Hz), 7.54 (2H, d, J = 8.3 Hz), 7.23-7.21 (5H, m), 5.71 (1H, s), 4.38 (2H, 13 q, J = 7.1 Hz), 3.95 (3H, s), 2.41 (3H, s), 1.49 (6H, s), 1.40 (3H, t, J =
7.1 14 Hz).
3-(4-methoxy-phenylsulfanvl)-3-methvl-butvric acid (Compound 216) 16 A heavy-walled screw cap tube was charged with 3-methyl-2-butenoic 17 acid (13.86g, 138.4 mmol), 4-methoxy thiophenol (20.0g, 138.4 mmol), and 18 piperidine (3.45 g, 41.6 mmol). This mixture was heated to 105 C for 32 19 hours, cooled to room temperature and dissolved in EtOAc (700mL). The 2o resulting solution was washed with 1M aqueous HCI, H2O, and saturated 21 aqueous NaCI before being dried over NaZSO4. Concentration of the dry 22 solution under reduced pressure afforded an oil which upon standing in the 23 freezer provided a crystalline solid. The title compound was isolated as pale-24 yellow crystals by washing the crystalline solid with pentane. (27.33 g, 82%).
iH NMR (300 MHz, CDC13) 8: 7.48 (2H, d, J = 9.0 Hz), 6.89 (2H, d, J = 8.9 26 Hz), 3.83 (3H, s), 2.54 (2H, s), 1.40 (6H, s).
27 3-t4-m ethoxy-phenvlsulfanyl, -3-methyl-but royl chloride (Compound 217) 28 To a solution of 3-(4-methoxy-phenylsulfanyl)-3-methyl-butyric acid 29 (Compound 216, 20.0 g, 83.2 mmol) in 250 mL of benzene at room 1 temperature was added a solution of oxalyl chloride (15.84g, 124.8 mmol) in 2 10 mL of benzene over 30 minutes. After 4 hours the solution was washed 3 with ice cold 5% aqueous NaOH (CAUTION: a large volume of gas is 4 released during this procedure), followed by ice cold H20, and finally saturated aqueous NaCI. The solution was dried (Na2SO4) and concentrated s under reduced pressure to give a clear yellow oil. This material was used 7 without further purification in the next step. 1H NMR (300 MHz, CDC13) S:
8 7.45 (2H, d, J = 8.8 Hz), 6.90 (2H, d, J = 8.8 Hz), 3.84 (3H, s), 3.12 (2H, s), 9 1.41 (6H, s).
6-Methoxy-2.2-dimethvl-thiochroman-4-one (Compound 218) 11 To a solution of the acyl chloride (Compound 217, 21.5g, 83.2 mmol) 12 in 250 mL of CH2C12 at 0 C was added dropwise a solution of SnCl4 (21.7g, 13 83.2 mmol) in 30 mL of CH2Cl2. After 2 hours the reaction was quenched by 14 slow addition of 150 mL HZO. The organic layer was washed with 1M
aqueous HCI, 5% aqueous NaOH, HZO, and finally saturated aqueous NaCl 16 before being dried over MgSO4. Concentration under reduced pressure and 17 vacuum distillation of the residual oil (Bulb-to-bulb, 125-135 C, 5 mm/Hg) ls afforded 14.48 g (78%) of the title compound as a pale-yellow oil. zH NMR
19 (300 MHz, CDC13) 8: 7.62 (1H, d, J = 2.9 Hz), 7.14 (1H, d, J = 8.6 Hz), 7.03 2o (1H, dd, J = 2.8, 8.3 Hz), 3.83 (3H, s), 2.87 (211, s), 1.46 (6H, s).
21 6-Hvdroxy-2s2-dimethylthiochroman-4-one(Compound 219) 22 To a solution of 6-methoxy-2,2-dimethyl-thiochroman-4-one 23 (Compound 218, 6.0 g, 27 mmol) in 50 mL CHZCl2 cooled to -23 C was 24 added BBr3 (20.0 g, 80.0 mmol; 80.0 mL of a 1M solution in CHZCIZ) over a 20 minute period. After stirring for 5 hours at -23 C the solution was cooled 26 to -78 C and quenched by the slow addition of 50 mL of H20. Upon 27 warming to room temperature the aqueous layer was extracted with CH2C12 28 and the combined organic layers were washed with saturated aqueous 29 NaHCO3, HZO, and saturated aqueous NaCI before being dried over MgSO4.

1 Removal of the solvents under reduced pressure gave a green-brown solid 2 which upon recrystalization (Et20 / hexanes) afforded 2.25 g (40%) of the 3 title compound as a light brown solid. IH NMR (300 MHz, CDC13) 8:7.63 4 (1H,d,J = 2.8Hz),7.15 (1H, d, J = 8.5 Hz), 7.01 (1H,dd,J = 2.8, 8.5 Hz), 2.87 (2H, s), 1.46 (6H, s).
6 2.2-Dimethyl-4-oxo-thiochroman-6:y1 trifluoromethanesulfonate ,Compound 7 ~
8 To a solution of 6-hydroxy-2,2-dimethylthiochroman-4-one (Compound 9 219, 165.0 mg, 0.79 mmol) in 5.0 mL of anhydrous pyridine at 0 C was lo added trifluoromethanesulfonic anhydride (245.0 mg, 0.87 mmol). After 4 11 hours at 0 C the solution was concentrated and the residual oil dissolved in 12 Et20, washed with H20 followed by saturated aqueous NaCI, and dried over 13 MgSO4. Removal of the solvents under reduced pressure and column 14 chromatography (5% EtOAc / hexanes) afforded 126.0 mg (47%) of the title i s compound as a colorless solid. 1H NMR (300 MHz, CDCl3) 8: 7.97 (1H, s), 16 7.32 (2H, s), 2.90 (2H, s), 1.49 (6H, s).
17 2.2-Dimethyl-6-trimethylsilan ly ethyMI-thiochroman-4-one (Compound 221) 18 A solution of 2,2-d'nnethyl-4-oxo-thiochroman-6-yl 19 trifluoromethanesulfonate (Compound 220, 2.88 g, 8.50 mmol) in 10 mL Et3N
2o and 20.0 mL DMF was sparged with argon for 10 minutes. To this solution 21 was added trimethylsilylacetylene (4.15 g, 42.0 mmol) and 22 bis(triphenylphosphine)-palladium(II) chloride (298.0 mg, 0.425 mmol). The 23 solution was heated to 95 C for 5 hours, cooled to room temperature, and 24 diluted with H20. Extraction with EtOAc was followed by washing the 25 combined organic layers with H20 and saturated aqueous NaCI and drying 26 over MgSO4. Concentration of the dry solution under reduced pressure and 27 isolation of the product by column chromatography (3% EtOAc / hexanes) 28 afforded 2.23 g (91%) of the title compound as an orange oil. 1H NMR (300 29 MHz, CDC13) S: 8.18 (1H, d, J= 1.9 Hz), 7.34 (1H, dd, J = 1.9, 8.1 Hz), 7.15 WO 99l33821 PCT/US98/27314 t(1H, d, I= 8.1 Hz), 285 (2H, s), 1.45 (6H, s), 0.23 (911, s).
2 6-Ethymyl-2.2-dimethvlthiochroman-4-one (CoMpound 222) 3 A solution of 2,2-dimethyl-6-trimethylsilanylethynyl-thiochroman-4-one 4 (Compound 221, 110.0 mg. 0.38 mmol) and I{zCO, (40.0 mg, 0.29 mmol) in s 10.0 mL MeOH was stirred overnight at room temperature. The solution 6 was diluted with H=O and extracted with Et20. The combined organic layers 7 were washed with H~O and saturated aqueous Na(3 and dried over MgSO4.
a Removal of the sohrent under reduced pressure afforded 81 mg (99%) of the 9 title compound as an orange oil. IH NMR (300 MHz, CDC13) 8:8.20 (1H, d, J
to = 1.9 Hz), 7.46 (1H, dd, J= 1.9, 8.1 Hz), 7.18 (1H, d, J= 8.1 Hz), 3.08 (iH, ii s), 2.86 (2H, s), 1.46 (6H, s).
12 Ethyl 4-j( 2-d'une 1-4-oxo-thiochroman-6-yl ethmvll- nzoate (Compou d 13 =
14 A solution of 6-eth3myl-2,2-dimethylthiochroman-4-one (Compound is 222, 82.0 mg, 0.38 mmol) and ethyl4-iodobenzoate (104.9 mg, 0.38 mmol) in ls 5.0 mL Et3N was purged with argon for 10 minutes. To this solution were 17 added bis(triphenylphosphine)-palladium(II) chloride (88.0 mg, 0.12 mmol) is and copper(I) iodide (22.9 mg, 0.12 mmol). After sparging for an additional i9 5 minutes with argon, the solution was stirred overnight at room temperature.
2o The reaction mixture was filtered through a pad of Celite using an EtzO
21 wash. Concxntration of the filtrate under reduced pressure, followed by 22 column chromatography of the residual solid, afforded 100 mg (72%) of the 2s title compound as a yeAow solid. IH NMR (300 MHz, CDC13) 8: 8.25 (iH, d, 24 J = 1.8 Hz), 8.00 (2H, d, J = 8.4 Hz), 7.55 (2H, d, J = 8.4 Hz), 7.53 (1H, dd, 25 J = 1.8,8.2Hz),7.21(1H,d,J=8.2Hz),4.37(214,q,J=7.114z),2.88 26 (2H, s), 1.47 (6H, s), 1.39 (31L t, J= 7.1 Hz).
27 EtbA 4:(l 2-dimet X(-4-trifluoromethanesulfonvloxv-(M;tbochromen-6-2s y1)et13Y1ayi1- nzaate ,Qm,FQund 2241, 29 A solution of sodium bis(trimethylsilyl)amide (1.12 g, 6.13 mmol) in * Trade-mark 1 16.2 mL of THF was cooled to -78 C and a solution of ethyl 4-(2,2-dimethyl-2 4-oxo-thiochroman-6-ylethynyl)-benzoate (Compound 223, 1.86g, 5.10 mmol) 3 in 15.0 mL was added slowly. After 30 minutes a solution of 2-[N,N-4 bis(trifluoromethanesulfonyl)amino]-5-pyridine (2.40 g, 6.13 mmol) in 10 mL
of THF was added. After 5 minutes the solution was warmed to room 6 temperature and stirred overnight. The reaction was quenched by the 7 addition of saturated aqueous NH4C1 and extracted with EtOAc. The s combined organic layers were washed with 5% aqueous NaOH and H20 9 before being dried (MgSO4) and concentrated under reduced pressure. The 1 o title compound, 1.53 g (61%), was isolated by column chromatography (2%
ii EtOAc / hexanes) as a yellow solid. 1H NMR (300 MHz, CDC13) 8: 8.03 (2H, 12 d, J = 8.4 Hz), 7.61 (1H, d, J = 1.8 Hz), 7.59 (2H, d, J = 8.4 Hz), 7.41 (1H, 13 dd, J = 1.8,8.1Hz),7.29(1H,d,J=8.1Hz),5.91(1H,s),4.39(2H,q,J=
14 7.1Hz),1.53(6H,s),1.41(3H,t,J=7.1Hz).
Ethyl 4-[[4-phenyl-2.2-dimethyl-(2...~I,)-thiochromen-6-yl]-ethvnyl]-benzoate 1 s (Compound 225) 17 A solution of bromobenzene (190.0 mg, 1.18 mmol) in 2.0 mL of THF
18 was cooled to -78 C and tert-butyllithium (151.2 mg, 2.36 mmol, 1.4 mL of a 19. 1.7M solution in pentane) was added to give a yellow solution. After 30 minutes a solution of ZnC12 (225.0 mg, 1.4 mmol) in 4.0 mL THF was slowly 21 added via cannula. The resulting solution was warmed to room temperature 22 and transferred via cannula to a solution of ethyl4-(2,2-dimethyl-4-23 trifluoromethanesulfonyloxy-(2H)-thiochromen-6-ylethynyl)-benzoate 24 (Compound 224, 200.0 mg, 0.40 mmol) and tetrakis(triphenylphosphine)palladium(0) (24.0 mg, 0.02 mmol) in 2.0 mL
26 THF. This solution was heated to 50 C for 2 hours, cooled to room 27 temperature, and the reaction quenched by the addition of saturated aqueous 28 NH4C1. The solution was extracted with EtOAc and the combined organic 29 layers were washed with HZO and saturated aqueous NaCI before being dried 1 (MgSO4) and concentrated under reduced pressure. The title compound, 2 155.0 mg (91%), was isolated by column chromatography (5% EtOAc /
3 hexanes) as a colorless solid. 1H NMR (300 MHz, CDC13) 8: 7.99 (2H, d, J
4 8.4 Hz), 7.52 (2H, d, J = 8.4 Hz), 7.42-7.24 (8H, m), 5.78 (1H, s), 4.38 (2H, q, J = 7.1 Hz), 1.50 (6H, s), 1.40 (3H, t, J = 7.1 Hz).
6 Ethv14-[[4-(4-methvlplienyl)-2.2-dimethvl-(2H)-thiochromen-6-vl]-ethynyll-7 benzoate (,Com , ound =
8 A solution of 4-methylbromobenzene (203.0 mg, 1.15 mmol) in 2.0 mL
9 of THF was cooled to -78 C and tert-butyllithium (147.4 mg, 2.30 mmol, 1.4 io ml of a 1.7M solution in pentane) was added to give a yellow solution.
After >> 30 minutes a solution of ZnCl2 (219.4 mg, 1.4 mmol) in 4.0 mL THF was 12 slowly added via cannula. The resulting solution was warmed to room 13 temperature and transferred via cannula to a solution of ethyl 4-(2,2-14 dimethyl-4-trifluoromethanesulfonyloxy-(2H)-thiochromen-6-ylethynyl)-1s benzoate (Compound 224, 230.0 mg, 0.46 mmol) and 16 tetrakis(triphenylphosphine)palladium(0) (24.0 mg, 0.02 mmol) in 2.0 mL
17 THF. This solution was heated to 50 C for 2 hours, cooled to room is temperature, and the reaction quenched by the addition of saturated aqueous 19 NH4Cl. The solution was extracted with EtOAc and the combined organic 2o layers were washed with H20 and saturated aqueous NaCI before being dried 21 (MgSO4) and concentrated under reduced pressure. The title compound, 22 165.0 mg (82%), was isolated by column chromatography (5% EtOAc /
23 hexanes) as a colorless solid. 1H NMR (300 MHz, CDC13) 8: 7.98 (2H, d, J
.24 8.4 Hz), 7.51 (2H, d, J = 8.4 Hz), 7.35-7.21 (7H, m), 5.84 (1H, s), 4.38 (2H, 25 q, J = 7.1 Hz), 2.42 (3H, s), 1.48 (6H, s), 1.40 (3H, s).
26 Eth,}l 4-[j4-(4-eth,}~lphenXl)-2.2-dimeth,yl-(2H)-thiochromen-6-yl]-ethvnvll 27 benzoate (Compound 2M
28 A solution of 4-ethylbromobenzene (670.9 mg, 3.63 mmol) in 4.0 mL
29 of THF was cooled to -78 C and tert-butyllithium (464.5 mg, 7.25 mmol, 4.26 1 mL of a 1.7M solution in pentane) was added to give a yellow solution.
2 After 30 minutes a solution of ZnCl2 (658.7 mg, 4.83 mmol) in 8.0 mL THF
3 was slowly added via cannula. The resulting solution was warmed to room 4 temperature and transferred via cannula to a solution of ethyl 4-(2,2-dimethyl-4-trifluoromethanesulfonyloxy-(2H)-thiochromen-6-ylethynyl)-s benzoate (Compound 224, 1.20 g, 2.42 mmol) and 7 tetrakis(triphenylphosphine)palladium(0) (111.7 mg, 0.097 mmol) in 8.0 mL
8 THF. This solution was heated to 50 C for 1 hour, cooled to room 9 temperature, and the reaction quenched by the addition of saturated aqueous io NH4C1. The solution was extracted with EtOAc and the combined organic 11 layers were washed with H20 and saturated aqueous NaCI before being dried 12 (MgSO4) and concentrated under reduced pressure. The title compounds 13 was isolated by column chromatography (5% EtOAc / hexanes) as a colorless 14 oil. 1H NMR (300 MHz, CDC13) 8: 7.99 (2H, d, J = 8.2 Hz), 7.52 (2H, d, J
is 8.4 Hz), 7.40 (5H, m), 7.35 (2H, m), 5.85 (1H, s), 4.38 (2H, q, J = 7.1 Hz), t s 2.72 (2H, q, J = 7.6 Hz), 1.48 (6H, s), 1.40 (3H, t, J 7.1 Hz), 1.30 (3H, t, J
17 = 7.6 Hz).
18 Ethvl4-[[4-(4-isopropyl,pheMl)-2.2-dimethyl-(2H)-thiochromen-6-~Ll]-ethvn,vll,-1s benzoate (Compound 228) 20 A solution of 4-isopropylbromobenzene (161.0 mg, 0.81 mmol) in 2.0 21 mL of THF was cooled to -78 C and tert-butyllithium (103.8 mg, 1.62 mmol, 22 0.95 ml of a 1.7M solution in pentane) was added to give a yellow solution.
23 After 30 minutes a solution of ZnCl2 (175.5 mg, 1.3 mmol) in 4.0 mL THF
24 was slowly added via cannula. The resulting solution was warmed to room 25 temperature and transferred via cannula to a solution of ethyl 4-(2,2-26 dimethyl-4-trifluoromethanesulfonyloxy-(2H)-thiochromen-6-ylethynyl)-27 benzoate (Compound 224, 160.0 mg, 0.32 mmol) and 28 tetrakis(triphenylphosphine) palladium(0) (24.0 mg, 0.02 mmol) in 2.0 mL
29 THF. This solution was heated to 50 C for 2 hours, cooled to room 1 temperature, and the reaction quenched by the addition of saturated aqueous 2 NH4Cl. This solution was extracted with EtOAc and the combined organic 3 layers were washed with H20 and saturated aqueous NaC1 before being dried 4(MgSO4) and concentrated under reduced pressure. The title compound, 128.0 mg (85%), was isolated by column chromatography (5% EtOAc /
6 hexanes) as a colorless oil. 1H NMR (300 MHz, CDC13) S: 7.99 (2H, d, J
7 8.4 Hz), 7.53 (2H, d, J = 8.1 Hz), 7.38-7.22 (7H, m), 5.86 (1H, s), 4.38 (2H, 8 q, J = 7.1 Hz), 2.98 (1H, hept, 7.0 Hz), 1.49 (6H, s), 1.43 (3H, t, J = 7.1 Hz), 9 1.32 (6H, d, J = 7.0 Hz).
1 o Ethyl 4-([4-(4-tert-butvlphenyl)-2.2-dimethvl-(2H)-thiochromen-6-vl]-ethxn~-Il 11 benzoate (Compound 229) 12 A solution of 4-tert-butylbromobenzene (149.0 mg, 0.70 mmol) in 2.0 13 mL of THF was cooled to -78 C and tert-butyllithium (92.6 mg, 1.44 mmol, 14 0.85 ml of a 1.7M solution in pentane) was added to give a yellow solution.
After 30 minutes a solution of ZnC12 (133.0 mg, 0.98 mmol) in 4.0 mL THF
16 was slowly added via cannula. The resulting solution was warmed to room 17 temperature and transferred via cannula to a solution of ethyl 4-(2,2-18 dimethyl-4-trifluoromethanesulfonyloxy-(2H)-thiochromen-6-ylethynyl)-1s benzoate (Compound 224, 140.0 mg, 0.28 mmol) and tetrakis(triphenylphosphine) palladium(0) (24.0 mg, 0.02 mmol) in 2.0 mL
21 THF. This solution was heated to 50 C for 2 hours, cooled to room 22 temperature, and the reaction quenched by the addition of saturated aqueous 23 NH4C1. The solution was extracted with EtOAc and the combined organic 24 layers were washed with H20 and saturated aqueous NaCI before being dried (MgSO4) and concentrated under reduced pressure. The title compound, 26 94.0 mg (70%), was isolated by column chromatography (5% EtOAc /
27 hexanes) as a colorless solid. 'H NMR (300 MHz, CDC13) S: 7.98 (2H, d, J
28 8.3 Hz), 7.52 (2H, d, J = 8.3 Hz), 7.43-7.22 (7H, m), 5.86 (1H, s), 4.38 (2H, 29 q, J = 7.1 Hz), 1.47 (6H, s), 1.40 (3H, t, J = 7.1 Hz), 1.38 (9H, s).

i EthY14-[[4-(5-methvl-thiophen-2-vl)-2 2-dimethyl-(2I )-thiochromen-6-YI1 2 ethynyl]-benzoate (,Compound 230) 3 A solution of 2-methylthiophene (100.0 mg, 1.00 mmol) in 2.0 mL of 4 THF was cooled to -78 C and n-butyllithium (64.0 mg, 1.00 mmol, 0.63 ml of a 1.6M solution in hexanes) was added and the solution warmed to 0 C
6 during 1.5 hours. A solution of ZnC12 (218.0 mg, 1.60 mmol) in 3.0 mL THF
7 was slowly added via cannula. The resulting solution was warmed to room 8 temperature, stirred for 40 minutes, and transferred via cannula to a solution s of ethyl 4-[(2,2-dimethyl-4-trifluoromethanesulfonyloxy-(2H)-thiochromen-6-i o yl)ethynyl]-benzoate (Compound 224, 200.0 mg, 0.40 mmol) and 11 tetrakis(triphenylphosphine) palladium(0) (24.0 mg, 0.02 mmol) in 2.0 mL
12 THF. This solution was heated to 50 C for 2 hours, cooled to room 13 temperature, and the reaction quenched by the addition of saturated aqueous 14 NH4C1. The solution was extracted with EtOAc and the combined organic layers were washed with H20 and saturated aqueous NaCI before being dried 18 (MgSO4) and concentrated under reduced pressure. The title compound, 17 170.0 mg (96%), was isolated by column chromatography (5% EtOAc /
18 hexanes) as a pale-yellow solid. 1H NMR (300 MHz, CDC13) 8: 8.01 (2H, d, J
is = 8.3 Hz), 7.63 (1H, s), 7.55 (2H, d, J = 8.3 Hz), 7.35 (2H, s), 6.82 (1H, d, J
2o = 3.5 Hz), 6.72 (1H, m), 6.00 (1H, s), 4.38 (2H, q, J = 7.1 Hz), 2.52 (3H, s), 21 1.46 (6H, s), 1.40 (2H, t, J = 7.1 Hz).
22 Ethy-I 4-[[4-(5-etb1-y thiophen-2-xl)-2.2-dimethyl-(2I ,)-thiochromen-6-vll-2s gthynyl]-benzoate (Comnound 231) 24 A solution of 2-ethylthiophene (112.0 mg, 1.00 mmol) in 2.0 mL of THF was cooled to -78 C and n-butyllithium (64.0 mg, 1.00 mmol, 0.63 ml of 26 a 1.6M solution in hexanes) was added and the solution warmed to 0 C
27 during 1.5hours. A solution of ZnC12 (218.0 mg, 1.60 mmol) in 3.0 mL THF
28 was slowly added via cannula. The resulting solution was warmed to room 29 temperature, stirred for 40 minutes, and transferred via cannula to a solution 1 of ethyl 4-[(2,2-dimethyl-4-trifluoromethanesulfonyloxy-(2H)-thiochromen-6-2 yl)ethynyl]-benzoate (Compound 224, 200.0 mg, 0.40 mmol) and 3 tetrakis(triphenylphosphine) palladium(0) (24.0 mg, 0.02 mmol) in 2.0 mL
4 THF. This solution was heated to 50 C for 2 hours, cooled to room temperature, and the reaction quenched by the addition of saturated aqueous 6 NH4C1. The solution was extracted with EtOAc and the combined organic 7 layers were washed with H20 and saturated aqueous NaCI before being dried 8 (MgSO4) and concentrated under reduced pressure. The title compound, 9 29.0 mg (16%), was isolated by HPLC (1.25% EtOAc / hexanes) as a pale-1 o yellow solid. 1H NMR (300 MHz, CDC13) 8: 8.01 (2H, d, J = 8.4 Hz), 7.65 11 (1H, s), 7.55 (2H, d, J = 8.4 Hz), 7.35 (2H, s), 6.84 (1H, d, J = 3.5 Hz), 6.77 12 (1H,m),6.02(1H,s),4.39(2H,q,J=7.1Hz),2.88(2H,q,J=7.6Hz), 13 1.46 (6H, s), 1.41 (2H, t, J = 7.1 Hz), 1.35 (2H, t, J 7.6 Hz), 14 Ethyl 4-[[~5-tert-butvl-thiophen-2 y,~)-2 2-dimethy~2H)-thiochromen-6-vll-ethyn,yl]-benzoate (Compound 232) 16 A solution of 2-tert-butylthiophene (105.0 mg, 0.75 mmol) in 2.0 mL of 17 THF was cooled to -78 C and n-butyllithium (48.1 mg, 0.75 mmol, 0.47 ml of 18 a 1.6M solution in hexanes) was added and the solution warmed to 0 C
19 during 1.5 hours. A solution of ZnC12 (163.2 mg, 1.20 mmol) in 3.0 mL THF
was slowly added via cannula. The resulting solution was warmed to room 21 temperature, stirred for 40 minutes, and transferred via cannula to a solution 22 of ethyl 4-[(2,2-dimethyl-4-trifluoromethanesulfonyloxy-(2H)-thiochromen-6-2a yl)ethynyl]-benzoate (Compound 224, 200.0 mg, 0.40 mmol) and 24 tetrakis(triphenylphosphine) palladium(0) (20.0 mg, 0.02 mmol) in 2.0 mL
THF. This solution was heated to 50 C for 2 hours, cooled to room 26 temperature, and the reaction quenched by the addition of saturated aqueous 27 NH4C1. The solution was extracted with EtOAc and the combined organic 28 layers were washed with H20 and saturated aqueous NaCI before being dried 29 (MgSO4) and concentrated under reduced pressure. The title compound, 1 110.0 mg (76%), was isolated by column chromatography (5% EtOAc /
2 hexanes) as a colorless solid. 1H NMR (300 MHz, CDC13) 8: 8.01 (2H, d, J
3 8.3 Hz), 7.68 (1H, s), 7.56 (2H, d, J = 8.3 Hz), 7.36 (2H, m), 6.82 (2H, m), 4 6.04 (1H, s), 4.39 (2H, q, J = 7.1 Hz), 1.46 (6H, s), 1.43 (9H, s), 1.41 (2H, t, J=7.1Hz).
6 Ethv14-((4-(6-methvl-pyridin-3 vl)-2 2-dimethyl-(2H)-thiochromen-6-vl-7 ethVnyll-benzoate (Compound 233) 8 A solution of 4-bromo-2-methylpyridine (220.0 mg, 1.28 mmol) in 2.0 9 mL of THF was cooled to -78 C and tert-butyllithium (164.0 mg, 2.56 mmol, 1 o 1.5 ml of a 1.7M solution in pentane) was added to give a red-orange 11 solution. After 30 minutes a solution of ZnCl2 (348.0 mg, 2.56 mmol) in 4.0 12 mL THF was slowly added via cannula. The resulting solution was warmed 13 to room temperature, stirred for 40 minutes, and transferred via cannula to a 14 solution of ethyl 4-(2,2-d'unethyl-4-trifluoromethanesulfonyloxy-(2H)-thiochromen-6-ylethynyl)-benzoate (Compound 224, 172.0 mg, 0.35 mmol) 16 and tetrakis(triphenylphosphine) palladium(0) (24.0 mg, 0.02 mmol) in 2.0 17 mL THF. This solution was heated to 50 C for 2 hours, cooled to room 1 s temperature, and the reaction quenched by the addition of saturated aqueous 1 s NH4C1. The solution was extracted with EtOAc and the combined organic layers were washed with H20 and saturated aqueous NaCI before being dried 21 (MgSO4) and concentrated under reduced pressure. The title compound, 22 109.0 mg (72%) was isolated by column chromatography (5% EtOAc /
23 hexanes) as a pale-yellow solid. 1H NMR (300 MHz, CDC13) 8: 8.45 (1H, d, J
24 = 2.0 Hz), 8.00 (2H, d, J = 8.5 Hz), 7.52 (2H, d, J= 8.5 Hz), 7.49 (1H, dd, J
= 2.3, 8.1 Hz), 7.36 (2H, m), 7.20 (1H, d, J = 8.1 Hz), 7.16 (1H, d, J = 1.5 26 Hz), 5.86 (1H, s), 4.38 (2H, q, J = 7.1 Hz), 2.63 (3H, s), 1.50 (6H, s), 1.40 27 (3H, t, J = 7.1 Hz).
28 Ethyl 4[(2 2-d'unethyl-4-oxo-thiochroman-6-y_, ethavl]-2-fluorobenzoate 29 (Compound 234) 1 A solution of 6-ethynyl-2,2-dimethylthiochroman-4-one (Compound 2 222, 1.37g, 6.34 mmol) and ethyl 2-fluro-4-iodobenzoate (1.86g, 6.34mmol) in 3 40.0 mL Et3N was purged with argon for 20 minutes. To this solution was 4 added bis(triphenylphosphine)-palladium(II) chloride (1.05 g, 1.5 mmol) and copper(I) iodide (286.0 mg, 1.5 mmol). After sparging for an additional 10 6 minutes with argon, the solution was stirred overnight at room temperature.
7 The reaction mixture was filtered through a pad of Celite using an Et20 8 wash. Concentration of the fliltrate under reduced pressure, followed by 9 column chromatography (5% EtOAc / hexanes) of the residual solid afforded 1 o 1.88 g (78%) the title compound as a yellow solid. 1H NMR (300 MHz, 11 CDC13)8:8.27(1H,d,J = 1.9Hz),7.92(1H,d,J = 7.8Hz),7.52(1H,dd,J
12 = 1.9, 8.2 Hz), 7.36-7.24 (3H, m), 4.41 (2H, q, J = 7.1 Hz), 2.90 (2H, s), 1.50 13 (6H, s), 1.41 (3H, t, J = 7.1 Hz).
14 Ethyl 4-(2 2-dimethyl-4-trifluoromethanesulfonyloxy-(2H)-thiochromen-6-1 s vleth r~nvl)-2-fluorobenzoate (Compound 2351 16 A solution of sodium bis(trimethylsilyl)amide (0.82g, 5.90 mmol) in 17 16.2 mL of THF was cooled to -78 C and a solution of ethyl 4-[(2,2-18 dimethyl-4-oxo-thiochroman-6-yl)ethynyl]-2-flourobenzoate (Compound 234, 19 1.88g, 4.91 mmol) in 15.0 mL was added slowly. After 30 minutes a solution 20 of 2-[N,N-bis(trifluoromethanesulfonyl)amino]-5-pyridine (2.32 g, 5.90 mmol) 21 in 10 mL of THF was added. After 5 minutes the solution was warmed to 22 room temperature and stirred overnight. The reaction was quenched by the 23 addition of saturated aqueous NH4C1 and extracted with EtOAc. The 24 combined organic layers were washed with 5% aqueous NaOH and H20 25 before being dried (MgSO4) and concentrated under reduced pressure. The 26 title compound, 1.80 g (71%), was isolated by column chromatography (5%
27 EtOAc / hexanes) as a yellow solid. 1H NMR (300 MHz, CDC13) 8: 7.93 (1H, 28 d, J = 7.8 Hz), 7.61 (1H, d, J = 1.6 Hz), 7.43-7.27 (4H, m), 5.92 (1H, s), 4.41 29 (2H, q, J = 7.1 Hz), 1.53 (6H, s), 1.42 (3H, t, J = 7.1 Hz).

1 Ethyl 4-([4-(4-methvlphenyl)-2.2-dimethyl-(,2H)-thiochromen-6-A]-et )Myl]-2-2 fluorobenzoate (Compound 2361 3 A solution of 4-methylbromobenzene (280.0 mg, 1.64 mmol) in 2.0 mL
4 of THF was cooled to -78 C and tert-butyllithium (209.5 mg, 3.27 mmol, 1.9 mL of a 1.7M solution in pentane) was added to give a yellow solution.
s After 30 minutes a solution of ZnCl2 (680.0 mg, 5.0 mmol) in 4.0 mL THF
7 was slowly added via cannula. The resulting solution was warmed to room 8 temperature and transferred via cannula to a solution of ethyl 4-(2,2-9 d'unethyl-4-trifluoromethanesulfonyloxy-(2H)-thiochromen-6-ylethynyl)-2-1 o fluorobenzoate (Compound 234, 200.0 mg, 0.39 mmol) and 11 tetrakis(triphenylphosphine)palladium(0) (24.0 mg, 0.02 mmol) in 2.0 mL
12 THF. This solution was heated to 50 C for 1 hour, cooled to room 13 temperature, and the reaction quenched by the addition of saturated aqueous 14 NH4C1. The solution was extracted with EtOAc and the combined organic layers were washed with HZO and saturated aqueous NaC1 before being dried 16 (MgSO4) and concentrated under reduced pressure. The title compound, 17 177.0 mg (79%), was isolated by column chromatography (5% EtOAc /
1s hexanes) as a pale-yellow solid. 1H NMR (300 MHz, CDC13) 8: 7.88 (1H, d, J
19 = 7.8 Hz), 7.28-7.18 (9H, m), 5.84 (1H, s), 4.39 (2H, q, J = 7.1 Hz), 2.42 (3H, s), 1.48 (6H, s), 1.40 (3H, t, J = 7.1 Hz).
21 Ethyl 4-[[4-(4ethvlphenvll-2.2-dimethy~2H)-thiochromen-6-yl]-eth,~n~]-2-22 fluorobenzoate (Compound 237) 23 A solution of 4-ethylbromobenzene (185.0 mg, 1.00 mmol) in 2.0 mL
24 of THF was cooled to -78 C and tert-butyllithium (128.5 mg, 2.0 mmol, 1.2 mL of a 1.7M solution in pentane) was added to give a yellow solution.
26 After 30 minutes a solution of ZnCl2 (204.5 mg, 1.5 mmol) in 4.0 mL THF
27 was slowly added via cannula. The resulting solution was warmed to room 28 temperature and transferred via cannula to a solution of ethyl 4-(2,2-29 d'unethyl-4-trifluoromethanesulfonyloxy-(2H)-thiochromen-6-ylethynyl)-2-1 fluorobenzoate (Compound 234, 200.0 mg, 0.39 mmol) and 2 tetrakis(triphenylphosphine)palladium(0) (24.0 mg, 0.02 mmol) in 2.0 mL
3 THF. This solution was heated to 50 C for ihour, cooled to room 4 temperature, and the reaction quenched by the addition of saturated aqueous NH4C1. The solution was extracted with EtOAc and the combined organic 6 layers were washed with H20 and saturated aqueous NaCI before being dried 7 (MgSO4) and concentrated under reduced pressure. The title compound, a 158.0 mg (86%), was isolated by column chromatography (5% EtOAc /
9 hexanes) as a pale-yellow solid. 1H NMR (300 MHz, CDC13) 8: 7.88 (1H, d, J
1 o = 7.8 Hz), 7.39-7.20 (9H,m), 5.86 (1H, s), 4.40 (2H, q, J = 7.1 Hz), 2.72 (2H, ii q, J = 7.6 Hz), 1.49 (6H, s), 1.40 (3H, t, J = 7.1 Hz), 1.30 (3H, t, J =
7.6 12 Hz).
13 Ethyl 6-((2.2-dimeth1-4-oxo-thiochroman-6-A)ethy~11-nicotinate(Compound A solution of 6-ethynyl-2,2-dimethylthiochroman-4-one (Compound 16 222, 510.0 mg, 2.36 mmol) and ethyl 6-iodonicotinate (654.0 mg, 2.36 mmol) 17 in 20.0 mL Et3N was purged with argon for 20 minutes. To this solution was 1 e added bis(triphenylphosphine)-palladium(II) chloride (425.0 mg, 0.60 mmol) 1s and copper(I) iodide (115.0 mg, 0.60 mmol). After sparging for an additional 10 minutes with argon, the solution was stirred overnight at room 21 temperature. The reaction mixture was diluted with EtOAc (20 mL) and 22 filtered through a pad of Celite using an Et20 wash. Concentration of the 23 filtrate under reduced pressure, followed by column chromatography (10 to 24 20% EtOAc / hexanes) of the residual solid afforded 675 mg (78%) of the title compound as an orange solid. 1H NMR (300 MHz, CDC13) S: 9.20 (1H, 26 d, J = 1.0 Hz), 8.34 (1H, d, J = 1.8 Hz), 8.29 (1H, dd, J = 2.2, 8.2 Hz), 7.60 27 (2H, m), 7.25 (1H, d, J = 8.4 Hz), 4.43 (2H, q, J = 7.1 Hz), 2.90 (2H, s), 1.49 28 (6H, s), 1.43 (3H, t, J = 7.1 Hz).
29 Ethyl 6-(2.2-dimethyl-4-trifluoromethanesulfon,-ox,y:2H1-thiochromen-6------ -------i ly ethynyI)_nicotinate (Compound 239) 2 A solution of sodium bis(trimethylsilyl)amide (284.2 mg, 1.55 mmol) in 3 3.6 mL of THF was cooled to -78 C and a solution of ethyl 6-(2,2-dimethyl-4 4-oxo-thiochroman-6-ylethynyl)-nicotinate (Compound 238, 480.0 mg, 1.31 s mmol) in 3.0 mL THF was added slowly. After 30 minutes a solution of 2-s [N,N-bis(trifluoromethanesulfonyl)amino]-5-pyridine (608.0 mg, 1.55 mmol) 7 in 3.0 mL of THF was added. After 5 minutes the solution was warmed to 8 room temperature and stirred overnight. The reaction was quenched by the 9 addition of saturated aqueous NH4C1 and extracted with EtOAc. The io combined organic layers were washed with 5% aqueous NaOH and H20 11 before being dried (MgSO4) and concentrated under reduced pressure. The 12 title compound was isolated by column chromatography (20% EtOAc /
13 hexanes) as a yellow solid, 566.0 mg (87%). 1H NMR (300 MHz, CDC13) 8:
14 9.21(1H,d,J=2.0Hz),8.30(1H,dd,J=2.0,8.1Hz),7.68(1H,d,J= 1.5 15 Hz),7.61(1H,d,J=8.2Hz),7.49(1H,dd,J= 1.7,8.1Hz),7.31(1H,d,J
le = 8.2 Hz), 5.92 (1H, s), 4.41 (2H, q, J = 7.1 Hz), 1.53 (6H, s), 1.43 (3H, t, J
17 = 7.1 Hz).
18 Ethy16-[(4-(4-meth-lphenyl)-2.2-dimethA-(2H)-thiochromen-6-yl]-ethyn,tirl]-1s nicotinate (CoWound 240) 20 A solution of 4-methylbromobenzene (280.0 mg, 1.64 mmol) in 2.0 mL
21 of THF was cooled to -78 C and tert-butyllithium (209.5 mg, 3.27 mmol, 1.9 22 ml of a 1.7M solution in pentane) was added to give a yellow solution.
After 23 30 minutes a solution of ZnCl2 (680.0 mg, 5.0 mmol) in 4.0 mL THF was 24 slowly added via cannula. The resulting solution was warmed to room 25 temperature and transferred via cannula to a solution of ethyl6-(2,2-2s dimethyl-4-trifluoromethanesulfonyloxy-(2H)-thiochromen-6-ylethynyl)-27 nicotinate (Compound 239, 120.0 mg, 0.24 mmol) and 28 tetrakis(triphenylphosphine)palladium(0) (20.0 mg, 0.02 mmol) in 2.0 mL
29 THF. This solution was heated to 50 C for 2 hours, cooled to room 1 temperature, and the reaction quenched by the addition of saturated aqueous 2 NH4C1. The solution was extracted with EtOAc and the combined organic 3 layers were washed with H20 and saturated aqueous NaCI before being dried 4(MgSO4) and concentrated under reduced pressure. The title compound, 80.0 mg (76%), was isolated by column chromatography (10 to 15% EtOAc /
6 hexanes) as a colorless solid. 1H NMR (300 MHz, CDC13) 8: 9.15 (1H, d, J
7 2.2 Hz), 8.23 (1H, dd, J = 2.1, 8.0 Hz), 7.51 (1H, d, J = 8.1 Hz), 7.40-7.32 8 (3H, m), 7.18 (4H, m), 5.83 (1H, s), 4.40 (2H, q, J = 7.1 Hz), 2.40 (3H, s), 9 1.47 (6H, s), 1.40 (3H, t, J = 7.1 Hz).
1 o Ethv16-[[(4-ethylphenvl)-2 2-dimethyl-(2H)-thiochromen-6-ylj-ethvnyll-11 nicotinate (Compound 241) 12 A solution of 4-ethylbromobenzene (300.0 mg, 1.62 mmol) in 2.0 mL
13 of THF was cooled to -78 C and tert-butyllithium (207.6 mg, 3.24 mmol, 1.9 14 mL of a 1.7M solution in pentane) was added to give a yellow solution.
After 30 minutes a solution of ZnC12 (408.0 mg, 3.0 mmol) in 4.0 mL THF
1 s was slowly added via cannula. The resulting solution was warmed to room 17 temperature and transferred via cannula to a solution of ethyl 6-(2,2-18 dimethyl-4-trifluoromethanesulfonyloxy-(2H)-thiochromen-6-ylethynyl)-1s nicotinate (Compound 239, 178.0 mg, 0.36 mmol) and 2o tetrakis(triphenylphosphine)palladium(0) (24 mg, 0.02 mmol) in 2.0 mL THF.
21 This solution was heated to 50 C for 1 hour, cooled to room temperature, 22 and the reaction quenched by the addition of saturated aqueous NH4Cl. This 23 solution was extracted with EtOAc and the combined organic layers were 24 washed with H20 and saturated aqueous NaCI before being dried (MgSO4) and concentrated under reduced pressure. The title compound was isolated 26 by column chromatography (10 to 15% EtOAc / hexanes) and recrystalization 27 from CH3CN to give 136.0 mg (83%) of pale yellow crystals. 'H NMR (300 28 MHz,CDC13)8:9.15(1H,s),8.23(1H,dd,J=2.2,8.2Hz),7.51(1H,d,J=
29 8.1 Hz), 7.49-7.34 (3H, m), 7.24-7.17 (4H, m), 5.83 (1H, s), 4.40 (2H, q, J

1 7.1 Hz), 2.70 (2H, q, J= 7.6 Hz), 1.47 (6H, s), 1.40 (3H, t, J = 7.1 Hz), 1.29 2 (3H, t, J = 7.1 Hz).
3 4-[[4-phenyl-2.2-dimethyl-(2H)-thiochromen-6-yl]-ethnyl]-benzoic acid 4 (Compound 242) To a solution of ethyl 4-[[4-phenyl-2,2-dimethyl-(2H)-thiochromen-6-6 yl]-ethynyl]-benzoate (Compound 225, 105.0 mg, 0.247 mmol) in 3.0 mL THF
7 and 3.0 mL EtOH was added NaOH (120.0 mg, 3.0 mmol, 3.0 mL of a 1M
8 aqueous solution). The resulting solution was heated to 45 C and stirred 9 overnight. Upon cooling to room temperature the reaction mixture was lo acidified with 10% aqueous HCl and extracted with EtOAc. The combined ii organic layers were washed with H20, saturated aqueous NaCI, and dried 12 (NaZSO4) before removing the solvent under reduced pressure. The residual 13 solid was recrystallized from CH3CN to give the title compound as a pale-14 yellow solid. 1H NMR (300 MHz, d6-acetone) 8: 8.00 (2H, d, J = 8.4 Hz), 7.57 (2H, d, J= 8.4 Hz), 7.48-7.29 (7H, m), 7.18 (1H, d, J = 1.3 Hz), 5.96 le (1H, s), 1.48 (6H, s).
17 4-[[4-(4-methvlphenvl)-2,2-dimelhyl-(2H)-thiochromen-6-vl]-ethynyl]-benzoic 18 acid (Compound 243) 19 To a solution of ethyl4-[[4-(4-methylphenyl)-2,2-dimethyl-(2H.)-2o thiochromen-6-yl]-ethynyl]-benzoate (Compound 226, 126.0 mg, 0.287 mmol) 21 in 3.0 mL THF and 3.0 mL EtOH was added NaOH (160.0 mg, 4.0 mmol, 22 2.0 mL of a 2M aqueous solution). The resulting solution was heated to 45 23 C and stirred overnight. Upon cooling to room temperature the reaction 24 mixture was acidified with 10% aqueous HCl and extracted with EtOAc. The combined organic layers were washed with HZO, saturated aqueous NaCl, and 2s dried (Na2SO4) before removing the solvent under reduced pressure. The 27 residual solid was recrystallized from CH3CN to give 85.0 mg (72%) of the 28 title compound as a pale-yellow solid. 1H NMR (300 MHz, d6-acetone) 8:
29 8.00 (2H, d, J = 8.3 Hz), 7.58 (2H, d, J= 8.3 Hz), 7.45-7.38 (2H, m), 7.28-1 7.17 (5H, m), 5.92 (1H, s), 2.37 (3H, s), 1.47 (6H, s).
2 4-[L4-(4-ethylphenyl)-2.2-dimethy1-(2H)-thiochromen-6-yfl-ethvnxl]-benzoic 3 acid (Compound 244) 4 To a solution of ethyl 4-[[4-(4-ethylphenyl)-2,2-dimethyl-(2H)-thiochromen-6-yi]-ethynyl]-benzoate (Compound 227, 940.0 mg, 2.08 mmol) 6 in 10.0 mL THF and 5.0 mL EtOH was added NaOH (416.0 mg, 10.4 mmol, 7 5.2 mL of a 2M aqueous solution). The resulting solution was stirred 8 overnight at room temperature. The reaction mixture was acidified with 10%
9 aqueous HCl and extracted with EtOAc. The combined organic layers were lo washed with H20, saturated aqueous NaCl, and dried (Na2SO4) before 11 removing the solvent under reduced pressure. The residual solid was 12 recrystallized from CH3CN to give 786.0 mg (89%) of the title compound as 13 a colorless solid. 1H NMR (300 MHz, d6-acetone) 8: 8.01 (2H, d, J = 8.3 Hz), 14 7.60 (2H, d, J = 8.5 Hz), 7.42 (2H, m), 7.29 (2H, m), 7.22 (3H, m), 5.94 (1H, s), 2.69 (2H, q, J = 7.7 Hz), 1.47 (6H, s), 1.25 (3H, t, J = 7.7 Hz).
te 4-[(4-(,4-isoproõpylphenxl)-2.2-dimethyl-(2H)-thiochromen-6-yl]-ethyoyl)-17 benzoic acid Compound 245) 18 To a solution of ethyl4-[[4-(4-isopropylphenyl)-2,2-dimethyl-(2H)-1g thiochromen-6-yl]-ethynyl)-benzoate (Compound 228, 89.0 mg, 0.19 mmol) in 2o 3.0 mL THF and 3.0 mL EtOH was added NaOH (80.0 mg, 2.0 mmol, 2.0 21 mL of a 1M aqueous solution). The resulting solution was heated to 45 C
22 and stirred overnight. Upon cooling to room temperature the reaction 23 mixture was acidified with 10% aqueous HCl and extracted with EtOAc. The 24 combined organic layers were washed with H20, saturated aqueous NaC1, and dried (Na2SO4) before removing the solvent under reduced pressure. The 2e residual solid was recrystallized from CH3CN to give 78.0 mg (93%) of the 27 title compound as a pale-yellow solid. 1H NMR (300 MHz, d6-acetone) 8:
28 8.00 (2H, d, J = 8.4 Hz), 7.58 (2H, d, J = 8.4 Hz), 7.45-7.21 (7H, m), 5.93 29 (1H, s), 2.95 (1H, hept, J = 7.0 Hz), 1.47 (6H, s), 1.25 (6H, d, J = 7.0 Hz).

1 4-[[4-(4-te rt-butvlphenv_l)-2.2-dimethyl-(2H)-thiochromen-6-yl]-ethynyl]-benzoic 2 acid (Compound 246) 3 To a solution of ethyl 4-[[4-(4-tert-butylphenyl)-2,2-dimethyl-(2H)-4 thiochromen-6-yl]-ethynyl]-benzoate (Compound 229, 65.0 mg, 0.135 mmol) in 2.0 mL THF and 2.0 mL EtOH was added NaOH (80.0 mg, 2.0 mmol, 2.0 s mL of a 1M aqueous solution). The resulting solution was heated to 45 C
7 and stirred overnight. Upon cooling to room temperature the reaction 8 mixture was acidified with 10% aqueous HCl and extracted with EtOAc. The 9 combined organic layers were washed with H20, saturated aqueous NaCl, and 1o dried (Na2SO4) before removing the solvent under reduced pressure. The ii residual solid was recrystallized from CH3CN to give 33.0 mg (54%) of the 12 title compound as a pale-yellow solid. 1H NMR (300 MHz, d6-acetone) 8:
13 8.01 (2H, d, J = 8.4 Hz), 7.57 (2H, d, J = 8.4 Hz), 7.45 (4H, m), 7.24 (3H, 14 m), 5.93 (1H, s), 1.47 (6H, s), 1.34 (9H, s).
1 s 4-[[4 (5-methyl-thioohen-2-vl)-2.2-dimeth,yl-(2H)-thiochromen-6-A]-ethyntil]-1s benzoic acid (Compou nd 247) 17 To a solution of ethyl 4-[[4-(5-methylthiophen-2-yl)-2,2-dimethyl-(2H)-18 thiochromen-6-yl]-ethynyl]-benzoate (Compound 230, 143.0 mg, 0.322 mmol) 19 in 3.0 mL THF and 3.0 mL EtOH was added NaOH (160.0 mg, 4.0 mmol, 2o 4.0 mL of a 1M aqueous solution). The resulting solution was stirred 21 overnight at room temperature. The reaction mixture was acidified with 10%
22 aqueous HCl and extracted with EtOAc. The combined organic layers were 23 washed with HZO, saturated aqueous NaCl, and dried (Na2SO4) before 24 removing the solvent under reduced pressure. The residual solid was 25 recrystallized from CH3CN to give 110.0 mg (82%) of the title compound as 26 a pale-yellow solid. 1H NMR (300 MHz, d6-acetone) 8: 8.03 (2H, d, J = 8.4 27 Hz), 7.63 (2H, d, J = 8.3 Hz), 7.60 (1H, s), 7.44 (2H, s), 6.87 (1H, d, J =
3.5 28 Hz), 6.79 (1H, m), 6.10 (1H, s), 2.49 (3H, s), 1.45 (6H, s).
29 4[[4 f5-ethyl-thio hn en-2~Ll,1-2 2-dimethyl-_ (2H)-thiochromen-6-vl]-ethvnvll-i benzoic acid (Compound 248) 2 To a solution of ethyl 4-[[4-(5-ethylthiophen-2-yl)-2,2-dimethyl-(2H)-3 thiochromen-6 yl]-ethynyl]-benzoate (Compound 231, 23.0 mg, 0.05 mmol) in 4 1.0 mL THF and 1.0 mL EtOH was added NaOH (40.0 mg, 1.0 mmol, 1.0 mL of a 1M aqueous solution). The resulting solution was stirred overnight 6 at room temperature. The reaction mixture was acidified with 10% aqueous 7 HCl and extracted with EtOAc. The combined organic layers were washed 8 with H20, saturated aqueous NaCI, and dried (NaZSOa) before removing the 9 solvent under reduced pressure. The residual solid 'was recrystallized from CH3CN to give 15.5 mg (72%) of the title compound as an orange solid. 1H
11 NMR (300 MHz, d6-acetone) S: 8.03 (2H, d, J = 8.3 Hz), 7.63 (2H, d, J =
12 8.3 Hz), 7.61 (1H, s), 7.44 (2H, s), 6.90 (1H, d, J= 3.5 Hz), 6.83 (1H, d, J
13 3.5 Hz), 6.10 (1H, s), 2.86 (2H, q, J = 7.6 Hz), 1.45 (6H, s), 1.30 (3H, t, J
14 7.6 Hz).
4-[[4-(5-tert-bu lty thiopen-2 Xl)-2.2-dimethyl-(2H)-thiochromen-6 yl]-ethy.1w]-1s benzoic acid (Compound M
17 To a solution of ethyl 4-[[4-(5-tert-butylthiophen-2 yl)-2,2-dimethyl-1 s (2H)-thiochromen-6-yl]-ethynyl]-benzoate (Compound 232, 88.0 mg, 0.181 is mmol) in 2.0 mL THF and 2.0 mL EtOH was added NaOH (160.0 mg, 4.0 mmol, 4.0 mL of a 1M aqueous solution). The resulting solution was stirred 21 overnight at room temperature. The reaction mixture was acidified with 22 10% aqueous HCl and extracted with EtOAc. The combined organic layers 23 were washed with HZO, saturated aqueous NaCI, and dried (NaZSO4) before 24 removing the solvent under reduced pressure. The residual solid was recrystallized from CH3CN to give 68.0 mg (82%) of the title compound as a 26 pale-yellow solid. 1H NMR (300 MHz, d6-acetone) 6: 8.03 (2H, d, J = 8.3 27 Hz), 7.63 (2H, d, J = 8.3 Hz), 7.62 (1H, s), 7.44 (2H, d, J = 1.2 Hz), 6.87 28 (2H, s), 6.11 (1H, s), 1.45 (6H, s), 1.39 (9H, s).
29 4-[j4-(6-methylp.)tidin_3_yl)-2 2-dimethyl-(2H)-thiochromen-6-yl -ethvnvll 1 benzoic acid (Compound 250) 2 To a solution of ethyl 4-[[4-(6-methylpyridin-3-yl)-2,2-dimethyl-(2H)-3 thiochromen-6-yl]-ethynyl]-benzoate (Compound 233, 70.0 mg, 0.159 mmol) 4 in 3.0 mL THF and 3.0 mL EtOH was added NaOH (120.0 mg, 3.0 mmol, s 3.0 mL of a 1M aqueous solution). The resulting solution was stirred 6 overnight at 35 C. Upon cooling to room temperature, the reaction mixture 7 was acidified with 10% aqueous HCl and extracted with EtOAc. The 8 combined organic layers were washed with H20, saturated aqueous NaCI, and 9 dried (NazSO4) before removing the solvent under reduced pressure. The 1 o residual solid was recrystallized from acetone to give 60.0 mg (92%) of the 11 title compound as a colorless solid. iH NMR (300 MHz, d6-acetone) 8: 8.36 12 (1H, d, J = 2.1 Hz), 7.91 (2H, d, J = 8.3 Hz), 7.60 (2H, d, J = 8.3 Hz), 7.56 13 (1H,dd,J=2.18.0Hz),7.45(2H,m),7.31(1H,d,J=8.0Hz),7.06(1H, 14 s), 6.06 (1H, s), 2.51 (3H, s), 1.44 (6H, s).
15 4-jj4 (4-methX,lphenxll-2.2-dimethy1:12H)-thiochromen-6-yl]-eth3Myl]-2-1e fluorobenzoic acid (Compound 251) 17 To a solution of ethyl 4-[[4-(4-methylphenyl)-2,2-dimethyl-(2H)-18 thiochromen-6-yl]-ethynyl]-2-fluorobenzoate (Compound 236, 100.0 mg, 0.219 19 mmol) in 3.0 mL THF and 3.0 mL EtOH was added NaOH (80.0 mg, 2.0 20 mmol, 2.0 mL of a 1M aqueous solution). The resulting solution was heated 21 to 40 C and stirred overnight. Upon cooling to room temperature the 22 reaction mixture was acidified with 10% aqueous HCl and extracted with 23 EtOAc. The combined organic layers were washed with H20, saturated 24 aqueous NaCI, and dried (Na2SO4) before removing the solvent under 2s reduced pressure. The residual solid was recrystallized from CH3CN to 26 give78.0 mg (83%) of the title compound as a pale-yellow solid. 1H NMR
27 (3001VIHz, d6-acetone) 8: 7.94 (1H, d, J = 7.8 Hz), 7.43-7.17 (9H, m), 5.93 28 (1H, s), 2.38 (3H, s), 1.47 (6H, s).
29 4-[[4-(4-ethvlphe 11-2 2-dimethyl-(2H)-thiochromen-6-yl]-ethynyl]-2-1 fluorobenzoic acid (Compound 252) 2 To a solution of ethyl4-[[4-(4-ethylphenyl)-2,2-dimethyl-(2H)-s thiochromen-6-yl]-ethynyl]-2-fluorobenzoate (Compound 237, 125.0 mg, 0.266 4 mmol) in 3.0 mL THF and 3.0 mL EtOH was added NaOH (120.0 mg, 3.0 mmol, 3.0 mL of a 1M aqueous solution). The resulting solution was stirred e overnight at room temperature. The reaction mixture was acidified with 10%
7 aqueous HCl and extracted with EtOAc. The combined organic layers were 8 washed with H20, saturated aqueous NaCI, and dried (Na2SO4) before 9 removing the solvent under reduced pressure. The residual solid was 1 o recrystallized from CH3CN to give 100.0 mg (86%) of the title compound as 11 a pale-yellow solid. 1H NMR (300 MHz, d6-acetone) 8: 7.94 (1H, d J = 7.8 12 Hz), 7.43-7.20 (9H, m), 5.93 (1H, s), 2.68 (2H, q, J = 7.6 Hz), 1.47 (6H, s), 13 1.24(3H,t,J = 7.6 Hz).
14 6-[[4-(.4-methXlp leMl)-2.2-dimethyl-(,2H)-hiochromen-6-yl]-ethynyl]-nicotinic acid (Compound 25 Z
16 To a solution of ethyl 6-[[4-(4-methylphenyl)-2,2-dimethyl-(2H)-17 thiochromen-6-yl]-ethynyl]-nicotinate (Compound 240, 66.0 mg, 0.15 mmol) 18 in 2.0 mL THF and 2.0 mL EtOH was added NaOH (80.0 mg, 2.0 mmol, 2.0 19 mL of a 1M aqueous solution). The resulting solution was heated to 45 C
2o and stirred overnight. Upon cooling to room temperature the reaction 21 mixture was acidified with 10% aqueous HCl and extracted with EtOAc. The 22 combined organic layers were washed with H20, saturated aqueous NaCl, and 23 dried (Na2SO4) before removing the solvent under reduced pressure to give 24 the title compound as a yellow solid, 50.0 mg (81%). 1H NMR (300 MHz, d6-acetone) 8: 9.09 (1H, d, J = 1.4 Hz), 8.30 (1H, dd, J = 2.0, 8.1 Hz), 7.65 26 (1H, d, J = 8.1 Hz), 7.46 (2H, s), 7.24 ( 5H, m), 5.94 (1H, s), 2.37 (3H, s), 27 1.48 (6H, s).
28 6-[[4-(4-et yjnhenvll-2 2-dimetl~yl-(2H~ thiochromen-6-~]-ethy~nyl]-ni_, cotinic 29 acid (Compound 254) I To a solution of ethyl 6-[[4-(4-ethylphenyl)-2,2-dimethyl-(2H)-2 thiochromen-6-yl]-ethynyll-nicotinate (Compound 241, 110.0 mg, 0.24 mmol) 3 in 3.0 mL THF and 3.0 mL EtOH was added NaOH (120.0 mg, 3.0 mmol, 4 3.0 mL of a 1M aqueous solution). This mixture was heated to 40 C for 1 hour, cooled to room temperature, and the resulting solution was stirred 6 overnight. The reaction mixture was acidified with 10% aqueous HCI and 7 extracted with EtOAc. The combined organic layers were washed with H20, 8 saturated aqueous NaCI, and dried (Na2SO4) before removing the solvent 9 under reduced pressure to give 100 mg (96%) of the title compound as a to colorless solid. iH NMR (300 MHz, d6-acetone) 8: 9.08 (1H, d, J = 2.0 Hz), 11 8.30 (1H, dd, J = 2.2, 8.2 Hz), 7.66 (1H, d, J = 8.2 Hz), 7.46 (2H, s), 7.31-12 7.21 (5H, m), 5.95 (1H, s), 2.69 (2H, q, J = 7.6 Hz), 1.48 (6H, s), 1.25 (3H, t, 13 J=7.6Hz).
14 4-[4,(4-methvl^ uhenXl)-2.2-dimeth l-~ 7-fluoro- ,2H)-thiochromen-6-yl]-eth~nXl~
benzoic acid (Compound 255) 16 To a solution of ethyl 4-[[4-(4-methylphenyl)-2,2-dimethyl-7-fluoro-17 (2H)-thiochromen-6-yl]-ethynyl]-benzoate (Compound 213, 108.0 mg, 0.236 18 mmol) in 2.0 mL THF and 2.0 mL EtOH was added NaOH (120.0 nlg, 3.0 19 mmol, 3.0 mL of a 1M aqueous solution). The resulting solution was heated to 45 C and stirred overnight. Upon cooling to room temperature the 21 reaction mixture was acidified with 10% aqueous HCI and extracted with 22 EtOAc. The combined organic layers were washed with H20, saturated 23 aqueous NaCl, and dried (Na2SO4) before removing the solvent under 24 reduced pressure. The residual solid was recrystallized from CH3CN to give 85 mg (80%) of the title compound as a pale-yellow solid. 1H NMR (300 26 MHz, d6-acetone) 8: 8.03 (2H, d, J = 8.3 Hz), 7.61 (2H, d, J = 8.3 Hz), 7.31-27 7.18 (6H, m), 5.90 (1H, s), 2.37 (3H, s), 1.48 (6H, s).
28 4-[[4-(4-meth3lohenyll-2.2-dimethyl-7-methoxy_(2H)-thiochromen-6-vl]-29 ethMI]-benzoic acid f,Compound 256) 1 To a solution of ethyl 4-[[4-(4-methylphenyl)-2,2-dimethyl-7-methoxy-2(2H)-thiochromen-6-yl]-ethynyl]-benzoate (Compound 215, 64.0 mg, 0.113 3 mmol) in 2.0 mL THF and 2.0 mL EtOH was added NaOH (80.0 mg, 2.0 4 mmol, 2.0 mL of a 1M aqueous solution). The resulting solution was heated to 45 C and stirred overnight. Upon cooling to room temperature the e reaction mixture was acidified with 10% aqueous HCl and extracted with 7 EtOAc. The combined organic layers were washed with HZO, saturated 8 aqueous NaCI, and dried (Na2SO4) before removing the solvent under 9 reduced pressure. The residual solid was recrystallized from CH3CN to give io 50 mg (83%) of the title compound as a colorless solid. 1H NMR (300 MHz, 11 d6-acetone) 8: 8.00 (2H, d, J = 8.2 Hz), 7.56 (2H, d, J= 8.2 Hz), 7.28-7.14 12 (5H, m), 7.06 (1H, s), 5.76 (1H, s), 3.96 (3H, s), 2.36 (3H, s), 1.47 (6H, s).
13 4-([4 (5-methyl-2-thienydl-2.2-dimethyI-7-fluoro-(2H)-thiochromen-6--14 eth3M,yl]-benzoic acid (Compound 257) To a solution of ethyl4-[[4-(5-methyl-2-thienyl)-2,2-dimethyl-7-fluoro-1s (2H)-thiochromen-6-yl]-ethynyl]-benzoate (Compound 214, 58.0 mg, 0.125 17 mmol) in 2.0 mL THF and 2.0 mL EtOH was added NaOH (80.0 mg, 2.0 18 mmol, 2.0 mL of a 1M aqueous solution). The resulting solution was heated 19 to 45 C and stirred overnight. Upon cooling to room temperature the 2o reaction mixture was acidified with 10% aqueous HCl and extracted with 21 .-EtOAc. The combined organic layers were washed with H20, saturated 22 aqueous NaCI, and dried (Na2SO4) before removing the solvent under 23 reduced pressure. The residual solid was recrystallized from CH3CN to give 24 50 mg (90%) of the title compound as a pale-yellow solid. IH NMR (300 MHz, d6-acetone) 8: 8.05 (2H, d, J = 8.3 Hz), 7.56 (2H, d, J = 8.3 Hz), 7.61 26 (1H,d,J=7.4Hz),7.31(1H,d,J=9.6Hz),6.89(1H,d,J=3.5Hz),6.79 27 (1H, d, J = 3.5 Hz), 6.06 (1H, s), 2.49 (3H, s), 1.46 (6H, s).
26 4-Bromophenylacetate (Compound 258) 29 To a solution of 4-bromophenol (20.0 g, 0.121 mol) in 200 mL CH2Cl.2 1 was added triethylamine (14.0 g, 0.139 mol) followed by acetyl chloride (9.53 2 g, 0.121 mol) in small portions at room temperature. After stirring for 21 3 hours the mixture was poured into 200 mL H20. The organic layer was 4 washed with 5% aqueous NaOH, H20, and dried over MgSO4. Removal of the solvent under reduced pressure and distillation of the residue afforded 20.6 g 6 (83%) of the product as a colorless oil, bp 93-95 C / 2 mm.
7 2-Hydroxv-5-bromo-acetonhen ne (Compound 259) 8 A mixture of A1C13 (6.20 g, 46.50 mmol) and 4-bromophenylacetate s(Compound 258, 10.0 g, 46.50 mmol) was heated to 150 C for 30 minutes while a stream of argon was passed over the molten solid. Upon cooling to 11 -room temperature the orange solid was carefully treated with 100 mL of H20 12 and 100 mL of 10% aqueous HCI. The resulting mixture was extracted with 13 EtOAc and the combined organic layers were washed with HZO and saturated 14 aqueous NaCI before being dried over Na2SO4. Concentration of the solution under reduced pressure afforded a tan solid which after recrystallization from i-1 e PrOH provided 7.18 g (72%) of the title compound as an off-white crystalline 17 solid. 1H NMR (300 MHz, CDC13) 8: 12.17 (1H, s), 7.84 (1H, d, J = 2.6 Hz), 18 7.55 (1H, dd, J = 2.5, 9.0 Hz), 6.90 (1H, d, J = 8.9 Hz), 2.64 (3H, s).
19 2.2-Dimethyl-6-bromo-chroman-4-one (Compound 260) To a solution of piperidine (2,83 g, 33.25 mmol) in 45 mL benzene was 21 added trifluoroacetic acid (344.6 mg, 3.02 mmol) as a solution in 4.0 mL of 22 benzene. Acetone ( 8.78 g, 151.3 mmol) was added followed by 2-hydroxy-5-23 bromoacetophenone (Compound 259, 6.50 g, 30.23 mmol). The resulting 24 solution was heated to reflux in a flask fitted with a Dean-Stark trap.
After 24 hours, an additional 2.5 equivalents of acetone and 0.1 equivalents of 26 trifluoroacetic acid were added. After a total of 43 hours the reaction was 27 cooled to room temperature and diluted with EtOAc. The solution was 28 washed with 1M aqueous HCI, H20, and saturated aqueous NaCl before being 29 dried (MgSO4) and concentrated under reduced pressure. Distillation of the 3o residual oil (bulb-to-bulb) afforded 4.80g (62%) of the title compound as a 1 clear yellow oil, bp 105-115 C / 5 mm. The reaction conditions here are a 2 moderate modification of the synthesis of the title compound described by 3 Buckle et al. in J. Med. Chem. 1990 3028-3034. 1H NMR (300 MHz, CDC13) S
4 : 7.97 (1H, d, J = 2.6 Hz), 7.54 (1H, dd, J = 2.6, 8.7 Hz), 6.84 (1H, d, J =
8.8 Hz), 2.72 (2H, s), 1.46 (6H, s).
6 2.2-Dimethyl-6-trimethvlsilanxlethynyl-chroman-4-one l,Com oR und 261) 7 A solution of 2,2-dimethyl-6-bromo-chroman-4-one (Compound 260, 8 4.30 g, 16.85 mmol) and copper (I) iodide (321 mg, 0.17 mmol) in 55.0 mL
9 Et3N was sparged with argon for 30 minutes. To this solution were added trimethylsilylacetylene (4.66 g, 50.56 mmol) and bis(triphenylphosphine)-11 palladium(II) chloride (1.18g, 0.17 mmol). The mixture was heated to 70 C
12 for 26 hours, cooled to room temperature, and diluted with Et20 (100 mL).
13 The resulting mixture was filtered through a pad of Celite using an EtzO
wash 14 and the filtrate washed with H20, a solution of NH4OH/ saturated aqueous NH4C1 (9:1), H2O, 1M aqueous HCI, H2O and saturated aqueous NaCI before 16 being dried (Na2SO4) and concentrated under reduced pressure. Column 17 chromatography (5% EtOAc - hexanes) of the residual oil afforded 4.09 g 18 (89%) of the product as a yellow waxy solid. iH NMR (300 MHz, CDC13) S:
19 7.99(1H,d,J=2.1Hz),7.53(1H,dd,J=2.2,8.6Hz),6.86(1H,d,J=8.6 Hz), 2.72 (2H, s), 1.45 (6H, s), 0.24 (9H, s).
21 2.2-Dimethyl-6- yl-chroman-4-one (Compound 2621 22 To a solution of 2,2-dimethyl-6-trimethylsilanylethynyl-chroman-4-one 23 (Compound 261, 4.05 g, 14.87 mmol) in 60.0 mL of MeOH was added K2C03 24 (410.9 mg, 2.97 mmol) The resulting mixture was stirred at room temperature for 24 hours, diluted with EtOAc (100mL) and washed with H20 and saturated 26 aqueous NaCI, and dried over Na2SO4. Concentration of this solution under 27 reduced pressure afforded 2.71 g (91%) of the product as a tan solid. 1H
NMR
28 (300 MHz, CDC13) S: 8.01 (1H, d, J = 2.2 Hz), 7.56 (1H, dd, J = 2.2, 8.6 Hz), 29 6.90 (1H, d, J = 8.6 Hz), 3.02 (1H, s), 2.74 (2H, s), 1.47 (6H, s).
3o Ethx 4-[(2 2-dimethyl-4-oxo-chroman-6-yl)ethvnyl]-benzoate (Compound 263) 1 A solution of 6-ethynyl-2,2-dimethylchroman-4-one (Compound 262, 2 2.60 g, 13.0 mmol) and ethyl4iodobenzoate (3.6 g, 13.0 mmol) in 50.0 mL
s Et3N was purged with argon for 15 minutes. To this solution was added 4 bis(triphenylphosphine)-palladium(II) chloride (1.82 g, 2.6 mmol) and s copper(l) iodide (496 mg, 2.6 mmol). After sparging for an additional 10 s minutes with argon, the solution was atirred overnight at room temperature.
7 The reaction mixture was filtered through a pad of Celite*using an EtzO
a wash. Concentration of the fdtrate under reduced pressure, followed by a column chromatography (2-5% EtOAc-heaanes) of the residual solid afforded lo 2.28 g(50Rfo) the title compound as an oraage solid. IH NMR (300 MHz, it CD(3s) 8:&06 (1H, d, J= 2.2 Hz), 8.02 (2H, d, J= 8.5 Hz), 2.61 (1H, dd, J
12 =2.2,8.6Hz),7.55(2H,d,J=8.5Hz),6.93(1H,d,J=8.6Hz),4.39(2H, ta q, J= 7.1 Hz), 2.75 (3H, s), 1.48 (6H, s), 1.41 (t, J= 7.1 Hz).
14 ELT(2 ?rd;lnethyl4-trifluoromethanesulfonvIM(2H1-chromen-6-15 y ojhm,yj)- te (Comoound M
ta A solution of sodium bis(trimethylsilyl)amide (1.56g, 8.5 mmol) in 18.0 17 mL of THF was cooled to -78 C and a solution of ethyl4((2,2-dimethyl-4-ta oao-chroman-6-yl)ethynyl)-benzoate (Compound 263, 2.26 g, 6.49 mmol) in ia 15.0 niL was added slowly. After 30 minutes a solution of 2-[N,N-2o bis(tri8uoromethanesulfonyl)amino]-5-pyridine (3.10 g, 7.79 mmol) in 10 mL
21 of THF was added slowly. After 5 minutes the solution was warmed to room 22 temperature and stirred overnight. The reaction was quenched by the 2a addition of saturated aqueous NH4Q and eatracted with EtOAc. The 24 eombined organic layers were washed with 5% aqueous NaOH and 1160 25 before being dried (MgSO4) and concentrated under reduced pressure. The 2g title compound, 2.0 g(649b), was isolated by column chromatography (5%
27 EtOAFhexanes) as a colorless solid. IH NMR (300 MHz, CDCl3) 8: 8.03 29 (2H,d,J=8.3Hz),7.57(2H,d,J=8.3Hz),7.45-7.21(2H,m),6.85(1H,d, 28 J=8.4HZ),5.69(1H,8),4.40(2H,q,J=7.1HZ),1.55(6H,s),1.4(3H,t,J
* Trade-mark 1 =7.1Hz).
2 Ethyl4-([4-phenvl-2.2-dim1ethyl-(2H)-chromen-6-yl]-ethvnxll-benzoate 3 (Compound 265) 4 A solution of bromobenzene (250.0 mg, 1.59 mmol) in 3.0 mL of THF
was cooled to -78 C and tert-butyllithium (203.7 mg, 3.18 mmol, 1.9 ml of a 6 1.7M solution in pentane) was added to give a yellow solution. After 30 7 minutes a solution of ZnC12 (440.0 mg, 3.18 mmol) in 5.0 mL THF was slowly 8 added via cannula. The resulting solution was warmed to room temperature 9 and transferred via cannula to a solution of ethyl 4-(2,2-dimethyl-4-lo trifluoromethanesulfonyloxy-(2H)-chromen-6-ylethynyl)-benzoate (Compound 11 264, 150.0 mg, 0.31 mmol) and tetrakis(triphenylphosphine)palladium(0) (24.0 12 mg, 0.02 mmol) in 2.0 mL THF. This solution was heated to 50 C for 2 13 hours, cooled to room temperature, and the reaction quenched by the 14 addition of saturated aqueous NH4Ci. This solution was extracted with EtOAc and the combined organic layers were washed with H20 and saturated 16 aqueous NaCl before being dried (MgSO4) and concentrated under reduced 17 pressure. The title compound, 110.0 mg (87%), was isolated by column is chromatography (2.5% EtOAc / hexanes) as a colorless solid. 1H NMR (300 1s MHz, CDC13) S: 7.99 (2H, d, J = 8.3 Hz), 7.51 (2H, d, J= 8.3 Hz), 7.47-7.35 2o (6H,m),7.21(1H,d,J=2.0Hz),6.88(1H,d,J=8.4Hz),5.66(1H,s), 21 4.38 (2H, q, J = 7.1 Hz), 1.52 (6H, s), 1.40 (3H, t, J = 7.1 Hz).
22 Ethy14-j(4-(4-methylphenYl)-2.2-dimethyl-(2H)-chromen-6-yl]-ethvnyl]-23 benzoate (Compound 266) 24 A solution of 4-methylbromobenzene (237.0 mg, 1.38 mmol) in 3.0 mL
of THF was cooled to -78 C and tert-butyllithium (177.5 mg, 2.77 mmol, 1.6 26 ml of a 1.7M solution in pentane) was added to give a yellow solution.
After 27 30 minutes a solution of ZnCIZ (377.0 mg, 2.77 mmol) in 5.0 mL THF was 28 slowly added via cannula. The resulting solution was warmed to room 29 temperature and transferred via cannula to a solution of ethyl 4-(2,2-1 dimethyl-4-trifluoromethanesulfonyloxy-(2H)-chromen-6-ylethynyl)-benzoate 2 (Compound 264, 150.0 mg, 0.31 mmol) and 3 tetrakis(triphenylphosphine)palladium(0) (24.0 mg, 0.02 mmol) in 2.0 mL
4 THF. This solution was heated to 50 C for 2 hours, cooled to room s temperature, and the reaction quenched by the addition of saturated aqueous 6 NH4C1. This solution was extracted with EtOAc and the combined organic 7 layers were washed with H20 and saturated aqueous NaC1 before being dried 8(MgSOa) and concentrated under reduced pressure. The title compound, 9 120.0 mg (92%), was isolated by column chromatography (2.5% EtOAc /
hexanes) as a colorless solid. 1H NMR (300 MHz, CDC13) 8: 7.98 (2H, d, J
11 8.5 Hz), 7.50 (2H, d, J 8.5 Hz), 7.34 (1H, dd, J = 2.1, 8.3 Hz), 7.25-7.21 12 (5H,m),6.85(1H,d,J=8.3Hz),5.62(1H,s),4.36(2H,q,J 7.1Hz),2.41 13 (3H, s), 1.49 (6H, s), 1.39 (3H, t, J = 7.1 Hz).
14 Ethyl 4-([4-(4-ethylphenyl)-2.2-dimethvl-(2H)-chromen-6-yl]-eth rtinLI]-benzoate (Compound 267) 16 A solution of 4-ethylbromobenzene (280.0 mg, 1.51 mmol) in 3.0 mL
17 of THF was cooled to -78 C and tert-butyllithium (198.6 mg, 3.10 mmol, 1.9 18 ml of a 1.7M solution in pentane) was added to give a yellow solution.
After 19 30 minutes a solution of ZnC12 (408.0 mg, 3.10 mmol) in 5.0 mL THF was 2o slowly added via cannula. The resulting solution was warmed to room 21 temperature and transferred via cannula to a solution of ethyl 4-(2,2-22 dimethyl-4-trifluoromethanesulfonyloxy-(2H)-chromen-6-ylethynyl)-benzoate 23 (Compound 264, 150.0 mg, 0.31 mmol) and 24 tetrakis(triphenylphosphine)palladium(0) (24.0 mg, 0.02 mmol) in 2.0 mL
2s THF. This solution was heated to 50 C for 2 hours, cooled to room 26 temperature, and the reaction quenched by the addition of saturated aqueous 27 NH4,CI. This solution was extracted with EtOAc and the combined organic 28 layers were washed with H20 and saturated aqueous NaC1 before being dried 29 (MgSO4) and concentrated under reduced pressure. The title compound, 1 123.0 mg (91%), was isolated by column chromatography (2.5% EtOAc /
2 hexanes) as a pale-yellow solid. 1H NMR (300 MHz, CDC13) S: 8.00 (2H, d, J
3 = 8.4 Hz), 7.52 (2H, d, J = 8.4 Hz), 7.36 (1H, dd, J = 2.0, 8.3 Hz), 7.31-7.25 4 (5H, m), 6.88 (1H, d, J = 8.3 Hz), 5.65 (1H, s), 4.38 (2H, q, J = 7.1 Hz), 2.73 (2H, q, J = 7.6 Hz), 1.52 (6H, s), 1.41 (3H, t, J= 7.1 Hz), 1.31 (2H, t, J
6 7.6 Hz).
7 Ethy14-[j4-{4-(l-methylethyll~henyl-)-2.2-dimeth,y~2H)-chromen-6,Y1-s ethynyl]-benzoate (Compound 268) s A solution of 4-iso-propylbromobenzene (250.0 mg, 1.25 mmol) in 3.0 mL of THF was cooled to -78 C and tert-butyllithium (160.8 mg, 2.51 mmol, 11 1.5 ml of a 1.7M solution in pentane) was added to give a yellow solution.
12 After 30 minutes a solution of ZnCk (345.0 mg, 2.53 mmol) in 5.0 mL THF
13 was slowly added via cannula. The resulting solution was warmed to room 14 temperature and transferred via cannula to a solution of ethyl4-((2,2-dimethyl-4-trifluoromethanesulfonyloxy-(2H)-chromen-6-yl)ethynyl)-benzoate 16 (Compound 264, 150.0 mg, 0.31 mmol) and tetrakis(triphenylphosphine) 17 palladium(0) (24.0 mg, 0.02 mmol) in 2.0 mL THF. This solution was heated 18 to 50 C for 2 hours, cooled to room temperature, and the reaction quenched 19 by the addition of saturated aqueous NH4C1. This solution was extracted with EtOAc and the combined organic layers were washed with HZO and saturated 21 aqueous NaC1 before being dried (MgSO4) and concentrated under reduced 22 pressure. The title compound, 100.0 mg (72%), was isolated by column 23 chromatography (2.5% EtOAc / hexanes) as a colorless solid. 1H NMR (300 24 MHz, CDC13) 8: 7.99 (2H, d, J = 8.3 Hz), 7.52 (2H, d, J = 8.3 Hz), 7.36 (1H, dd, J = 2.1, 8.3 Hz), 7.30-7.26 (5H, m), 6.88 (1H, d, J = 8.3 Hz), 5.64 (1H, 26 s), 4.38 (2H, q, J = 7.1 Hz), 2.98 (1H, hept, J = 7.0 Hz), 1.51 (6H, s), 1.40 27 (3H, t, J = 7.1 Hz), 1.31 (6H, d, J = 7.0 Hz).
28 Ft x14-[[4 (4 (1 1-dimethylethyll12hewl)-2 2-dimethxl_(2H)-chromen-6.y11-2s ethynyl]-benzQate (Compound 269) , . h WO 99133821 PCTlUS98/27314 1 A solution of 4-ten-butylbromobenzene (280.0 mg, 1.61 mmol) in 3.0 2 mL of THF was cooled to -78 C and rerr-butyllithium (206.3 tng, 3.22 mmol, s 1.7 ml of a 1.7M solution in pentane) was added to give a yellow solution.
4 After 30 minutes a solution of ZnC4 (380.0 mg, 3.22 mmol) in 5.0 mL THF
6 was slowly added via-cannula. The resulting solution was warmed to room s temperature and transferred via cannula to a solution of ethyl 4-((2,2-7 dimethyl-4-trifluoromethanesulfonyloxy-(2H)-chromen-6-yl)ethynyl)-benzoate s(C.bmpound 264, 150.0 mg, 0.31 mmol) and tetrakis(triphenylphosphine) a paDadium(0) (24.0 mg, 0.02 mmol) in 2.0 mL THF. This solution was heated 1o to 50 C for 2 hours, cooled to raom temperature, and the reaction quenched t t by the addition of saturated aqueous NH4C1. This solution was extracted with 12 EtOAc and the combined organic layers were washed with HZO and saturated 13 aqueous NaQ before being dried (MgSO4) and concentrated under reduced 14 pressure. The tffle compound, 85.0 mg (59%), wgs isolated by column i5 chromatography (2.59''o EtOAc / hexanes) as a eolorless solid. 1H NMR (300 is Mf3z,CDC1,)5:7.99(2H,d,J=8.3Hz),7.53(2H,d,J= 8.3Hz),7.45(iH, ty d,J=8.4Hz),7.37(1H,dd,J=20,8.3Hz),7.32-7.28(3H,m),6.89(1H,d, 18 J= 8.3 Hz), 5.66 (1H, s), 4.39 (2H, q, J= 7.1 Hz),152 (6H, s), 1.40 (3H, t, 19 Jm 7.1 Hz), 1.39 (9H, s).
zo Ethv1 2-fluoro-4-ff2.2-dimgft:4-oxo-chroman-6-yl, et yMj]-benzoate m c(anmRonad 2701 22 A solution of 6-ethynyl-2,2-dimethylchroman-4-one (Compound 262, a, 314.0 mg, 1.57 mmol) and ethyl 2-fluoro-4iodobenzoate (440.0 mg, 1.50 24 . mmol) in 10.0 mL Et3N was purged with argon for 15 minutes. To this 25 solntion was added bis(trlphenylphosphine)-palladium(II) chloride (275.0 mg, 2s 0.39 mmol) and oopper(I) iodide (75.0 mg, 0.39 mmol). After sparging for an 27 additional 10 minutes with argon, the solution was stkred overnight at room 2e temperature. The reaction mixture was filtered through a pad of Celite using 29 an Bt,,O wash. Concentration of the filtrate under reduccd pressure, foqowcd * Trade-mark 1 by column chromatography (3-5% EtOAc-hexanes) of the residual solid 2 afforded 400.0 mg (69%) the title compound as an orange solid. 1H NMR
3 (300MHz,CDC13)5:8.05(1H,d,J=2.1Hz),7.90(1H,d,J=7.8Hz), 4 7.60(1H,dd,J=2.28.5Hz),7.29(1H,dd,J= 1.5, 8.2 Hz), 7.25 (1H, d, J
s= 11.4 Hz), 6.93 (1H, d, J = 8.5 Hz), 4.39 (2H, q, J = 7.1 Hz), 2.75 (2H, s), 6 1.48 (6H, s), 1.40 (3H, t, J = 7.1 Hz).
7 Ethyl 2-fluoro-4-((2.2-dimethy,l-4-trifluoromethanesulfonyloxv-f 2H)-chromen-s 6-gl ethvnvl)-benzoate (Compound 271) s A solution of sodium bis(trimethylsilyl)amide (238.0 mg, 1.30 mmol) in to 3.0 mL of THF was cooled to -78 C and a solution of ethyl 2-fluoro-4-((2,2-ii dimethyl-4-oxo-chroman-6-yl)ethynyl)-benzoate (Compound 270, 400.0 mg, 12 1.09 mmol) in 2.0 mL was added slowly. After 30 minutes a solution of 2-13 [N,N-bis(trifluoromethanesulfonyl)aminoJ-5-pyridine (510.0 mg, 1.30 mmol) 14 in 1.0 mL of THF was added. After 5 minutes the solution was warmed to is room temperature and stirred overnight. The reaction was quenched by the 16 addition of saturated aqueous NH4C1 and extracted with EtOAc. The 17 combined organic layers were washed with 5% aqueous NaOH and HZO
18 before being dried (MgSO4) and concentrated under reduced pressure. The is title compound, 315.0 mg (58%), was isolated by column chromatography 20 (5% EtOAc / hexanes) as a colorless solid. 1H NMR (300 MHz, CDC13) 8:
21 7.92 (1H, d, J = 7.8 Hz), 7.44-7.26 (4H, m), 6.85 (1H, d, J = 8.7 Hz), 5.70 22 (1H, s), 4.31 (2H, q, J = 7.1 Hz), 1.55 (6H, s), 1.41 (3H, t, J = 7.1 Hz).
23 Ethy12-fluoro-4-[[4-(4-methvlQh~e A)-2.2-dimethy-(,2H)-chromen-6-ylL-24 ethynyl]-benzoate ,Compound 272) 25 A solution of 4-methylbromobenzene (140.0 mg, 0.80 mmol) in 3.0 mL
26 of THF was cooled to -78 C and tert-butyllithium (102.5 mg, 1.60 mmol, 0.94 27 ml of a 1.7M solution in pentane) was added to give a yellow solution.
After 28 30 minutes a solution of ZnC12 (174.0 mg, 1.28 mmol) in 5.0 mL THF was 29 slowly added via cannula. The resulting solution was warmed to room 1 temperature and transferred via cannula to a solution of ethyl 2-fluoro-4-(2,2-2 d'unethyl-4-trifluoromethanesulfonyloxy-(2H)-chromen-6-ylethynyl)-benzoate 3 (Compound 271, 150.0 mg, 0.31 mmol) and 4 tetrakis(triphenylphosphine)palladium(0) (20.0 mg, 0.02 mmol) in 2.0 mL
THF. This solution was heated to 50 C for 2 hours, cooled to room e temperature, and the reaction quenched by the addition of saturated aqueous 7 NH4C1. This solution was extracted with EtOAc and the combined organic s layers were washed with HZO and saturated aqueous NaCl before being dried s(MgSO4) and concentrated under reduced pressure. The title compound, 1 o 86.0 mg (62%), was isolated by column chromatography (5% EtOAc /
11 hexanes) as a colorless solid. iH NMR (300 MHz, CDC13) 8: 7.88 (1H, d, J
12 7.8 Hz), 7.35 (1H, d, J 7.8 Hz), 7.35 (1H, dd, J = 2.0, 8.4 Hz), 7.28-7.19 13 (7H,m),6.88(1H,d,J=8.3Hz),5.64(1H,s),4.40(2H,q,J=7.1Hz),2.43 14 (3H, s), 1.51 (6H, s), 1.41 (3H, t, J = 7.1 Hz).
Ethy12-fluoro-4-([4-(4-ethylphenyl)-2.2-dimethy-l-(2H)-chromen-6-vlj-eth r}, nyl]-1 s benzoate f,~omnound 2731 17 A solution of 4-ethylbromobenzene (150.0 mg, 0.80 mmol) in 3.0 mL
1e of 'IBF was cooled to -78 C and tert-butyllithium (102.5 mg, 1.60 mmol, 0.94 19 ml of a 1.7M solution in pentane) was added to give a yellow solution.
After 2o 30 minutes a solution of ZnC12 (218.0 mg, 1.60 mmol) in 5.0 mL THF was 21 slowly added via cannula. The resulting solution was warmed to room 22 temperature and transferred via cannula to a solution of ethyl 2-fluoro-4-(2,2-23 dimethyl-4-trifluoromethanesulfonyloxy-(2H)-chromen-6-ylethynyl)-benzoate 24 (Compound 271, 160.0 mg, 0.32 mmol) and tetrakis(triphenylphosphine)palladium(0) (20.0 mg, 0.02 mmol) in 2.0 mL
26 THF. This solution was heated to 50 C for 2 hours, cooled to room 27 temperature, and the reaction quenched by the addition of saturated aqueous 28 NH4C1. This solution was extracted with EtOAc and the combined organic 29 layers were washed with H20 and saturated aqueous NaCI before being dried 1(MgSO4) and concentrated under reduced pressure. The title compound, 2 115.0 mg (79%), was isolated by column chromatography (5% EtOAc /
3 hexanes) as a colorless solid. 1H NMR (300 MHz, CDC13) 8: 7.88 (1H, d, J
4 7.8 Hz), 7.35 (1H, dd, J = 2.0, 8.3 Hz), 7.28-7.20 (7H, m), 6.88 (1H, d, J =
8.3 Hz), 5.65 (1H, s), 4.39 (2H, q, J = 7.1 Hz), 2.73 (2H, q, J = 7.6 Hz), 1.52 6 (6H, s), 1.40 (3H, t, J = 7.1 Hz), 1.31 (3H, t, J = 7.6 Hz).
7 4-[[4-phenyl-2.2-dimethyl-(2H)-chromen-6-yl]-ethynyl-benzoic acid s (Compound 274) 9 To a solution of ethyl 4-[[4-phenyl-2,2-dimethyl-(2H)-chromen-6-yl]-1 o ethynyll-benzoate (Compound 265, 70.0 mg, 0.171 mmol) in 3.0 mL THF and 11 3.0 mL EtOH was added NaOH (120.0 mg, 3.0 mmol, 3.0 mL of a 1M
12 aqueous solution). The resulting solution was heated to 35 C, cooled to 13 room temperature and stirred overnight. The reaction was acidified with 14 10% aqueous HCl and extracted with EtOAc. The combined organic layers were washed with H20, saturated aqueous NaCI, and dried (Na2SO4) before 1 e the solvent was removed under reduced pressure. The residual solid was 17 recrystallized from CH3CN to give 48.0 mg (74%) of the title compound as a 18 colorless solid. 1H NMR (300 MHz, d6-acetone) S: 8.00 (2H, d, J = 8.2 Hz), 1 a 7.57 (2H, d, J = 8.2 Hz), 7.51-7.37 (6H, d), 7.14 (1H, d, J = 2.0 Hz), 6.91 2o (1H, d, J = 8.3 Hz), 5.80 (1H, s), 1.50 (6H, s), 21 4-[[4-(4-methWphelW)-2.2-dimetlW-(2i,~-chromen-6-yl]-ethyul]-benzoic acid 22 (Cgmpound 275) 23 To a solution of ethyl 4-[[4-(4-methylphenyl)-2,2-dimethyl-(2H)-24 chromen-6-yl]-ethynyl]-benzoate (Compound 266, 90.0 mg, 0.213 mmol) in 2s 3.0 mL THF and 3.0 mL EtOH was added NaOH (120.0 mg, 3.0 mmol, 3.0 26 mL of a 1M aqueous solution). The resulting solution was heated to 35 C, 27 cooled to room temperature and stirred overnight. The reaction mixture was 28 acidified with 10% aqueous HCl and extracted with EtOAc. The combined 29 organic layers were washed with H20, saturated aqueous NaCI, and dried 1 (Na2SO4) before the solvent was removed under reduced pressure. The 2 residual solid was recrystallized from CH3CN to give 70.0 mg (83%) the title 3 compound as a colorless solid. 1H NMR (300 MHz, d6-acetone) 8: 8.01 (2H, 4 d, J = 8.3 Hz), 7.57 (2H, d, J = 8.3 Hz), 7.40 (1H, dd, J = 2.1, 8.3 Hz), 7.30-7.20 (4H, m), 7.16 (1H, d, J = 2.0 Hz), 6.90 (1H, d, J = 8.3 Hz), 5.67 (1H, s), 6 2.38 (3H, s), 1.49 (6H, s).
7 4-([4-(4-ethylpheny1)-2.2-dimethyl-(2H)-chromen-6-yl]-eth3nyl]-benzoic acid a (Compound 276) 9 To a solution of ethyl4-[[4-(4-ethylphenyl)-2,2-d'unethyl-(2H)-chromen-io 6-yl]-ethynyl]-benzoate (Compound 267, 82.0 mg, 0.188 mmol) in 3.0 mL
>> THF and 3.0 mL EtOH was added NaOH (120.0 mg, 3.0 mmol, 3.0 mL of a 12 1M aqueous solution). The resulting solution was heated to 35 C, cooled to 13 room temperature and stirred overnight. The reaction mixture was acidified 14 with 10% aqueous HCl and extracted with EtOAc. The combined organic layers were washed with H20, saturated aqueous NaC1, and dried (Na2SO4) 16 before the solvent was removed under reduced pressure. The residual solid 17 was recrystallized from CH3CN to give 65.0 mg (85%) the title compound as 18 a colorless solid. 1H NMR (300 MHz, d6-acetone) 8: 8.01 (2H, d, J = 8.4 Hz), 19 7.57 (2H, -d, J = 8.4 Hz), 7.40 (1H, dd, J = 2.0, 8.3 Hz), 7.35-7.25 (4H, m), 2o 7.17(1H,d,J=2.0Hz),6.90(1H,d,J=8.3Hz),5.77(1H,s),2.69(2H,q,J
21 = 7.6 Hz), 1.49 (6H, s), 1.24 (2H, t, J = 7.6 Hz).
22 4-[[4-(4- 1-meth, l,y ethvl)p enXl)-2.2-dimethy-l-(2H)-chromen-6-vl]-ethvnyl]-2s benzoic acid (Compound 277) 24 To a solution of ethyl4-[[4-(4-(1-methylethyl)phenyl)-2,2-dimethyl-2s (2H)-chromen-6-yl]-ethynyl]-benzoate (Compound 268, 95.0 mg, 0.210 mmol) 26 in 3.0 mL THF and 3.0 mL EtOH was added NaOH (120.0 mg, 3.0 mmol, 27 3.0 mL of a 1M aqueous solution). The resulting solution was heated to 35 28 C, cooled to room temperature and stirred overnight. The reaction mixture 29 was acidified with 10% aqueous HCl and extracted with EtOAc. The t combined organic layers were washed with HZO, saturated aqueous NaCI, and 2 dried (Na2SO4) before the solvent was removed under reduced pressure. The 3 residual solid was recrystallized from CH3CN to give 75.0 mg (84%) the title 4 compound as a colorless solid. 1H NMR (300 MHz, CDC13) 8: 8.05 (2H, d, J
s= 8.4 Hz), 7.55 (2H, d, J = 8.4 Hz), 7.36 (1H, dd, J = 2.0, 8.3 Hz), 7.30-7.26 6 (5H, m), 6.88 (1H, d, J = 8.3 Hz), 5.64 (1H, s), 2.98 (1H, hept, J = 6.9 Hz), 7 1.51 (6H, s), 1.31 (6H, d, J = 6.9 Hz).
s 4[[4-((1.1-dimeth lY ethvl)phenvl)-2.2-dimethyl-(2H)-chromen-6 yl]-ethl]-s benzoic acid (Compound 278,) To a solution of ethyl4-[[4-(4-(1,1-dimethylethyl)phenyl)-2,2-dimethyl-t i (2H)-chromen-6-yl]-ethynyl]-benzoate (Compound 269, 72.0 mg, 0.155 mmol) 12 in 3.0 mL THF and 3.0 mL EtOH was added NaOH (120.0 mg, 3.0 mmol, 13 3.0 mL of a 1M aqueous solution). The resulting solution was heated to 35 14 C, cooled to room temperature and stirred overnight. The reaction mixture was acidified with 10% aqueous HC1 and extracted with EtOAc. The is combined organic layers were washed with H20, saturated aqueous NaCI, and 17 dried (Na2SO4) before the solvent was removed under reduced pressure. The 18 residual solid was recrystallized from CH3CN to give 50.0 mg (74%) the title is compound as a colorless solid. 1H NMR (300 MHz, d6-acetone) 8: 8.00 (2H, 2o d, J = 8.3 Hz), 7.57 (2H, d, J = 8.3 hz), 7.51 (2H, d, J = 8.3 Hz), 7.41 (1H, 21 dd,J=2.0,8.3Hz),7.31(2H,d,J=8.3Hz),7.19(1H,d,J=2.0Hz),6.91 22 (1H, d, J = 8.3 Hz), 5.78 (1H, s), 1.49 (6H, s), 1.35 (9H, s).
23 2-Fluoro-4-[[4-(4-methylohenyl)-2.2-dimethy1-(2H)-chromen-6-yl]-ethvnyll 24 benzoic acid (Compound 2M
To a solution of ethyl2-fluoro-4-[[4-(4-methylphenyl)-2,2-dimethyl-2s (2H)-chromen-6-yl]-ethynyl]-benzoate (Compound 272, 60.0 mg, 0.136 mmol) 27 in 2.0 mL THF and 1.0 mL EtOH was added NaOH (120.0 mg, 3.0 mmol, 2s 3.0 mL of a 1M aqueous solution). The resulting solution was heated to 35 2s C, cooled to room temperature and stirred overnight. The reaction mixture 1 was acidified with 10% aqueous HCl and extracted with EtOAc. The 2 combined organic layers were washed with H20, saturated aqueous NaCI, and 3 dried (Na2SO4) before the solvent was removed under reduced pressure. The 4 residual solid was recrystallized from CH3CN to give 53.0 mg (94%) the title compound as a colorless solid. 1H NMR (300 MHz, d6-acetone) 8: 7.93 (1H, s d,J=7.8Hz),7.43-7.16(8H,m),6.91(1H,d,J=8.3Hz),5.77(1H,s),2.38 7 (3H, s), 1.49 (6H, s).
8 2-Fluoro-4-[[4-(4-ethYlphenyl)-2 2-dimethA-(2H)-chromen-6-vl]-ethy~yl]-s benzoic aci d(Compound 280) To a solution of ethyl 2-fluoro-4-[[4-(4-ethylphenyl)-2,2-dimethyl-(2H)-11 chromen-6-yl]-ethynyl]-benzoate (Compound 273, 82.0 mg, 0.180 mmol) in 12 2.0 mL THF and 2.0 mL EtOH was added NaOH (120.0 mg, 3.0 mmol, 3.0 13 mL of a 1M aqueous solution). The resulting solution was heated to 35 C, 14 cooled to room temperature and stirred overnight. The reaction mixture was acidified with 10% aqueous HCl and extracted with EtOAc. The combined 1 e organic layers were washed with H20, saturated aqueous NaCI, and dried 17 (Na2SO4) before the solvent was removed under reduced pressure. The 18 residual solid was recrystallized from CH3CN to give 70.0 mg (91%) the title 19 compound as a pale-yellow solid. 1H NMR (300 MHz, d6-acetone) 8: 7.93 2o (1H, d, J = 7.8 Hz), 7.41 (1H, dd, J = 2.1, 8.3 Hz), 7.37-7.25 (6H, m), 7.18 21 (1H,d,J=2.0Hz),6.91(1H,d,J=8.3Hz),5.77(1H,s),2.69(2H,q,J=
22 7.6 Hz), 1.49 (6H, s), 1.24 (3H, t, J 7.6 Hz).
23 Method of Potentiating Nuclear Receptor Agonists 24 Overview and introduction We have discovered that a subset of retinoid antagonists which exhibit 26 negative hormone activity can be used for potentiating the biological activities 27 of other retinoids and steroid receptor superfamily hormones. These other 28 retinoids and steroid receptor superfamily hormones can be either 29 endogenous hormones or pharmaceutical agents. Thus, for example, when 1 used in combination with a retinoid negative hormone, certain activities of 2 pharmaceutical retinoid agonists can be rendered more active in eliciting 3 specific biological effects. Advantageously, this combination approach to 4 drug administration can minimize undesirable side effects of pharmaceutical retinoids because lower dosages of the pharmaceutical retinoids can be used 6 with improved effectiveness.
7 More particularly, we have discovered that AGN 193109, a synthetic 8 retinoid having the structure shown in Figure 1, exhibits unique and 9 unexpected pharmacologic activities. AGN 193109 exhibits high affinity for lo the RAR subclass of nuclear receptors without activating these receptors or it stimulating transcription of retinoid responsive genes. Instead, AGN 193109 12 inhibits the activation of RARs by retinoid agonists and therefore behaves as 13 a retinoid antagonist.
14 Additionally, we have discovered that retinoid negative hormones can be used without coadministration of a retinoid agonist or steroid hormone to 16 control certain disease symptoms. More specifically, the retinoid negative 17 hormone disclosed herein can down-regulate the high level basal transcription 18 of genes that are responsive to unliganded RARs. If, for example, 19 uncontrolled cellular proliferation results from the activity of genes 2o responsive to unliganded RARs, then that gene activity can be reduced by the 21 administration of a retinoid negative hormone that inactivates RARs.
22 Consequently, cellular proliferation dependent on the activity of unliganded 23 RARs can be inhibited by the negative hormone. Inhibition of unliganded 24 RARs cannot be achieved using conventional antagonists.
Significantly, we have discovered that AGN 193109 can both repress 26 RAR basal activity and can sometimes potentiate the activities of other 27 retinoid and steroid receptor superfamily hormone agonists. In the context 28 of the invention, a hormone agonist is said to be potentiated by a negative 29 hormone such as AGN 193109 if, in the presence of the negative hormone, a 1 reduced concentration of the agonist elicits substantially the same 2 quantitative response as that obtainable with the agonist alone. The 3 quantitative response can, for example, be measured in a reporter gene assay 4 in vitro. Thus, a therapeutic retinoid that elicits a desired response when used at a particular dosage or concentration is potentiated by AGN 193109 if, 6 in combination with AGN 193109, a lower dosage or concentration of the 7 therapeutic retinoid can be used to produce substantially the same effect as a 8 higher dosage or concentration of the therapeutic retinoid when that 9 therapeutic retinoid is used alone. The list of agonists that can be -1 o potentiated by coadministration with AGN 193109 includes RAR agonists, 11 vitamin D receptor agonists, glucocorticoid receptor agonists and thyroid 12 hormone receptor agonists. More particularly, specific agonists that can be 13 potentiated by coadministration include: ATRA, 13-cis retinoic acid, the 14 synthetic RAR agonist AGN 191183, 1,25-dihydroxyvitamin D3, dexamethasone and thyroid hormone (3,3',5-triiodothyronine). Also disclosed 18 herein is a method that can be used to identify other hormones that can be 17 potentiated by coadministration with AGN 193109.
18 Thus, AGN 193109 behaves in a manner not anticipated for a simple 1 s retinoid antagonist, but as a negative hormone that can potentiate the 2o activities of various members of the family of nuclear receptors. We also 21 disclose a possible mechanism that can account for both negative hormone 22 activity and the ability of AGN 193109 to potentiate the activities of other 23 nuclear receptor ligands. This mechanism incorporates elements known to 24 participate in retinoid-dependent signalling pathways and additionally incorporates a novel negative regulatory component.
26 Those having ordinary skill in the art will appreciate that RARs, which 27 are high affinity targets of AGN 193109 binding, are transcription factors that 28 regulate the expression of a variety of retinoid responsive genes. Cis-29 regulatory DNA binding sites for the RARs have been identified nearby i genes that are transcriptionally regulated in a retinoid-dependent fashion.
2 RAR binding to such DNA sites, known as retinoic acid response elements 3(RA]LtEs), has been well defined.. Importantly, the RAREs bind . . . . =:z , 4 heterodimers consisting of one RAR and one RXR. The RXR component of the.heterodimer functions to, promote a high affinity interaction between 6 the RAR/RXR heterodimer and the RARE (Mangelsdorf et al. The Retinoid I Receptors in The Retinoids: BioloV, Chemist and Medic'ne 2nd edition, 8 eds. Sporn et_ al., Raven Presst Pd., New York 1994..) s As detailed below, our fii;dings related. to the negative hormone 11 activity of AGN 193109 are consistent with a mechanism involving the . - - . - , - 4.
12 interaction. of a putative Negative Coactivator Protein. (NCP) vvith the RAR.
13 According. to: the. proposed.:mechanism, this interaetion is stab'iUied by AGN
14 193109.
Our results further indicated that AGN 193109 can modulate 16 intracellular availability. of NCP for interaction vvith, .nuclear receptors other 17 than RARs that are occupied by AGN 193109. It follows that- AGN 193109 u ca.n. potentiate transcri~tional regulatory pathways'involving nuclear receptors t_ ... , , ts that share with the. RARs . :the ability to bind the NCP. ' In tliis regard, AGN
2o : 193109 .exhibits the ability to modulate a variety of nuclear receptor pathways, 21an activity that would not be predicted for a conventional retinoid antagonist.
22 Accordingly, AGN 193109 is useful as an agent for potentiatilig tile activity of 23 nuclear receptor ligands, including both endogenous horlnones : and 24 prescribed therapeutics. This specific embodiment illustrates the Ynore general _principle that any nuclear receptor negative hormone will poteritiate 26 the activity of other nuctear receptors that competitively bind the NCP:
27 Although other materials and methods'. similar or equivalent to those 28 described herein can be used in the practice or testirig of the present 29 invention, the preferred methods and materials.;arenow ~described 'General WU'99i3382 f PCT/US98/273I4 1 references for methods that can be used to perform the various nucleic acid 2 m:anipulations and procedures described herein can be found in Molecular 3 Cloning: A Laboratory Manual (Sambrook et at. eds.. Cold Spring Harbor Lab 4 Publ. 1989) and Current Protocols in Molecular Biology (Ausubel et al. eds., Greene Publishing Associates and Wiley-Interscience 1987).

7 description' of t'he experiinents and results that led to the creation of the s present_ inventiori follows.
9 Example 6 describes the methods used to: demonstrate that AGN
193109 bound; each of Akee F:ARs with hig4 affinity but failed to activate 111 retiinoid 4ependent gene expression.

12 Example 6 RARs With Hi~ h lAffinitv But Does Not 13 A~I~I 193109 t~nds "
. . . . . .. . . . , 4~. . , . ' . ..
14 Transacdvatd Retinoid-Denendenx Gene F.a` rcp ession ~s Hiaman RAR-a, ~AR-j6 and RAR-y r =ptors were separately 16 expresSed .aS -3eroIllbinait vrOteins;uSing a ba6u:IovirLis'expression system . _ , 17 essenti a~iy a~cording to'ahe method described by Aiiegretto et a1. in J.
BioL
W. Chem. 20 2.6625 (1993),. The recombinant reeeptQr proteins were separately 19 employe4}for determming ;AGbi 193109binding:'s.ffinities using the [3H]-2o. ATRA:d-tsplacement assay described by Iieyhan et al, in Ce1168:397 (1992).
21 Dissociation constants (K~ds) were tletermined: according to the procedure ;:.
22 : des+cnbe~ by Cheng et al. in Braochern~cal Pharmacology 22:3099 (1973).
23 AGN 193109 was` also tested for..its ability to transactivate RARs in 24 CV-1 celts transiently cotransfected with: RAR expression vectors and a 2s retinold responsive reporter gene constriict.. Receptor expression vectors . ,_ 26, pRShRAR-a (Giguere et aL Nature 330:624 (1987)), pRShRAR E3 (Benbrook 27 et al )-1'nture 333:669 (1988)) and pRShRAR-y'.(Ishikawa et al. Mol 28 Endocrinol. 4:837 (1990)) were separately cotransfected with the AMTV-29 TREp-huc reporter plasmid. Use of this 'Iuciferase reporter plasmid has -been I disclosed by Heyman et al. in Cell 68:397 (1992). The OIVITV-TREp-Luc 2 plasmid is essentially identical to the AMTV-TREp-CAT reporter construct 3 described by Umesono et al. in Nature 336:262 (1988), except that the 4 chloramphenicol acetyltransferase (CAT) reporter gene was substituted by a polynucleotide sequence encoding firefly luciferase. Transfection of green 6 monkey CV-1 cells was carried out using the calcium phosphate 7 coprecipitation method described in Molecular Cloning: A Laboratory Manual 8 (Sambrook et al. eds. Cold Spring Harbor Lab Publ. 1989). CV-1 cells were 9 plated at a density of 4 X 104/well in 12 well multiwell plates and transiently lo transfected with a calcium phosphate precipitate containing 0.7 g of reporter 11 plasmid and 0.1 g of receptor plasmid according to standard laboratory 12 procedures. Cells were washed after 18 hours to remove the precipitate and 13 refed with Dulbecco's modified Eagle's medium (DMEM) (Gibco), 14 containing 10% activated charcoal extracted fetal bovine serum (Gemini Bio-Products). Cells were treated with vehicle alone (ethanol) or AGN 193109 16 (10-9 to 10-6 M) for 18 hours. Cell lysates were prepared in 0.1 M KPO4 (pH
17 7.8), 1.0% TRITON X-100, 1.0 mM DTT, 2 mM EDTA. Luciferase activity 18 was measured as described by de Wet et al. in MoL Cell. Biol. 7:725 (1987), is using firefly luciferin (Analytical Luminescence Laboratory) and an EG&G
2o Berthold 96-well plate luminometer. Reported luciferase values represented 21 the mean SEM of triplicate determinations.
22 The results presented in Table 11 indicated that AGN 193109 bound 23 each of RAR-a, RAR -0 and RAR-y with high affinity, but did not activate 24 retinoid-dependent gene expression. More specifically, AGN 193109 bound .2s each of the three receptors with Kd values in the 2 - 3 nM range. Despite 26 this tight binding, AGN 193109 failed to activate gene expression when 27 compared with inductions stimulated by ATRA. Accordingly, the half-28 maximal effective concentration of AGN 193109 (EC50) was unrneasurable.
29 Although not presented in the Table, we also found that AGN 193109 had no 1 measurable affinity for the RXRs.

3 AGN 193109 Binding and Transactivation of the RARs 4 RAR-a RAR-P RAR-y EC., (nM) No Activity No Activity No Activity s I{d (nM) 2 2 3 8 Example 7 describes the methods used to demonstrate that AGN
9 193109 is an antagonist of ATRA dependent gene expression.
Example 7 11 AGN 193109-Dependent Inhibition of RAR Transactivation 12 bv ATRA
13 The ability of AGN 193109 to antagonize ATRA mediated RAR
14 activation was investigated in CV-1 cells cotransfected by the calcium phosphate coprecipitation method of Sambrook et al. (Molecular Cloning.= A
16 Laboratory Manual Cold Spring Harbor Lab Publ. 1989). Eukaryotic 17 expression vectors pRShRAR-a (Giguere et al. Nature 330:624 (1987)), 18 pRShRAR-/3 (Benbrook et al. Nature 333:669 (1988)) and pRShRAR-y ts (Ishikawa et al. Mol. EndocrinoL 4:837 (1990)) were cotransfected with the MTV-Luc reporter plasmid described by Hollenberg et al. (Cell 55:899 21 (1988)). Notably, the reporter plasmid contained two copies of the TRE-22 palindromic response element. Calcium phosphate transfections were carried 23 out exactly as described in Example 6. Cells were dosed with vehicle alone 24 (ethanol), ATRA (10-9 to 10-6 M), AGN 193109 (10-9 to 1& M), or 10-$ M
ATRA in combination with AGN 193109 (10-9 to 10-6 M) for 18 hours. Cell 26 lysates and luciferase activity measurements were also performed as in 27 Example 6.
28 The results of these procedures are presented in Figures 2A through 29 2F where luciferase values represent the mean SEM of triplicate 3o determinations. More specifically, the results presented in Figures 2A, 2C

I and 2E indicated that stimulation of transfected cells with ATRA led to dose 2 responsive increases in luciferase activity. This confirmed that ATRA
3 activated each of the three RARs in the experimental system and provided a 4 comparative basis for detecting the activity of an antagonist. The graphic results presented in Figures 2B, 2D and 2F indicated that cotreatment of 6 transfected cells with 10 nM ATRA and increasing concentrations of AGN
7 193109 led to an inhibition of luciferase activity. In particular, equal doses of 8 AGN 193109 and ATRA gave greater than 50% inhibition relative to ATRA
s alone for all three RAR subtypes. Comparison of the ATRA dose response lo in the presence of different concentrations of AGN 193109 indicated that i I ATRA was competitively inhibited by AGN 193109. Notably, the horizontal 12 axes on all of the graphs shown in Figure 2 represents the log of the retinoid 13 concentration. These results proved that AGN 193109 was a potent RAR
14 antagonist.
is We next performed experiments to elucidate the mechanism underlying 18 the antagonist activity of AGN 193109. Those having ordinary skill in the art 17 will appreciate that nuclear receptor activation is believed to involve a 18 conformational change of the receptor that is induced by ligand binding.
is Indeed, the results of protease protection assays have confirmed that nuclear 2o hormone agonists and antagonists cause receptor proteins to adopt different 21 conformations (Keidel et al. MoL CelL Biol. 14:287 (1994); Allan et al. J.
Biol 22 Chem. 267:19513 (1992)). We used such an assay to determine if AGN
23 193109 and ATRA caused RAR-a to adopt different conformations. AGN
24 193583, an RAR-a-selective antagonist, was included as a positive control 25 that is known to confer an antagonist-specific pattern of protease sensitivity.
26 Example 8 describes one method that was used to detect 27 conformational changes in RAR-a resulting from AGN 193109 binding. As 28 presented below, the results of this procedure unexpectedly indicated that 29 AGN 193109 led to a pattern of trypsin sensitivity that was substantially 1 identical to that induced by ATRA, an RAR agonist, and unlike that induced 2 by a model RAR antagonist. This finding suggested that AGN 193109 3 possessed properties distinct from other retinoid antagonists.
4 Example 8 Protease Protection An lvsis 6 A plasmid constructed in the vector pGEM3Z (Pharmacia) and 7 containing the RAR-a cDNA (Giguere et al. Nature 330:624 (1987)), was 8 used in connection with the TNT-coupled reticulocyte lysate in vitro 9 transcription-translation system (Promega) to prepare [35S]-methionine 1 o labeled RAR-a. Limited proteolytic digestion of the labeled protein RAR-a 11 was carried out according to the method described by Keidel et al. in Mol.
12 CelL Biol 14:287 (1994). Aliquots of reticulocyte lysate containing [35S]-13 methionine labeled RAR-a were incubated with either ATRA, AGN 193583 14 or AGN 193109 on ice for 45 minutes in a total volume of 9 l. The retinoid final concentration for all trials was 100 nM for ATRA and AGN 193109, 16 and 1000 nM for AGN 193583. The difference between the final 17 concentrations of the retinoids was based on the approximate 10-fold 18 difference in relative affinities of ATRA and AGN 193109 (having Kd at 19 RAR-a of 2 and 10 nM, respectively) and AGN 193583 (having Kd at RAR-2o a of a100 nM). After ligand binding, 1 l of appropriately concentrated 21 trypsin was added to the mixture to give final concentrations of 25, 50 or 22 g/ml. Samples were incubated at room temperature for 10 minutes and 23 trypsin digestion stopped by addition of SDS-sample buffer. Samples were 24 subjected to polyacrylamide gel electrophoresis and autoradiographed according to standard procedures.
26 Both the agonist and antagonist led to distinct patterns of trypsin 27 sensitivity that were different from the result obtained by digestion of the 28 unliganded receptor. Autoradiographic results indicated that trypsin 29 concentrations of 25, 50 and 100 g/ml completely digested the radiolabeled 1 RAR-a in 10 minutes at room temperature in the absence of added retinoid.
2 Prebinding of ATRA led to the appearance of two major protease resistant 3 species. Prebinding of the RAR-a-selective antagonist AGN 193583 gave rise 4 to a protease resistant species that was of lower molecular weight than that s resulting from ATRA prebinding. This result demonstrated that a retinoid 6 agonist and antagonist led to conformational changes detectable by virtue of 7 altered trypsin sensitivities. Surprisingly, prebinding of AGN 193109 gave rise 8 to a protease protection pattern that was indistinguishable from that 9 produced by prebinding of ATRA.
The results presented above confirmed that AGN 193109 bound the 11 RAR-a and altered its conformation. Interestingly, the nature of this 12 conformational change more closely resembled that which resulted from 13 binding of an agonist (ATRA) than the alteration produced by antagonist 14 (AGN 193583) binding. Clearly, the mechanism of AGN 193109 dependent antagonism was unique.
16 We considered possible mechanisms that could account for the 17 antagonist activity of AGN 193109. In particular, we used a standard gel shift 18 assay to test whether AGN 193109 perturbed RAR/RXR heterodimer 19 formation or inhibited the interaction between RAR and its cognate DNA
2o binding site.
21 Example 9 describes a gel electrophoretic mobility-shift assay used to 22 demonstrate that AGN 193109 neither inhibited RAR/RXR dimerization nor 23 inhibited binding of dimers to a target DNA.
24 Example 9 Gel Shift AnaW
26 In vitro translated RAR-a was produced essentially as described under 27 Fxample 8, except that 35S-labeled methoinine was omitted. In vitro 28 translated RXR-a was similarly produced using a pBluescript(II)(SK)-based 29 vector containing the RXR-a cDNA described by Mangelsdorf, et al. in 1 Nature 345:224-229 (1990) as the template for generating in vitro transcripts.
2 The labeled RAR-a and RXR-a, alone or in combination, or prebound with 3 AGN 193109 (10-6 M) either alone or in combination, were allowed to 4 interact with an end-labeled DR-5 RARE double-stranded probe having the sequence 5'-TCAGGTCACCAGGAGGTCAGA-3' (SEQ ID NO:1). The 6 binding mixture was electrophoresed on a non-denaturing polyacrylamide gel 7 and autoradiographed according to standard laboratory procedures. A single 8 retarded species appearing on the autoradiograph that was common to all the 9 lanes on the gel represented an undefined probe-binding factor present in the 1 o reticulocyte lysate. Only the RARlRXR combination gave rise to a retinoid 11 receptor-specific retarded species. Neither RAR alone nor RXR alone 12 bound the probe to produce this shifted species. The presence of AGN
13 193109 did not diminish this interaction.
14 These results indicated that AGN 193109 did not substantially alter either the homo- or hetero-dimerization properties of RAR-a. Further, 16 AGN 193109 did not inhibit the interaction of receptor dimers with a DNA
17 segment containing the cognate binding site.
ts In view of the unique properties which characterized AGN 193109, we 19 proceeded to investigate whether this antagonist could additionally inhibit the 2o activity of unliganded RARs. The receptor/reporter system used to make this 21 determination advantageously exhibited high level constitutive activity in the 22 absence of added retinoid agonist. More specifically, these procedures 23 employed the ER-RAR chimeric receptor and ERE-tk-Luc reporter system.
24 The ERE-tk-Luc plasmid includes the region -397 to -87 of the estrogen responsive 5'-flanking region of the Xenopus vitellogenin A2 gene, described 2s by Klein-Hitpass, et al. in Cell 46:1053-1061 (1986), ligated upstream of the 27 HSV thymidine kinase promoter and luciferase reporter gene of plasmid tk-28 Luc. The ER-RAR chimeric receptors consisted of the estrogen receptor 29 DNA binding domain fused to the "D-E-F"' domain of the RARs. Those I having ordinary skill in the art appreciate this "D-E-F" domain functions to 2 bind retinoid, to provide a retinoid inducible transactivation function and to 3 provide a contact site for heterodimerization with RXR. Thus, luciferase 4 expression in this reporter system was dependent on activation of the transfected chimeric receptor construct.
6 Example 10 describes the method used to demonstrate that AGN
7 193109 inhibited basal gene activity attributable to unliganded RARs. These 8 procedures were performed in the absence of added retinoid agonist. The 9 results presented below provided the first indication that AGN 193109 io exhibited negative hormone activity.
11 Example 10 12 Repression of Basal Gene Activity of a Retinoid-Regulated 13 Reporter in Transiently Cotransfected Cell Lines 14 CV-1 cells were co-transfected with the ERE-tk-Luc reporter plasmid and either ER-RAR-a, ER-RAR -0 or ER-RAR-y expression plasmids. The ls ERE-tk-Luc plasmid contained the estrogen-responsive promoter element of 17 the Xenopus laevis vitellogenin A2 gene and was substantially identical to the 18 reporter plasmid described by Klein-Hitpass et al. in Cell 46:1053 (1986), 19 except that the CAT reporter gene was substituted by a polynucleotide 2o sequence encoding luciferase. The ER-RAR-a, ER-RAR-0 and ER-RAR-y 21 chimeric receptor-encoding polynucleotides employed in the co-transfection 22 have been described by Graupner et al. in Biochern. Biophys. Res. Comm.
23 179:1554 (1991). These polynucleotides were ligated into the pECE
24 expression vector described by Ellis et al. in Cell 45:721 (1986) and expressed under transcriptional control of the SV-40 promoter. Calcium phosphate 26 transfections were carried out exactly as described in Example 6 using 0.5 27 g/well of reporter plasmid and either 0.05 g, 0.10 g or 0.2 g/well of 28 receptor plasmid. Cells were dosed with vehicle alone (ethanol), ATRA (10-9 29 to 10-6 M), or AGN 193109 (10-9 to 10-6 M) for 18 hours. Cell lysates and I luciferase activity measurements were performed as described in Example 6.
2 The results presented in Figures 3A, 4A and 5A confirmed that ATRA
3 strongly induced luciferase expression in all transfectants. Basal level 4 expression of luciferase for the three transfected chimeric RAR isoforms ranged from approximately 7,000 to 40,000 relative light units (rlu) and was e somewhat dependent on the amount of receptor plasmid used in the 7 transfection. Thus, the three chimeric receptors were activatable by ATRA, s as expected. More specifically, all three receptors bound ATRA and 9 activated transcription of the luciferase reporter gene harbored on the ERE-io tk-Luc plasmid.
11 Figures 3B, 4B and 5B present AGN 193109 dose response curves 12 obtained in the absence of any exogenous retinoid agonist. Interestingly, 13 ER-RAR-a (Figure 3B) was substantially unaffected by AGN 193109, while 14 the ER-RAR-0 and ER-RAR-y chimeric receptors (Figures 4B and 5B, respectively) exhibited an AGN 193109 dose responsive decrease in luciferase le reporter activity.
17 We further investigated the negative hormone activity of AGN 193109 18 by testing its ability to repress gene expression mediated by a chimeric RAR-ls y receptor engineered to possess a constitutive transcription activator domain. More specifically, we used a constitutively active RAR-y chimeric 21 receptor fused to the acidic activator domain of HSV VP-16, called RAR-y-22 VP-16, in two types of luciferase reporter systems. The first consisted of the 23 ERE-tk-Luc reporter cotransfected with ER-RARs and ER-RXR-a. The 24 second utilized the AMTV-TREp-Luc reporter instead of the ERE-tk-Luc reporter.
26 Example 11 describes the method used to demonstrate that AGN
27 193109 -could suppress the activity of a transcription activator domain of an 28 RAR. The results presented below proved that AGN 193109 could suppress 29 RAR-dependent gene expression in the absence of an agonist and confirmed 1 that AGN 193109 exhibited negative hormone activity.
2 Example 11 s Re,pression of RAR-VP-16 Activitv in Transiently 4 Transfected Cells CV-1 cells were transiently, cotransfected according to the calcium s phosphate coprecipitation technique described under Example 6 using 0.5 7,Aglwell of the ERE-tk-Luc luciferase reporter plasmid, '0.1 g/well of the ER-a RXR-a chimeric reporter.expression plasmid, and either 0 g or 0.1 pg/well .a of the RAR-y-VP-16 expression pinsmid. The chimeric receptor ER-RXR-a consisted of the hormone binding domain (amino acids 181 to 458) of 'RXR-1.1 a(Mangelsdorf; et al. Nature .345:224-229 (1990)) fused to the estrogen 12 receptor DNA .binding domain (Graupner, et al. Biochem. Biophys. Res.
13 Comm. 179:1554 (1991)) and was expressed from the SV-40 based expression 14 vector pECE described by Ellis;' et al. in Cell 45:72I. (1986).. RAR-Y-VP-16 is is identical to the VP16RAR-yl.expr.ession-plasmid deseribed by Nagpal etal.
16- -in EMBLf:J. 12:2349';(19 i7 and encod..e.s. a chimerie.protein having the activation.domain.of . 1a the VP-16 protein of HSV fused to the -ainino-terminus .of full IenBth.
RAR-y.
is At eighteen hours post-transfection, cells were rinsed with phosphate 2o .buffered :.saline (PBS) and fed.tiv~,h DMEM (Gibco-BRL) containing 10%
21 FBS (Gemini Bio-Products) that had. been extracted with charcoal.to remove 22 retinoids. Cells were dosed with~an appropriate dilution of AGN 193109 or 23 ATRA iia ethanol vehicle or ethanol alone, for 18 hours, then rinsed with PBS
24 and Iysed using 0.1 M KPQ¾.(pH 7.8), 1.0% TRITON X-100, 1.0 mM DTT, 2 25 mM EDTA. Luciferase activity was measured according to_ the method 26 described by de Wet, et al, in MoL CeM Biol. 7:725 (1987using firefly 27 luciferin (Analytical Luminescence I:aboratory) and an EG&G Berthold 96-28 well plate luminometer. Lueiferase. values represented the..mean SEM of 29 triplica#e. determinations.

t As shown in Figure 6, CV-1 cells transfected with the ERE-tk-Luc 2 reporter construct and the ER-RAR-a chimeric expression plasmid exhibited 3 a weak activation of luciferase activity by ATRA, likely due to isomerization 4 of ATRA to 9C-RA, the natural ligand for the RXRs (Heyman et al. Cell 68:397 (1992). Cells transfected with the same mixture of reporter and e chimeric receptor plasmids but treated with AGN 193109 did not exhibit any 7 effect on luciferase activity. As AGN 193109 does not bind to the RXRs, 8 this latter result was expected. CV-1 cells similarly transfected with the ERE-9 tk-Luc reporter but with substitution of an ER-RAR chimeric receptor io expression plasmid for ER-RXR-a exhibited a robust induction of luciferase 11 activity following ATRA. treatment.
12 In contrast, inclusion of the RAR-y-VP-16 expression plasmid with the 13 ER-RXR-a and ERE-tk-Luc plasmids in the transfection mixture resulted in 14 a significant increase in the basal luciferase activity as measured in the absence of any added retinoid. This increase in basal luciferase activity te observed for the ER-RXR-a/RAR-y-VP-16 cotransfectants, when compared 17 to the result obtained using cells transfected with ER-RXR-a alone, indicated ls that recombinant ER-RXR-a and RAR-y-VP-16 proteins could 19 heterodimerize. Interaction of the heterodimer with the cis-regulatory 2o estrogen responsive element led to a targeting of the VP-16 activation 21 domain to the promoter region of the ERE-tk-Luc reporter. Treatment of 22 such triply transfected cells with ATRA led to a modest increase of luciferase 23 activity over the high basal level. However, treatment of the triple 24 transfectants with AGN 193109 resulted in a dose dependent decrease in luciferase activity. Importantly, Figure 6 shows that AGN 193109 treatment 26 of cells cotransfected with ER-RXR-a and RAR-y-VP-16 led to repression 27 of luciferase activity with maximal inhibition occurring at approximately 10-$
28 M AGN 193109.
29 Our observation that AGN 193109 repressed the constitutive 1 transcriptional activation function of RAR-y-VP-16 in the presence of an 2 RXR was explained by a model wherein binding of AGN 193109 to the RAR
3 induced a conformational change in the RAR which stabilizes a negative 4 conformation that facilitates the binding of a trans-acting negative coactivator protein. When the AGN 193109/RAR complex is bound by the NCP, the 6 RAR is incapable of upregulating. transcription of genes that are ordinarily 7 responsive to activated RARs. Our model further proposes that the 8 intracellular reservoir of NCP is in limiting concentration in certain contexts 9 and can be depleted by virtue of AGN 193109 stimulated complexation with lo RARs.
11 The results presented in Figure 6 additionally indicated that even at 10-12 6 M AGN 193109, the ER-RXR-a and RAR-y-VP-16 proteins could interact 13 to form heterodimers competent for activating transcription of the reporter 14 gene. More specifically, cells transfected with ER-RXR-a and RAR-y-VP-16 and treated with AGN 193109 at a concentration (10-8-10-6 M) sufficient to 16 provide maximal inhibition, gave luciferase activity readings of approximately 17 16,000 rlu. Conversely, cells transfected only with ER-RXR-a and then 18 treated with AGN 193109 at a concentration as high as 10-6 M exhibited is luciferase expression levels of only approximately 8,000 rlu. The fact that a 2o higher level of luciferase activity was obtained in cells that expressed both 21 ER-RXR-a and RAR-y-VP-16, even in the presence of 10-6 M AGN 193109 22 demonstrated the persistence of an interaction between the two recombinant 23 receptors. The repression of RAR-Y-VP-16 activity by AGN 193109 24 suggested that modulation of NCP interaction can be codominate with VP-16 activation. Accordingly, we realized that it may be possible to modulate the 26 expression of genes which are not ordinarily regulated by retinoids in an 27 AGN 193109 dependent manner.
28. Candidates for AGN 193109 regulatable genes include those that are 29 activated by transcription factor complexes which consist of non-RAR
factors y that associate or heterodimerize with RARs, wherein the non-RAR factor 2 does not require an RAR agonist for activation. While stimulation with an 3 RAR agonist may have substantially no effect on the expression of such 4 genes, administration with AGN 193109 can promote formation of inactive s transcription complexes comprising AGN 1931091RAR/NCP. Consequently, 6 addition of the AGN 193109 retinoid negative hormone can down-regulate 7 transcription of an otherwise retinoid-insensitive gene.
s lhis. same mechanism can account for the observation that AGN
9 193109 can repress the activity of the tissue,transglutaminase (TGase) gene in lo HL-60 cells. A retinoid response element consisting of three canonical ii retinoid half sites spaced by 5 and 7 base pairs has been identified in the 12 transcription control region of this gene. While TGase can be induced by 13 RXR-selective agonists, it is not responsive to RAR-selective agonists. The 14 TGase retinoid response elenlent is bound by an RAR/RXR heterodimer 15 Nagy, L., J. of Biol. Chem., 271(8):4355-4365 (1996). Interestingly, AGN
193109 is able to repress TGase 16 activity induced by RXR agonists. This AGN 193109 mediated repression 17 can be accounted for by the ability of this negative hormone to sequester is NCPs to the RAR component of the heterodimer, thereby repressing the i s activity of the associated RXR:
20 We have also obtained results which support conclusions identical to 21 those presented under Exarhple 11 by employing RAR-y-VP-16 and 22 expression constructs and the OMTV-TREp-Luc reporter plasmid instead of 23 the RAR-y-VP-16 and ER-RXR-a expression constructs in combination with 24 the ERE-tk-Luc reporter plasmid. Consistent with the results presented 2s above, we found that RAR-Y-VP-16 activity at the AMTV-TREp-Luc 26 reporter was inhibited by AGN 193109. Therefore, AGN 193109 repressed 27 RAR-y-VP-16 activity when this chimeric receptor was direetly bound to a 28 retinoic acid receptor response element instead of indirectly bound to an 29 estrogen response element in the promoter region of the reporter plasmid:

1 These findings demonstrated that an assay for identifying agents having 2 negative hormone activity need not be limited by the use of a particular 3 reporter plasmid. Instead, the critical feature embodied by an experimental 4 system useful for identifying retinoid negative hormones involves detecting the ability of a compound to repress the activity of an RAR engineered to 6 contain a constitutive transcription activation domain.
7 Generally, retinoid negative hormones can be identified as the subset 8 of retinoid compounds that repress within a transfected cell the basal level 9 expression of a reporter gene that is transcriptionally responsive to direct or 1 o indirect binding by a retinoid receptor or a chimeric receptor that includes at ii least the domains of the retinoid receptor located C-terminal to the DNA
12 binding domain of that receptor. This approach has been adapted to a 13 screening method useful for identifying retinoid negative hormones. In the 14 various embodiments of the invented screening method, the structure of the receptor for which a negative hormone is sought is variable. More 1 e specifically, the retinoid receptor can be either of the RAR or the RXR
17 subtype. The receptor can optionally be engineered to include a constitutive ls transcription activator domain. The retinoid receptor used to screen for 1s negative hormones optionally contains a heterologous DNA binding domain 2o as a substitute for the DNA binding domain endogenous to the native 21 receptor. However, when a second receptor is used in the screening method, 22 and where the second receptor can dimerize with the retinoid receptor for 23 which a negative hormone is sought, then that retinoid receptor may not 24 require a DNA binding domain because it can be linked to the transcription control region of the reporter gene indirectly through dimerization with the 2s second receptor which is itself bound to the transcription control region.
27 In the practice of the screening method, the ability of a compound to 28 repress the basal expression of a reporter is typically measured in an in vitro 29 assay. Basal expression represents the baseline level of reporter expression in 1 transfected cells under conditions where no exogenously added retinoid 2 agonist is present. Optionally, steps may be taken to remove endogenous 3 retinoidligands from the environment of the transfected cells via procedures 4 such as charcoal extraction of the serum that is used to culture cells in vitro.
Examples of reporter genes useful in connection with the screening 6 method include those encoding luciferase, beta galactosidase, 7 chloramphenicol acetyl transferase or cell surface antigens that can be 8 detected by lnmmunochemlcal means. In practice, the nature of the reporter 9 gene is not expected to be critical for the operability of the method.
t o However,, the transcriptional regulatory region of the reporter construct must 11 include one or more cis-regulatory elements that are targets of transcription 12 factors for which negative hortnones are being sought. For example, if one 13 desires to identify RAR negative hormones, then the transcriptional 14 regulatory region of the reporter construct could contain a cis-regulatory element that can be bound -by an RAR=containing protein. In this example, 16 there should be correspondence between the DNA binding domain of the 17 RAR and the cis-regulatory element of the'transcriptional regulatory region le of.the reporter construct. Thus, if a-chimeric RAR having a constitutive is transcription activator domain and a DNA binding domain that can bind cis-2o regulatory estrogen responsive elements is employed in the screening method, 21 then the -transcriptional regulatory region of the reporterconstruct should 22 contain an estrogen responsive eiement.
23 Examples of cis-regulatory elements that directly bind retinoid 24 receptors (1ZAREs) 'useful in connection with the reporter assay are disclosed by Mangelsdorf et at: in The Retinoid Receptors in The Retinoids: Biolo,.g,y, 26 Chemistry and'Medicine, 2nd edition, eds. Sporn et al., Raven Press, Ltd., 27 New`York (1994)õ
28 Examples of cis-regulatory elements that indirectly bind 29 chiirieric receptors include DNA binding sites for any DNA binding protein I for which the DNA binding domain of the protein can be incorporated into a 2 chimeric receptor consisting of this DNA binding domain attached to a 3 retinoid receptor. Specific examples of heterologous DNA binding domains 4 that can be engineered into chimeric receptors and that will recognize heterologous cis-regulatory elements include those recognizing estrogen 6 responsive elements. Thus, the retinoid receptor portion of a chimeric 7 receptor useful in connection with the screening method need not contain the 8 DNA binding of the retinoid receptor but must contain at least the ligand 9 binding domain of the retinoid receptor.
A further example of indirect retinoid receptor binding to the cis-11 regulatory element includes the use of a protein that can bind the cis-12 regulatory element and dimerize with a retinoid receptor. In this case, the 13 retinoid receptor associates with the cis-regulatory element only by 14 association with the protein responsible for DNA binding. An example of such a system would include the use of a fusion protein consisting of a 16 heterologous DNA binding domain fused to an RXR, containing at least the 17 domain of the RXR responsible for dimerization with RARs. Cointroduced is RARs can dimerize with such a fusion protein bound to the cis-regulatory 19 element. We anticipate that any cis-regulatory element-binding protein that dimerizes with RARs to result in an indirect association of the RAR with the 21 cis-regulatory element will also be suitable for use with the negative hormone 22 screening method.
23 In a preferred embodiment of the screening method, retinoid negative 24 hormones are identified as those retinoids that repress basal expression of an 2s engineered RAR transcription factor having increased basal activity.
26 Although not essential for operability of the screening method, the 27 engineered RAR employed in the following Example included a constitutive 28 transcription activating domain. Use of this chimeric receptor advantageously 29 provided a means by which the basal expression of a reporter gene could be 1 elevated in the absence of any retinoid. Although we have employed 2 transient transfection in the procedures detailed above, stably transfected cell 3 lines constitutively expressing the chimeric receptor would also be useful in 4 connection with the screening method.
As disclosed in the following Example, a chimeric retinoid receptor 6 having a constitutive transcription activator domain was heterodimerizable 7 with a second receptor engineered to contain a DNA binding domain specific 8 for an estrogen responsive cis-regulatory element. In this case the chimeric 9 retinoid receptor having a constitutive transcription activator domain 1 o associates with the cis-regulatory region controlling reporter gene expression 11 indirectly via binding to a second receptor that binds a DNA target sequence.
12 More particularly, the second receptor was engineered to contain a DNA
13 binding domain that recognized an estrogen responsive element.
14 Advantageously, the reporter gene having an estrogen responsive element in the upstream promoter region was unresponsive to retinoid agonists in the 16 absence of the transfected chimeric receptor having the constitutive 17 transcription activator domain. Accordingly, all reporter gene activity was 18 attributed to the transfected receptors. The combination use of the estrogen 19 responsive element DNA binding domain and the estrogen responsive zo element cis-regulatory element are intended to be illustrative only. Those 21 having ordinary skill in the art will realize that other combinations of 22 engineered receptors having specificity for non-RARE cis-regulatory elements ra will also be useful in the practice of the invented screening method.
24 Cells useful in connection with the screening method will be eukaryotic cells that can be transfected. The cells may be animal cells such as human, 26 primate or rodent cells. We have achieved very good results using CV-1 27 cells, but reasonably expect that other cultured cell lines could also be used 28 successfully. Any of a number of conventional transfection methods known 29 in the art can be used to introduce an expression construct encoding the 1 chimeric retinoid receptor having a constitutive transcription activator 2 domain.
3 The constitutive transcription activator domain will consist of a 4 plurality of amino acids which will likely have an overall acidic character as represented by a negative charge under neutral pH conditions. For example, 6 the constitutive transcription activator domain may have an amino acid 7 sequence which is also found in viral transcription factors. One example of a 8 viral transcription factor having a constitutive transcription activator domain 9 is the herpes simplex virus 16. However, other viral or synthetic transcription t o activator domains would also be useful in the construction of expression 11 constructs encoding the chimeric retinoid receptor having a constitutive 12 transcription activator domain.
13 As described below, we have developed a generalized screening 14 method useful for identifying retinoid negative hormones. This screening method provides a means for distinguishing simple antagonists from negative 16 hormones. Table 12 lists several retinoid compounds which exhibit potent 17 affinity for RAR-y yet, with the exception of ATRA, did not transactivate 18 this receptor in a transient cotransfection transactivation assay. We therefore 19 tested these compounds to determine which were RAR-y antagonists and 2o which, if any, of these antagonists exhibited negative hormone activity.
21 Example 12 describes the method used to identify retinoid compounds 22 that were antagonists, and the subset of antagonists that exhibited negative 23 hormone activity.
24 Example 12 Assay for Retinoid Negative Hormones 26 4 X 104 CV-1 cells were transfected by the calcium phosphate 27 coprecipitation procedure described in Molecular Cloning: A Laboratory 28 Manual (Sambrook et al. eds. Cold Spring Harbor Lab Publ. 1989) using 0.5 29 g ERE-tk-Luc reporter plasmid and 0.1 g ER-RAR-y (Graupner et al.

1 Biochem. Biophys. Res. Comm. 179:1554 (1991)) chimeric expression plasmid.
2 After 18 hours, cells were rinsed with PBS and fed with DMEM (Gibco-3 BRL) containing 10% activated charcoal extracted FBS (Gemini Bio-4 Products). Cells were treated with 104 M ATRA in ethanol or ethanol alone.
In addition, ATRA treated cells were treated with 10-9, 10-8, 10-7 or 10-6 M
of 6 the compounds listed in Table 12. After 18 hours, cells were rinsed in PBS
7 and lysed in 0.1 M KPO4 (pH 7.8), 1.0% TRITON X-100, 1.0 mM DTT, 2 8 mM EDTA. Luciferase activities were measured as described by deWet et al.
9 in MoL Cell. BioL 7:725 (1987).

11 Compound Kd (nM) @ R.AR-y4 EC50 (nM) @ RAR-ya 13 AGN 193109 6 na 14 (Compound 60) AGN 193174 52 na 16 (Compound 34a) 17 AGN 193199 30 na 18 AGN 193385 25 na 1 s (Compound 23) 2o AGN 193389 13 na 21 (Compound 25) 22 AGN 193840 40 na 23 AGN 193871 30 na 24 (Compound 50) 26 a Relative affinity (Kd) determined by competition of 3H-ATRA
27 binding to baculovirus expressed RAR-y and application of the Cheng-28 Prussof equation.
29 b EC.50 measured in CV-1 cells transiently cotransfected with 3o AMTV-TREp-Luc and RS-RAR-y. "na" denotes no activity.

1 As indicated by the results presented in part in Figure 7 and in Table 2 12,. with the exception of ATRA, all of the compounds listed in Table 12 3 were retinoid antagonists at RAR-y.
4 The RAR-y antagonists identified in Table 12 were next screened to determine which, if any, were also retinoid negative hormones. 4 X 104 CV-1 6 cells were transfected according to the calcium phosphate procedure 7 described in Molecular Cloning: A Laboratory Manual (Sambrook et al. eds.
e Cold Spring Harbor Lab Publ. 1989) using 0.5 g ERE-tk-Luc reporter 9 plasmid and 0.1 g ER-RXR-a (Graupner et al. Biochem. Biophys. Res.
to Comm. 179:1554 (1991)) and 0.2 g RAR-y-VP-16 (Nagpal et al. EMBO J.
11 12:2349 (1993)) chimeric expression plasmids. After 18 hours, cells were 12 rinsed with PBS and fed with DMEM (Gibco-BRL) containing 10% activated 13 charcoal extracted FBS (Gemini Bio-Products). Cells were treated with 10-9, 14 10-8, 10-7 or 10-6 M of each of the compounds listed in Table 12. Treatment with ethanol vehicle alone served as the negative control. After 18 hours, 16 cells were rinsed in PBS and lysed in 0.1 M KPO4 (pH 7.8), 1.0% TRITON
17 X-100, 1.0 mM DTT, 2 mM EDTA. Luciferase activities were measured as 18 previously by deWet et al. in Mol Cell. BioL 7:725 (1987).
19 As shown in Figure 8, the retinoid antagonists of Table 12 could be 2o separated into two classes -by virtue of their effect on the constitutive 21 transcription activation function of the RAR-y-VP-16 chimeric retinoid 22 receptor. One group, which included AGN 193174, AGN 193199 and AGN
23 193840, did not repress RAR-y-VP-16 activity even though they were ATRA
24 antagonists. In contrast AGN 193109, AGN 193385, AGN 193389 and AGN
193871 exhibited a dose dependent repression of RAR-y-VP-16 constitutive 2s activity. Therefore, while the compounds of both groups were RAR-y 27 antagonists, only those of the second group exhibited negative hormone 28 activity. This assay advantageously distinguished retinoid negative hormones 29 from simple retinoid antagonists.

I The foregoing experimental results proved that AGN 193109 met the 2 criteria that define a negative hormone. More specifically, the results 3 presented under Example 11 demonstrated that AGN 193109 had the 4 capacity to exert inhibitory activity at the RARs even in the absence of exogenously added retinoid ligands. As such, this compound possessed 6 biological activities that did not depend upon blockade of the interaction 7 between the RARs and agonists such as ATRA and AGN 191183. These 8 findings led us to conclude that AGN 193109 stabilized interactions between 9 RARs and NCPs. As diagrammed in Figure 9, NCP/RARlPCP interactions lo exist in an equilibrium state. An agonist serves to increase PCP
interactions ii and decrease NCP interactions, while an inverse agonist or negative hormone 12 stabilizes NCP and decreases PCP interactions. As indicated previously, our 13 experimental results suggested that the intracellular availability of NCP
for 14 other receptors can be modulated by AGN 193109 administration. More specifically, we discovered that AGN 193109 can promote complexation of le NCP with RARs, thereby reducing the intracellular reservoir of NCP
17 available for interaction with transcription factors other than the RARs.
t s We next examined the effect of AGN 193109 on agonist-mediated 19 inhibition of AP-1 dependent gene expression. In Endocr. Rev. 14:651 (1993), 2o Pfhal disclosed that retinoid agonists can down-regulate gene expression by a 21 mechanism that involved inhibition of AP-1 activity. We postulated that 22 AGN 193109 could have had either of two effects when used in combination 23 with a retinoid agonist in a model system designed to measure AP-1 activity.
24 First, AGN 193109 could conceivably have antagonized the effect of the agonist, thereby relieving the agonist-dependent inhibition of AP-1 activity.
2s Alternatively, AGN 193109 could have potentiated the agonist's activity, 27 thereby exaggerating the agonist-dependent inhibition of AP-1 activity.
28 Example 13 describes the methods used to demonstrate that AGN
29 193109 potentiated the anti-AP-1 activity of a retinoid agonist. As disclosed 1 below, the AGN 191183 retinoid agonist wealdy inhibited AP-1 dependent 2 gene expression. The combination of AGN i93109 and the retinoid r agonist . . . . . .. . - p ' : . ' - - . . . . 3 strongly inhibited AP-1 dependent gene expression.' By itself, AGN 193109 4 had substantially no anti-AP-1 activity.
Exairiple 13 6 AGN 193109 Potentiates the Anti-AP-1 ActivitY
7 of a Retiffoid A onist 8 HeLa cells were transfected with 1 g of the Str-AP1-CAT reporter 9 gene construct and 0.2 g of plasmid pRS-hRA.Ra, described by Giguere et io al. in Nature 33:624 (1987), usmg LIPOpECTAMINE (Life Technologies, t t Ine.). Str-AP1-CAT was prepared by cloning a DNA fragment corresponding 12 to positions -84 to + 1 of the rat stromelysin-1 promater (Matrisiari et al., 13 MoL .-Cell. BioL 6:1679'.(1986)) between .the HindIII-'BamHI sites of . _ . . . :, . .
14 pBLCAT3 (Luckow et al:, Nud Aculs Res 15:5490 (1987)): > This sequence.9f ys the stromelysin-1 promoter contains an AP1. motif as its sole. enhancer . :.. . .
16 element (Nicholson et a1., EMBO J. 9:4443 (1990)). The pronioter sequence was prepared by annealing two synthetic 'oligonucleotide. s having sequences:
18 5'-' . . . _ : . . i. ' - ' .
i 9 AGAAGCITATGGAAGCAATTATGAGTCAGTTTGCGGGTGACT'CTG

21 GAGGATCCAG-3' , ,.- .
22 (SEQ ID NO:2), and :5'-23 CTG GATCCTCGAGGTCCA:CCTTI'CTGAGCCCAACrITTATAGAGTG

.,~.. .
25 TAAG(."ITCT-3' (SEQ ID N0:3): Procedures involving .transfectiori, 26 treatment with appropr'iate compounds and measurement of CAT activity 27 were carried out as described by Nagpal et al. in J. Biol. Chern. 270:923 28 (1995).
29 The results of these procedures indicated that AGN 193109 1 potentiated the anti-AP-1 activity of the retinoid agonist, AGN 191183.
2 More specifically, in the concentration range of from 10-12 to 10-10 M, AGN
3 191183 did not inhibit the TPA-induced Str-AP1-CAT expression. Treatment 4 with AGN 193109 in the concentration range of from 10-10 to 10-8 M did not substantially inhibit AP-1 mediated reporter activity. However, the results 6 presented in Figure 10 indicated that stimulation of the transfectants with the 7 combination of AGN 193109 (104 M) and AGN 191183 in the concentration 8 range of from 10-12 to 10-10 M substantially inhibited TPA-induced Str-AP1-9 CAT expression by an amount of from 12% to 21%. Therefore, AGN
lo 193109 potentiated the anti-AP-1 activity of AGN 191183 under conditions i i where this retinoid agonist ordinarily did not inhibit AP-1 activity.
12 We reasoned that AGN 193109 potentiated the agonist-mediated 13 repression of AP-1 activity by a mechanism that likely involved AGN 193109-14 dependent sequestration of NCPs onto RARs. RARs belong to a t s superfamily of nuclear receptors that also includes receptors for 1,25-16 dihydroxyvitamin D3, glucocorticoid, thyroid hormone, estrogen and 17 progesterone. It was a reasonable assumption that the ability to bind NCPs 18 may be shared among different members of the nuclear receptor superfamily.
19 This led us to speculate that AGN 193109 could potentiate the anti-AP-1 2o activity of one or more of the ligands that interact with this superfamily of 21 nuclear receptors.
22 The results presented in the preceding Example clearly indicated that 23 AGN 193109 potentiated the anti-AP-1 activity of a retinoid agonist. More 24 specifically, AGN 193109 lowered the threshold dose at which the anti-AP-1 2s activity of AGN 191183 could be detected. Since AGN 193109 has 26 substantially no anti-AP-1 activity by itself, its effect on nuclear receptor 27 agonists was synergistic. We also found that the AGN 193109 negative 28 hormone potentiated the anti-AP-1 activity of 1,25-dihydroxyvitamin D3, the 29 natural ligand for the vitamin D3 receptor.

I The observed synergy between AGN 193109 and AGN 191183 in the 2 preceding Example necessarily implied that the anti-AP-1 activity of the 3 retinoid agonist and the AGN 193109-mediated potentiation of that activity 4 must result from different mechanisms. If the mechanisms of action of the two agents were identical, then it follows that the effectiveness of the 6 combination of AGN 193109 and the agonist would have been additive.
7 Instead, the combination was shown to be more effective than either agent 8 alone, an effect that could not have been predicted in advance of this finding.
9 Significantly, the AGN 193109-mediated potentiation of the RAR
lo agonist was performed using an approximately 100-fold molar excess of AGN
>> 193109 over that of the retinoid agonist. Accordingly, the majority of RARs 12 should have been bound by AGN 193109 leaving very few RARs available for 13 agonist binding. In spite of this fact, the population of RARs that were not 14 bound by AGN 193109 were able to bind retinoid agonist and vigorously stimulate an agonist-dependent response measurable as an inhibition of ie reporter gene expression. Thus, our data suggested possible heterogeneity of 17 RARs that are induced by AGN 193109.
ie The negative hormone activity of AGN 193109, attributed to its ability is to promote the interaction of RARs and NCPs, provided a basis for 2o understanding the synergy between AGN 193109 and retinoid agonists. Our 21 results were fully consistent with a model in which AGN 193109 treatment of 22 cells promoted binding of RARs and NCPs, thereby reducing the number of 23 free NCP and free RAR within the cell. This results in the generation of two 24 populations of RARs that are functionally distinct. The first population is represented by RARs associated with NCPs. Such AGN 193109/RAR/NCP
26 complexes cannot be activated by retinoid agonists. The second population 27 consists of RARs that are not bound by NCP, and that remain available for 28 interaction with agonists. This latter population is designated "RAR*" to 29 indicate free RARs in an environment substantially depleted of NCP.

1 The RAR*s have decreased probabilities of association with NCP
2 through equilibrium binding and have an increased sensitivity to retinoid 3 agonists measurable, for example, as anti-AP-1 activity. This is so because, 4 while the intracellular reservoir of NCP is depleted by virtue of AGN 193109 administration, the reservoir of PCP has not been depleted. Accordingly, 6 free RAR*s can bind a retinoid agonist and interact with PCP factors in an 7 environment substantially depleted of NCP. The ability of AGN 193109 to a increase the sensitivity of other nuclear receptors to their respective agonists 9 can be attributed to the ability of these different nuclear receptors to interact 1 o with the same NCPs that interact with AGN 193109/RAR complexes. This ii model of 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 AGN
16 193109 potentiated the activity of 1,25-dihydroxyvitamin D3 in an in vitro 17 transactivation assay.
18 Example 14 describes the methods used to demonstrate that AGN
19 193109 enhanced the activity of 1,25-dihydroxyvitamin D3 in a transactivation 2o assay.
21 F.xample 14 22 AGN 193109 Potentiates 1.25-Dihydroxyvitamin D3 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 plates and 26 grown in DMEM supplemented with 10% FBS. Cells were cotransfected in 27 serum-free medium using 2 g/well of LIPOFECTAMINE reagent (Life 2s Technologies, Inc.) with 0.7 g of the reporter plasmid MTV-VDRE-Luc, 29 containing two copies of the 1,25-dihydroxyvitamin D3 response element 1 5'-GTACAAGGTTCACGAGGTTCACGTCTTA-3' (SEQ ID NO:4) from 2 the mouse osteopontin gene (Ferrara et al. J. Biol. Chern. 269:2971 (1994)) 3 ligated into the reporter plasmid OMTV-Luc (Heyman et al. in Cell 68:397 4 (1992)), and 0.3 g of the plasmid pGEM3Z (Pharmacia, Inc.) as carrier s DNA to bring the final concentration of DNA to 1.0 g per well. After six e hours of transfection, cells were fed with growth medium containing charcoal 7 extracted FBS at a final concentration of 10%. Eighteen hours after s transfection cells were treated with vehicle alone (ethanol) or AGN 193109 in 9 ethanol at a final concentration of either 104 or 10-7M. Six hours later 1,25-1o dihydroxyvitamin D3 was added in ethanol to a final concentration of from 10=
11 lo to 10-7 M. Cells were lysed and harvested eighteen hours following 1,25-12 dihydroxyvitamin D3 treatment. Luciferase activity was measured as described 13 above. This experimental system allowed a convenient method of monitoring 14 and quantitating 1,25-dihydroxyvitamin D3-dependent gene expression.
15 The results presented in Figure 12 indicated that, when compared with 16 the result obtained using 1,25-dihydroxyvitamin D3 alone, AGN 193109 17 coadministered with 1,25-dihydroxyvitamin D3 shifted the dose response curve 18 to the left. This confirmed that AGN 193109 potentiated the effectiveness of 19 1,25-dihydroxyvitamin D3 in the in vitro transactivation assay. More 2o specifically, Figure 12 graphically illustrates that an AGN 193109 21 concentration as low as 10 - 100 nM rendered the 1,25-dihydroxyvitamin D3 22 approximately 10 fold more active. While a 1,25-dihydroxyvitamin D3 23 concentration of 104 M was required to produce a luciferase expression of 24 approximately 2,000 rlu,=only one-tenth as much 1,25-dihydroxyvitamin D3 25 was required to produce the same luciferase output when the vitamin was 26 coadministered with AGN 193109 at a concentration of 10-8 - 10-7 M.
27 Although not shown on the graph in Figure 12, substantially identical results 28 were obtained using AGN 193109 concentrations of 10-9 M and 10$ M. Thus, 29 coadministration with AGN 193109 substantially reduced the amount of 1,25-1 dihydroxyvitamin D3 that was required to produce a similar effect in the 2 absence of the negative hormone.
3 Interestingly, when the above procedure was repeated with 4 cotransfection of a vitamin D receptor (VDR) expression plasmid, there was a coincident decrease in the ability of AGN 193109 to potentiate the activity 6 of 1,25-dihydroxyvitamin D3. We interpreted this result as indicating that 7 over-expression of VDRs could affect the ability of AGN 193109 to 8 potentiate the activity of 1,25-dihydroxyvitamin D3. Thus, the intracellular 9 concentration of a ligand receptor, which may differ in a tissue-specific 1 o fashion, can influence the ability of AGN 193109 to potentiate the activity of 11 a ligand that binds the receptor. This was again consistent with a model in 12 which titratable NCPs contributed to the regulation of the Vitamin D3 13 response, and supported the model set forth above.
14 As illustrated in the following Example, we also confirmed that AGN
193109 potentiated the anti-AP-1 activity of 1,25-dihydroxyvitamin D3. Our 16 model for the activity of AGN 193109 action explains this observation by 17 invoking that NCPs avidly associate with RARs in the presence of this drug.
18 Endogenous vitamin D receptors present in HeLa cells likely were rendered 19 more sensitive to the 1,25-dihydroxyvitamin D3 ligand, with the consequence 2o of exaggerating the ability of this ligand to inhibit expression from the Str-21 APl-CAT reporter.
22 Example 15 describes the methods used to demonstrate that AGN
23 193109 potentiated the anti-AP-1 activity of 1,25-dihydroxyvitamin D3.
24 Example 15 AGN 193109 Potentiates the Anti-AP-1 Activitx 26 of 1.25-Dihvdxxvitamin D3 27 HeLa cells were transfected with 1 g of Str-AP1-CAT using 28 LIPOFECTAMINE according to the method described by Nagpal et al. in J.
29 BioL Chem. 270:923 (1995). Transfected cells were treated with AGN 193109 1 alone (10-9 to 10-7 M), 1,25-dihydroxyvitamin D3 alone (10-12 to 10-7 M) or 2 1,25-dihydroxyvitamin D3 (10-12 to 10-7 M) in the presence of 10-8 M AGN
3 193109.
4 The results of these procedures indicated that AGN 193109 potentiated the ability of 1,25-dihydroxyvitamin D3 to inhibit TPA-induced 6 AP-1 activity. When used alone in the concentration range of from 10-9 to 7 10-7 M, AGN 193109 had no detectable anti-AP-1 activity. The results 8 presented in Figure 13 indicated that 1,25-dihydroxyvitamin D3 repressed 9 TPA-stimulated activity only in the 104 and 10-1 M concentration range.
1 o Analysis of 1,25-dihydroxyvitamin D3 mediated repression of TPA stimulated 11 CAT activity in the presence of 10-8 M AGN 193109 indicated that anti-AP-1 12 activity was detectable at 10-10 and 10-9 M 1,25-dihydroxyvitamin D3 and an 13 increase in activity at 10-8 and 10-7 M doses compared to 1,25-14 dihydroxyvitamin D3 treatment alone. This AGN 193109 dependent modulation of 1,25-dihydroxyvitamin D3 mediated anti-AP-1 activity was 16 consistent with our model in which NCP sequestration to RARs made the 17 NCP unavailable for interaction with other nuclear receptor family members.
18 Accordingly, the receptors were rendered more sensitive to the 1,25-19 dihydroxyvitamin D3 treatment.
The mechanisms underlying RAR mediated transactivation and anti-21 AP-1 activity are likely different. This conclusion was based on our 22 observation that high doses of AGN 193109 completely inhibited 23 transactivation without substantially inhibiting anti-APl activity. We 24 therefore wished to gain additional evidence to support our model for RAR*
formation mediated by AGN 193109 treatment. To accomplish this, we 26 investigated whether AGN 193109 could potentiate the activity of the RAR
27 specific agonist AGN 191183 in an in vitro transactivation assay.
28 Example 16 describes the methods used to demonstrate that AGN
29 193109 potentiated the activity of the RAR specific agonist, AGN 191183.

1 The results of this procedure indicated that, under particular circumstances, 2 AGN 193109 enhanced the potency of the RAR specific retinoid, and 3 provided strong evidence that AGN 193109 promoted RAR* formation.
4 Example 16 Potentiation of Retinoid Effectiveness by 6 AGN 193109 Coadministration 7 Hela cells were transfected using the cationic liposome-mediated a transfection procedure described by Felgner et al. in Proc. NatL Acad Sci.
9 USA 84:7413 (1987). 5 X 104 cells were plated in 12 well multiwell plates and 1o grown in DMEM supplemented with 10% FBS. Cells were cotransfected in 11 serum free medium using LIPOFECTAMINE reagent (2 ug/well, Life 12 Technologies, Inc.) with 0.7 g of the reporter plasmid 1ViTV-TREp-Luc, 13 containing two copies of the TREpal response element 14 5'-TCAGGTCATGACCTGA-3' (SEQ ID NO:5) inserted into the reporter plasmid AMTV-Luc (Heyman et al. in Cell 68:397 (1992)), and 0.1 g of the 16 RAR-y expression plasmid pRShRAR-y (Ishikawa et al. MoL Endocrinol.
17 4:837 (1990)). After six hours of transfection, cells were fed with growth 18 medium containing charcoal extracted FBS at a final concentration of 10%.
19 Eighteen hours after transfection, cells were treated with vehicle alone (ethanol) or AGN 193109 in ethanol at a final concentration of from 10-11 to 21 10 M. Six hours later, AGN 191183 was added in ethanol to a final 22 concentration of either 0, 10-10 or 10-9 M. Cells were harvested after eighteen 23 hours of AGN 191183 treatment and luciferase activity was measured as 24 described above.
Preliminary experiments indicated that 10-9 M AGN 193109 was 26 relatively ineffective at inhibiting the response to of 10-9 M AGN 191183 in 27 HeLa cells. This contrasted with the ability of 10-9 M AGN 193109 to inhibit 28 10$ M ATRA in CV-1 cells (Figure 2).
29 The results presented in Figure 14 supported the prediction that AGN

1 193109 stimulated the formation of RAR*. Consistent with our 2 characterization of the antagonist and negative hormone activities of AGN
3 193109, treatment with AGN 193109 resulted in a biphasic dose response 4 curve. The lowest doses of AGN 193109 (10-11 and 10-10 M) resulted in a stimulation of luciferase activity over that of AGN 191183 alone. This effect s suggests that RAR*s are generated by AGN 193109. Curiously, this was also 7 seen for AGN 193109 treatment alone, suggesting that RAR*'s can respond 8 to an endogenous ligand. AGN 191183 is a synthetic retinoid agonist and, 9 like ATRA, activates transcription through the RARs. Substitution of AGN
io 191183 for ATRA in Example 7 would give qualitatively similar results (i.e., ii AGN 193109 would antagonize the effect of 10 nM AGN 191183). Example 12 16 illustrates that, while AGN 193109 can function as an antagonist of RAR
13 agonists, dosing conditions could easily be identified wherein AGN 193109 14 coadministration potentiated activation mediated by an RAR agonist. It is important to note that the doses of the compounds used in Example 16 are 16 substantially lower than the doses employed in the procedure described under 17 Example 7. We proposed that AGN 193109 treatment could lead to RAR
18 heterogeneity RARs versus RAR*s. The apparent heterogeneity (i.e., ability 19 to potentiate) appears to have different windows in transactivation versus 2o AP-1 repression. The reason that the curves are biphasic is because, with 21 increasing amounts of AGN 193109, there is proportionately less RAR
22 available to bind the agonist. This doesn't appear to be the case for AP-1 23 repression and we are left to speculate that this difference must reflect two 24 distinct mechanisms for transactivation and AP-1 repression by the same receptor species.
26 Clinical results have confirmed that some retinoids are useful for 27 inhibiting the growth of premalignant and malignant cervical lesions.
28 Exemplary studies supporting this conclusion have been published by Graham 29 et al. in West. J. Med 145: 192 (1986), by Lippman et al. in J. Natl.
Cancer 1 Inst. 84:241 (1992), and by Weiner et al. in Invest. New Drugs 4:241 (1986)).
2 Similar conclusions are supported by the results of in vitro studies that 3 used cultured cells to quantitate the antiproliferative effects of various 4 retinoids. More specifically, Agarwal et al. in Cancer Res. 51:3982 (1991) employed the ECE16-1 cell line to model the early stages of cervical 6 dysplasia and demonstrated that retinoic acid could inhibit epidermal growth 7 factor (EGF) dependent cellular proliferation.
8 Example 17 describes the methods used to demonstrate that AGN
9 193109 can antagonize the activity of the AGN 191183 retinoid agonist which 1o inhibited proliferation of the ECE16-1 cell line.

11 Example 17 12 AGN 193109 Antagonizes the Antiproliferative Effect 13 of Retinoids in ECE16-1 Cells 14 ECE16-1 cells were seeded at a density of 1 x 104 cells per cm2 in complete medium containing DMEM:F12 (3:1), nonessential amino acids, 5%
16 FBS, 5 g/ml transferrin, 2 nM of 3,3,5 triiodothyronine (thyroid hormone or 17 'T3'), 0.1 nM cholera toxin, 2 mM L-glutamine, 1.8 x 104 M adenine and 10 18 ng/ml EGF. Cells were allowed to attach to plates overnight and then shifted 19 to defined medium containing DMEM:F12 (3:1), 2 mM L-glutamine, 2o nonessential amino acids, 0.1% bovine serum albumin, 1.8 x 10-4 M adenine, 21 5 g/ml transferrin, 2 nM T3, 50 g/ml ascorbic acid, 100 ug/mi streptomycin, 22 100 units/ml penicillin and 50 g/ml gentamicin. Defined medium (DM) was 23 supplemented with 10 ng/ml EGF. EGF treated cells received 10 nM of the 24 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. After three days 26 of treatment, cells were harvested as described by Hembree et al. in Cancer 27 Res. 54:3160 (1994) and cell numbers determined using a COULTER

28 counter.
29 The results presented in Figure 15 demonstrated that ECE16-1 cells 1 proliferated in response to EGF but not in defined medium alone. This 2 confirmed the findings published by Andreatta-van Leyen et al. in J. Cell.
3 Physio. 160:265 (1994), and by Hembree et al. in Cancer Res. 54:3160 (1994).
4 Addition of 10 nM AGN 191183 and 0 nM AGN 193109 completely inhibited EGF mediated proliferation. Thus, AGN 191183 was a potent s antiproliferative retinoid. Increasing the AGN 193109 concentration from 0 7 nM to 10 nM antagonized the AGN 191183 mediated growth inhibition by 8 approxiinately 50%. A ten-fold molar excess of AGN 193109 completely 9 reversed the antiproliferative effect of AGN 191183. Treatment of cells with i o 1000 nM AGN 193109 alone had no effect on the EGF mediated t i proliferation increase. These results proved that AGN 193109 antagonized 12 the antiproliferative effect of a retinoid but had substantially no 13 antiproliferative activity of its own when used to treat cells representing 14 cervical epithelium that is sensitive to growth inhibition by retinoids such as AGN 191183. Notably, there was no evidence that AGN 193109 potentiated ie the antiproliferative activity of the AGN 191183 agonist using the ECE16-1 17 model system.
ts In contrast to the model system represented by the ECE16-1 cell line, 19 there are other examples where cellular proliferation associated with cervical 2o dysplasia cannot be inhibited by retinoid agonists. For example, Agarwal et 21' al. in Cancer Res. 54:2108 (1994) described the use of CaSki cells as a model 22 for cervical tumors that are unresponsive to retinoid therapy. As disclosed 23 below, rather than inhibiting cell proliferation, retinoid treatment had 24 substantially no effect on the growth rate of CaSki cells. The following Example addressed the effect of the AGN 193109 negative hormone on the 26 proliferation rates of this cell line. The results unexpectedly proved that 27 AGN 193109 can inhibit the proliferation of cervical tumor cells that are 28 unresponsive to the antiproliferative effects of retinoid agonists.
29 Example 18 describes the methods used to demonstrate that AGN

1 193109 inhibited the growth of a cervical tumor cell line that did not respond 2 to the antiproliferative effects of other retinoids such as AGN 191183.
3 Significantly, AGN 193109 displayed antiproliferative activity in the absence 4 of added retinoid Example 18 6 AGN 193109 Inhibits the Proliferation Rate of CaSki 7 Cervical Carcinoma-Derived Cell Line 8 We tested the effect of EGF on CaSki cell proliferation, either alone 9 or in combination with the AGN 191183 retinoid agonist and/or the AGN
lo 193109 negative hormone at a concentration of 1& M. Cell proliferation ii assays were performed as described above for studies involving ECE16-1 12 cells. EGF was added to the retinoid treated cultures to give a final 13 concentration of 20 ng/ml. Cells were treated with AGN 191183 (10=10 to 10-14 M) in the presence or absence of 10-6 M AGN 193109 for a total of three days. The media was replaced with fresh media and each of the two retinoid ie compounds, as appropriate, every day. Cell numbers were determined using 17 a COULTER counter as described above.
18 The results presented in Figure 16 indicated that CaSki cells were is substantially refractory to the effects of a retinoid agonist and that AGN
2o 193109 exhibited antiproliferative activity in the absence of added retinoid.
21 The presence of EGF in the culture media stimulated CaSki cell growth.
22 This conclusion was based on comparison of the stripped bar representing no 23 AGN 191183 and the open bar representing defined growth media ("DM") 24 alone. AGN 191183 treatment had no antiproliferative activity on the CaSki tumor cell line. We discounted any slight increase in the cellular 26 proliferation rate associated with the retinoid agonist, because a ten thousand 27 fold increase in the retinoid agonist concentration was associated with only 28 roughly a 20% increase in the proliferation rate. Thus, the AGN 191183 29 agonist had substantially no effect on the proliferation rate of CaSki cells.

1 The results presented in Figure 16 also indicated that AGN 193109 2 inhibited proliferation of the CaSki cervical epithelial cell line. This 3 conclusion was based on comparison of the measurements appearing as the 4"0" AGN 191183 black bar and the "0" AGN 191183 stripped bar. Thus, AGN 193109 was capable of stimulating a biological response in the absence 6 of added retinoid agonist when used to treat cervical tumor cells that were 7 not growth inhibited by retinoid agonists such as AGN 191183.
8 Our discovery that the AGN 193109 negative hormone could inhibit 9 cellular proliferation was consistent with a model in which unliganded RAR
1o mediated the expression of genes that were required for proliferation.
While 11 an RAR agonist such as AGN 191183 had substantially no effect, or perhaps 12 promoted cellular proliferation slightly, AGN 193109 had an antiproliferative 13 effect. The AGN 193109 negative hormone likely bound RARs thereby 14 promoting NCP association and causing the RARs to adopt an inactive 1 s conformation. According to our model, this repressed gene activity that was 1 e positively regulated by unliganded RARs. This ability of AGN 193109 to 17 down-regulate the activity of unliganded RARs likely resulted from its ability 18 to promote the association of RARs and NCPs:
19 Those having ordinary skill in the art will appreciate that some retinoid 2o agonists are useful for controlling the undesirable consequences of cell 21 growth that follows retinal detachment. After retinal detachment the retinal 22 pigment epithelium (RPE) dedifferentiates, proliferates and migrates into the 23 subretinal space. This process can negatively impact the success of surgical 24 procedures aimed at retinal reattachment. Campochiaro et al. in Invest.
25 Opthal & V. Scf. 32:65 (1991) have demonstrated that RAR agonists such as 2s ATRA exhibited an antiproliferative effect on the growth of primary human 27 RPE cultures. Retinoid agonists have also been shown to decrease the 28 incidence of retinal detachment following retinal reattachment surgery 29 (Fekrat et al. Opthamology 102:412 (1994)). As disclosed in the following 1 Example, we analyzed the ability of the AGN 193109 negative hormone to 2 suppress growth in primary human RPE cultures.
3 Example 19 describes the methods used to demonstrate that AGN
4 193109 potentiated the antiproliferative effect of a retinoid antagonist in a primary culture of human retinal pigment epithelium.
6 Example 19 7 AGN 193109 Potentiates the Antiproliferative Activity of ATRA
a Primary cultures of human retinal pigment epithelium (RPE) were 9 established according to the method described by Campochiaro et al. in 1o Invest. Opthal & Vis. Sci. 32:65 (1991). 5 x 104 cells were plated in 16-mm 11 wells of 24-well multiwell plates in DMEM (Gibco) containing 5% FBS.
12 Cells were mock treated with ethanol vehicle alone, ATRA (10-10 to 10-6 M) 13 in ethanol, AGN 193109 (10-10 to 10-6 M) in ethanol, or ATRA (10-10 to 10-6 14 M) and 10-6 M AGN 193109. Cells were fed with fresh media containing the appropriate concentrations of these compounds every two days for a total of 16 five days of treatment. Cells were removed from the plates by gentle 17 digestion with trypsin and the number of cells was counted with an electronic 18 cell counter.
19 The results presented in Figure 17 indicated that AGN 193109 2o dramatically potentiated the antiproliferative activity of ATRA on RPE
cells.
21 Treatment of primary RPE cells with ATRA led to a dose dependent 22 decrease in RPE cell proliferation with an approximately 40% decrease at 10-23 6 M ATRA relative to control cultures. AGN 193109 treatment did not 24 substantially alter the growth rate of the RPE cells at any concentration tested in the procedure. Unexpectedly, the combination of ATRA (10-11 to 26 10-6 M) and 10-6M AGN 193109 had a stronger antiproliferative activity than 27 ATRA alone. Thus, AGN 193109 cotreatment potentiated the 28 antiproliferative effect of ATRA. More specifically, the results shown in the 29 Figure indicated that the antiproliferative effect of 10-8 M ATRA was 1 obtainable using only 10-10 M ATRA in combination with 10-' M AGN
2 193109. Thus, the AGN 193109 negative hormone advantageously enhanced 3 the antiproliferative activity of ATRA by approximately 100 fold.
4 In an independent experiment, comparisoln of the antiproliferative effect of ATRA (10-11 to 10-6 M) with that of ATRA and 10-6 M AGN 193109 s again demonstrated the apparent increase in sensitivity of primary RPE cells 7 to ATRA in the presence of AGN 193109. In this system, AGN 193109 8 neither functioned as a retinoid antagonist nor exhibited an antiproliferative 9 effect when used alone. However, AGN 193109 coadministration potentiated 1 o the antiproliferative activity of the retinoid agonist.
11 AGN 193109 was tested for its ability to potentiate the anti-12 proliferative effect of 13-cis retinoic acid (13-cis RA) in primary RPE
cultures 13 using conditions and techniques to measure RPE cell proliferation described 14 above. Notably, 13-cis RA is clinically significant. More particularly, 13-cis RA is useful in the treatment of several disease states, including acne (Peck te et al. N. EngL J. Med 300:329 (1977); Jones et al. Br. J. DermatoL 108:333 17 (1980)), and squamous cell carcinoma of the skin and cervix in combination 1 s treatment with interferon 2a (Lippman et al. J. NatL Cancer Inst. 84:241 19 (1992); Moore et al. Seminars in Hematology 31:31 (1994)).
The results presented in Figure 18 indicated that both 13-cis RA (10,12 21 to 10-6M) and ATRA (10-12 to 10-6M) effectively inhibited RPE cell growth.
22 Notably, the 13-cis isomer was approximately two orders of magnitude less 23 effective in this assay when compared with ATRA. Similar to the results 24 obtained using coadministration of AGN 193109 and ATRA (above), coadministration of AGN 193109 (either 104 or 10-6M) with 13-cis RA (10-12 26 to 106M) dramatically increased the potency of 13-cis RA in mediating 27 repression of RPE cell proliferation. In contrast to treatment with 13-cis RA
28 alone, coadministration of AGN 193109 enhanced the potency of 13-cis RA.
29 Thus, AGN 193109 potentiated the antiproliferative activity of 13-cis RA.

1 We next tested the ability of AGN 193109 to potentiate the activities 2 of other nuclear receptor hormones in primary RPE cell cultures.
3 Dexamethasone, a synthetic glucocorticoid receptor agonist, is one member 4 of a class of compounds that have been used clinically for their potent anti-inflammatory and immunosuppressive properties. Thyroid hormone (T3;
6 3,3',5'-Triiodothyronine) is a natural thyroid hormone receptor agonist used 7 primarily for hormone replacement therapy in the treatment of s hypothyroidism. Methods used in these experiments were identical to those s described above for procedures employing ATRA and 13-cis RA.
The results of these procedures indicated that coadministration of 11 AGN 193109 and the nuclear receptor agonists potentiated the 12 antiproliferative activities of the nuclear receptor agonists. More specifically, 13 the results presented in Figure 19 showed that single-agent treatment of RPE
14 cells with either dexamethasone (10=11 to 10-6M) or ATRA (10-12 to 10-6M) was substantially unable to inhibit RPE cell proliferation. However, le treatment of RPE cells with dexamethasone (10-11 to 10-6M) and either 10-8 or 17 10-6M AGN 193109 repressed RPE cell proliferation to an extent that 18 approximated the inhibition caused by treatment with ATRA. Similarly, the 19 results presented in Figure 20 indicated that AGN 193109 potentiated the 2o antiproliferative activity of thyroid hormone. Similar to the results obtained 21 using dexamethasone, the proliferation of RPE cells was refractory to single-22 agent treatment with thyroid hormone (10-11 to 10-6M). However, co-23 treatment of RPE cells with thyroid hormone (10-11 to 10-6M) and AGN
24 193109 (either 10-8 or 10-6M) inhibited RPE cell proliferation in a thyroid hormone dependent manner. We concluded that AGN 193109 rendered 26 primary RPE cultures sensitive to the anti-proliferative effects of these 27 nuclear receptor agonists. The mechanism by which AGN 193109 mediated 28 these effects likely involved modulation of NCP/RAR interactions.
29 We additionally examined the effect of AGN 193109 on the expression 1 of marker genes in other experimental systems that were sensitive to retinoid 2 agonists. Both the MRP8 and stromelysin genes are known to be inhibited 3 by retinoid agonists in a variety of biological systems. For example, 4 Wilkinson et al. in J. Cell Sci. 91:221 (1988) and Madsen et al. in J.
Invest.
DermatoL 99:299 (1992) have disclosed that MRP8 gene expression was 6 elevated in psoriasis. Conversely, MRP8 gene expression was repressed by 7 the retinoid agonist AGN 190168 in human psoriatic skin (Nagpal et al., 8 submitted 1995), in human keratinocyte raft cultures (Chandraratna et al. J.
9 Invest. Dermatol. 102:625 (1994)) and in cultured human newborn foreskin 1o keratinocytes (Thacher et al. J. Invest. Dennatol 104:594 (1995)). Nagpal et 11 al. in J. Biol. Chem. 270:923 (1995) have disclosed that stromelysin mRNA
12 levels were repressed by retinoid agonists such as AGN 190168 in cultured 13 human newborn foreskin keratinocytes. We analyzed the regulated 14 expression of these genes following treatment of cultured human newborn foreskin keratinocytes with either the AGN 191183 retinoid agonist or AGN
1e 193109.
17 Example 20 describes the methods used to demonstrate that AGN
18 193109 inhibited MRP-8 expression in cultured keratinocytes.
19 Example 20 AGN193109 Inhibits MRP-8 Exvression 21 in Keratino es 22 Primary foreskin keratinocytes were isolated according to the 23 procedure described by Nagpal et al. in J. Biol. Chem. 270:923 (1995) and 24 cultured in keratinocyte growth medium (KGM) that was purchased from Clonetics. After 3 days of treatment with AGN 191183 (10-7 M) or AGN
26 193109 (10-6 M), total cellular RNA was isolated from treated and control 27 keratinocytes according to standard methods. The mRNA was reverse 28 transcribed into cDNA which then served as the template in a PCR
29 amplification protocol using primers specific for either the glyceraldehyde I phosphate dehydrogenase (GAPDH) housekeeping gene or MRP-8. The 2 GAPDH primers had the sequences 5'-3 CCACCCATGGCAAATTCCATGGCA-3' (SEQ ID NO:6) and 5'-4 TCTAGACGGCAGGTCAGGTCCACC-3' (SEQ ID NO:7). The MRP-8 primers had the sequences 5'-ACGCGTCCGGAAGACCTGGT-3' (SEQ ID
6 NO:8) and 5'-ATTCTGCAGGTACATGTCCA-3' (SEQ ID NO:9). An 7 aliquot from the MRP-8 amplification reaction (10 l) was removed after 8 every cycle of PCR amplification starting from 12 cycles and ending at 21 9 cycles. Similarly, an aliquot of the GAPDH amplification reaction was 1 o removed after every PCR cycle starting at 15 cycles and ending at 24 cycles.
ii The samples were electrophoresed on 2% agarose gels and the separated 12 amplification products detected by ethidium bromide staining. The staining 13 intensity of the amplification products served as a quantitative measure of the 14 amount of starting mRNA specific for the given primer set.
The results of this procedure indicated that both AGN 191183 and 16 AGN 193109 independently inhibited MRP-8 expression in keratinocytes.
17 The intensity of the stained GAPDH amplification product was substantially 18 equivalent in the lanes of the gel representing starting material isolated from 1 s control, AGN 191183, and AGN 193109 treated keratinocytes. Weak bands 2o representing the GAPDH amplification product were first detectable in lanes 21 corresponding to samples removed after 18 cycles of PCR amplification. The 22 equivalent staining intensities among the various lanes of the gel indicated 23 that equivalent masses of starting material were used for all samples.
24 Accordingly, differences in the intensities of stained bands representing MRP-8 amplification products were indicative of differences in MRP-8 26 mRNA expression among the various starting samples. As expected, the 27 MRP-8 amplified signal was inhibited in AGN 191183 (10-7 M) treated 28 cultures relative to an untreated control. AGN 193109 (10-6 M) treatment of 29 cultured keratinocytes also repressed MRP8 expression as judged by lower I intensity of stained amplificatioa product.
2 As illustrated in the following Example, AGN 193109 also inhibited 3 expression of a second marker gene' in keratinocytes. Nagpal et al. in J.
Biol. 4 Chem: 270:923 (1995) disclosed that. stromelysin mRNA expression was down-regulated by RAR specific agonists in cultured newborn human 6 foreskin keratinocytes. Nicholson et al. (EMBO J. 9:4443 (1990)) disclosed 7 that an AP-1 promoter element played a role in . the retinoid-dependent 8 negative regulation of the stromelysin-l gene. Thus, it was of interest to 9 determine whether AGN 193109 could alter the expression of this gene.
Exaxnple 21 describes the methods used to demonstrate that AGN
11 193109 inhibited stron.zelysin-1 gene expression in.the absence of'an 12 exogenously added retinoid agonist. :
1s Example 21 14 AGN 193109 Inhibits Strom~elvsin=1 Expression in CZl~tured Keratinocvtes 16 Primary foreskin keratinoeytes were either mock treated or treated for -17 24 hours with the RAR agonist AGN.191183 (10-7 M), or AGN 193109 (10-6 18 M. Total RNA prepared from mock=treated 'and retinoid-tTeated 19 keratinocytes was teverse transcribed and the resulting cbNA was PCR
:
2o amplified using %3-actin_ or stromelysin-1 oligo primers exactly as described by hTagpal et al: in J. Biol. Chem. 270:923 (1995).
.22 A sample (10 111) from the PCR
23 amplification reaction was removed after every three cycles starting from 24 cycles of PCR amplification. 'The "sample "was electrophoresed on a 2%
z~ agarose gel and detected after ethidium bromide` staining."
: zs Results of these procedures indicated that AGN 193109 inhibited 27 stromelysin-l gene expression in the absence of an exogenously added 26 .retinoid 'agonist. More specihcally, ethidium-stained bands representing (3- "
2s . actin amplification products were easily detectable the agarose gels after 18 i cycles of PCR. While all band intensities increased witli additional cycles of 2 the amplification reaction, stained bands were somewhat less intense in 3 samples representing AGN 191183 treated cells. This indicated that a slightly 4 lesser amount of RNA must have been present in the starting samples corresponding to cells treated with AGN 191183. The results also indicated 6 that stromelysin-1 mRNA was detectable in mock-treated keratinocytes 7 starting at 33 cycles of PCR amplification. As expected, stromelysin-1 8 mRNA expression was inhibited after AGN 191183 (10-7 M) treatment as 9 judged by the weaker band intensity on when compared with samples derived 1 o from mock-treated samples. When normalized to the intensities of the ~-i1 actin amplification products, and consistent with the results obtained in 12 measurements of MRP-8 expression, AGN 193109 (10-6 M) treatment of 13 keratinocytes resulted in down-regulation of stromelysin-1 mRNA levels.
14 Indeed, the down-regulation stimulated by AGN 193109 treatment was indistinguishable from the down-regulation caused by treatment of 16 keratinocytes with the RAR agonist AGN 191183.
17 As disclosed herein, AGN 193109 can have any of three possible 18 effects with respect to modulating the activity of a coadministered steroid 1 s superfamily agonist. First, AGN 193109 may have no effect. Second, AGN
2o 193109 may antagonize the effect of the agonist, thereby leading to a 21 decrease in the activity of the agonist. Finally, AGN 193109 may potentiate 22 the activity of the agonist, thereby leading to a stimulation of the measured 23 effect produced by the agonist.
24 Compounds having activities that can be modulated by AGN 193109 include retinoid receptor agonists and agonists which bind to other members 26 of the steroid receptor superfamily. This latter category of agonists includes 27 vitamin D receptor agonists, glucocorticoid receptor agonists and thyroid 2s hormone receptor agonists. Peroxisome proliferator-activated receptors, 29 estrogen receptor and orphan receptors having presently unknown ligands 1 may also be potentiated by AGN 193109. In the case where the steroid 2 superfamily agonist is an RAR agonist, AGN 193109 may either antagonize 3 or potentiate the activity of that agonist. In the case where the agonist used 4 in combination with AGN 193109 is a compound that can bind to a nuclear receptor other than an RAR, coadministration of AGN 193109 will either 6 have no effect or will sensitize of the system to the agonist so that the activity 7 of the agonist is potentiated.
8 A generalized exemplary procedure for determining which of the three 9 possible activities AGN 193109 will have in a particular system follows.
This lo description illustrates each of the possible outcomes for AGN 193109 ii coadministration with a steroid receptor superfamily agonist. Biological 12 systems useful for assessing the ability of AGN 193109 to modulate the 13 activity of a nuclear receptor agonist include but are not limited to:
14 established tissue culture cell lines, virally transformed cell lines, ex-vivo primary culture cells and in vivo studies utilizing living organisms.
16 Measurement of the biological effect of AGN 193109 in such systems could 17 include determination of any of a variety of biological endpoints. These 18 endpoints include: analysis of cellular proliferation, analysis of programmed is cell death (apoptosis), analysis of the differentiation state of cells via gene 2o expression assays, analysis of the ability of cells to form tumors in nude mice 21 and analysis of gene expression after transient or stable introduction of 22 reporter gene constructs.
23 For illustrative purposes, an mRNA species designated as mRNA "X"
24 is expressed from gene "X" in primary cultured "Y" cells isolated from the organ "Z." Under standard culture conditions, where several "Y' cell genetic 26 markers are maintained, including expression of gene "X", addition of a 27 retinoid agonist leads to a decrease in the abundance of "X" mRNA.
28 Analysis of gene X expression can be assessed via isolation of cellular mRNA
29 and measurement of the abundance of X mRNA levels via polymerase chain 1 reaction, ribonuclease protection or RNA blotting procedures such as 2 Northern analyses. After isolation from organ Z, primary Y cells are 3 cultured in an appropriate growth medium. The primary cultures are then 4 plated into tissue culture plates for expansion of the cell population. This s step facilitates separation of the cells into four sample groups so that various s doses of the retinoid agonist and AGN 193109 can be delivered. The first 7 group will be a control, receiving vehicle only. The second group will receive 8 the RAR agonist, retinoic acid, delivered in ethanol, in amounts sufficient to 9 provide final concentrations in the range of from 10-11 to 10-6 M. The lowest 1o dose may need to be empirically determined depending on the sensitivity of 11 the system. Such determinations fall within the scope of routine 12 experimentation for one having ordinary skill in the art. The third group will 13 receive both the nuclear receptor agonist at the same doses used for treating 14 the cells of group 2, and a constant dose of AGN 193109. The dose of AGN
15 193109 used for treating the cells of group 3 will also need to be determined 1s empirically, but should approximate the affinity constant (Kd) of AGN
17 193109 for the RAR subtypes (i.e., at least 10-8 M). The fourth group will 1 s receive AGN 193109 at doses minimally including that used for agonist 1 s coadministration in group 3. An alternative to this dosing regimen would 20 substitute AGN 193109 for the retinoid agonist described in the foregoing 21 example, as specified in group 2, and a constant dose of retinoid agonist in 22 place of AGN 193109, as specified in groups 3 and 4. After a suitable 23 incubation period, cells should be harvested in a manner suitable for 24 determination of the biological endpoint being measured as an indicator of 2s agonist activity.
26 For example, analysis of the effect of AGN 193109 on retinoic acid 27 dependent regulation of gene expression would involve comparison of the 28 abundance of the mRNA species X in the mRNA pool harvested from cells 29 treated according to each of the four protocols described above. RNA

1 derived from control cells will serve to determine the baseline expression of 2 X mRNA and will represent a condition corresponding to no repression.
3 Comparison of this level with that measured in the mRNA pool derived from 4 cells treated with retinoic acid will allow for determination of the effect of this agonist on gene expression. Quantitated levels of the repression of 6 specific mRNAs resulting from retinoic acid treatment can then be compared 7 with mRNA abundances from cells treated in parallel with either AGN
8 193109 alone or AGN 193109 in combination with retinoic acid. While this 9 generalized example illustrates an analysis of the effect of coadministered lo AGN 193109 on the expression of a gene repressed by a retinoid agonist, the i t example could alternatively have described analysis of the effect of 12 coadministered AGN 193109 on a gene that was induced by a retinoid 13 agonist. The critical feature for determining whether AGN 193109 will 14 behave as an agonist, as a negative hormone or have no effect in a particular system will involve quantitative comparison of the magnitude of the effect in 16 the presence and absence of AGN 193109.
17 An example in which AGN 193109 potentiated the activity of a 18 coadministered agonist would be a case in which AGN 193109 cotreatment 19 with retinoic acid resulted in a level of X mRNA expression that is further 2o repressed relative to the level measured in cells treated with retinoic acid 21 alone. More specifically, comparison of the dose response curve of the 22 biological effect (i.e., repression of X mRNA abundance) plotted on the Y-23 axis versus the dose of the agonist (logarithmic scale) on the X-axis would 24 allow comparison of agonist-mediated repression of X mRNA abundance in the presence and absence of AGN 193109 cotreatment. The ability of AGN
26 193109 to sensitize the biological response to the agonist, thereby potentiating 27 the activity of the agonist, will be indicated by a leftward shift in the dose 28 response curve. More specifically, in the presence of AGN 193109 less 29 agonist would be required to obtain the same biological effect obtainable 1 using the agonist alone.
2 An example of AGN 193109 mediating antagonism of a coadministered 3 agonist would be a case in which AGN 193109 cotreatment with retinoic acid 4 resulted in a level of X mRNA expression that is less repressed compared to that measured in cells treated with retinoic acid alone. Comparison of dose e response curves of X mRNA repression versus log dose of agonist in the 7 presence and absence of AGN 193109 will demonstrate a shift to the right in 8 the dose response curve. More specifically, in the presence of AGN 193109, 9 more agonist will be necessary to obtain the same biological effect obtainable 1 o with single agent treatment with the agonist alone.
11 The above examples wherein AGN 193109 mediates either antagonism 12 or potentiation describe experimental outcomes for coadministration of AGN
13 193109 with a retinoid agonist. If, however, the agonist coadministered with 14 AGN 193109 is an agonist capable of binding and activating a member of the steroid receptor superfamily other than an RAR, then instead of antagonizing t e the agonist, it becomes possible that AGN 193109 would have no effect on 17 the activity of the agonist. If AGN 193109 cotreatment with such an agonist 1s results in a level of mRNA expression which is equal to that measured in is cells treated with agonist alone, then AGN 193109's ability to affect the 2o availability of NCPs via promotion of RAR:NCP associations will be silent in 21 this system. This would be an example wherein AGN 193109 has no effect 22 on a coadministered agonist.
23 Example of Antagonism 24 The method disclosed in the above generalized example for determining the effect of AGN 193109 coadministered with a retinoid agonist ' 26 is exemplified by the procedure described under Example T CV-1 cells 27 cotransfected with one of the three retinoic acid receptors and the retinoid 28 agonist inducible MTV-TREp-Luc reporter construct were dosed with either 29 ethanol (control, group 1), AGN 193109 at final concentrations of from 10-9 . ' ~

DEMANDES OU BREVETS VOLUMINEUX

LA PRESENTE PARTIE DE CBTTE DEMANDE OU CE BREVET
COMPREND PLUS D'UN TOME.

CECI EST LE TOME _~__~DE

NOTE: Pour les tomes additionels, veuiliez contacter le Bureau canadien des brevets - ~.

JUMBO APPLICATIONS/PATENTS

THIS SECTION OF THE APPLICATION/PATENT CONTAINS MORE
THAN ONE VOLUME

THIS IS VOLUME `-OF
~., ~

NOTE: For additional voiumes -ptease contac2'the Canadian Patent Office .:. , _ _ . . . ._ , . . . - .

Claims (46)

WHAT IS CLAIMED IS:
1. A compound of the formula where X1 is S or O;
X2 is CH or N;
R2 is H, F, CF3 or alkoxy of 1 to 6 carbons;
R2* H, F, or CF3;
R8 is H, or lower alkyl of 1 to 6 carbons;
R14 is unsubstituted phenyl, thienyl or pyridyl, or phenyl, thienyl or pyridyl substituted with one to three R15 groups, where R15 is lower alkyl of to 6 carbons, chlorine, CF3, or alkoxy of 1 to 6 carbons, or a pharmaceutically acceptable salt of said compound.
2. A compound in accordance with Claim 1 where X1 is S.
3. A compound in accordance with Claim 2 where X2 is CH.
4. A compound in accordance with Claim 2 where R14 is phenyl, 2-thienyl, or 3-pyridyl unsubstituted or substituted with one lower alkyl group having 1 to 6 carbons.
5. A compound in accordance with Claim 2 where R2 is H or F.
6. A compound in accordance with Claim 2 where R2* is H or F.
7. A compound in accordance with Claim 2 where X2 is N.
8. A compound in accordance with Claim 1 where X1 is O.
9. A compound in accordance with Claim 8 where X2 is CH.
10. A compound in accordance with Claim 8 where R14 is phenyl, 2-thienyl, or 3-pyridyl unsubstituted or substituted with one lower alkyl group having 1 to 6 carbons.
11. A compound in accordance with Claim 8 where R2 is H or F.
12. A compound in accordance with Claim 8 where R2 * is H or F.
13. A compound of the formula wherein X2 is CH or N;
R2 is H, F or OCH3;
R2* is H or F;
R8 is H, or lower alkyl of 1 to 6 carbons, and R14 is selected from the group consisting of phenyl, 4-(lower-alkyl)phenyl, 5-(lower alkyl)-2-thienyl, and 6-(lower alkyl)-3-pyridyl where lower alkyl has 1 to 6 carbons, or a pharmaceutically acceptable salt of said compound.
14. A compound in accordance with Claim 13 where X2 is CH.
15. A compound in accordance with Claim 14 where R2 is H.
16. A compound in accordance with Claim 15 where R2* is H.
17. A compound in accordance with Claim 16 where the R14 group is selected from phenyl, 4-methylphenyl, 4-ethylphenyl, 4-i-propylphenyl, 4-t-butylphenyl, 5-methyl-2-thienyl, 5-ethyl-2-thienyl, 5-1-butyl-2-thienyl and 6-methyl-3-pyridyl.
18. A compound in accordance with Claim 17 where R8 is ethyl or H, or a pharmaceutically acceptable salt of said compound.
19. A compound in accordance with Claim 15 where R2* is F.
20. A compound in accordance with Claim 19 where the R14 group is selected from 4-methylphenyl and 4-ethylphenyl.
21. A compound in accordance with Claim 20 where R8 is ethyl or H, or a pharmaceutically acceptable salt of said compound.
22. A compound in accordance with Claim 14 where R2 is F and R2*
is H.
23. A compound in accordance with Claim 22 where the R14 group is selected from 4-methylphenyl and 5-methyl-2-thienyl.
24. A compound in accordance with Claim 22 where R8 is ethyl or H, or a pharmaceutically acceptable salt of said compound.
25. A compound in accordance with Claim 14 where R2 is OCH3, R2* is H and R14 is 4-methylphenyl.
26. A compound in accordance with Claim 25 where R8 is ethyl or H, or a pharmaceutically acceptable salt of said compound.
27. A compound in accordance with with Claim 13 where X2 is N, R2 is H, R2* is H, and R14 is selected from the group consisting of 4-methylphenyl and 4-ethylphenyl.
28. A compound in accordance with Claim 27 where R8 is ethyl or H, or a pharmaceutically acceptable salt of said compound.
29. A compound of the formula where R2* is H or F;
R8 is H, or lower alkyl of 1 to 6 carbons, and R14 is selected from the group consisting of phenyl, and 4-(lower-alkyl)phenyl, where lower alkyl has 1 to 6 carbons, or a pharmaceutically acceptable salt of said compound.
30. A compound in accordance with Claim 29 where R2* is H.
31. A compound in accordance with Claim 30 where the R14 group is selected from phenyl, 4-methylphenyl, 4-ethylphenyl, 4-i-propylphenyl, and 4-tbutylphenyl.
32. A compound in accordance with Claim 31 where R8 is ethyl or H, or a pharmaceutically acceptable salt of said compound.
33. A compound in accordance with Claim 29 where R2* is F.
34. A compound in accordance with with Claim 33 where R14 is selected from the group consisting of 4-methylphenyl and 4-ethylphenyl.
35. A compound in accordance with Claim 34 where R8 is ethyl or H, or a pharmaceutically acceptable salt of said compound.
36. A compound of the formula where R8 is H or lower alkyl of 1 to 6 carbons, or a pharmaceutically acceptable salt of said compound.
37. A compound in accordance with Claim 36 where R8 is H, or a pharmaceutically acceptable salt of said compound.
38. A compound in accordance with Claim 36 where R8 is ethyl.
39. Use of a retinoid antagonist or negative hormone which binds to a retinoic acid receptor subtype selected from the group consisting of RAR.alpha., RAR.beta. and RAR.gamma.
to treat in a mammal a pathological condition caused by a retinoid or vitamin A or vitamin A precursor induced toxicity or side effects, wherein said antagonist or negative hormone is a compound in accordance with Claim 1.
40. Use of a retinoid antagonist or negative hormone which binds to a retinoic acid receptor subtype selected from the group consisting of RAR.alpha., RAR.beta. and RAR.gamma.
in the manufacture of a medicament to treat in a mammal a pathological condition caused by a retinoid or vitamin A or vitamin A precursor induced toxicity or side effects, wherein said antagonist or negative hormone is a compound in accordance with Claim 1.
41. Use according to Claim 39 or 40 wherein the retinoid antagonist or negative hormone is used in order to cure or ameliorate a pre-existing pathological condition caused by intake of a retinoic drug or vitamin A or vitamin A precursors by the mammal.
42. Use according to Claim 39 or 40 wherein the retinoid antagonist or negative hormone is used topically to block or ameliorate undesired topical side effects of a retinoic drug administered for a therapeutic purpose.
43. Use according to Claim 39 or 40 wherein the retinoid antagonist or negative hormone is used topically to block or ameliorate undesired topical side effects of a retinoic drug administered systemically for a therapeutic purpose.
44. Use according to Claim 39 or 40 wherein the retinoid antagonist or negative hormone is used topically to treat a pre-existing condition or side effect caused by a retinoid drug or vitamin A.
45. Use according to Claim 39 or 40 wherein the retinoid antagonist or negative hormone is used systemically to treat a pre-existing condition or side effect caused by a retinoid drug or vitamin A.
46. Use according to Claim 39 or 40 wherein the retinoid antagonist or negative hormone is used to systemically to block or ameliorate bone toxicity caused by coadministration of a retinoid drug or vitamin A.
CA002316351A 1997-12-24 1998-12-21 Benzopyran and benzothiopyran derivatives having retinoid antagonist-like activity Expired - Lifetime CA2316351C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/998,319 US5958954A (en) 1995-09-01 1997-12-24 Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities
US08/998,319 1997-12-24
PCT/US1998/027314 WO1999033821A1 (en) 1997-12-24 1998-12-21 Benzopyran and benzothiopyran derivatives having retinoid antagonist-like activity

Publications (2)

Publication Number Publication Date
CA2316351A1 CA2316351A1 (en) 1999-07-08
CA2316351C true CA2316351C (en) 2009-03-17

Family

ID=25545052

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002316351A Expired - Lifetime CA2316351C (en) 1997-12-24 1998-12-21 Benzopyran and benzothiopyran derivatives having retinoid antagonist-like activity

Country Status (9)

Country Link
US (1) US5958954A (en)
EP (1) EP1042313B1 (en)
JP (1) JP4118511B2 (en)
AT (1) ATE265447T1 (en)
AU (1) AU744757B2 (en)
CA (1) CA2316351C (en)
DE (1) DE69823553T2 (en)
HK (1) HK1030167A1 (en)
WO (1) WO1999033821A1 (en)

Families Citing this family (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003523166A (en) 1998-09-29 2003-08-05 ガミダ セル リミテッド Methods for controlling proliferation and differentiation of stem and progenitor cells
US6521641B1 (en) * 1998-10-08 2003-02-18 Allergan, Inc. Male anti-fertility agents
EP1119350B1 (en) * 1998-10-08 2005-02-23 Allergan, Inc. Rar antagonists as male anti-fertility agents
CA2254429A1 (en) 1998-11-19 2000-05-19 Tully Michael Underhill Compositions for promoting chondrogenesis
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
AU4314000A (en) 1999-04-28 2000-11-17 Institute Of Medicinal Molecular Design. Inc. Heterocyclic carboxylic acid derivatives
WO2001007028A2 (en) * 1999-07-23 2001-02-01 Allergan Sales, Inc. The use of retinoid receptor antagonists in the treatment of prostate carcinoma
MXPA02000828A (en) * 1999-08-02 2002-07-30 Hoffmann La Roche Retinoids for the treatment of emphysema.
US6479670B1 (en) * 1999-08-25 2002-11-12 Hoffmann-La Roche Inc. Selective retinoid acid receptor agonists
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
US20020193403A1 (en) * 2001-05-03 2002-12-19 Allergan Sales, Inc. Methods of treating hyperlipidemia
CA2469435A1 (en) 2001-12-21 2003-07-24 X-Ceptor Therapeutics, Inc. 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
EP1465982A4 (en) * 2002-01-25 2006-06-07 Gamida Cell Ltd Methods of expanding stem and progenitor cells and expanded cell populations obtained thereby
US7105566B2 (en) * 2002-10-22 2006-09-12 Allergan, Inc. Methods of treatment during vascular procedures
US20050026958A1 (en) * 2003-07-30 2005-02-03 Allergan, Inc. Methods of therapeutic treatment using amounts of retinoid components
US20050026951A1 (en) * 2003-07-30 2005-02-03 Allergan, Inc. Methods of therapeutic treatment using retinoids with reduced drug interaction
US20050026949A1 (en) * 2003-07-30 2005-02-03 Allergan, Inc. Methods of therapeutic treatment using amounts of retinoids without regard to body weight
US20050026950A1 (en) * 2003-07-30 2005-02-03 Allergan, Inc. Methods of therapeutic treatment using retinoids to achieve consistent bioavailability
US20070224278A1 (en) 2003-11-12 2007-09-27 Lyons Robert T Low immunogenicity corticosteroid compositions
US20050101582A1 (en) 2003-11-12 2005-05-12 Allergan, Inc. Compositions and methods for treating a posterior segment of an eye
EP1701934B1 (en) * 2003-12-26 2009-02-25 Allergan, Inc. DISUBSTITUTED CHALCONE OXIMES HAVING RAR(Gamma)-RETINOID RECEPTOR ANTAGONIST ACTIVITY
ES2318453T3 (en) 2004-01-20 2009-05-01 Allergan, Inc. COMPOSITIONS FOR LOCALIZED EYE THERAPY, WHICH INCLUDE, PREFERENCE, ACETONID TRIAMCINOLONE AND HIALURONIC ACID.
BRPI0510485A (en) 2004-04-30 2007-11-13 Allergan Inc biodegradable intravitreal tyrosine kinase inhibitor implants
US9498457B2 (en) 2004-04-30 2016-11-22 Allergan, Inc. Hypotensive prostamide-containing biodegradable intraocular implants and related implants
US7799336B2 (en) 2004-04-30 2010-09-21 Allergan, Inc. Hypotensive lipid-containing biodegradable intraocular implants and related methods
US7993634B2 (en) 2004-04-30 2011-08-09 Allergan, Inc. Oil-in-oil emulsified polymeric implants containing a hypotensive lipid and related methods
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
US8722097B2 (en) 2004-04-30 2014-05-13 Allergan, Inc. Oil-in-water method for making polymeric implants containing a hypotensive lipid
US20050244462A1 (en) * 2004-04-30 2005-11-03 Allergan, Inc. Devices and methods for treating a mammalian eye
US8591885B2 (en) * 2004-04-30 2013-11-26 Allergan, Inc. Carbonic anhydrase inhibitor sustained release intraocular drug delivery systems
US8425929B2 (en) 2004-04-30 2013-04-23 Allergan, Inc. Sustained release intraocular implants and methods for preventing retinal dysfunction
US20050244471A1 (en) * 2004-04-30 2005-11-03 Allergan, Inc. Estradiol derivative and estratopone containing sustained release intraocular implants and related methods
US8119154B2 (en) 2004-04-30 2012-02-21 Allergan, Inc. Sustained release intraocular implants and related methods
US8147865B2 (en) 2004-04-30 2012-04-03 Allergan, Inc. Steroid-containing sustained release intraocular implants and related methods
US20050244465A1 (en) * 2004-04-30 2005-11-03 Allergan, Inc. Drug delivery systems and methods for treatment of an eye
US8455656B2 (en) 2004-04-30 2013-06-04 Allergan, Inc. Kinase inhibitors
US8673341B2 (en) 2004-04-30 2014-03-18 Allergan, Inc. Intraocular pressure reduction with intracameral bimatoprost implants
US20050244461A1 (en) * 2004-04-30 2005-11-03 Allergan, Inc. Controlled release drug delivery systems and methods for treatment of an eye
US20050244463A1 (en) 2004-04-30 2005-11-03 Allergan, Inc. Sustained release intraocular implants and methods for treating ocular vasculopathies
US20050244478A1 (en) * 2004-04-30 2005-11-03 Allergan, Inc. Anti-excititoxic 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
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
GB0516723D0 (en) * 2005-08-15 2005-09-21 Novartis Ag Organic compounds
CA2623638A1 (en) 2005-09-30 2007-04-12 Vitae Pharmaceuticals, Inc. Treatment of cancer with specific rxr agonists
US8846393B2 (en) 2005-11-29 2014-09-30 Gamida-Cell Ltd. Methods of improving stem cell homing and engraftment
TR201819157T4 (en) * 2006-05-16 2019-01-21 Io Therapeutics Llc Rar Antagonist Or Inverse Agonist For Use In The Treatment Of Side Effects Of Chemotherapy And 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
KR101292188B1 (en) * 2009-03-27 2013-08-02 한국생명공학연구원 Benzopyrane derivatives, pharmaceutically acceptable salts thereof, preparation method thereof and pharmaceutical composition for preventing or treating inflammatroy diseases containing the same as an active ingredient
CN102724951A (en) 2009-11-09 2012-10-10 阿勒根公司 Compositions and methods for stimulating hair growth
US8772273B2 (en) 2011-10-04 2014-07-08 Quretino Therapeutics, Inc. Formulations and uses of retinoic acid receptor selective agonists
AU2012352149B2 (en) 2011-12-13 2017-06-01 Io Therapeutics, Inc. Autoimmune disorder treatment using RXR agonists
CA2863795A1 (en) 2012-02-13 2013-08-22 Gamida-Cell Ltd. Culturing of mesenchymal stem cells
US9567569B2 (en) 2012-07-23 2017-02-14 Gamida Cell Ltd. Methods of culturing and expanding mesenchymal stem cells
US9175266B2 (en) 2012-07-23 2015-11-03 Gamida Cell Ltd. Enhancement of natural killer (NK) cell proliferation and activity
CA2901280A1 (en) 2013-02-15 2014-08-21 Allergan, Inc. Sustained drug delivery implant
PL3368080T3 (en) 2015-10-31 2023-09-11 Io Therapeutics, Inc. Treatment of nervous system disorders using combinations of rxr agonists and thyroid hormones
EP4098257A1 (en) 2015-11-25 2022-12-07 IO Therapeutics, Inc. Use of cyp26-resistant rar alpha selective agonists in the treatment of cancer
NZ745704A (en) 2016-03-10 2019-04-26 Io Therapeutics Inc Treatment of muscular disorders with combinations of rxr agonists and thyroid hormones
CN115252789A (en) 2016-03-10 2022-11-01 Io治疗公司 Application of RXR agonist and thyroid hormone in preparation of medicine for treating autoimmune diseases
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
CN109310763B (en) 2016-06-10 2022-10-21 Io治疗公司 Receptor selective retinoid and rexinoid compounds and immunomodulators for cancer immunotherapy
AU2018301810A1 (en) 2017-07-13 2020-01-23 Io Therapeutics, Inc. Receptor subtype and function selective retinoid and rexinoid compounds in combination with immune modulators for cancer immunotherapy
MX2020002262A (en) 2017-08-31 2020-07-20 Io Therapeutics Inc Rar selective agonists in combination with immune modulators for cancer immunotherapy.
WO2019060600A1 (en) 2017-09-20 2019-03-28 Io Therapeutics, Inc. Treatment of disease with esters of selective rxr agonists
US10966950B2 (en) 2019-06-11 2021-04-06 Io Therapeutics, Inc. Use of an RXR agonist in treating HER2+ cancers
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 (109)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4085091A (en) * 1976-12-16 1978-04-18 E. I. Dupont De Nemours And Company Thermally stable, rigid polyesters from thermally stable, rigid dibasic acids and aromatic dihydroxy compounds
US4326055A (en) * 1977-12-22 1982-04-20 Hoffmann-La Roche Inc. Stilbene derivatives
DE2920947A1 (en) * 1979-05-23 1980-12-04 Bayer Ag 2-PHENYL-ALKEN-1-YL-CYCLOPROPANE-CARBONIC ACID ESTER, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE AS INSECTICIDES AND ACARICIDES AND INTERMEDIATE PRODUCTS FOR THEIR PRODUCTION
EP0047817A3 (en) * 1980-08-14 1982-05-26 F. HOFFMANN-LA ROCHE & CO. Aktiengesellschaft Hydrogenated naphthalines, their preparation and application, and mixtures containing such naphthalines
CH651034A5 (en) 1982-05-12 1985-08-30 Hoffmann La Roche CHROMAN, THIOCHROMAN OR 1,2,3,4-TETRAHYDROCHINOLIN DERIVATIVES AND THEIR USE AS MEDICINAL ACTIVE SUBSTANCES.
DK158947C (en) 1982-07-06 1991-01-21 Hoffmann La Roche TETRAHYDRONAPHTHALIN, BENZOFURAN AND BENZOTHIOPHENDER DERIVATIVES, PREPARATION AND USE THEREOF, AND RODENTICID CONTAINING SUCH DERIVATIVES
US4539154A (en) * 1983-06-15 1985-09-03 Hoffmann-La Roche Inc. Polyene compounds
EP0130795B1 (en) 1983-07-05 1988-11-23 Pfizer Inc. Carboxylic acid derivatives useful for inhibiting the degradation of cartilage
US4826984A (en) * 1984-04-09 1989-05-02 The Board Of Regents For The Oklahoma Agricultural And Mechanical College Acting For And On Behalf Of Oklahoma State University Heteroarotinoid compounds as anticancer agents
JPS60222445A (en) * 1984-04-19 1985-11-07 Yoshitomi Pharmaceut Ind Ltd Phthalic acid compound
JPS6122046A (en) 1984-07-07 1986-01-30 Koichi Shiyudo Stilbene derivative
LU85544A1 (en) * 1984-09-19 1986-04-03 Cird AROMATIC HETEROCYCLIC DERIVATIVES, THEIR PREPARATION PROCESS AND THEIR APPLICATION IN THE THERAPEUTIC AND COSMETIC FIELDS
DE3434947A1 (en) 1984-09-22 1986-04-03 Basf Ag, 6700 Ludwigshafen ISOXAZOLIC CARBONIC ACID DERIVATIVES, THEIR PRODUCTION AND USE
DE3434942A1 (en) 1984-09-22 1986-04-03 Basf Ag, 6700 Ludwigshafen TETRALINE DERIVATIVES, THEIR PRODUCTION AND USE
IL80270A0 (en) * 1985-10-11 1987-01-30 Cird Naphthalene derivatives,their preparation and pharmaceutical compositions containing them
DE3602473A1 (en) 1986-01-28 1987-07-30 Basf Ag VINYLPHENOL DERIVATIVES, THEIR PRODUCTION AND USE
LU86351A1 (en) 1986-03-12 1987-11-11 Oreal BENZOPYRANNYL AND BENZOTHIOPYRANNYL BENZOIC COMPOUNDS, PROCESS FOR THEIR PREPARATION AND THEIR USE IN COSMETICS AND HUMAN AND VETERINARY MEDICINE
DE3615157A1 (en) * 1986-05-05 1987-11-12 Schwabe Willmar Gmbh & Co 5-ARYLALKYL-4-ALKOXY-2 (5H) -FURANONE, INTERMEDIATE PRODUCTS AND METHOD FOR THE PRODUCTION THEREOF AND THEIR USE AS THERAPEUTIC ACTIVE SUBSTANCES
ZW6587A1 (en) 1986-05-13 1987-12-02 Hoffmann La Roche Tetrahydro naphthanline derivatives
ZW7487A1 (en) 1986-05-23 1987-12-16 Hoffmann La Roche Tetrahydronaphthaline and indane derivatives
ZW10287A1 (en) 1986-07-15 1988-01-13 Hoffmann La Roche Tetrahydronaphthaline and indane derivatives
FR2601670B1 (en) * 1986-07-17 1988-10-07 Cird NOVEL AROMATIC BICYCLIC DERIVATIVES, THEIR PREPARATION PROCESS AND THEIR USE IN HUMAN AND VETERINARY MEDICINE AND IN COSMETICS
US4739098A (en) * 1986-09-22 1988-04-19 Allergan, Inc. Ethynylphenyl-containing retinoic acid derivatives
NZ222968A (en) 1986-12-24 1990-05-28 Allergan Inc Ethynylheteroaromatic derivatives and medicaments
US4923884A (en) * 1986-12-24 1990-05-08 Allergan, Inc. Ethynylheteroaromatic-acids having retinoic acid-like activity
US4927947A (en) * 1986-12-24 1990-05-22 Allergan, Inc. Ethynylheteroaromatic-acids having retinoic acid-like activity
US5149705A (en) * 1987-03-13 1992-09-22 Allergan, Inc. Acetylenes disubstituted with a heteroaromatic group and a tetralin group and having retinoid like activity
CA1305480C (en) 1987-03-20 1992-07-21 Roshantha A.S. Chandraratna Disubstituted acetylenes bearing heteroaromatic and heterobicyclic groups having retinoid like activity
US5264578A (en) 1987-03-20 1993-11-23 Allergan, Inc. Disubstituted acetylenes bearing heterobicyclic groups and heteroaromatic or phenyl groups having retinoid like activity
US5089509A (en) * 1988-09-15 1992-02-18 Allergan, Inc. Disubstituted acetylenes bearing heteroaromatic and heterobicyclic groups having retinoid like activity
US5234926A (en) * 1987-03-20 1993-08-10 Allergan, Inc. Disubstituted acetylenes bearing heteroaromatic and heterobicyclic groups having retinoid like activity
US5602130A (en) * 1987-03-20 1997-02-11 Allergan Disubstituted acetylenes bearing heteroaromatic and heterobicyclic groups having retinoid like activity
US4810804A (en) * 1987-03-26 1989-03-07 Allergan, Inc. Acetylenes disubstituted with a phenyl group and a heterobicyclic group having retinoid-like activity
IL85896A (en) 1987-04-06 1993-01-14 Riker Laboratories Inc Substituted di-tert.- butylphenols and pharmaceutical compositions containing them
DE3726806A1 (en) 1987-08-12 1989-02-23 Basf Ag ARYLPHOSPHOR DERIVATIVES, THEIR PRODUCTION AND USE
ZA885192B (en) 1987-08-19 1989-04-26 Hoffmann La Roche Pharmaceutical preparations
CA1298309C (en) 1987-11-06 1992-03-31 Michael Klaus Benzocycloheptene derivatives
KR0139216B1 (en) * 1988-04-11 1998-05-01 제임스 엠. 캐내지 Tetralin esters of phenols or benzoic acids having retinoic like activity
US5231113A (en) * 1988-04-11 1993-07-27 Allergan, Inc. Tetralin esters of phenols or benzoic acids having retinoid like activity
US5015658A (en) * 1988-06-29 1991-05-14 Allergan, Inc. Thiochroman esters of phenols and terephthallates having retinoid-like activity
US4895868A (en) * 1988-06-29 1990-01-23 Allergan, Inc. Thiochroman esters of phenols and terephthallates having retinoid-like activity
AU626881B2 (en) * 1988-07-14 1992-08-13 F. Hoffmann-La Roche Ag Benzofused heterocyclics used as pharmaceuticals
US5217867A (en) * 1988-11-30 1993-06-08 The Salk Institute For Biological Studies Receptors: their identification, characterization, preparation and use
EP0451206A4 (en) * 1988-12-23 1992-07-22 The Salk Institute For Biological Studies Receptor transcription-repression activity compositions and methods
US4992468A (en) * 1989-07-26 1991-02-12 Allergan, Inc. Phenylethenyl compounds having retinoid-like activity
US5068252A (en) * 1989-07-26 1991-11-26 Allergan, Inc. Methods of using phenylethenyl compounds having retinoid-like activity
DE3926148A1 (en) 1989-08-08 1991-02-28 Basf Ag DIARYLACETYLENE, THEIR MANUFACTURE AND USE
US5272156A (en) * 1989-09-19 1993-12-21 Allergan, Inc. Acetylenes disubstituted with a heteroaromatic group and a 2-substituted 1,2,3,4-tetrahydroquinolinyl group having retinoid-like activity
US5023341A (en) * 1989-09-19 1991-06-11 Allergan, Inc. Compounds having a disubstituted acetylene moiety and retinoic acid-like biological activity
US4980369A (en) * 1989-09-19 1990-12-25 Allergan, Inc. Acetylenes disubstituted with a phenyl group and a 2-substituted chromanyl or thiochromanyl group having retinoid-like activity
US5045551A (en) * 1989-09-19 1991-09-03 Allergan, Inc. Acetylenes disubstituted with a heteroaromatic group and a 2-substituted chromanyl, thiochromanyl or 1,2,3,4-tetrahydroquinolinyl group having retinoid-like activity
US5183827A (en) * 1989-09-19 1993-02-02 Allergan, Inc. Acetylenes disubstituted with a heteroaromatic group and a 2-substituted chromanyl, thiochromanyl or 1,2,3,4-tetrahydroquinolinyl group having retinoid-like activity
US5053523A (en) * 1989-09-19 1991-10-01 Allergan, Inc. Ethynyl-chroman compounds
US5162546A (en) * 1989-09-19 1992-11-10 Allergan, Inc. Acetylenes disubstituted with a phenyl group and a 2-substituted chromanyl, thiochromanyl or 1,2,3,4-tetrahydroquinolinyl group having retinoid-like activity
US5248777A (en) * 1989-09-19 1993-09-28 Allergan, Inc. Process and intermediates for preparing compounds having a disubstituted acetylene moiety and retinoic acid-like biological activity
US5399561A (en) * 1989-09-19 1995-03-21 Allergan, Inc. Acetylenes disubstituted with a phenyl or heteroaryl group and a 2-oxochromanyl, 2-oxothiochromanyl or 2-oxo-1,2,3,4-tetrahydro-quinolinyl group having retinoid-like biological activity
US5013744B1 (en) * 1989-12-29 1994-09-20 Allegran Inc Acetylenes disubstituted with a pyridinyl group and a substituted phenyl group having retinoid like activity
US5006550A (en) * 1989-12-29 1991-04-09 Allergan, Inc. Chroman esters of phenols and benzoic acids having retinoid-like activity
US5264456A (en) * 1989-12-29 1993-11-23 Allergan, Inc. Acetylenes disubstituted with a furyl group and a substituted phenyl group having retinoid like activity
US5175185A (en) * 1989-12-29 1992-12-29 Allergan, Inc. Acetylenes disubstituted with a heteroaromatic group and a substituted phenyl group having retinoid like activity
US5202471A (en) * 1990-02-06 1993-04-13 Allergan, Inc. Alkyl, alkoxy and thioalkoxy substituted diphenyl acetylenes having retinoid like activity
US5214202A (en) 1990-03-20 1993-05-25 Shionogi & Co., Ltd. Method for preparing benzoic acid derivatives
US5563292A (en) * 1990-10-17 1996-10-08 Schering Corporation Lipoxygenase inhibitors
WO1992014725A1 (en) * 1991-02-13 1992-09-03 Allergan, Inc. Chroman and thiochromans with phenylethynyl substituents at the 7-position having retinoid-like biological activity
JP3157523B2 (en) * 1991-03-26 2001-04-16 アラーガン、インコーポレイテッド Chromans and thiochromans having a heteroarylethynyl substituent at position 7 with retinoid-like biological activity
US5134159A (en) * 1991-03-26 1992-07-28 Allergan, Inc. 7-chromanyl esters of phenols and benzoic acids having retinoid-like activity
FR2676440B1 (en) 1991-05-15 1993-07-30 Cird Galderma NOVEL AROMATIC COMPOUNDS DERIVED FROM IMINE, THEIR PREPARATION PROCESS AND THEIR USE IN HUMAN AND VETERINARY MEDICINE AND IN COSMETICS.
AU2579692A (en) * 1991-09-17 1993-04-27 Salk Institute For Biological Studies, The Receptor of the thyroid/steroid hormone receptor superfamily
US5326898A (en) * 1992-02-11 1994-07-05 Allergan, Inc. Substituted phenylethenyl compounds having retinoid-like biological activity
AU3659293A (en) * 1992-02-11 1993-09-03 Allergan, Inc. Heteroaryl substituted phenylethenyl compounds having retinoid-like biological activity
US5420145A (en) * 1993-03-22 1995-05-30 Koichi Shudo Carboxylic acid derivative
EP0617020A1 (en) 1992-04-02 1994-09-28 Shudo, Koichi, Prof. Dr. Carboxylic acid derivatives having retinoic acid-like activity
US5324840A (en) * 1992-06-11 1994-06-28 Allergan, Inc. Method of treatment with compounds having retinoid-like activity and reduced skin toxicity and lacking teratogenic effects
US5466861A (en) * 1992-11-25 1995-11-14 Sri International Bridged bicyclic aromatic compounds and their use in modulating gene expression of retinoid receptors
US5455265A (en) * 1993-02-11 1995-10-03 Allergan, Inc. Method of treatment with compounds having selective agonist-like activity on RXR retinoid receptors
US5399586A (en) * 1993-03-11 1995-03-21 Allergan, Inc. Treatment of mammals afflicted with tumors with compounds having RXR retinoid receptor agonist activity
EP0619116A3 (en) 1993-04-05 1994-11-23 Hoechst Japan Use of synthetic retinoids for osteopathy.
US5344959A (en) * 1993-05-18 1994-09-06 Allergan, Inc. Tetrahydronaphthyl and cyclopropyl substituted 1,3-butadienes having retinoid-like activity
US5475022A (en) 1993-10-18 1995-12-12 Allergan, Inc. Phenyl or heteroaryl and tetrahydronaphthyl substituted diene compounds having retinoid like biological activity
FR2713635B1 (en) 1993-12-15 1996-01-05 Cird Galderma New bi-aromatic propynyl compounds, pharmaceutical and cosmetic compositions containing them and uses.
US5470999A (en) * 1993-12-30 1995-11-28 Allergan, Inc. Cyclohexene and bicyclic aromatic substituted ethyne compounds having retinoid-like biological activity
US5451605A (en) * 1993-12-30 1995-09-19 Allergan, Inc. 1,2-epoxycyclohexanyl and bicyclic aromatic substituted ethyne compounds having retinoid-like biological activity
US5426118A (en) * 1993-12-30 1995-06-20 Allergan, Inc. [4-(1,2-epoxycyclohexanyl)but-3-en-1-ynyl]aromatic and heteroaromatic acids and derivatives having retinoid-like biological activity
CA2137997A1 (en) 1994-01-03 1995-07-04 John E. Starrett, Jr. Retinoid-like compounds
US5648385A (en) * 1994-01-03 1997-07-15 Bristol-Myers Squibb Co. Retinoid-like compounds
CA2138000A1 (en) 1994-01-03 1995-07-04 John E. Starrett, Jr. Retinoid-like compounds
US5498755A (en) * 1994-08-23 1996-03-12 Chandraratna; Roshantha A. Disubstituted aryl and heteroaryl imines having retinoid-like biological activity
IL116259A (en) 1994-12-19 2000-07-16 American Cyanamid Co Analogs of 9-cis retinoic acid and their use
US5599967A (en) * 1994-12-29 1997-02-04 Allergan Acetylenes disubstituted with a 5 substituted tetrahydronaphthyl group and with an aryl of heteroaryl group having retinoid-like biological activity
US5489584A (en) * 1994-12-29 1996-02-06 Allergan, Inc. Acetylenes disubstituted with a 5-amino or substituted 5-amino substituted tetrahydronaphthyl group and with an aryl or heteroaryl group having retinoid-like biological activity
US5556996A (en) * 1994-12-29 1996-09-17 Allergan Oxiranyls disubstituted with a phenyl group and a substituted chromanyl or tetrahydroquinolinyl group having retinoid like activity
US5498795A (en) * 1994-12-29 1996-03-12 Allergan, Inc. Acetylenes disubstituted with hydroxyaryl and aryl or heteroaryl groups having retinoid-like biological activity
US5543534A (en) * 1994-12-29 1996-08-06 Allergan Acetylenes disubstituted with a 5 substituted tetrahydronaphthyl group and with an aryl or heteroaryl groups having retinoid-like biological activity
US5618943A (en) * 1994-12-29 1997-04-08 Allergan Acetylenes disubstituted with a 5 OXO substituted tetrahydronaphthyl group and with an aryl or heteroaryl group having retinoid-like biological activity
US5648514A (en) * 1994-12-29 1997-07-15 Allergan Substituted acetylenes having retinoid-like biological activity
US5618931A (en) * 1994-12-29 1997-04-08 Allergan Acetylenes disubstituted with a 5 substituted dihydronaphthyl group and with an aryl or heteroaryl group having retinoid-like biological activity
US5534641A (en) 1994-12-29 1996-07-09 Allergan Acetylenes disubstituted with 2-tetrahydropyranoxyaryl and aryl or heteroaryl groups having retinoid-like biological activity
US5514825A (en) * 1994-12-29 1996-05-07 Allergan, Inc. Acetylenes disubstituted with a 5 substituted tetrahydronaphthyl group and with an aryl or heteroaryl group having retinoid-like biological activity
US5559248A (en) * 1995-04-05 1996-09-24 Bristol-Myers Squibb Co. Retinoid-like heterocycles
US5616712A (en) * 1995-05-16 1997-04-01 Allergan Acetylenes disubstituted with a phenyl or heteroaryl group and a 2-thio-1,2,3,4-tetrahdroquinolinyl, 2-alkylthio-3,4-dihydroquinolinyl or 2-alkoxy-3,4-dihydroquinolinyl group having retinoid-like biological activity
US5675033A (en) 1995-06-06 1997-10-07 Allergan 2,4-pentadienoic acid derivatives having retinoid-like biological activity
US5776699A (en) * 1995-09-01 1998-07-07 Allergan, Inc. Method of identifying negative hormone and/or antagonist activities
AU7598596A (en) 1995-11-01 1997-05-22 Allergan, Inc. Sulfides, sulfoxides and sulfones disubstituted with a tetrahydronaphthalenyl, chromanyl, thiochromanyl or tetrahydroquinolinyl and substituted phenyl or heteroaryl group, having retinoid-like biological activity
US5663357A (en) 1995-11-22 1997-09-02 Allergan Substituted heteroarylamides having retinoid-like biological activity
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
US5688957A (en) 1995-12-29 1997-11-18 Allergan (3"-thioxacyclohex-1"-enyl)!-but-3'-ene-1'-ynyl!aryl and (3"-thioxacyclohex-1"-enyl)!-but-3'-ene-1'-ynyl!heteroaryl carboxylic acids and esters having retinoid-like biological activity
US5723666A (en) 1996-06-21 1998-03-03 Allergan Oxime substituted tetrahydronaphthalene derivatives having retinoid and/or retinoid antagonist-like biological activity
US5739338A (en) 1996-11-05 1998-04-14 Allergan N-aryl substituted tetrahydroquinolines having retinoid agonist, retinoid antagonist or retinoid inverse agonist type biological activity
US5728846A (en) 1996-12-12 1998-03-17 Allergan Benzo 1,2-g!-chrom-3-ene and benzo 1,2-g!-thiochrom-3-ene derivatives

Also Published As

Publication number Publication date
EP1042313B1 (en) 2004-04-28
US5958954A (en) 1999-09-28
JP4118511B2 (en) 2008-07-16
EP1042313A1 (en) 2000-10-11
AU1940699A (en) 1999-07-19
DE69823553T2 (en) 2005-05-25
HK1030167A1 (en) 2001-04-27
AU744757B2 (en) 2002-03-07
CA2316351A1 (en) 1999-07-08
JP2001527071A (en) 2001-12-25
DE69823553D1 (en) 2004-06-03
ATE265447T1 (en) 2004-05-15
WO1999033821A1 (en) 1999-07-08

Similar Documents

Publication Publication Date Title
CA2316351C (en) Benzopyran and benzothiopyran derivatives having retinoid antagonist-like activity
US5877207A (en) Synthesis and use of retinoid compounds having negative hormone and/or antagonist activities
EP0853610B1 (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
MKEX Expiry

Effective date: 20181221