Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
THROMBIN RECEPTOR ANTAGONISTS
Document Type and Number:
WIPO Patent Application WO/2005/030712
Kind Code:
A2
Abstract:
A series of compounds represented by the structural formulas and pharmaceutically acceptable isomers, salts, solvates and polymorphs thereof are disclosed. Also disclosed are pharmaceutical compositions containing said compounds and their use as thrombin receptor antagonists and binders to cannabinoid receptors.

Inventors:
CHACKALAMANNIL SAMUEL (US)
CHELLIAH MARIAPPAN V (US)
XIA YAN (US)
Application Number:
PCT/US2004/031495
Publication Date:
April 07, 2005
Filing Date:
September 23, 2004
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
SCHERING CORP (US)
CHACKALAMANNIL SAMUEL (US)
CHELLIAH MARIAPPAN V (US)
XIA YAN (US)
International Classes:
C07D405/06; C07D405/14; C07D413/14; C07D417/14; C07D521/00; (IPC1-7): C07D/
Domestic Patent References:
WO2001096330A22001-12-20
Foreign References:
US6063847A2000-05-16
Other References:
CHACKALAMANNIL ET AL., J. AM. CHEM SOC., vol. 118, 1996, pages 9812 - 9813
Attorney, Agent or Firm:
REINHARDT, Gerard, E. (2000 Galloping Hill RoadPatent Department, K-6-1 199, Kenilworth NJ, US)
Download PDF:
Claims:
We claim : 1. A compound represented by any of the following structural formulas

or a pharmaceutical acceptable isomer, salt, solvate or polymorph thereof.

2. A pharmaceutical composition comprising an effective amount of at least one compound of Claim 1 and at least one pharmaceutical acceptable carrier.
3. A method of inhibiting thrombin receptors comprising administering to a mammal in need of such treatment an effective amount of at least one compound of Claim 1.
4. A method of inhibiting cannabinoid receptors comprising administering to a mammal in need of such treatment an effective amount of at least one compound of Claim 1.
5. A method of treating thrombosis, platelet aggregation, coagulation, cancer, inflammatory diseases or respiratory diseases comprising administering to a mammal in need of such treatment an effective amount of at least one compound of Claim 1.
6. A method of treating atherosclerosis, restenosis, hypertension, angina pectoris, arrhythmia, heart failure, myocardial infarction, glomerulonephritis, thrombotic stroke, thromboembolytic stroke, peripheral vascular diseases, cerebral ischemia, rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, diabetes, osteoporosis, renal ischemia, cerebral stroke, cerebral ischemia, nephritis, inflammatory disorders of the lungs and gastrointestinal tract, reversible airway obstruction, chronic asthma or bronchitis comprising administering to a mammal in need of such treatment an effective amount of at least one compound of Claim 1.
7. A compound represented by the following structural formula :

or a pharmaceutical acceptable isomer, salt or solvate thereof.

8. A compound represented by the following structural formula

or a pharmaceutical acceptable isomer, satt or solvate thereof.

9. A compound represented by the following structural formula

or a pharmaceutical acceptable isomer, salt or solvate thereof.

10. A compound represented by the following structural formula

or a pharmaceutical acceptable isomer, salt or solvate thereof.

11. A compound represented by the following structural formula

or a pharmaceutical acceptable isomer, salt or solvate thereof.

12. A compound represented by the following structural formula

or a pharmaceutically acceptable isomer, salt or solvate thereof.

13. A compound represented by the following structural formula

or a pharmaceutical acceptable isomer, salt or solvate thereof.

14. A compound represented by the following structural formula

or a pharmaceuticaliy acceptable isomer, salt or solvate thereof.

15. A compound represented by the following structural formula

or a pharmaceutical acceptable isomer, sait or solvate thereof.

16. A compound represented by the following structural formula

or a pharmaceutical acceptable isomer, salt or solvate thereof.

Description:

THROMBIN RECEPTOR ANTAGONISTS

BACKGROUND OF THE INVENTION The present invention relates to nor-seco himbacine derivatives useful as thrombin receptor antagonists in the treatment of diseases associated with thrombosis, atherosclerosis, restenosis, hypertension, angina pectoris, arrhythmia, heart failure, cerebral ischemia, stroke, neurodegenerative diseases and cancer.

Thrombin receptor antagonists are also known as protease activated receptor (PAR) antagonists. The compounds of the invention also bind to cannabinoid (CB2) receptors and are useful in the treatment of rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, diabetes, osteoporosis, renal ischemia, cerebral stroke, cerebral ischemia, nephritis, inflammatory disorders of the lungs and gastrointestinal tract, and respiratory tract disorders such as reversible airway obstruction, chronic asthma and bronchitis. The invention also relates to pharmaceutical compositions containing said compounds.

Thrombin is known to have a variety of activities in different cell types and thrombin receptors are known to be present in such cell types as human platelets, vascular smooth muscle cells, endothelial cells and fibroblasts. It is therefore expected that thrombin receptor antagonists will be useful in the treatment of thrombotic, inflammatory, atherosclerotic and fibroproliferative disorders, as well as other disorders in which thrombin and its receptor play a pathological role.

Thrombin receptor antagonist peptides have been identified based on structure-activity studies involving substitutions of amino acids on thrombin receptors.

In Bernatowicz et al., J. Med. Chem., 39 (1996), p. 4879-4887, tetra-and pentapeptides are disclosed as being potent thrombin receptor antagonists, for example N-trans-cinnamoyl-p-fluoroPhe-p-guanidinoPhe-Leu-Arg-NH2 and N-trans- cinnamoyl-p-fluoroPhe-p-guanidinoPhe-Leu-Arg-Arg-NH2. Peptide thrombin receptor antagonists are also disclosed in WO 94/03479, published February 17,1994.

Cannabinoid receptors belong to the superfamily of G-protein coupled receptors. They are classified into the predominantly neuronal CB1 receptors and the predominantly peripheral CB2 receptors. These receptors exert their biological actions by modulating adenylate cyclase and Ca+2 and K+ currents. While the effects of CB1 receptors are principally associated with the central nervous system, CB2 receptors are believed to have peripheral effects related to bronchial constriction, immunomodulation and inflammation. As such, a selective CB2 receptor binding

agent is expected to have therapeutic utility in the control of diseases associated with rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, diabetes, osteoporosis, renal ischemia, cerebral stroke, cerebral ischemia, nephritis, inflammatory disorders of the lungs and gastrointestinal tract, and respiratory tract disorders such as reversible airway obstruction, chronic asthma and bronchitis (R. G.

Pertwee, Curr. Med. Chem. 6 (8), (1999), 635).

Himbacine, a piperidine alkaloid of the formula

has been identified as a muscarinic receptor antagonist. The total synthesis of (+)- himbacine is disclosed in Chackalamannil et al., J. Am. Chem Soc., 118 (1996), p.

9812-9813.

Tricyclic himbacine-related compounds have been disclosed as thrombin receptor antagonists in US 6,063, 847.

SUMMARY OF THE INVENTION The present invention relates to thrombin receptor antagonists represented by the formula I

or a pharmaceutical acceptable isomer, salt, solvate or polymorph thereof, wherein: Ruz (CH2) <i (CH2) n< Z is- (CH2) n- ; ' ; '', when R10 is absent ; or Dw (CH2) n (S, ", when R3 is absent ;

the single dotted line represents an optional double bond; the double dotted line represents an optional single bond; n is 0-2; R1 and R2 are independently selected from the group consisting of H, C1-C6 <BR> <BR> alkyl, fluoro (C1-C6) alkyl, difluoro (C-C6) alkyl, trifluoro- (C1-C6) alkyl, C3-C7 cycloalkyl, C2-C6 alkenyl, aryl (C1-C6) alkyl, aryl (C2-C6) alkenyl, heteroaryl (C-C6) alkyl,

heteroaryl (C2-C6) alkenyl, hydroxy- (C1-C6) alkyl, (C1-C6)alkoxy(C1-C6)alkyl, amino- (C1-C6) alkyl, aryl and thio (Ci-Ce) alkyl ; or R1 and R2 together form a =0 group; R3 is H, hydroxy, C1-C6 alkoxy, -NR18R19, -SOR16, -SO2R17, -C(O)OR17, -C(O)NR18R19, C1-C6 alkyl, halogen, fluoro(C1-C6)alkyl, difluoro(C1-C6)alkyl, trifluoro (C1-C6) alkyl, C3-C7 cycloalkyl, C2-C6 alkenyl, aryl (C1-C6) alkyl, aryl (C2- C6) alkenyl, heteroaryl (Ci-Ce) alkyl, heteroaryl (C2-C6) alkenyl, hydroxy (Cl-C6) alkyl, amino (C1-C6) alkyl, aryl, thio (C1-C6) alkyl, (Ci-C6) alkoxy (C1-C6) alkyl or (C1-C6) alkylamino(C1-C6)alkyl ; R34 is (H, R3), (H, R43), =O or =NoR17 when the optional double bond is absent; R34 is R44 when the double bond is present; Het is a mono-, bi-or tricyclic heteroaromatic group of 5 to 14 atoms comprised of 1 to 13 carbon atoms and 1 to 4 heteroatoms independently selected from the group consisting of N, O and S, wherein a ring nitrogen can form an N-oxide or a quaternary group with a C1-C4 alkyl group, wherein Het is attached to B by a carbon atom ring member, and wherein the Het group is substituted by 1 to 4 substituents, W, independently selected from the group consisting of H; C1-C6 alkyl ; fluoro (C1-C6) alkyl ; difluoro(C1-C6)alkyl ; trifluoro-(C1-C6)-alkyl ; C3-C7 cycloalkyl ; heterocycloalkyl ; heterocycloalkyl substituted by C1-C6 alkyl, C2-C6 alkenyl, OH- (Ci- C6) alkyl, or =0 ; C2-C6 alkenyl ; R21-aryl (C1-C6) alkyl ; R21-aryl-(C2-C6)-alkenyl; R21- aryloxy ; R21-aryl-NH-; heteroaryl(C1-C6)alkyl ; heteroaryl (C2-C6)-alkenyl ; heteroaryloxy ; heteroaryl-NH- ; hydroxy (Ci-C6) alkyl ; dihydroxy (Ci-C6) alkyl ; amino (C1- C6) alkyl ; (Ci-C6) alkylamino-(C1-C6)alkyl ; di-((C1-C6)alkyl)-amino(C1-C6)alkyl ; thio (C1- C6) alkyl ; C1-C6 alkoxy ; C2-C6 alkenyloxy ; halogen ;-NR4R5 ; -CN;-OH ;-COOR17 ; <BR> <BR> <BR> - COR16 ;-OS02CF3 ; -CH20CH2CF3; (C1-C6) alkylthio ; -C (O) NR4R5 ;-OCHR6-phenyl ; phenoxy-(C1-C6)alkyl; -NHCOR16; -NHSO2R16 ; biphenyl ; -OC (R6) 2COOR7 ; - (R6) 2C (O) NR4R5 ; (C1-C6) alkoxy ; -C (=NOR17)R18 ; C1-C6 alkoxy substituted by (C1-C6) alkyl, amino, -OH, COOR17,-NHCOOR17, -CONR4R5, aryl, aryl substituted by 1 to 3 substutuents independently selected from the group consisting of halogen,-CF3, C1-Cg alkyl, C1-C6 alkoxy and-COOR17, aryl <BR> <BR> <BR> wherein adjacent carbons form a ring with a methylenedioxy group, -C (O) NR4R5 or heteroaryl ; R21-aryl ; aryl wherein adjacent carbons form a ring with a methylenedioxy group; R41-heteroaryl ; and heteroaryl wherein adjacent carbon atoms form a ring with a C3-C5 alkylene group or a methylenedioxy group; R4 and R5 are independently selected from the group consisting of H, C1-C6 alkyl, phenyl, benzyl and C3-C7 cycloalkyl, or R4 and R5 together are- (CH2) 4-, -(CH2)5- or -(CH2)2NR7-(CH2)2- and form a ring with the nitrogen to which they are attached;

R6 is independently selected from the group consisting of H, C1-C6 alkyl, phenyl, (C3-C7) cycloalkyl, (C3-C7)cycloalkyl(C1-C6)alkyl, (C1-C6)alkoxy(C1-C6)alkyl, hydroxy (C1-C6) alkyl and amino (Ci-C6) alkyl ; R7 is H or (C1-C6) alkyl ; R8, Rio and R11 are independently selected from the group consisting of R1 and-OR1, provided that when the optional double bond is present, Rio is absent; R9 is H, OH, C1-C6 alkoxy, halogen or halo (C1-C6) alkyl ; B is -(CH2)n3-, -CH2-O-, -CH2S-, -CH2-NR6-, -C(O)NR6-, -NR6C(O)-, wherein n3 is 0-5, n4 and n5 are independently 0-2, and R12 and Rua are independently selected from the group consisting of H, Ci-Ce alkyl and halogen ; X is-O-or-NR6-when the double dotted line represents a single bond, or X is H, -OH or-NHR20 when the bond is absent; Y is =0, =S, (H, H), (H, OH) or (H, Ci-Ce alkoxy) when the double dotted line represents a single bond, or when the bond is absent, Y is =O, =NoR17, (H, H), (H, OH), (H, SH), (H, C1-C6 alkoxy) or (H,-NHR45) ; R15 is absent when the double dotted line represents a single bond; R's is H, C1-C6 alkyl,-NR18R19 or-OR17 when the bond is absent; or Y is s -2 and R15 is H or C1-C6 alkyl ; 1 6 Y R16 is C1-C6 lower alkyl, phenyl or benzyl ; R17, R18 and R19 are independently selected from the group consisting of H, C1-C6 alkyl, phenyl, benzyl ; R20 is H, Ci-Ce alkyl, phenyl, benzyl,-C (O) R6 or-S02R6 ; R21 is 1 to 3 substutuents independently selected from the group consisting of hydrogen, CN,-CF3,-OCF3, halogen, -NO2, C1-C6 alkyl, C1-C6alkoxy, (C1- C6) alkylamino, di-((C1-C6) alkyl) amino, amino (C1-C6) alkyl, (C1-C6)-alkylamino (Cl- C6) alkyl, di-((C1-C6) alkyl)-amino (C1-C6) alkyl, hydroxy-(C1-C6) alkyl,-COOR17,- COR17,-NHCOR16 -NHSO2R16, -NHSO2CH2CF3, heteroaryl or-C (=NOR") R'8 ; R22 and R23 are independently selected from the group consisting of hydrogen, R24-(C1-C10)alkyl, R24-(C2-C10)alkenyl, R24-(C2-C10)alkynyl, R27-hetero-cycloalkyl, R25- aryl, R25-aryl (C1-C6) alkyl, R29-(C3-C7) cycloalkyl, R29-(C3-C7) cycloalkenyl,-OH, -OC(O)R30, -C(O)OR30, -C(O)R30, -C(O)NR30R31, -NR30R31, -NR30C(O)R31, -NR30C(O)NR31R32, -NHSO2R30, -OC(O)NR30R31, R24-(C1-C10)alkoxy, R24-(C2-C10)- alkenyloxy, R24-(C2-C10)alkynyloxy, R27-heterocycloalkyloxy, R29-(C3-C7)cycloalkyloxy, R29-(C3-C7)cyclo-alkenyloxy, R29-(C3-C7)cycloalkyl-NH-, -NHSO2NHR16 and

-CH(=NOR17) ; or R22 and R10 together with the carbon to which they are attached, or R23 and R"together with the carbon to which they are attached, independently form a R42- substituted carbocyclic ring of 3-10 atoms, or a R42-substituted heterocyclic ring of 4- 10 atoms wherein 1-3 ring members are independently selected from the group consisting of-O-,-NH-and-SOo-2-, provided that, when R22 and Rlo form a ring, the optional double bond is absent; R24 is 1,2 or 3 substituents independently selected from the group consisting of hydrogen, halogen,-OH, (C1-C6) alkoxy, R35-aryl, (C1-C10)-alkyl-C (O) -, (C2-C1o)- alkenyl-C(O)-, (C2-C10)alkynyl-C(O)-, heterocycloalkyl, R26-(C3-C7)cycloalkyl, R26- (C3-C7) cycloalkenyl, -OC(O)R30, -C(O)OR30, -C(O)R30, -C(O)NR30R31, -NR30R31, -NR30C(O)R31, -NR3-C(O)NR31R32, -NHSO2R30, -OC(O)NR30R31, R24-(C2-C10)- <BR> <BR> alkenyloxy, R24- (C2-C1o) alkynyloxy, R27-heterocycloalkyloxy, R29- (C3-C7)-cycloalkyloxy, R29-(C3-C7)cyclo-alkenyloxy, R29-(C3-C7)cycloalkyl-NH-, -NHSO2NHR16 and -CH(=NOR17) ; R25 is 1,2 or 3 substituents independently selected from the group consisting of hydrogen, heterocycloalkyl, halogen,-COOR36,-CN,-C (O) NR37R,-NR39C (O) R -OR36, (C3-C7)cycloalkyl, (C3-C7)cycloalkyl-C1-C6)alkyl, (C1-C6)alkyl(C3-C7)cycloalkyl- (C1-C6) alkyl, halo (C1-C6) alkyl (C3-C7) cycloalkyl (C1-C6) alkyl, hydroxy (C1-C6) alkyl, (C1- C6) alkoxy (C1-C6) alkyl, and R41-heteroaryl ; or two R25 groups on adjacent ring carbons form a fused methylenedioxy group; R26 is 1,2, or 3 substituents independently selected from the group consisting of hydrogen, halogen and (C1-C6) alkoxy ; R27 is 1,2 or 3 substituents independently selected from the group consisting of hydrogen, R28-(C1-C10)alkyl, R28-(C2-C10)alkenyl, R28-(C2-C10)alkynyl, R28 is hydrogen, -OH or (Ci-C6) alkoxy ; R29 is 1,2 or 3 substituents independently selected from the group consisting of hydrogen, (C1-C6) alkyl,-OH, (C1-C6) alkoxy and halogen ; R30, R31 and R32 are independently selected from the group consisting of hydrogen, (C1-C10)-alkyl, (Ci-C6) alkoxy(C1-C10)-alkyl, R25-aryl(C1-C6)-alkyl, R33-(C3- C7) cycloalkyl, R34-(C3-C7) cycloalkyl (C1-C6) alkyl, R25-aryl, heterocycloalkyi, heteroaryl, heterocycloalkyl (C1-C6) alkyl and heteroaryl (Ci-Ce) alkyl ; R33 is hydrogen, (C1-C6) alkyl, OH-(C1-C6)alkyl or (Ci-Ce) alkoxy ; R35 is 1 to 4 substituents independently selected from the group consisting of hydrogen, (C1-C6) alkyl,-OH, halogen,-CN, (C1-C6) alkoxy, trihalo (Ci-C6) alkoxy, (C1- C6) alkylamino, di ((C1-C6)alkyl)amino, -OCF3, OH-(C1-C6)alkyl, -CHO, -C(O)(C1-C6)- alkylamino,-C (O) di ((C1-C6)alkyl)amino, -NH2, -NHC(O)(C1-C6)alkyl and-N ( (Ci- C6) alkyl) C (O) (C1-C6) alkyl ;

R36 is hydrogen, (C1-C6) alkyl, halo (C,-C6) alkyl, dihalo (C1-C6) alkyl or trifluoro(C1-C6)alkyl, R37 and R38 are independently selected from the group consisting of hydrogen, (Ci-C6) alkyl, aryl(C1-C6)alkyl, phenyl and (C3-C1s) cycloalkyl, or R37 and R38 together are -(CH2)4-, -(CH2)5- or -(CH2)2-NR39-(CH2)2- and form a ring with the nitrogen to which they are attached; R39 and R40 are independently selected from the group consisting of hydrogen, (CI-C6) alkyl, aryl (C1-C6) alkyl, phenyl and (C3-C15)-cycloalkyl, or R39 and R40 in the group-NR39C (O) R40, together with the carbon and nitrogen atoms to which they are attached, form a cyclic lactam having 5-8 ring members; R41 is 1 to 4 substituents independently selected from the group consisting of hydrogen, (C1-C6) alkyl, (C1-C6) alkoxy, (Ci-C6) alkylamino, di ( (Ci-C6) alkyl) amino, - OCFs, OH- (Cl-C6) alkyl,-CHO and phenyl ; R42 is 1 to 3 substituents independently selected from the group consisting of hydrogen, -OH, (C1-C6) alkyl and (Ci-Ce) alkoxy ; R43 is -NR30R31, -NR30C(O)R31, -NR30C(O)NR31R32, -NHSO2R30 or - NHCOOR" ; R44 is H, C1-C6 alkoxy, -SOR16, -SO2R17, -C (O) OR17,-C (O) NR18R19, C1-C6 alkyl, halogen, fluoro (C1-C6) alkyl, difluoro (C1-C6) alkyl, trifluoro (C1-C6) alkyl, C3-C7 cycloalkyl, C2-C6 alkenyl, aryl(C1-C6)alkyl, aryl(C2-C6)alkenyl, heteroaryl(C1-C6)alkyl, heteroaryl (C2-C6) alkenyl, hydroxy (C1-C6) alkyl, amino (C1-C6) alkyl, aryl, thio (C1- C6) alkyl, (C1-C6) alkoxy (C1-C6) alkyl or (C1-C6) alkylamino (C1-C6) alkyl ; and R45 is H, C1-C6 alkyl, -COOR16 or -SO2.

R2, R8, Rio and Rl 1 are each preferably hydrogen. R3 preferably is hydrogen, OH, C1-C6 alkoxy,-NHR'8 or C1-C6 alkyl. The variable n is preferably zero. R9 is preferably H, OH or alkoxy. Rl is preferably C1-C6 alkyl, more preferably methyl.

The double dotted line preferably represents a single bond; X is preferably-O-and Y is preferably =O or (H, -OH). B is preferably trans-CH=CH-. Het is preferably pyridyl, substituted pyridyl, quinolyl or substituted quinolyl. Preferred substituents (W) on Het are R2'-aryl, R41-heteroaryl or alkyl. More preferred are compounds wherein Het is 2- pyridyl substituted in the 5-position by R21-aryl, R41-heteroaryl or alkyl, or 2-pyridyl substituted in the 6-position by alkyl. R34 is preferably (H, H) or (H, OH).

R22 and R23 are preferably selected from OH, (C1-C10)alkyl, (C2-C10)alkenyl, (C2-C10)-alkynyl, trifluoro(C1-C10)alkyl, trifluoro(C2-C10)-alkenyl, trifluoro(C2-C10)alkynyl, (C3-C7)-cycloalkyl, R25-aryl, R25-aryl(C1-C6)alkyl, R25-arylhydroxy(C1-C6)alkyl, R25-aryl- alkoxy-(C1-C6)alkyl, (C3-C7)cycloalkyl-(C1-C6)alkyl, (C1-C10)alkoxy, (C3- C7) cycloalkyloxy, (C1-C6)alkoxy(C1-C6)alkyl, OH-(C1-C6)alkyl, trifluoro(C1-C10)alkoxy and R27-heterocyclo-alkyl (C1-C6) alkyl. More preferred are compounds wherein R22 and R23 are independently selected from the group consisting of (C1-Cio) alkyl and OH-(C1-C6)alkyl.

More preferably, the present invention relates to thrombin receptor antagonists represented by any of the following structural formulas :

or a pharmaceutical acceptable isomer, salt, solvate or polymorph thereof.

Thrombin receptor antagonist compounds of the present invention can have anti-thrombotic, anti-platelet aggregation, antiatherosclerotic, antirestenotic and anti- coagulant activity. Thrombosis-related diseases treated by the compounds of this invention are thrombosis, atherosclerosis, restenosis, hypertension, angina pectoris, arrhythmia, heart failure, myocardial infarction, glomerulonephritis, thrombotic and thromboembolytic stroke, peripheral vascular diseases, other cardiovascular diseases, cerebral ischemia, inflammatory disorders and cancer, as well as other disorders in which thrombin and its receptor play a pathological role.

The compounds of the invention which bind to cannabinoid (CB2) receptors can be useful in the treatment of rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, diabetes, osteoporosis, renal ischemia, cerebral stroke, cerebral ischemia, nephritis, inflammatory disorders of the lungs and gastrointestinal tract, and respiratory tract disorders such as reversible airway obstruction, chronic asthma and bronchitis.

This invention also relates to a method of using at least one compound of formula I in the treatment of thrombosis, platelet aggregation, coagulation, cancer, inflammatory diseases or respiratory diseases, comprising administering a compound of formula I to a mammal in need of such treatment. In particular, the present invention relates to a method of using at least one compound of formula I in the treatment of thrombosis, atherosclerosis, restenosis, hypertension, angina pectoris, arrhythmia, heart failure, myocardial infarction, glomerulonephritis, thrombotic stroke, thromboembolytic stroke, peripheral vascular diseases, cerebral ischemia, cancer, rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, diabetes, osteoporosis, renal ischemia, cerebral stroke, cerebral ischemia, nephritis, inflammatory disorders of the lungs and gastrointestinal tract, reversible airway obstruction, chronic asthma or bronchitis. It is contemplated that a compound of this invention may be useful in treating more than one of the diseases listed.

In another aspect, the invention relates to a pharmaceutical composition comprising a therapeutical effective amount of at least one compound of formula I in at least one pharmaceutical acceptable carrier.

DETAILED DESCRIPTION : Unless otherwise defined, the term"alkyl"or"lower alkyl"means straight or branched alkyl chains of 1 to 6 carbon atoms and"alkoxy"similarly refers to alkoxy groups having 1 to 6 carbon atoms.

Fluoroalkyl, difluoroalkyl and trifluoroalkyl mean alkyl chains wherein the terminal carbon is substituted by 1,2 or 3 fluoroatoms, e. g.,-CF3,-CH2CF3,- CH2CHF2 or-CH2CH2F. Haloalkyl means an alkyl chain substituted by 1 to 3 halo atoms.

"Alkenyl"means straight or branched carbon chains of carbon atoms having one or more double bonds in the chain, conjugated or unconjugated. Similarly, "alkynyl"means straight or branched carbon chains of carbon atoms having one or more triple bonds in the chain. Where an alkyl, alkenyl or alkynyl chain joins two other variables and is therefore bivalent, the terms alkylene, alkenylene and alkynylene are used. Unless otherwise defined, alkenyl and alkynyl chains comprise 1 to 6 carbon atoms.

Substitution on alkyl, alkenyl and alkynyl chains depends on the length of the chain, and the size and nature of the substituent. Those skilled in the art will appreciate that while longer chains can accommodate multiple substituents, shorter alkyl chains, e. g., methyl or ethyl, can have multiple substitution by halogen, but otherwise are likely to have only one or two substituents other than hydrogen.

Shorter unsaturated chains, e. g., ethenyl or ethynyl, are generally unsubstituted or substitution is limited to one or two groups, depending on the number of available carbon bonds.

"Cycloalkyl"means a saturated carbon ring of 3 to 7 carbon atoms, while "cycloalkylene"refers to a corresponding bivalent ring, wherein the points of attachment to other groups include all positional and stereoisomers."Cycloalkenyl" refers to a carbon ring of 3 to 7 atoms and having one or more unsaturated bonds, but not having an aromatic nature.

"Heterocycloalkyl"means saturated rings of 5 or 6 atoms comprised of 4 to 5 carbon atoms and 1 or 2 heteroatoms selected from the group consisting of-O-,-S- and-NR7-joined to the rest of the molecule through a carbon atom. Examples of heterocycloalkyl groups are 2-pyrrolidinyl, tetrahydrothiophen-2-yl, tetrahydro-2- furanyl, 4-piperidinyl, 2-piperazinyl, tetrahydro-4-pyranyl, 2-morpholinyl and 2- thiomorpholinyl.

"Halogen"refers to fluorine, chlorine, bromine or iodine radicals.

When R4 and R5 join to form a ring with the nitrogen to which they are attached, the rings formed are 1-pyrrolidinyl, 1-piperidinyl and 1-piperazinyl, wherein the piperazinyl ring may also be substituted at the 4-position nitrogen by a group R7.

"hydroxy (C1-C6) alkyl" refers to an alkyl chain substituted by two hydroxy groups on two different carbon atoms.

"Aryl"means phenyl, naphthyl, indenyl, tetrahydronaphthyl or indanyl.

"Heteroaryl"means a single ring or benzofused heteroaromatic group of 5 to 10 atoms comprised of 2 to 9 carbon atoms and 1 to 4 heteroatoms independently selected from the group consisting of N, O and S, provided that the rings do not include adjacent oxygen and/or sulfur atoms. N-oxides of the ring nitrogens are also included, as well as compounds wherein a ring nitrogen is substituted by a C1-C4 alkyl group to form a quaternary amine. Examples of single-ring heteroaryl groups are pyridyl, oxazolyl, isoxazolyl, oxadiazolyl, furanyl, pyrrolyl, thienyl, imidazolyl, <BR> <BR> <BR> pyrazolyl, tetrazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyrazinyl, pyrimidyl, pyridazinyl and triazolyl. Examples of benzofused heteroaryl groups are indolyl, quinolyl, isoquinolyl, phthalazinyl, benzothienyl (i. e., thionaphthenyl), benzimidazolyl, benzofuranyl, benzoxazolyl and benzofurazanyl. All positional isomers are contemplated, e. g., 2-pyridyl, 3-pyridyl and 4-pyridyl. W-substituted heteroaryl refers to such groups wherein substitutable ring carbon atoms have a substituent as defined above, or where adjacent carbon atoms form a ring with an alkylen group or a methylenedioxy group, or where a nitrogen in the Het ring can be substituted with R2-aryl or an optionally substituted alkyl substituent as defined in W.

The term"Het"is exemplified by the single ring, the ring substituted with another ring (which can be the same or different), benzofused heteroaryl groups as defined immediately above, as well as tricyclic groups such as benzoquinolinyl (e. g., 1,4 or 7,8) or phenanthrolinyl (e. g., 1,7 ; 1,10 ; or 4,7). Het groups are joined to group B by a carbon ring member, e. g., Het is 2-pyridyl, 3-pyridyl or 2-quinolyl.

Examples of heteroaryl groups wherein adjacent carbon atoms form a ring with an alkylen group are 2, 3-cyclopentenopyridine, 2, 3-cyclohexenopyridine and 2,3- cycloheptenopyridine.

The term"optional double bond"refers to the bond shown by the single dotted line in the middle ring of the structure shown for formula 1. The term"optional single bond"refers to the bond shown by the double dotted line between X and the carbon to which Y and R15 are attached in the structure of formula 1.

The above statements, wherein, for example, R4 and R5 are said to be independently selected from a group of substituents, means that R4 and R5 are independently selected, but also that where an R4 or R5 variable occurs more than once in a molecule, those occurrences are independently selected. Those skilled in

the art will recognize that the size and nature of the substituent (s) will affect the number of substituents which can be present.

It should also be noted that any formula, compound, moiety or chemical illustration with unsatisfied valences in the present specification and/or claims herein is assumed to have sufficient hydrogen atom (s) to satisfy the valences.

Compounds of the invention have at least one asymmetrical carbon atom and therefore all isomers, including diastereomers and rotational isomers are contemplated as being part of this invention. The invention includes (+)-and (-)- isomers in both pure form and in admixture, including racemic mixtures. Isomers can be prepared using conventional techniques, either by reacting optically pure or optically enriched starting materials or by separating isomers of a compound of formula 1.

"Polymorph"means a crystalline form of a substance that is distinct from another crystalline form but that shares the same chemical formula.

Prodrugs and solvates of the compounds of the invention are also contemplated herein. The term"prodrug", as employed herein, denotes a compound that is a drug precursor which, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield a compound of formula I or a salt and/or solvate thereof (e. g. , a prodrug on being brought to the physiological pH or through enzyme action is converted to the desired drug form). A discussion of prodrugs is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems (1987) Volume 14 of the A. C. S. Symposium Series, and in Bioreversible Carriers in Drug Design, (1987) Edward B. Roche, ed. , American Pharmaceutical Association and Pergamon Press, both of which are incorporated herein by reference thereto.

"Solvate"means a physical association of a compound of this invention with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid."Solvate"encompasses both solution-phase and isolatable solvates. Non-limiting examples of suitable solvates include ethanolates, methanolates, and the like."Hydrate"is a solvate wherein the solvent molecule is H20.

Compounds of the invention with a carboxylic acid group can form pharmaceutical acceptable esters with an alcohol. Examples of suitable alcohols include methanol and ethanol.

Abbreviations which are used in the preparative exampies, schemes and examples include the following : DBAD: Di-tert-butyl azodicarboxyfate DBU: 1, 8-Diazabicyclo [5.4. 0] undec-7-ene DCC: Dicyclohexylcarbodiimide DCM: Dichloromethane DIBAL : Diisobutylaluminum hydride DMAP: 4-Dimethyl aminopyridine DMF : NN-Dimethylformamide DMSO: Methyl sulfoxide EDCI : 1- (3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride HMPA : Hexamethylphosphoramide HOBt : Hydroxybezotriazole LAH: Lithium aluminum hydride LHMDS: Lithium bis (trimethylsilyl) amide NMO: 4-Methylmorphine N-oxide TBAF: Tetrabutylammonium fluoride TFA: Trifluoroacetic acid THF: Tetrahydrofuran TMSI : Trimethylsilyl iodide TPAP: Tetrapropylammonium perruthenate Typical preferred compounds of the present invention have the following stereochemistry: with compounds having that absolute stereochemistry being more preferred.

Those skilled in the art will appreciate that for some compounds of formula 1, one isomer will show greater pharmacological activity than other isomers.

Compounds of the invention with a basic group can form pharmaceutically acceptable salts with organic and inorganic acids. Examples of suitable acids for salt formation are hydrochloric, sulfuric, phosphoric, acetic, citric, oxalic, malonic, salicylic, malic, fumaric, succinic, ascorbic, maleic, methanesulfonic and other mineral and carboxylic acids well known to those in the art. The salt is prepared by contacting the

free base form with a sufficient amount of the desired acid to produce a salt. The free base form may be regenerated by treating the salt with a suitable dilute aqueous base solution such as dilute aqueous sodium bicarbonate. The free base form differs from its respective salt form somewhat in certain physical properties, such as solubility in polar solvents, but the salt is otherwise equivalent to its respective free base forms for purposes of the invention.

Certain compounds of the invention are acidic (e. g., those compounds which possess a carboxyl group). These compounds form pharmaceutical acceptable salts with inorganic and organic bases. Examples of such salts are the sodium, potassium, calcium, aluminum, lithium, gold and silver salts. Also included are salts formed with pharmaceutically acceptable amines such as ammonia, alkyl amines, hydroxyalkylamines, N-methylglucamine and the like. Bisulfate salts of the compounds of the invention are preferred embodiments.

Compounds of the present invention are generally prepared by processes known in the art, for example by the processes described below.

Compounds of formula IA, wherein n is 1, the optional double bond is not present, the single bond is present between X and the carbon to which Y is attached, X is-O-, Y is =O, B is-CH=CH-, Het is W-substituted pyridyl, R3, R8, R9, Rio and R are each hydrogen, and R'and R2 are as defined above can be prepared by condensing an aldehyde of formula 11, wherein the variables are as defined above, with a phosphonate of formula 111, wherein W is as defined above: Similar processes may be used to prepare compounds comprising other optionally substituted Het groups. Those skilled in the art will also recognize that the processes are equally applicable to preparing optically active or racemic compounds.

Compounds of formula IA can be converted to the corresponding compounds wherein R3 is OH by treatment with Davis reagent ( S)-(+)-(1 0-camphorsulfonyl)- oxaziridine) and LHMDS (Lithium bis (trimethylsilyl) amide).

Aldehydes of formula 11 can be prepared from dienoic acids, for example compounds of formula Ila, wherein R1 is methyl and R2 is H can be prepared according to the following reaction scheme.

Scheme 1: 22 0 0 (Et0) 2PCH2CO2Et R22 Et0 NaH, THF R 2 rus 22 KOH) 2, DCM R THF-MeOH-H20 R 3 R23 >, Et3N CO2Bn 5 C02Bn obi OBn H Lindlar cat., H2 O/ (1) Toluene, 185 °C O H R Quinoline, THF R23 2) DBU, rt = R R eCO2Bn mRn237 H H R22 0 H H 22 H H R22 1) Pd-C, H2 1) (COCI) 2, DCM H CH 2) Pt02, H2 23 2) Pd cat., BU3SnH 23 R Ils

The alkyne of formula 4, prepared by known methods, is esterified with the dienoic acid of formula 3 using standard conditions to yield the ester 5. Selective reduction of the triple bond of 5 using Lindlar catalyst under hydrogen gives the intermediate 6, which upon thermal cyclization at about 185°C, followed by base treatment, gives the intermediate 7. The ester 7 is subjected to hydrogenation in the presence of platinum oxide to generate the intermediate saturated carboxylic acid, treatment of which with oxalyl chloride gives the corresponding acid chloride which is converted to the aldehyde Ila by reduction using tributyltin hydride in the presence of Palladium catalyst.

Dienoic acids of formula 3 are commercially available or are readily prepared.

Aldehydes of formula 11 also can be prepared by a thiopyran ring opening, for example compounds of formula Ila as defined above can be prepared according to the following reaction scheme.

Scheme 2: o On /S 1) Toluene, 195 °C S BBr3, DCM p II O H 2) DBU, rt 4C02Bn 02 H H 0 0 H s H2, PtO2, H s 1) (COCI) 2, DCM MeOH-AcOH 2) Pd cat., Bu3SnH O H O H4- H H H H H H 17 0 H H 0 H H 0 H S 1) NaBH4 0 H TPAP, NMO 2) Ra-Ni /H H 18 KOH

The alkyne of formula 4, is reduced to the alkene 13 using Lindlar catalyst under hydrogen. The alkene 13 is esterified with the dienoic acid of formula 12 using standard conditions to yield the ester 14. Thermal cyclization at about 185°C, followed by base treatment, gives the intermediate 15. The ester 15 is converted to the intermediate carboxylic acid, and the double bond is reduced by hydrogenation in the presence of a platinum catalyst. The acid is then treated with oxalyl chloride to obtain the corresponding acid chloride, which is converted to the aldehyde 18 by reduction using tributyltin hydride in the presence of Palladium catalyst. The aldehyde moiety on 18 is treated with a reducing agent such as NaBH4, and the sulfur-containing ring is then opened by treatment with a reagent such as Raney nickel to obtain the alcohol 19. The alcohol is then oxidized to the aldehyde, Ila, using tetrapropylammonium perruthenate (TPAP) in the presence of 4- methylmorpholine N-oxide (NMO).

Phosphonates of formula III wherein W is aryl or R21-aryl can be prepared by a process similar to that described immediately below for preparing the trifluoromethy- phenyl-substituted compound, Illa. B (OH) 2 1 (EtO) 20Pß i N n-BuLi, -- I'I i-Pr2NH N Tf20, Py w CF3 w OH OTf Suzuki n ClPO (OEt) 2 Illa CFg"CFs CF3 Commercially available hydroxypyridine derivative is converted to the corresponding triflate using triflic anhydride, which is then coupled with commercially available boronic acid in the presence of Pd (0) under Suzuki conditions. The resulting product is converted to the phosphonate by treatment with n-butyllithium followed by quenching with diethylchlorophosphonate.

Alternatively, compounds of formula I wherein W is optionally substituted aryl can be prepared from compounds of formula I wherein W is-OH using a triflate intermediate. For example, 3-hydroxy-6-methylpyridine is treated with triisopropylsilyl chloride, and the resultant hydroxy-protected compound is converted to the phosphonate as described above for preparing intermediate Illa. The triisopropylsilyl- protected intermediate is then reacted with intermediate 11 and the protecting group is removed under standard conditions. The resultant compound of formula I wherein W is OH is then treated with triflic anhydride at room temperature in a solvent such as CH2CI2 ; the triflate is then reacted with an optionally substituted arylboronic acid, e. g., optionally substituted phenylboronic acid, in a solvent such as toluene, in the presence of Pd (PPh3) 4 and a base such a K2CO3 at elevated temperatures and under an inert atmosphere.

Compounds of formula I wherein W is a substituted hydroxy group (e. g., benzyloxy) can be prepared from compounds of formula I wherein W is hydroxy by refluxing in a suitable solvent such as acetone with a halogen-substituted compound such as optionally substituted benzyl bromide in the presence of a base such as K2C03.

Compounds of formula I wherein Het is substituted by W through a carbon atom (e. g. , wherein W is alkyl, alkenyl or arylalkyl) or a nitrogen atom (i. e.,-NR4R5) can be prepared as shown in Scheme 3 using a compound of formula I wherein W is chloroalkyl as an intermediate. Compounds of formula I wherein W is a polar group such as hydroxy alkyl, dihydroxyalkyl,-COOH, dimethylamino and-COH can be prepared as shown in Scheme 4, wherein the starting material is a compound of formula I wherein W is alkenyl. The following Schemes 3 and 4 show well-know reaction conditions for preparing various W-substituted compounds wherein X is-O-, Y is =O, R15 is absent, Rl is methyl, R2, R3, R9, R10 and R11 are each H, B is - CH=CH-, and Het is 2-pyridyl.

Scheme 3 '""t PhB (OH) 2, Pd (PPh3) 4 J. K2CO3, toluene 120 °C, 16 h n-Bu (OH) 2, jus Pd (PPh3) 4 r N K2C03, toluene Bu 120 °C, 16 h WSnBu3, Pd (PPh3) 4 W = phenyl ) LR 'LjjL W=viny ! THF, 120 °C, 16 h W W-_ all II R23 ""' W=Et W-i-Bu W = n-Pr W = n-Hex N Pd (PPh3) 4, WZnHaI N W-i_pr W = benzyl Cl THF, 120 °C W W = n-Bu m ffi ffi amines N neat, 190C N NMe NH NH if , IVA 6 NH3/CH30H N heat NH2 Scheme 4 Rua p Os04, NMO, Na104 R'100% 0 OH N OH Oh zizi H2°2 ! T HsQs f i N a i N H i N 100% 1 0 C02H OH Me2NH, ulli NaCNBH3 CN 34% Ors04, NMO t N MS N OH NMe2 \ N OH OU

Those skilled in the art will appreciate that similar reactions to those described in the above schemes may be carried out on other compounds of formula I as long as substituents present would not be susceptible to the reaction conditions described.

Compounds of formula I wherein the optional single bond (represented by the double dotted line) is absent, X is OH, Y is OH, R15 is H and the remaining variables are as defined above can be prepared by treating corresponding compounds wherein

the optional single bond is present, X is-O-, Y is =O and R15 is absent, with a reducing agent such as LAH.

Compounds of formula I wherein the optional single bond is present, X is-O-, Y is (H, OH), R5 is absent and the remaining variables are as defined above can be prepared by treating corresponding compounds wherein the optional single bond is present, X is-O-, Y is =O and R15 is absent, with a reagent such as DIBAL. The resultant compounds wherein Y is (H, OH) can be converted to the corresponding compounds wherein Y is (H, alkoxy) by reacting the hydroxy compound with an appropriate alkanol in the presence of a reagent such as BF3OEt2. A compound wherein Y is (H, OH) can also be converted to the corresponding compound wherein Y is (H, H) by treating the hydroxy compound with BF3-OEt2 and Et3SiH in an inert solvent such as CH2CI2 at low temperatures.

Compounds of formula I wherein R9 is hydrogen can be converted to the corresponding compound wherein R9 is hydroxy by heating with an oxidizing agent such as Se02.

Compounds of formula IB, wherein R2 is H, R3 is H or OH, and W'is R21-aryl, R41-heteroaryl, amino or hydroxylamino derivatives, are prepared from compounds of formula 1A wherein W is 5-bromo (compounds of formula 23 or 24) using a variety of standard chemical transformations, e. g. the Suzuki reaction, Stille coupling, and Buchwald amination. Reaction Scheme 5 shows the process from the 2,5- dibromopyridine: Scheme 5 : Br OH 0 OEt H H 1) BuLi 1) MsCl, Et3N P, OEt R, 2) DMF N 2) HP (O) (OEt) 2. N = 3) NaBH4 NaH 2) aldehyde Ila 20 r 21 Br 22 g 23 ßN zu ° OH H R Suzuki, Stille coupling, Br 1) LHMDS H Buchwald amination etc.. 2) Davis reagent 24 N 0 H-R R3 H, OH 24 N H R3 = H, OH H H 2 Br IB wl The phosphonate 22 is prepared from the known alcohol 21 by a two step transformation: the alcohol is treated with CH3SO2CI to provide the mesylate, which is then displaced with sodium diethylphosphite to provide 22. Intermediate 23 can also be a-hydroxylated using Davis reagent to provide alcohol 24. Both 23 and 24 can be converted into diverse analogs as shown in Scheme 6: Scheme 6: w' Method 1 1-i) ArB (OH) 2, Pd (PPh3) 4 I 2) subsequent transformation Ar Method 2 ArSnBu3, Pd (PPh3) 4 Ar ArZnCI, Pd (PPh3) 4 Method 3 Ar 1) NNR4R5, Pd cat. +, R22 Method 4 R4, No 5 H 2) subsequent transformation o" H H 1) CH2=CHOPr, Pd cat., N Method 5 then HCI i NOR 1 2) subsequent transformation Bu (CuOTf) 2. PhH, Imidazoles N. Ra1 23 or 24 Method 6 R = H/OH 1) Zn (CN) 2, Pdcat. ! Method 7 N'-NH 2) NaN3, NH4CI N=N

As shown in Scheme 6, the bromide (23 or 24) can be coupled with boronic acids under palladium catalysis condition (method 1). If the boronic acid possesses a functional group, it can be subsequently transformed. Similarly, aryl-tin compounds (method 2), aryl-zinc compounds (method 3) and amines (method 4) can be coupled.

Heck reaction with vinyl ethers can introduce a keto-group, which can be subsequently functionalized (method 5). Imidazoles can be coupled using Copper (l) triflate as catalyst (method 6). The bromide can also be converted to a cyanide which can be subsequently transformed, for example to a tetrazol (method 7).

Using a Diels-Alder strategy as shown in Scheme 7, a variety of dienoic acids 3 can be coupled with alcohol 25 and the ester 26 can be subjected to thermal cyclization to provide the Diels-Alder product IC : Scheme 7: 0 R22 1) Acid, (COC !) 2 22 O HA 3 23 2 Alcohol, Et3N,/R o H R22 DMAP p 1) xylene, 185 C w R R23 2) DBU, rt Acid, Alcohol, H Fi Hot DCC. DMAP 1 Het IC Het 25 Alcohol 25 is prepared as follows : TBDPSO TBDPSO OH TgDPSCI 1) BuLi Et3N, DMAP 2 1 H2 Lindlar cat. Et, Et3N, DMAP 2) (CH20) , 2) Dess-Martin reagent 27 28 OH 29 OH 1) HetCH2P (O) (OEt) 2, BuLi HO (2) TBAF 25 Het 130 25

Alcohol 25 is prepared from the readily available (R)- (+)-3-butyn-2-ol 27. The alcohol is protected as its TBDPS ether, the alkyne is deprotonated and quenched with paraformaldehyde to provide alcohol 29. The alkyne is reduced to cis-alkene using Lindlar catalyst in presence of quinoline and the allylic alcohol was oxidized to provide the aldehyde 30, which is converted to the alcohol 25.

Compounds of formula ID wherein R22 is-CH20C (O) CH3 or a derivative thereof, R23 is ethyl, R2 is H and the remaining variables are as defined for IA can be prepared from the corresponding tetrahydropyran analog by opening the ring. The compounds of formula ID can be converted to other compounds of formula 1, e. g. compounds of formula IE wherein R22 is-CH2OH, by well known methods. The reaction is shown in Scheme 8: Scheme 8 : 0 H H schema 1 p Ouzo H H H H 31 N 9 bzw

Tetrahydropyran analog 31 can be prepared starting from 3-formyl-5, 6-dihydro- 2H-pyran (known compound) and using the similar procedure used in Scheme 1. The ring can opened regioselectively using BBr3 and the alcohol can be protected to give the acetate ID. Bromide reduction with NaCNBH3, followed by acetate deprotection, furnishes alcohol IE.

Starting materials for the above processes are either commercially available, known in the art, or prepared by procedures well known in the art.

Reactive groups not involved in the above processes can be protected during the reactions with conventional protecting groups which can be removed by standard procedures after the reaction. The following Table A shows some typical protecting groups: Table A Group to be Group to be Protected and Protected Protecting Group -COOH-COOalkyl,-COObenzyl,-COOphenyl \ NH/NCOalkyl gNCObenzyl, _ NCOphenyl, nu NCH 20CH 2CH 2Si (CH 3) 3 NC (O) OC (CH 3) 3, CI 3 N-benzyl,/NSi (CH 3) 3, SI C (CH) 3 0 CH3 - NH2-N 0 ICH3 - OH-OCH 3,-OCH 20CH 3,-OSi (CH 3) 3-- OSi-C (CH) 3 or-OCH2phenyl

The present invention also relates to a pharmaceutical composition comprising a compound of formula I of this invention and a pharmaceutical acceptable carrier.

The compounds of formula I can be administered in any conventional oral dosage form such as capsules, tablets, powders, cachets, suspensions or solutions. The

formulations and pharmaceutical compositions can be prepared using conventional pharmaceutical acceptable excipients and additives and conventional techniques.

Such pharmaceutical acceptable excipients and additives include non-toxic compatible fillers, binders, disintegrants, buffers, preservatives, anti-oxidants, lubricants, flavorings, thickeners, coloring agents, emulsifiers and the like.

The daily dose of a compound of formula I for treatment of a disease or condition cited above is about 0.001 to about 100 mg/kg of body weight per day, preferably about 0.001 to about 10 mg/kg. For an average body weight of 70 kg, the dosage level is therefore from about 0.1 to about 700 mg of drug per day, given in a single dose or 2-4 divided doses. The exact dose, however, is determined by the attending clinician and is dependent on the potency of the compound administered, the age, weight, condition and response of the patient.

Following are examples of preparing starting materials and compounds of formula 1. In the procedures, the following abbreviations are used: room temperature (rt), tetrahydrofuran (THF), ethyl ether (Et2O), methyl (Me), ethyl (Et), ethyl acetate (EtOAc), dimethylformamide (DMF), 4-dimethylaminopyridine (DMAP), 1,8- diazabicyclo [5.4. 0] undec-7-ene (DBU), 1, 3-dicyclohexylcarbodiimide.

Preparation 1 Step 1 : See J. Org. Chem., 59 (17) (1994), p. 4789.

Step 2: To a suspension of 60% NaH (7.42 g, 185.5 mmol, 1.3 eq) in 300 mi THF at 0°C was added dropwise triethylphosphono acetate (37 ml, 186.5 mmol, 1.3 eq) and the mixture was stirred at 0°C for 30 min. The product of Step 1 (14.0 g, 142.7 mmol) was added and the mixture was stirred at 0°C for 30 min. The reaction was quenched by the addition of aq. NH4CI (500 ml), the THF was evaporated and the aqueous phase was extracted with 3x200 ml of Et20, the combined organic layer was washed with brine (300 ml), dried over MgS04, filtered and evaporated to give the crude mixture which was chromatographed (5% Et20-hexane) to give 18.38 g (77% yield) of liquid.

1H NMR (400 MHz, CD13) 7.29 (d, 1 H, J = 15.4), 5.86 (t, 1 H, J = 7.4), 5.76 (d, 1 H, J = 15. 4), 4.18 (q, 2H, J = 7.2), 2.22-2. 15 (m, 2H), 1.74 (d, 3H, J = 0. 7), 1.27 (t, 3H, J = 7. 2), 1.00 (t, 3H, J = 7. 7).

'3C NMR (100 MHz, CDC13) 167. 29,149. 38,143. 45,132. 04,115. 39,60. 08,22. 14, 14.42, 13.58, 12.05.

MS: 169 (MH+).

Step 3: To a solution of the product of Step 2 (6. 4 g, 38 mmol) in THF and MeOH (40 mi each) was added a solution of KOH (6. 4 g, 114 mmol, 3 eq) in H20 (40 ml). The mixture was stirred at rt for 2 h, cooled to 0°C and H20 (100 ml) and 1 N HCI (150 ml) were added. The mixture was extracted with EtOAc (3x100 ml), the combined organic layer was washed with H20 (150 ml) and brine (150 ml), dried over MgS04, filtered and evaporated to give 5.26 g (99% yield) of crystalline solid. oh NMR (400 MHz, CDCl3) 7.40 (d, 1H, J = 16), 5.95 (t, 1H, J = 7.2), 5.79 (d, 1H, J = 16), 2.26-2. 19 (m, 2H), 1.78 (s, 3H), 1.04 (t, 3H, J = 7.6).

Step 4: To a solution of the product of Step 3 (2.0 g, 14.3 mmol) in CH2CI2 (70 ml) was added oxalyl chloride (2.5 ml, 28.7 mmol, 2 eq.) followed by DMF (33 µl, 3 mol%.).

The mixture was stirred at rt for 1 h, then the solvent was evaporated to give the crude acid chloride which was dissolved in CH2C12 (70 ml) and cooled to 0°C. To this was added DMAP (175 mg, 1.43 mmol, 0.1 eq. ) and a solution of alcohol 4 (2.62 g, 12.8 mmol, 0.9 eq. ) in CH2Cl2 (5 ml) followed by Et2N (4 mi, 28.7 mmol, 2 eq. ). The mixture was stirred at 0°C for 2 h, diluted with Et20 (200 ml), washed with aq.

NaHC03 and brine (200 ml each), and dried over MgS04. The solution was filtered, concentrated and the resultant residue was chromatographed with 5% EtOAc-hexane to provide 3.56 g (85%) of pale-yellow resin.

1H NMR (400 MHz, CDCl3) 7.38-7. 33 (m, 6H), 5.93 (t, 1H, J = 7.4), 5.77 (d, 1H, J = 15.6), 5.62 (q, 1H, J = 6. 2), 5.20 (s, 2H), 2.25-2. 18 (m, 2H), 1.76 (d, 3H, J = 0. 4), 1. 58 (d, 3H, J = 6. 2), 1.03 (t, 3H, J = 7. 4).

Step 5:

To a solution of the product of Step 4 (3.19 g, 9.8 mmol) in THF (50 ml) was added Lindlar catalyst (320 mg, 10 wt%) and quinoline (230 ßI, 2.0 mmol, 0.2 eq. ).

The suspension was stirred under 1 atm. H2 until the starting material was consumed.

The solution was filtered through celte and evaporated. The resin was dissolved in EtOAc (250 ml) and washed with 1 N HCI (3x100 ml) and brine (100 ml). The solution was dried over MgS04, filtered and evaporated to give 3.17 g of crude alkene which was used directly in the next step.

Step 6: A solution of the product of Step 5 (3. 15 g, 9.6 mmol) in m-xylene (100 ml) was heated at 185°C for 10 h. The solution was cooled to rt and stirred for 1 hour with DBU (290, u1, 1.94 mmol, 0.2 eq. ). The solvent was evaporated and the crude was chromatographed with 10% EtOAc-hexane to provide 1.1 g (35%) of exo product.

'H NMR (400 MHz, CDC13) 7.38-7. 34 (m, 5H), 5.45 (br s, 1H), 5.14 (ABq, J = 12.0, 22.8, 2H), 4.52 (dq, J = 6.1, 8.1, 1 H), 3.26-3. 23 (m, 1 H), 2.87 (dd, J = 9.4, 4.6, 1 H), 2.62 (dt, J = 8.1, 4.5, 1 H), 2.54 (br s, 1 H), 1.71 (t, J = 1.2, 3H), 1.69-1. 60 (m, 1 H), 1.50-1. 44 (m, 1 H), 1.20 (d, J = 6.4, 3H), 0.77 (t, J = 7.4, 3H).

'3C NMR (100 MHz, CDCl3) 175.25, 173.04, 137.86, 135.00, 128.38, 128.34, 128.30, 116.54, 76.64, 66.70, 42.85, 42.14, 41.40, 37.27, 22.52, 21.65, 20.44, 8.98 [&alpha;] 22D =-64. 4 (c 1, CH2CI2).

HRMS: 329.1754, calculated 329.1753.

Step 7: To a solution of the product of Step 6 (1.35 g, 4.1 mmol) in EtOAc (30 ml) was added 10% Pd-C (140 mg, 10 wt%) and the suspension was stirred under H2 balloon for 5 h. The mixture was filtered through celte and concentrated. The crude material was dissolved in MeOH (30 ml), Pt02 (100mg) was added and the mixture

was shaken in a Parr vessel at 50 Psi H2 for 2 days. The mixture was filtered through celiteTM and evaporated to give 980 mg (99%) of the acid as foam.

'H NMR (400 MHz, CDCl3) 4.73-4. 66 (m, 1 H), 2.71 (dd, J = 11.8, 5.4, 1 H), 2.68-2. 62 (m, 1H), 2.53 (dt, J = 10.0, 6.4, 1H), 1.92, ddd, J = 13.4, 6.0, 2.6, 1H), 1.63-1. 57 (m, 1 H), 1.52-1. 20 (unresolved m, 3H), 1.30 (d, J = 5.9, 3H), 0.96 (d, J = 6.6, 3H), 0.93- 0.89 (m, 1 H), 0.80 (t, J = 7.5, 3H).

MS: 319.1 (MH+. DMSO).

Step 8: To a solution of the product of Step 7 (490 mg, 2.04 mmol) in CH2CI2 (20 ml) was added oxalyl chloride (360, ul, 4.13 mmol, 2 eq.) followed by 1 drop of DMF. The solution was stirred at rt for 1 hour and the solvent was removed to provide the crude acid chloride, which was dissolved in toluene (20 ml) and cooled to 0°C. To this was added Pd (PPh3) 4 (236 mg, 0.20 mmol, 0.1 eq. ) followed by Bu3SnH (825 µl, 3.07 mmol, 1.5 eq. ). The mixture was stirred for 3 hours at 0°C, concentrated and chromatographed with 25% EtOAc-hexane to provide the title compound 220 mg (48%) as a resin.

1H NMR (400 MHz, CDC13) 9.72 (d, J = 3. 6, 1 H), 4.70 (dq, J = 5. 7,9. 5, 1 H), 2.71-2. 64 (m, 2H), 2.56-2. 51 (m, 1H), 1.98 (ddd, J = 13.5, 6.1, 2.9, 1H), 1.68-1. 59 (m, 3H), 1.52-1. 37 (m, 1 H), 1.36 (d, J = 5.9, 3H), 1.32-1. 20 (m, 1 H), 1.00 (d, J = 6.2, 3H), 0.80 (d, J = 7. 3,3H).

Preparation 2 Step 1: The thiopyran enai was prepared according to the procedure of McGinnis and Robinson, J. Chem. Soc. , 404 (1941), 407.

Step 2: To a suspension of 60% NaH (6.3 g, 158 mmol, 1.3 eq. ) in THF (200 ml) at 0°C was added methyl diethylphosphonoacetate (29 ml, 158 mmol, 1.3 eq. ) and the

mixture was stirred at 0°C for 30 min. The solution was then transferred to a solution of the product of Step 1 (15.6 g, 122 mmol) in THF (100 ml) and stirred at 0°C for 1 h.

The reaction was quenched by the addition of aq. NH4CI (500 ml) and the THF was evaporated. The aqueous phase was extracted with Et20 (3x200 mi) and the combined organic layer was washed with H20 and brine (200 mi each). The solution was dried over MgS04, concentrated and the resultant residue was chromatographed with 5% EtOAc-hexane to provide 13.0 g (58%) of oil.'H NMR (400 MHz, CDCI3) 7.26 (d, J = 15.9 Hz, 1H), 6.26 (t, J =4. 4 Hz, 1H), 5.78 (dd, J = 15.9, 0.6 Hz, 1H), 3.75 (s, 3H), 3.25-3. 23 (m, 2H), 2.71 (t, J = 5.8 Hz, 2H), 2.57-2. 53 (m, 2H).

Step 3: To a solution of the product of Step 2 (13.0 g, 70.6 mmol) in THF and MeOH (50 ml each) was added a solution of KOH (11.9 g, 212 mmol, 3.0 eq. ) in H20 (50 ml).

The mixture was stirred at rt for 1 h, diluted with H20 (100 mi) and acidified with 1 N HCI. The aqueous phase was extracted with EtOAc (3x200 ml) and the combined organic layer was washed with H20 and brine (300 ml each). The solution was dried over MgS04, filtered and evaporated to give 11.66 g (97%) of pale-yellow solid.'H NMR (400 MHz, CDCI3) 7.34 (d, J = 15.6 Hz, 1 H), 6.32 (t, J = 4. 4 Hz, 1 H), 5.78 (d, J = 15.6 Hz, 1 H), 3.26 (d, J = 1.6 Hz, 2H), 2.72 (t, J = 5. 8 Hz, 2H), 2.59-2. 55 (m, 2H). Step 4 : Step H2, Lindlarcat./==\ HO- COBn EtOAc HO-- (C02Bn 4 To a solution of 4 (5.2 g) in EtOAc (120 ml) was added Lindlar catalyst (520 mg) and the suspension was stirred under 1 atm. H2. Another portion of catalyst (500 mg) was added after 45 min. and the mixture stirred for further 30 min. The mixture was filtered through a celiteTM pad and evaporated to provide 5.2 g (99%) of the desired alkene. 1H NMR (400 MHz, Ceci3) 7.38-7. 26 (m, 5H), 6.32 (dd, J = 11.9, 6.6 Hz, 1H), 5.86 (d, J = 12.0 Hz, 1H), 5.18 (s, 2H), 5.12-5. 07 (m, 1H), 3.20 (brs, 1H), 1.34 (d, J = 6. 6 Hz, 3H).

Step 5:

To a solution of the product of Step 3 (2.45 g, 14.39 mmol) in CH2C12 (60 ml) at 0°C was added DCC (3. 27 g, 15.85 mmol, 1.1 eq. ) followed by DMAP (352 mg, 2.88 mmol, 0.2 eq. ) and the mixture was stirred at 0°C for 30 min. To this was added a solution of 3.27 g (15.85 mmol, 1.1 eq.) of the alcohol of Step 4 in 10 ml of CH2CI2 and the mixture was stirred at 0 °C for 5 hr and at rt for 1 hr. The solution was diluted with 350 mlof Et20 and washed with 2x200 ml of aq. citric acid, 200 ml of aq.

NaHCO3 and 200 ml of brine. The solution was dried over MgS04, filtered, concentrated and the resultant residue was chromatographed with 6% EtOAc-hex to provide 2.1 g (41%) of resin.'H NMR (400 MHz, CDCI3) 7.38-7. 32 (m, 5H), 7.45 (d, J = 16.0 Hz, 1 H), 6.38-6. 34 (m, 1 H), 6.26 (t, J = 4.6 Hz, 1 H), 6.21 (d, J = 11.6 Hz, 1 H), 6.19 (d, J = 11.2 Hz, 1H), 5. 85 (dd, J = 11.6, 1.2 Hz, 1H), 5.76 (d, J = 16.0 Hz, 1H), 5.18 (d, J = 1. 2 Hz, 2H), 3. 24 (d, J = 2. 0 Hz, 2H), 2.71 (t, 2H, J = 5.6 Hz, 2H), 2.56- 2.52 (m, 2H), 1.41 (d, J = 6.4 Hz, 3H).

Step 6: A solution of the product of Step 5 (2.1 g, 5.85 mmol) in xylène (50 ml) was heated at 200°C for 6 hours in a sealed tube. The solution was cooled to rt and stirred with DBU (178 il, 1.19 mmol, 0.2 eq. ) for 1 h, concentrated and chromatographed with 15% EtOAc-hexane to provide 1. 44 g (69%) of the desired exo product NMR (400 MHz, CDCl3) 7.39-7. 35 (m, 5H), 5.46 (br s, 1 H), 5.16 (ABq, J = 21.6, 12.0 Hz, 2H), 4.42 (dq, J = 9.2, 6.0 Hz, 1 H), 3. 36-3. 33 (m 2H), 3.08 (dd, J = 14.4, 2.4 Hz, 1H), 2.85 (ddd, J = 13.9, 12.4, 2.5 Hz, 1H), 2.72-2. 57 (m, 4H), 2.27-2. 21 (m, 1H), 1.47-1. 25 (m, 1H), 1.12 (d, J = 6. 4 Hz, 3H).

Step 7: To a solution of the product of Step 6 (750 mg, 2.09 mmol) in CH2CI2 (10 ml) at -78°C was added BBr3 in CH2CI2 (4.2 ml of 1 M solution). The solution was stirred at- 78°C for 30 min. and at 0 °C for 30 min, then poured into aq. K2CO3 (100 ml). The aqueous phase washed with Et20 (2x50 ml) and the organic layer was back extracted with aq. K2CO3 (50 ml). The combined aqueous phase was acidified with 1 N HCI and extracted with EtOAc (3x50 ml). The EtOAc layer was washed with brine (50 ml), dried over MgS04, filtered and evaporated to provide 500 mg (89%) of acid.'H NMR

(400 MHz, CDCI3) 5.50 (br s, 1 H), 4.47 (dq, J = 9.6, 6.0 Hz, 1 H), 3.43-3. 39 (m, 1 H), 3.36 (d, J = 15.6 Hz, 1H), 3.10 (dd, J= 14.0, 2.4 Hz, 1H), 2.91-2. 84 (m, 1H), 2.82-2. 77 (m, 1 H), 2.70 (dd, J = 10. 6,4. 2 Hz, 1 H), 2.69-2. 63 (m, 1 H), 2.57-2. 52 (m, 1 H), 2.34- 2.29 (m, 1 H), 1.53-1. 42 (m, 1 H), 1.34 (d, J = 6.0 Hz, 3H).

Step 8: To a solution of the product of Step 7 (500 mg, 1.86 mmol) in MeOH (30 ml) was added AcOH (3 ml) and Pt02 (250 mg) and the suspension was shaken under 40 Psi H2 in a Parr vessel for 1.5 days. The catalyst was filtered off with a Celite pad, the solution was concentrated and the resultant residue was dissolved in AcOH- MeOH-CH2CI2 mixture (0.5 : 2: 97.5 vivivl) and filtered through a short Si02 column to provide 400 mg (79%) of the reduced product as a resin which solidified on standing.

1H NMR (400 MHz, CDC13) 4.68 (dq, J = 9.4, 5.9 Hz, 1H), 2.76-2. 69 (m, 2H), 2.60- 2.55 (m, 3H), 2.49 (d, J = 11.6 Hz, 1H), 2.10 (brs, 1H), 1.93 (ddd, J = 13.5, 6.0, 2.7 Hz, 1 H), 1.60-1. 48 (m, 2H), 1.45-1. 19 (m, 3H), 1.33 (d, J = 5.6 Hz, 3H).

Step 9: To a solution of the product of Step 8 (97 mg, 0.36 mmol) in CH2CI2 (4 ml) was added oxalyl chloride (94 lli) followed by 1 drop of DMF. The solution was stirred for 1 hour at rt and concentrated to provide the crude acid chloride which was dissolved in toluene (3 ml) and cooled to 0°C. Pd (PPh3) 4 (42 mg, 0.04 mmol, 0.1 eq. ) was added, followed by Bu3SnH (94 ZI). The mixture was stirred at 0° C for 3 h, concentrated and chromatographed with 25% EtOAc-hexane to provide 73 mg (80%) of aldehyde as white solid. 1H NMR (400 MHz, CDCI3) 9.75 (d, J = 2.8 Hz, 1 H), 4.62 (dq, J = 9.7, 6.0 Hz, 1 H), 2.8-2. 70 (m, 2H), 2.65-2. 55 (m, 3H), 2.50 (d, J = 7.2 Hz), 2.10 (ddd, J = 13.2, 6.4, 3.0 Hz, 1H), 1.94 (ddd, J = 13. 6,6. 0,3. 0, 1H), 1.69 (dq, J = 10.9 Hz, 3.00 Hz, 1 H), 1.58-1. 48 (m, 1 H), 1.42-1. 20 (m, 3H), 1.33 (d, J = 6. 4 Hz, 3H).

Step 10: To a solution of the product of Step 9 (90 mg, 0.35 mmol) in MeOH (10 ml) (4: 1 v/v) at 0 °C, excess NaBH4 was added and the mixture stirred for 15 min at 0°C.

The reaction was quenched with aq. NH4CI (50 ml) and extracted with EtOAc (3x20 ml). The combined organic layer was washed with brine (50 ml), dried over MgS04 and concentrated to provide the crude alcohol. A solution of the alcohol in MeOH- THF (6 ml, 1: 1 v/v) was added to a flask containing excess Raney nickel which was washed with dioxane and THF. The suspension was heated at reflux for 3 h, cooled, filtered, concentrated and chromatographed with 25% EtOAc-hex to provide 54 mg (67%) of title compound as a resin. 1H NMR (400 MHz, CDC13) 4.70 (dq, J = 9.7, 5.9 Hz, 1 H), 3.73 (dd, J = 10. 5, 3.4 Hz, 1 H), 3.62 (dd, J = 10.5, 7.6 Hz, 1 H), 2.60-2. 53 (m, 1 H), 2.46 (ddd, J = 9. 6,7. 2,5. 2 Hz, 1 H), 1.90 (ddd, J = 13. 5,6. 1,3. 1 Hz, 1 H), 1.87- 1.81 (m, 1 H), 1.77 (br s, 1 H), 1.66-1. 59 (m, 1 H), 1.50 (d, J = 6.0 Hz, 3H), 1.48-1. 36 (m, 2H), 1.25-1. 14 (m, 2H), 0.93 (d, J = 6.6 Hz, 3H), 0.78 (d, J = 7.5 Hz, 3H). toc NMR (100 MHz, CDCI3) 178.58, 77.63, 61.79, 45.10, 42.49, 39.37, 38.65, 33.44, 31. 96, 21.39, 19.91, 19.74, 7.26. Preparation 3 0 'I-OET Et I N Br Step 1 : Br OH ('N i i Br Br Prepared according to the procedure described in Wang et al., Tet. Lett, 41, (2000), p. 4335-4338.

Step 2: To a solution of the product of Step 1 (20 g, 106 mmol) and Et3N (17.8 mi, 128 mmol, 1.2 eq. ) in CH2CI2 (300 mi) kept-30°C was slowly added CH3SO2CI (9.1 ml, 118 mmol, 1.1 eq. ). The slurry was stirred for 1 hour while it warmed up to 0 °C. The reaction mixture was diluted with aq. NaHC03 (500 ml) and the organic layer was separated. The aqueous layer was extracted with Et20 (2x200 mi) and the combined organic layers were washed with aq. NaHC03 (2x300 ml) and brine (300 ml). The solution was dried over MgS04, filtered and evaporated to give the crude mesylate, which was used as such for the next step.

H NMR: 8.67 (d, J = 2.0 Hz, 1 H), 7.89 (dd, J = 8.4, 2.4 Hz, 1 H), 7.33 (d, J = 8.4 Hz, 1 H), 5.28 (s, 2H), 3.10 (s, 3H).

Step 3: To a suspension of 60% NaH (8.5 g, 212 mmol 2.0 eq. ) in THF (500 ml) at rt was added diethylphosphite (27.4 ml, 213 mmol, 2 eq, ) drop by drop and the mixture was stirred for 1 h. To this cloudy solution was added a solution of the product of Step 2 in THF (125 ml) and the mixture was stirred at rt for 1 h. The reaction was quenched by the addition of H20 (500 ml), the THF was evaporated and the aq. layer was extracted with EtOAc (4x150 mi). The combined organic layers were washed with aq. K2CO3 (2x300 ml), brine (300 ml), dried over MgS04, filtered, evaporated and the crude product was chromatographed with 5: 95 CH3OH-CH2C12 to give 31.7 g (97%) of oil.

H NMR: 8.59 (d, J = 2.0 Hz, 1 H), 7.76 (dd, J = 8.2, 2.1 Hz, 1 H), 7.29 (dd, J = 8.2, 2.2 Hz, 1 H), 4.12-4. 05 (m, 4 H), 3.36 (d, J = 22.0 Hz, 2H), 1.27 (t, J = 7.0 Hz, 6H). Preparation 4 H H OH H H H N N Br To a solution of the product of Preparation 3 (15 g, 49 mmol, 1.5 eq. ) in THF (100 ml) at 0 °C was added 1 M LHMDS in THF (49 ml, 49 mmol, 1.5 eq. ) and the solution was stirred for 30 min. To this was added Ti (O'Pr) 4 (14.4 ml, 49 mmol, 1.5 eq. ) followed by a solution of the product of Preparation 1 (7.3 g, 32 mmol) in THF (30 ml) and the mixture was stirred at rt for 45 min. The solution was diluted with aq. potassium sodium tartrate (300 ml) and the THF was evaporated. The slurry was extracted with EtOAc (4x100 ml) and the combined organic layer washed with brine (100 ml), dried over MgS04, filtered, concentrated and the resultant crude product was chromatographed with 15: 85 EtOAc-hexane to provide 11.8 g (96%) of foam.

'H NMR: 8.58 (d, J = 2. 4 Hz, 1 H), 7.74 (dd, J = 8.4, 2.8 Hz, 1 H), 7. 09 (d, J = 8.4 Hz, 1H), 6.55 (dd, J = 15.6, 10.0 Hz, 1H), 6.45 (d, J = 16.0 Hz, 1H), 4.75-4. 68 (m, 1H), 2.69-2. 56 (m, 2H), 2.32 (dt, J = 10.1, 6.5 Hz, 1H), 1.98 (ddd, J = 13.4, 6.6, 2.8 Hz, 1 H), 1.67-1. 59 (m, 1 H), 1.47-1. 39 (m, 2H), 1.37 (d, J = 5.9 Hz, 3H), 1.31-1. 20 (m, 2H), 0.98 (d, J = 6. 2 Hz, 3H), 0.73 (t, J = 7. 5 Hz, 3H).

Preparation 5

To a solution of the product of Preparation 4 (7.2 g, 19 mmol), in THF (100 ml) at-78 °C was added 1 M LHMDS in THF (23 ml, 23 mmol, 1.2 eq. ). The solution was stirred for 30 min at-78 °C, 30 min at 0 °C and cooled back to-78 °C. To this was added a solution of (1 S)- (+)- (10-camphorsulfonyl) oxaziridine (6.0 g, 26 mmol, 1.4 eq. ) in THF (50 ml) and the mixture was stirred for 1 hour at-78 °C and 1.5 hours at 0°C.

To the solution was added aq. NH4CI (300 ml), THF was evaporated and the aqueous layer was extracted with EtOAc (4x100 ml). The combined organic layer was washed with brine (100 ml), dried over MgS04, filtered, concentrated and the crude product was chromatographed with 15: 20: 65 EtOAc-CH2CI2-hex to provide 6.4 g (85%) of foam.

AH NOR : 8.56 (d, J = 2.0 Hz, 1 H), 7.72 (dd, J = 8.4 Hz, 1 H), 7.07 (d, J = 8.4 Hz, 1 H), 6.56 (dd, J = 15.6, 9.8 Hz, 1 H), 6.48 (d, J = 15.6 Hz, 1 H), 4.62-4. 55 (m, 1 H), 3.72 (br s, 1 H), 2.80-2. 74 (m, 1 H), 2.28 (dd, J = 9.6, 5.6 Hz, 1 H), 1.81-1. 78 (m, 2H), 1. 63-1.58 (m, 1 H), 1.44-1. 27 (m, 3H), 1.37 (d, J = 6.0 Hz, 3H), 0.94 (d, J = 6.4 Hz, 3H), 0.73 (t, J = 7.5 Hz, 3H). Preparation 6 OTBDPS OH Step 1: To a solution of (R)- (+)-3-butyn-2-ol (5 ml, 64 mmol) in CH2CI2 (100 ml) at rt was added DMAP (780 mg, 6.4 mmol, 0.1 eq. ), tert-butylchlorodiphenylsilane (17.4 ml, 67 mmol, 1.05 eq. ) and Et3N (9.8 ml, 70 mmol, 1.1 eq. ). The mixture was stirred overnight, diluted with Et20 (400 ml), washed with 1 N HCI (2x200 ml), aq. NaHCOs (200 ml), brine (200 ml), dried over MgS04, filtered and evaporated to give ~20 g of oil which was used as such for the next step.

Step 2: To a solution of the product of Step 1 in THF (200 ml) at-78°C was added 2.5M BuLi in hexanes (30.4 ml, 76 mmol, 1.1 eq. ), the solution was stirred for 1 hour and solid paraformaldehyde (4.15 g, 138 mmol, 2.0 eq. ) was added. The mixture was

stirred for 15 min at-78°C, 1 hour at rt, then quenched with the addition of aq. NH4CI (500 ml). The THF was evaporated and the aqueous layer was extracted with EtOAc (3x200 ml). The combined organic layers were washed with H20 (2x300 ml) and brine (300 ml), dried over MgS04, filtered, evaporated and the crude was chromatographed with 10% EtOAc-hex to provide 16.5 g (71 %) of resin.

'H NMR: 7.77-7. 74 (m, 2H), 7.71-7. 68 (m, 2H), 7.46-7. 36 (m, 6H), 4.53 (tq, J = 1.8, 6.5 Hz, 1 H), 4.08 (dd, J = 6.2, 1.8 Hz), 2.82 (d, J = 6.4 Hz, 3H), 1.07 (s, 9H). Example 1 H H H N F N zon 1 F F Preparation 1 +-eux. 1 'N P\O F EtO OEt To a solution of the phosphonate (650 mg, 2.01 mmol, 2 eq. ) in THF (8 mi) at 0°C was added BuLi in hexanes (790 ti of 2.5M solution, 2.0 mmol, 2 eq. ), the mixture was stirred for 10 min, then Ti (OiPr) 4 (590 pI, 2.0 mmol, 2 eq. ) was added and the solution was stirred at rt for 10 min. To this was added a solution of the product of Preparation 1 (220 mg, 0.98 mmol) in THF (3 ml) and the mixture was stirred at rt for 1.5 h. To the solution was added aq. Rochelles's salt (100 ml) and THF was evaporated. The aqueous phase was extracted with EtOAc (3x30 ml) and the combined organic layer was washed with brine (50 ml). The solution was dried over MgS04, concentrated and the resultant residue was chromatographed with 20% EtOAc-hexane to provide the title compound (240 mg, 62%) as a resin.

1HNMR (400MHz, CDCl3) 8.78 (d, J = 2. 0, 1 H), 7.82 (dd, J = 2. 4,8. 0, 1 H), 7. 44 (dt, J = 5.7, 8.1, 1 H), 7.36 (dt, J = 1. 2,7. 7,1 H), 7.30-7. 25 (m, 2H), 7.09 (ddt, J = 2.5, 1.0, 8.4, 1H), 6.61 (dd, J = 15.3, 8.6, 1H), 6.56 (d, J = 15.3, 1H), 4.78-4. 71 (m, 1H), 2.71- 2.61 (m, 2H), 2.36 (dt, J = 10. 0,6. 4, 1 H), 1.99 (ddd, J = 13. 5,6. 1,2. 9, 1H), 1.68-1. 61 (m, 1 H), 1.51-1. 44 (m, 2H), 1.42 (d, J = 5.9, 3H), 1.39-1. 22 (m, 2H), 0.99 (d, J = 6.6, 3H), 0.76 (t, J = 7.5, 3H).

FAB HRMS: 394. 2184, calculated: 394.2182.

Anal. calc'd for C25H28FN02. HCI : C, 69.84 ; H, 6.80 ; N, 3.26. Found: C, 71.00, H, 6.96 ; N, 3.19.

Using a similar procedure with the appropriate phosphonate, the following compound 1 A was prepared:

1H NMR (400 MHz, CDCI3) 8.73 (bs, 1 H), 7.84 (dt, J = 2.0, 8.0, 1 H), 7.44 (dt, J = 1.7, 7.7, 1 H), 7.40-7. 34 (m, 1 H), 7.30 (d, J = 8.0, 1H), 7.25 (dt, J = 7.6, 1.1, 1H), 7.18 (ddd, J = 10. 6,8. 4,1. 2, 1H), 6.62 (dd, J=15. 1,8. 6, 1 H), 6.56 (d, J = 15. 1, 1H), 4.79- 4.72 (m, 1H), 2.71-2. 61 (m, 2H), 2.36 (dt, J = 10.0, 6.5, 1H), 1.99 (ddd, J = 13.5, 6.1, 2.9, 1 H), 1.70-1. 57 (m, 1 H), 1.51-1. 44 (m, 2H), 1.42 (d, J = 5. 9,3H), 1.39-1. 22 (m, 2H), 0.99 (d, J = 6.6, 3H), 0.76 (t, J = 7.3, 3H).

FAB HRMS: 394.2184, calculated : 394.2182. Example 2 H H o H H gH N xi 1 tCF3 Preparation 2 +---------0-Ex. 2 N P=O F3C Etc OEt To a solution of the product of Preparation 2 (50 mg, 0.22 mmol) in CH2Ci2 (3 ml) was added NMO (78 mg, 0.67 mmol, 3 eq. ) and 4 A° molecular sieves (about 50 mg). After stirring for 10 min. , TPAP (8 mg, 0.02 mmol, 0.1 eq. ) was added and the stirring was continued for another 40 min. The mixture was diluted with Et20 (20 ml), filtered through celiteTM and concentrated to provide a residue. The residue was filtered through a short Si02 plug, eluting with 30% EtOAc-hexane to provide 38 mg of aldehyde.

In another flask containing the phosphonate (210 mg, 0.56 mmol, 3.3 eq. ) in THF (1.5 ml) at 0°C was added a 2. M solution of BuLi in hexanes (224 FI, 0.56 mmol, 3.3 eq. ) and the mixture was stirred for 20 min. A solution of the above aldehyde in 1.5 ml of THF was added and the mixture was stirred at 0°C for 1 h. The solution was diluted with EtOAc (20 ml), washed with H20 (2x20 ml) and brine (20 ml), dried over MgS04, filtered, concentrated and purified by preparative TLC using 25% EtOAc-hexane to provide 9 mg of the title compound.'H NMR (400 MHz, CDCl3)

8.79 (d, J = 2.4 Hz, 1H), 7.85 (dd, J = 8.4, 2. 6 Hz, 1H), 7.81 br s, 1H), 7.76 (d, J = 7.2 Hz, 1 H), 7.67-7. 58 (m, 2H), 7.31 (d, J=7. 6Hz, 1H), 6.63 (dd, J = 15. 6, 9. 2 Hz, 1 H), 6.57 (d, J = 15.6 Hz, 1H), 4.79-4. 72 (m, 1H), 2. 71-2. 61 (m, 2H), 2.37 (dt, J = 10. 0,6. 4 Hz, 1H), 2.00 (ddd, J = 13. 5,6. 3,2. 7 Hz, 1H), 1.64-1. 56 (m, 1H), 1.51-1. 23 (m 4H), 1.42 (d, J = 6.2 Hz, 3H), 1. 00 (d, J = 6.6 Hz, 3H), 0.77 (t, J = 7.5 Hz, 3H) FABHRMS: 446.2306 (MH+), Calculated 446.2280.

Using similar procedures, the following compounds were also prepared: Example 3

1H NMR (400 MHz, CDCl3) 8.62 (d, J = 2.0 Hz, 1H), 7.76 (dd, J = 8.0, 2.4 Hz, 1H), 7.51-7. 48 (m, 1 H), 7.37-7. 26 (m, 4H), 6.65-6. 55 (m, 2H), 4.78-4. 71 (m, 1 H), 2.71-2. 61 (m, 2H), 2.36 (dt, J = 10.0, 6.4 Hz, 1H), 1.99 (ddd, J = 13.7, 6.3, 2.9 Hz, 1H), 1.68- 1.61 (m, 1 H), 1.50-1. 45 (m, 2H), 1.43 (d, J = 5. 6 Hz, 3H), 1.33-1. 25 (m, 2H), 0.99 (d,

J = 6. 4 Hz, 3H), 0.76 (t, J = 7. 4 Hz, 3H).

[&alpha;]20D = +13. 2 °(c 0. 5, MeOH).

FAB HRMS: 410.1891 (MH+), Calculated 410. 1887.

Example 4

'H NMR (400 MHz, CDCl3) 8.75 (d, J = 2. 0 Hz, 1H), 7. 80 (dd, J = 8. 2,2. 0 Hz, 1 H), 7.54 br s, 1H), 7.46-7. 34 (m, 3H), 7.29 (d, J = 8.0 Hz, 1H), 6.61 (dd, J = 15.3, 9.0 Hz, 1H), 6.56 (d, J = 15.3 Hz, 1H), 4.78-4. 71 (m, 1H), 2.70-2. 60 (m, 2H), 2.31 (dt, J = 10.1, 6.5 Hz, 1H), 1.98 (ddd, J = 13.5, 6.4, 2.9 Hz, 1 H), 1. 71-1.64 (m, 1H), 1.49-1. 43

(m, 2H), 1.40 (d, J = 6.0 Hz, 3H), 1.33-1. 21 (m, 2H), 0.99 (d, J = 6.4 Hz, 3H), 0.75 (t, J = 7.4 Hz, 3H) <76504-097-A-H in 2A&gt; [a] 20D = +23. 1 ° (c 0.5, MeOH).

FAB HRMS: 410.1887 (MH+), Calculated 410.1887.

Example 5

1 H NMR (400 MHz, CDCI3) 8.58 (d, J = 2.0 Hz, 1 H), 7.72 (dd, J = 8.0, 2.0 Hz, 1 H), 7.50 (dd, J = 8.0, 1.6 Hz, 1 H), 7.31-7. 21 (m, 3H), 6.63 (dd, J = 15.5, 8.8 Hz, 1 H), 6.57 (d, J = 15.5 Hz, 1H), 4.78-4. 71 (m, 1H), 2.71-2. 61 (m, 2H), 2.36 (dt, J = 10.0, 6.4 Hz, 1 H), 1.99 (ddd, J = 13.6, 6.4, 2.8 Hz, 1 H), 1.68-1. 61 (m, 1 H), 1.50-1. 45 (m, 2H), 1.43

(d, J = 6.0 Hz, 3H), 1.35-1. 22 (m, 2H), 0.99 (d, J = 6.4 Hz, 3H), 0.76 (t, J=7.4 Hz, 3H) [a] 20D = +5.8 °(c 0. 4, MeOH).

FAB HRMS: 444.1491 (MH+), Calculated 444.1497. Example 6 OH H O H H Fi N F

To a solution of the product of Example 1 (540 mg, 1.37 mmol) in THF (8 ml) at - 78 °C was added 1 M LHMDS solution in THF (1.65 ml, 1.65 mmol, 1.2 eq. ). The solution was stirred at-78 °C for 15 min. and at 0 °C for 30 min. It was cooled back to-78 °C and a solution of (1 S)- (+)- (10-camphorsulfonyl) oxaziridine (475 mg, 2.10 mmol, 1.5 eq. ) in THF (4 mi) was added. The mixture was stirred at-78 °C for 15 min. then allowed to warm up slowly to rt. To the mixture was added aq. NH4CI (100 mi) and it was then extracted with EtOAc (3x30 ml). The combined organic layer was washed with 30 ml brine, dried over MgS04, concentrated and chromatographed with 15: 20: 65 EtOAc-CH2CI2-hexanes to provide 390 mg (69%) of resin.

1H NMR: 8.78 (d, J = 2.4 Hz, 1H), 7.82 (dd, J = 8.2, 2.6 Hz, 1 H), 7.44 (dt, J = 6.0, 8.0 Hz, 1 H), 7.37-7. 35 (m, 1 H), 7.29-7. 25 (m, 2H), 7.09 (ddt, J = 1.0, 2.4, 8.3 Hz, 1 H), 6.67-6. 58 (m, 2H), 4.67-4. 60 (m, 1 H), 2.85-2. 79 (m, 2H), 2.32 (dq, J = 1.5, 5.7 Hz, 1 H), 1.89-1. 82 (m, 1 H), 1.79-1. 75 (m, 1 H), 1.70-1. 61 (m, 2H), 1.54-1. 46 (m, 1 H), 1.45 (d, J=6.0 Hz, 3H), 1.43-1. 32 (m, 1 H), 0.99 (d, J = 6.6 Hz, 3H), 0.78 (t, J = 7.5 Hz, 3H).

The Suzuki coupling procedure is exemplified by heating a solution of a bromide of Preparation 4 or 5 with boronic acid (1.0 to 2.0 eq. ), K2CO3 (4 eq. ) and Pd (PPh3) 4 (5 to 10 mol%) in toluene : EtOH: H20 (4: 2: 1, v/v/v) at 100°C until the reaction is complete. The reaction mixture is diluted with H2O, extracted with EtOAc, and the organic layer is washed with brine, dried over MgS04, filtered, concentrated and purified by chromatography to provide the desired compounds.

Using the Suzuki coupling procedure described above, the following compounds were prepared: Example 7 9 OH H 0 H %-i u H H N I Me

'H NMR: 8.54 (dd, J = 2.2, 0.6 Hz, 1 H), 7.62 (dd, J = 8.0, 2.2 Hz, 1 H), 7.31-7. 25 (m, 4H), 7.22-7. 20 (m, 1 H), 6.65-6. 56 (m, 1 H), 4.67-4. 60 (m, 1 H), 3. 20 (br s, 1 H), 2.89- 2.80 (m, 1 H), 2.34 (ddd, J = 10. 1,5. 7,1. 5 Hz, 1 H), 2.30 (s, 3H), 1.91-1. 77 (m, 2H), 1.70-1. 64 (m, 1 H), 1.55-1. 43 (m, 2H), 1.45 (d, J = 6.0 Hz, 3H), 1.39-1. 25 (m, 1 H), 0.98 (d, J = 6.50, 3H), 0.79 (t, J = 7.5 Hz, 3H). Example 8 OH H X w 0L X H H N xi I

1H NMR : 8.80 (d, J = 2.0 Hz, 1H), 7.84 (dd, J = 8.2, 2.2 Hz, 1H), 7.58 (d, J = 7.6 Hz, 2H), 7.47 (t, J = 7.4 Hz, 2H), 7.39 (t, J = 7.2 Hz, 1 H), 7.29 (d, J = 8.0 Hz, 1 H), 6.65-

6.55 (m, 2H), 4.67-4. 60 (m, 1 H), 3.56 (br s, 1 H), 2.87-2. 81 (m, 1 H), 2.34 (dd, J = 9.6, 5.6 Hz, 1 H), 1. 87-1. 80 (m, 2H), 1.70-1. 63 (m, 1H), 1.53-1. 33 (m, 3H), 1.44 (d, J = 6. 0 Hz, 3H), 0.98 (d, J = 6.5 Hz, 3H), 0.79 (t, J = 7.4 Hz, 3H).

Also using the Suzuki coupling procedure with the appropriate reagents, compounds of the following structures were prepared: wherein R3, R22, R23 and W are as defined in the following table (Me is methyl, Et is ethyl and Ph is phenyl) : Ex. R R R W Analytical Data 8A H Me Et HRMS (MH+) CF3 444. 2165 8B H Me Et 1 HRMS fy (MHi F 394. 2184 8C H Me Et HRMS F (MH+) 394. 2184 8D H Me Et e HFIM5 (mu+) 410. 1891 8E H Me Et HRMS fj ! (MH ci 410. 1887 8F H Me Et ci HRMS , . C) ci 444. 1491 8G H H Ph HRMS (Y (MH F 428. 2026 8H H H Ph HRMS ry (MHi 428. 2027 81 H Me Et HRMS fj ! (MH 418. 2381 0 8J H Me Et HRMS (mu+) X 433M. 2H4) 90 N OH 8K H Me Et HRMS (j ! (MH') ¢4 447M. 2H648 N i OMe 8L H Me Et T HRMS C ° (MH+) N O ~ 483. 2319 H 0 8M H Me Et HRMS ( 390. 2441 390. 2441 8N H Me Et 1 HRMS (MH+) Me 390. 2437 80 H Me Et l cl HRMS (MH+) ci 444. 1490 8P Me Me Et HRMS (t, (MH+) F 408. 2346 8Q OH Me Et HRMS Me (MH+) fY (MH 406. 2380 8R OH Me Et HRMS fY (MH Me 406. 2376 8S OH Me Et HRMS (MHz LS 398. 1788 8T OH Me Et HRMS Qs (MH+) Cl 432. 1392 8U OH Me Et HRMS N (MH+) 393. 2181 8V OH Me Et HRMS ce 417. 2178 8W OH Me Et HRMS () (mu+) CN 417. 2178 8X OH Me Et HRMS (MH+) (y 434. 2330 0 8Y OH Me Et HRMS ("t (MH (y 449. 2440 N I OH 8ZA OH Me Et HRMS (mu+) i 463. 2599 N i OMe 8AA OH Me Et HRMS (MH+) 435. 2275 N I OH 8AB OH Me Et HRMS (mu+) 449. 2446 N N i OMe 8AC OH Me Et HRMS (mu+) (r 435. 2279 8AD OH Me Et HRMS N, OMe (MH+) 449. 2442 8AE OH Me Et HRMS (mu+) 422. 2332 OH 8AF OH Me Et ~ HRMS (MH 422. 2332 8AG H H Et "" HRMS (mu+) 380. 2028 8AH H Ph Me MS (MH+) if 442. 1 F 8AI H Ph Me MS (MH+) 4 458. 1 C. 8AJ OH Me Et HRMS (-1 (MH 463. 2589 OUT OEt OEt 8AK OH Me Et HRMS N, OEt (MH+) 463. 2593 8AL OH Me Et-HRMS (mu+) i 477. 2750 N OEt 8AM OH Me Et HRMS fj (MH 392. 2227 8AN OH Me Et HRMS (MH+) my 434. 2695 8AO OH Me Et _ HRMS (MH+) S 398. 1788 8AP OH Me Et HRMS MH+) 382. 2020 8AQ OH Me Et HRMS (mi-') 435. 2282 (3 8AR OH Me Et HRMS (mu+) Me 424. 0945 F 8AS OMe Me Et t~ MS (MH+) o 450. 1 8AT OH Me Et < MS (MH7 436. 1 i 8AU OMe Me Et MS (MH+) OH 436. 1 i 8AV OH Me Et HRMS rY (MHi 480. 2752 0 8AW OH Me Et HRMS (MH+) OH 436. 2489 OH 8AX OH Me Eut 0 HRMS (MH+) 434. 2325 8AY OH Me Et OH HRMS (MH+) 436. 2489 8AZ OH H Et Me MS (MH+) 392. 2 i 8BA OH H Et MS (MH+) 396. 3 F 8BB OH H Et MS (MH+) 368. 4 6n 8BC OH Me Et HRMS (MH+) 408. 2169 OH 8BD OH Me Et HRMS (mu+) _ Cl 456. 1941 8BE OH H Me ~ HRMS (mu+) F 382. 1813 8BF OH H Me ~ HRMS (mu+) ce 389. 1863 8BG OH H Me HRMS f (MH 365. 1871 8BH OH Me Et HRMS (mu+) Fob 440. 2243 8BI OH H Me HRMS Mu ( 378. 2064 8BJ OH H Me HRMS (mu+) 364. 1919 8BK OH Me Et HRMS (mu+) & 449. 2435 8BL OH Me Et HRMS \N'OMe (MH+) 463. 2604 8BM OH Me Et ~ HRMS (mu+) NOEt & 477. 2751 8BN OH Me Et HRMS (mu+) 450. 2640 OH More Compounds of Example 8

The following compounds were prepared using Suzuki type coupling procedures by using appropriate reagents. Ex. R3 R22 R23 W Analytical Data HRMS 8BP OH H Me (MH+) y 408. 2181 OU 8BQ OH H Me (MH+) 408. 2181 HARMS 8BR OH Me Et (MH+) 417. 2182 HRMS HRMS 8BS OH H Me (MH+) ""F 366. 1867 HRMS 8BT OH Me Et (MH+) OH 436. 2493 QH M HRMS 8BU OH Me Me O _ HRMS 378. 2075 HARMS 8BV OH H Me (MH+) 408. 2173 OH HRMS 8BW OH H Me (MH+) 408. 2169 OH HRMS 8BX OH Me Et (MH+) 436. 2492 OH HRMS 8BY OH Me Me (MH+) 392. 2231 MS (MH+) i MS (MH) 8BZ H Me Et 376. 1 HRMS 8CA OH Me Me (MH+) if 396. 1969 MUS 8CB OH Me Me con 403. 1 HUMS JL HRMS 8CC OH Me Me (MH+) 422. 2337 OH HRMS 8CD OH Me Et 422. 2336 HARMS 8CE OH Me Et (MH+) 422. 2331 o HRMS 8CF OH Me Et (MH+) 422. 2336 HRMS J. HRMS 8CG OH Me Et 0 (MH+) 611, NH2 s 471. 1961 0 HARMS 8CH OH Me Et (MH+) "0 440. 2234 HRMS 8CI OH Me Et (MH+) 466. 2600 JvW O MS (MH+) 8CJ OH Me Me zou MS (MH+) 8CK OH Me Me 409. 1 HRMS 8CL OH Me Me CN (MH+) 403. 2027 HRMS 8CM OH Me Me e M e (MH+) tu 422. 2336 MS (MH+) 8CN OH Me Me 422. 1 ' 8CO H Et Et h ! 408. 1 MS (MH+) 8CO H Et Et 408. 1 F MS (MH+) 8CP H Me Et u 401. 1 m CN.. MS (MH+) 8CQ OH Et Et 424. i MS (MH+) 8CR H Me Me 387. 1 i m i i MS (MH+) 8CS H Me Me tgCN 387. 1 i MS (MH+) 8CT H Et Et C 415. 1 i 8CU F MS (MH+) OH Me Me 396. 2 Example 9 OH H O H 02m, v H R N N I H N

To the product of Preparation 5 (0.127 mmol) in dry toluene (5 ml) was added aniline (0.254 mmol, 2 eq. ), potassium phosphate (0.380 mmol, 3 eq.), palladium acetate (6.5 mol%) and 2- (dicyclohexylphosphino) biphenyl (13 mol%). The mixture was bubbled with N2 for 2 min. then heated to 120 °C in a sealed tube. After 16 h, the reaction was cooled to rt, poured into water and extracted with Et20 (3x). The combined extracts were washed with brine, dried with MgS04, filtered and evaporated to dryness. Purification by flash chromatography (2-5% CH30H in CH2CI2) yielded the desired product in a 66% yield.

1H NMR: 8.31 (d, J = 2.8 Hz, 1 H), 7.40 (dd, J = 2.8, 8.5 Hz, 1 H), 7.30-7. 26 (m, 2H), 7.15 (d, J = 8.5 Hz, 1 H), 7.07 (dd, J = 0.9, 8.5 Hz, 1 H), 6.97 (t, J = 7.4 Hz, 1 H), 6.50 (d, J = 15.6 Hz, 1H), 6.25 (dd, J = 10.4, 15.6 Hz, 1H), 6.14 (s, 1H), 4.60-4. 56 (m, 1H), 4.43 (br s, 1 H), 2.79-2. 76 (m, 1 H), 2.31 (dd, J = 5.6, 9.2 Hz, 1 H), 1.91-1. 79 (m, 2H), 1.65-1. 58 (m, 1 H), 1.41-1. 35 (m, 2H), 1.39 (d, J = 6.0 Hz, 3H), 1.31-1. 25 (m, 1 H), 0.95 (d, J = 6. 4 Hz, 3H), 0.77 (t, J = 7. 4 Hz, 3H).

Using a similar procedure, compounds of the formula were prepared, wherein W is as defined in the table : Ex. W Analytical Data 9A N HRMS O (MH+) 385. 2490 9B N HRMS KOH (MH 415. 2601 9C 1 HRMS t \OH (MH+) 414. 2593 9D ° HRMS ) 399. 2278

Example 10 O H \ O H oX, H H N F Steps 1-3 : TBDPSO I--N N w F

Step 1: A suspension of the alkyne of Preparation 6 (3.1 g, 9.2 mmol), quinoline (215 pI, 1.8 mmol, 0.2 eq. ), and Lindlar catalyst (310 mg, 10 wt%) in EtOAc (50 ml) was stirred under 1 atm. H2 (balloon) and the reaction was monitored by NMR. After the reaction was completed, it was filtered through a celiteTM pad, washed with 1 N HCI and brine, dried over MgS04, filtered and evaporated to give-3. 4 g of resin which was used as such for the next step.

Step 2: Dess-Martin reagent (4.28 g, 10.1 mmol, 1.1 eq. ) was added to a mixture of the product of Step 1 and NaHC03 (1.54 g, 18.3 mmol, 2 eq. ) in CH2C12 (30 ml) at rt and stirred for 1 hr. The mixture was diluted with Et20 (60 ml) and a solution of Na2S203.5H20 (4.55 g, 18.3 mmol, 2 eq. ) and NaHC03 (1.54 g, 18.3 mmol, 2 eq. ) in H20 (100 ml) and stirred vigorously until the two layers became clear. The organic layer was separated and the aq. layer was extracted with Et20 (2x50 ml). The combined organic layers were washed with aq. Na2S203/NaHCO3 solution (100 ml), brine (100 ml), dried over MgS04, filtered and evaporated to give-3. 5 g of aldehyde, which was used as such for the next step.

Step 3: po N OEt To a solution of a phosphonate of the formula out (3. 9 g, 12. 1 mmol, 1.3 eq. ) in THF (30 ml) at 0 °C was added 60% NaH in mineral oil (480 mg, 12. 0 mmol, 1.3 eq. ) and the mixture was stirred for 20 min. To this was added a solution of the product of Step 2 in THF (15 ml), and after 1 hr of stirring at 0 °C, it was diluted with aq. NH4CI (200 ml). The THF was evaporated and the aq. layer was extracted with EtOAc (3x75 ml). The combined organic layers were washed with brine (100 ml), dried over MgS04, filtered, evaporated and the residue was chromatographed with 5% EtOAc-hex to provide 4.0 g (87%) of resin.

H NMR : 8.75 (d, J = 2.0 Hz, 1 H), 7.76 (dd, J = 8.0, 2.4 Hz, 1 H), 7.73-7. 66 (m, 4H), 7.47-7. 26 (m, 9H), 7.19 (d, J = 8.0 Hz, 1H), 7.09 (ddt, J = 1.1, 2.5, 8.4 Hz, 1H), 7.00 (ddd, J = 15.3, 11.5, 1.1 Hz, 1H), 6.52 (d, J = 15.2 Hz, 1H), 6.05-5. 99 (m, 1H), 5.74- 5.69 (m, 1 H), 4.93-4. 86 (m, 1 H), 1. 28 (d, J = 6.4 Hz, 3H), 1.06 (s, 3H). Step 4 : OH ZON I F To a solution of silyl ether (4.0 g, 7.88 mmol) in THF (30 mi) at 0 °C was added 1 M TBAF in THF (11.8 ml, 11.8 mmol, 1.5 eq. ) and the mixture was stirred at rt for 6

h. It was diluted with aq. NH4CI (150 ml), the THF was evaporated and the aq. layer was extracted with EtOAc (3x60 ml). The combined organic layers were washed with H20 (50 ml), brine (50 ml), dried over MgS04, filtered, evaporated and the residue was chromatographed with 30% EtOAc-hex to provide 2.0 g (94%) of resin.

H NMR: 8.80 (d, J = 2. 0 Hz, 1 H), 7. 81 (dd, J = 8.0, 2.4 Hz, 1 H), 7.64 (ddd, J = 15.1, 11.5, 1.1 Hz, 1 H), 7.44 (dt, J = 5.6, 7.9 Hz, 1 H), 7.38-7. 33 (m, 2H), 7.30-7. 26 (m, 1 H), 7.09 (ddt, J = 1. 0,2. 5,8. 3 Hz, 1 H), 6.67 (d, J = 7. 6 Hz, 1 H), 6.24 (t, J = 11. 2 Hz, 1 H), 5. 70-5. 65 (m, 1 H), 5.07-5. 00 (m, 1 H), 1.35 (d, J = 6.4 Hz, 3H).

Step 5 : To a solution of the alcohol of Step 4 (110 mg, 0.41 mmol) and the acid (85 mg, 0.61 mmol, 1.5 eq. ) in CH2CI2 (2 ml) was added DCC (130 mg, 0.63 mmol, 1.5 eq. ) and DMAP (10 mg, 0.08 mmol, 0.2 eq. ) and stirred at 0 °C until the reaction was complete. The mixture was diluted with Et20 (50 ml), washed with aq. NaHCO3 (2x20 ml) and brine (20 ml), dried over MgS04, filtered, concentrated and the residue was chromatographed with 10% EtOAc-hex to provide 135 mg (84%) of resin.

'H NMR : 8.79 (d, J = 2. 4 Hz, 1 H), 7.81 (dd, J = 8. 0,2. 4 Hz, 1H), 7.67 (ddd, J = 15. 3, 11.5, 1.2 Hz, 1 H), 7.47-7. 27 (m, 5H), 7.15 (ddt, J = 2.0, 1.0, 8.3 Hz, 1H), 6.71 (d, J = 15.6 Hz, 1H), 6.29 (dt, J = 0. 8,11. 4 Hz, 1H), 6.11-6. 00 (m, 1H), 5.88 (t, J = 7. 6 Hz, 1 H), 5.63 (t, J = 10.0 Hz, 1 H), 2.24-2. 16 (m, 2H), 7.76 (d, J = 0. 8 Hz, 3H), 1.43 (d, J = 6.4 Hz, 3H), 1.00 (t, J = 7.6 Hz, 3H).

Step 6: A solution of the tetraene of Step 5 (130 mg) in toluene (10 ml) was stirred in a sealed tube at 185 °C for 7 h, cooled to rt and stirred with 10) iL of DBU for 3 hr. The solution was concentrated and purified by preparative chromatography to afford 63 mg (49%) of resin.

1H NMR : 8.72 (d, J= 2.0 Hz, 1H), 7.77 (dd, J = 8.4, 2.4 Hz, 1H), 7.41 (dt, J = 6.0, 8.0 Hz, 1 H), 7.36-7. 31 (m, 2H), 7.26-7. 22 (m, 1 H), 7.06 (ddt, J = 1.0, 2.7, 8.3 Hz, 1 H), 6.66 (d, J = 16.0 Hz, 1H), 6.47 (dd, J = 15.8, 9.8 Hz, 1H), 5.62-561 (m, 1H), 4.55 (dq, J = 4. 0,6. 4 Hz, 1 H), 3.27-3. 24 (m, 1 H), 2.80-2. 75 (m, 1 H), 2.56-2. 52 (m, 1 H), 2.02- 1.97 (m, 1 H), 1.78 (d, J = 1.5 Hz, 3H), 1.69-1. 59 (m, 1 H), 1.50-1. 45 (m, 1 H), 1.41 (d, J = 6.4 Hz, 3H), 0.92 (t, J = 7.4 Hz, 3H).

Using a similar procedure, compounds of the following structure were prepared wherein R11, R22, R23 and W are as defined in the table (Me is methyl, Et is ethyl, Bn is benyl): Ex. R R R W HRMS (MH+) 10A H H H T 350. 1565 I F 1 OB Me-CH2OBn 1 484. 2299 H ¢AF F 10C Me H-CH20Bn 484. 2294 F F 10D Me H Et 1 392. 2021 ¢ ; F F 10E Me Me H 378. 1870 F F 1 OF Me H Me 378. 1870 F F lOG Me Me 1 3641714 uF 10H Me-CH20H H 394. 1821 _ uF F Example 11 OH H O H H H N N bV

A solution of Preparation 4 (100 mg), 2 (tri-n-butylstannyl) pyridine (292 mg) and Pd (PPh3) 4 (31 mg) in toluene (5 ml) in a sealed tube was bubbled with N2 and heated at 120 °C overnight. The mixture was diluted with aq. NH4C1, extracted with EtOAc, dried over MgS04, filtered, concentrated and the residue was chromatographed with 2% CH3OH-CH2CI2 to provide 83 mg of resin.

The resin was dissolved in THF (5ml), cooled to-78 °C, a solution of 1 M LHMDS in THF (290 tl) was added, stirred at 0 °C for 1 h, then cooled to-78 °C. To this was added a solution of (1 S)- (+)- (10-camphorsulfonyl) oxaziridine (76 mg) in THF.

After stirring for about 1.5 h, it was quenched by the addition of aq. NH4CI and extracted with EtOAc. The organic layer was washed with brine, dried over MgS04, filtered, concentrated and the residue purified by preparative TLC to afford 20 mg of the title compound. HRMS : 393.2185 (MH+), calculated 393.2178.

Using a similar procedure, the following compounds are also prepared: wherein W is as defined in the table : Ex. W HRMS (MH 'w\ 11A 1 394. 2127 N- N NJ 11 B N ; S 399. 1750 Nos I Example 12 H H OOH) H H /Ht H N N N'O \_I

Step 1 : To a solution of oxazole (75 ti, 1.1 mmol) in THF (2 ml) at-78 °C was added a solution of 2.5 M BuLi in hexanes (465 ul, 1.2 mmol, 2.2 eq. ) and the mixture was stirred for 30 min. To this was added 0.5 M ZnCI2 in Et2O (4.3 ml, 2.2 mmol, 4 eq.) and the mixture stirred for 30 min at-78 °C and 30 min. at 0 °C.

Step 2 : Separately, to a suspension of Pd (PPh3) 2CI2 (37 mg, 0.05 mmol) in THF at 0 °C was added 2.5 M BuLi in hexanes (43 pI, 0.11 mmol) and the suspension was stirred for 20 min. This solution was added to zincate of Step 1, followed by the product of Preparation 4 (200 mg, 0.5 mmol) and the mixture was refluxed overnight.

It was cooled, diluted with aq. NH4CI (60 ml) and extracted with EtOAc (3x20 ml).

The combined organic layer was washed with brine (20 ml), dried over MgS04, filtered, evaporated and purified by preparative TLC to provide 29 mg of resin.

HRMS : 367.2025 (MH+), calculated 367.2022. Example 13 OH H O H 0L X H Fi N ruz N'OH Step 1 : A solution of Preparation 5 (60 mg, 0.15 mmol), Et3N (26, ul, 0.19 mmol, 1.2 eq.), bis (diphenylphosphino) propane (3 mg, 7 limon, 5 mol%), Pd (OAc) 2 (1.7 mg, 7.6 µmol, 5 mol%) and vinyl n-propyl ether (85) J, 0.76 mmol, 5 eq. ) in DMF (1.5 ml) in a sealed tube was heated at 100 °C for 2 h, cooled to rt and stirred with 2N HCI (2 ml) for 2 h. The mixture was diluted with aq. NaHC03, extracted with EtOAc, dried over MgS04, filtered, concentrated and the residue was purified by preparative TLC to provide 25 mg of ketone.

Step 2: A solution of the product of Step 1 (13 mg, 36 µmol) and hydroxylamine hydrochloride (8 mg, 0.12 mmol) in pyridine (0.5 ml) was stirred overnight at rt. The

mixture was diluted with aq. NH4CI (30 ml) and extracted with EtOAc (2x10 ml), the combined organic layer was washed with brine (10 ml), dried over MgS04, filtered, concentrated and the residue was purified by preparative TLC to provide 13 mg of the title compound as a resin. HRMS: 373.2113 (MH), calculated 373.2127.

Using a similar procedure the following compound is prepared: Example 14 0 OH H 0 ! Hj ! =v H H N N N A mixture of Preparation 5 (100 mg, 0.25 mmol), imidazole (35 mg, 0.51 mmol, 2.0 eq. ), copper (l) trifluoromethanesulfonate benzene complex (13 mg, 0.026 mmol, 0. 1 eq. ), 1, 10-phenanthroline (46 mg, 0.26 mmol, 1 eq*), dibenzylideneacetone (6 mg, 0.026 mmol, 0.1 eq.) and CssCOs (125 mg, 0.38 mmol, 1.5 eq. ) in m-xylene (3ml) in a sealed tube was bubbled with argon and heated at 130 °C overnight. The mixture was cooled to rt, diluted with aq. NH4CI (40 ml) and extracted with CH2CI2 (3x10 ml).

The combined organic layer was washed with brine (10 ml), dried over MgS04, filtered, concentrated and the residue was purified by preparative TLC to provide 43 mg (44%) of the title compound. HRMS: 382.2133 (MH+), calculated 382. 2131.

Using a similar procedure, the following compound was prepared: Ex. 14-2: HRMS: 396.2286 (MH+)

Example 15 OH H ojHf _u H H N ZON i N NH N=N A mixture of Preparation 5 (1.0 g, 2.54 mmol), Zn (CN) 2 (300 mg, 2.56 mmol, 1 eq. ), Pd2 (dba) 3 (116 mg, 0.13 mmol, 5 mol%) and diphenylphosphinoferrocine (170 mg, 0.31 mmol, 12 mol%) in DMF (10 ml) and H20 (100 jut, 1 vol%) in a sealed tube was bubbled with argon and heated at 120 °C for 5 h. The mixture was cooled to rt, diluted with EtOAc (150 ml) and washed with H20 (3x50 ml), brine (50 ml), dried over MgS04, filtered, evaporated and the crude product was chromatographed with 30% EtOAc-hex to provide 800 mg (93%) of arylcyanide.

A mixture of the arylcyanide (100 mg, 0.29 mmol), NaN3 (115 mg, 1.77 mmol, 6 eq. ) and NH4CI (95 mg, 1.78 mmol, 6 eq. ) in DMF (2 ml) in a sealed tube was heated overnight at 120 °C. It was cooled to rt, diluted with H20 (10 ml), extracted with CH2C12, concentrated and the crude product was purified by preparative TLC to give 50 mg of the title compound as a solid. HRMS: 384. 2033 (MH+), calculated 384.2036. Example 16 H OAc 0 OH H H O H O H H H Br = XBr g rN r'N ç 16A ¢ 16B t F t F Step 1: To a solution of compound 31 a (wherein W is 3-fluorophenyl) (480 mg, 1.2 mmol) in CH2CI2 was added 1 M solution of BBr3 in CH2CI2 (11.7 ml, 11.7 mmol, 10 eq. ), and the mixture refluxed for 2.5 h, then diluted with aq. NaHC03 (100 ml). After stirring for about 30 min. the organic layer was isolated and the aqueous layer was extracted with CH2CI2 (2x40 ml). The combined organic layer was washed with aq.

NaHC03 (100 ml), brine (100 ml), dried over MgS04, filtered and evaporated to give the crude alcohol.

The crude alcohol was dissolved in CH2CI2 (12 ml), cooled to 0 °C, and Ac20 (225 pL, 2.4 mmol, 2 eq. ) was added followed by DMAP (27 mg, 0.24 mmol, 0.2 eq. ) and Et3N (0.5 ml, 3.6 mmol, 3 eq. ). After stirring for about 2 hrs. , the mixture was diluted with EtOAc (80 ml), washed with aq. NaHCOs (2x50 ml), and brine. The solution was dried over MgS04, filtered, evaporated and the residue was chromatographed with 40% EtOAc-hex to provide 350 mg (56%) of Example 16-A as a white foam.

HRMS: 530.1336, calculated 530.1342.

Step 2: A mixture of Example 16-A (53 mg, 0.1 eq. ), NaCNBHs (32 mg, 0.5 mmol, 5 eq. ) in HMPA (1 ml) was stirred at 80 °C for 4 h, cooled to rt, diluted with HzO (30 ml) and extracted with EtOAc (3x15 ml). The combined organic layer was washed with brine (20 ml), dried over MgS04, filtered, concentrated and purified by preparative TLC to provide 27 mg of resin. To this was added K2CO3 (32 mg) in CH30H-H20 mixture (2 ml of 9: 1 v/v) and the solution was stirred at rt for 1 hour. The mixture was diluted with H20 (30 ml), extracted with EtOAc (3x10 ml), and the combined organic layers were washed with brine (10 ml), dried over MgS04, filtered, concentrated and filtered through a short Si02 plug to provide 17 mg (72%) of Example 16-B as a resin.

HRMS: 410. 2126, calculated 410. 2131.

Using a similar procedure, the compounds with the following structure were prepared wherein R3, R22, R23 and W are as defined in the table (Me is methyl, Et is ethyl) : Ex. R R"R W HRMS (MH 16C H-CH20H Et'T. F 410. 2138 16D H-CH=N-OH p 423. 2090 16E H-CH=N-OMe"p 437. 2235 16F H-CH=N-OEt J Et t d F 451. 2396 | 16G OH-CH20H Et d F 426. 2075 Example 17: 7a-Carboxylic Acid and Amides

To a stirred solution of 2.5 g of compound 1 (6.59 mmol), in 50 ml of dry THF at 0 °C under argon, was added LHMDS (9.88 mmol, 9.9 ml of a 1.0 M solution in THF) and the mixture allowed to stir for 30 minutes. The temperature was lowered to - 78°C and 785p, L (9. 88mmol) of methylcyanoformate was added. After 2 hours, approximately 75 mL of an aqueous solution of ammonium iron (II) sulfate hexahydrate (10% w/v) was added and the mixture then extracted with three portions of ethyl acetate. The combined organic extracts were washed with brine, dried with magnesium sulfate, filtered and evaporated to dryness. Purification by flash chromatography using 15% ethyl acetate in hexanes yielded 2.47 g of compound 2.

MS (ESI) m/z424 (MH+).

To a stirred solution of 2.47 g of compound 2 (5.65 mmol) in 50 mL of dry THF at 0 °C under N2, was added boron tribromide (11.3 mmol) and the mixture allowed to stir for approximately 30 min. The reaction mixture was diluted with about 50 mL of dichloromethane and the pH adjusted, with aqueous sodium bicarbonate, to approximately pH=4 and the mixture extracted with three portions of dichloromethane. The combined organic extracts were washed with brine, dried with magnesium sulfate, filtered and evaporated yielding 2.32 g of compound 3.

MS (ESI) m/z424. 1 (MH+).

Ex. 17H A mixture of 4 (68 mg), EDCI (2 eq. ), HOBT (2 eq. ) and aq. NH3 (3 eq. ) in 2 mL DMF was stirred at rt for 16 hours.

It was diluted with EtOAc, washed with aq. NaHC03, dried over MgS04, filtered, concentrated and purified by preparative TLC to give 18 mg of 5, Ex. 17H.

MS: 419.1 (MH+).

The following compounds were prepared using an analogous procedure: Ex. R3 R22 R23 Data Data 17A 4 H Et m. MS (MH+) OMe \ OMe U 420. 3 O xw 17B Jk H Et MS (MH+) r. OH \ OH U 406. 1 17C A Me Et S (MH+) OH \ OH U 420. 1 17D n Me Me MS (MH+) oH 424. 1 17E Me Me MS (MH+) OMe \ OMe CF 438. 1 P 17F Jk Me Me X CN MS (MH+) \ OMe C 445. 1 17G Me Me CN MS (MH+) OH \ OH C 431. 1 Example 18: 7a Hvdroxymethyl

To 0.65g (1.71 mmol) of compound 1 in dry THF at-10°C under argon was added LHMDS (2.06 mmol) and the mixture allowed to stir for 30 min.

Benzylchloromethylether (2.57 mmol) was then added and, after 60 min. the mixture poured onto aqueous ammonium chloride and extracted with three portions of diethyl ether. The combined organic extracts were washed with brine, dried with magnesium sulfate, filtered and evaporated to dryness. Purification by flash chromatography yielded 0. 69g of compound 6.

MS (ESI) m/z500 (MH+).

To 2.19 g (4.38 mmol) of compound 6 in dry dichloromethane was added iodotrimethylsilane (87.6 mmol) and the mixture heated to reflux under a balloon of argon for 2.5 hours. The reaction mixture was cooled to room temperature, poured onto an aqueous solution of sodium bicarbonate and extracted with three portions of dichloromethane. The combined organic extracts were washed with aqueous sodium sulfite, dried with magnesium sulfate, filtered and evaporated to dryness. Purification by flash chromatography yielded compound 7.

MS (ESI) m/z 410. 1 (MH+).

The following compounds were prepared using a similar procedure: Ex. R3 R22 R23 W Analytical Data m CH20H H Et MS (MH+) 18A W 392. 2 CH20H Me Et MS (MH+) 18B 406. 1 CH20H Me Me MS (MH+) 18C W 392. 1 CH20H Me Me HRMS 18D (MH+) OMe 410. 2126 CH20H Me Me HRMS 18E ¢ ; (MH+) CN 417. 2174 Example 19: 7a-Hydroxymethyl to 7a-Aminomethy

To 0.15 g of compound 7 in 10 mL of dry dichloromethane at 0 °C was added 77 FL of triethylamine (1.5 eq. ) and 34 IlL of methanesulfonylchloride (1.2 eq. ). The mixture was stirred under N2 for one hour, diluted with dichloromethane, washed twice with aq. NaHC03, and once with brine. The organic phase was dried with MgS04, filtered and evaporated to dryness yielding 0.145g mesylate.

To this product in 10 mL of DMSO was added 0.290 g of sodium azide (15 eq.) and the mixture heated to 65 °C while stirring under N2for 3 days. The reaction mixture was poured onto H20 and extracted three times with ethylacetate. The combined extracts were washed with brine, dried with MgS04, filtered and evaporated to dryness yielding 65 mg of azide.

To a solution of this azide in 5 mL of ethylacetate and 501lL of H20 at 0 °C was added 300 gel of 1 M THF solution of trimethylphosphine (2 eq. ) and the mixture allowed to warm to room temperature while stirring under argon. After 24 hours, the reaction was evaporated to dryness and purified by flash chromatography yielding 0.053 g of amine 8.

MS (ESI) m/z409 (MH+).

Example 20: 7a-Amination Chemistry

To a solution of 9 (1.01 g, 2.57 mmol) in 20 ml THF at 0 °C was added a solution of 1 M LHMDS in THF (3.34 ml) and stirred for 20 min. It was cooled to-78 °C and a solution of di-tert- butylazodicarboxylate (890 mg, 3.87 mmol) in 2.5 ml THF was added. It was stirred at - 78 °C for 2 hours and at 0 °C for 1 hour and quenched by the addition of aqueous NH4CI. The aqueous layer was extracted with EtOAc and dried over MgS04 and concentrated.

The crude product was stirred with 5 mi DCM and 10 ml trifluoroacetic acid at 0 °C for 1 hour. It was concentrated and suspended in 100 ml of aq. K2CO3. The aqueous phase was extracted with DCM to provide the crude hydrazide.

This crude material was dissolved in 10 ml glacial acetic acid and 2 ml acetone. To this was added 2 g of Zn dust in portions. The suspension was stirred vigorously for 2 hours and filtered through a celiteT""pad and washed with plenty of DCM. The DCM layer was washed with water followed by aq. NaHC03 and brine. It was dried over MgS04, concentrated and purified by chromatography to give 500 mg of 10. MS: 409.2 (MH+).

The following compounds were prepared using a similar procedure: Ex. R3 Rz R23 Analytical Data Data 20A NH2 H Et MS (MH+) U 377. 1 20B NH2 Me Et HRMS (MH+) 6 391. 2384 20C NH2 Me Et F HRMS (MH+) 409. 2297 20D NH2 Me Et HRMS (MH+) 416. 2345 CN 20E NHz Me Me HRMS (MH+) U 377. 2227 20F NH2 Me Me HRMS (MH+) 395. 1296 20G NH2 Me Me HRMS (MH+) ))) 402. 2186 CN 20H NH2 Me Me < CN HRMS (MH+) 402. 2186 Example 21: 7a-Fluoro Analogs

To a solution of 11 (300 mg, 0.83 mmol) in 5 ml THF and 2 mi DMF at 0 °C was added a 1 M solution of LHMDS in THF (1.1 ml, 1.3 eq. ). The solution was stirred for 20 min at 0 °C, cooled to-78 °C and a solution of N-fluorobenzenesulfonamide (400 mg, 1.27 mmol, 1.5 eq. ) in THF was added. The mixture stirred overnight and allowed to warm to rt. It was

diluted with EtOAc, washed twice with aq. K2C03, and twice with H20 and brine. It was dried over MgS04, filtered, concentrated and chromatographed with 20% EtOAc in hexanes to provide 260 mg of 12.

HRMS: 380.2032 (MH+), calculated 380.2026.

Using a similar procedure, the following compound was prepared: Ex. 21B HRMS: 394.2188 (MH+), calculated 394. 218.

Further embodiments of the invention encompass the administration of compounds of Formula I along with at least one additional cardiovascular agent. The contemplated additional cardiovascular agent is one that differs in either atomic make up or arrangement from the compounds of Formula I. Additional cardiovascular agents that can be used in combination with the novel compounds of this invention include drugs which have anti-thrombotic, anti-platelet aggregation, antiatherosclerotic, antirestenotic and/or anti-coagulant activity. Such drugs are useful in treating thrombosis-related diseases including thrombosis, atherosclerosis, restenosis, hypertension, angina pectoris, arrhythmia, heart failure, myocardial infarction, glomerulonephritis, thrombotic and thromboembolic stroke, peripheral vascular diseases, other cardiovascular diseases, cerebral ischemia, inflammatory disorders and cancer, as well as other disorders in which thrombin and its receptor play a pathological role. Suitable cardiovascular agents are selected from the group consisting of thromboxane A2 biosynthesis inhibitors such as aspirin; thromboxane antagonists such as seratrodast, picotamide and ramatroban; adenosine diphosphate (ADP) inhibitors such as clopidogrel ; cyclooxygenase inhibitors such as aspirin, meloxicam, rofecoxib and celecoxib ; angiotensin antagonists such as valsartan, telmisartan, candesartran, irbesartran, losartan and eprosartan; endothelin antagonists such as tezosentan; phosphodiesterase inhibitors such as milrinoone and

enoximone; angiotensin converting enzyme (ACE) inhibitors such as captopril, enalapril, enaliprilat, spirapril, quinapril, perindopril, ramipril, fosinopril, trandolapril, lisinopril, moexipril and benazapril ; neutral endopeptidase inhibitors such as candoxatril and ecadotril ; anticoagulants such as ximelagatran, fondaparin and enoxaparin; diuretics such as chlorothiazide, hydrochlorothiazide, ethacrynic acid, furosemide and amiloride ; platelet aggregation inhibitors such as abciximab and eptifibatide; and GP Ilb/Illa antagonists.

Preferred types of drugs for use in combination with the novel compounds of this invention are thromboxane A2 biosynthesis inhibitors, cyclooxygenase inhibitors and ADP antagonists. Especially preferred for use in the combinations are aspirin and clopidogrel bisulfate.

When the invention comprises a combination of a compound of Formula I and another cardiovascular agent, the two active components may be co-administered simultaneously or sequentially, or a single pharmaceutical composition comprising a compound of Formula I and another cardiovascular agent in a pharmaceutically acceptable carrier can be administered. The components of the combination can be administered individually or together in any conventional dosage form such as capsule, tablet, powder, cachet, suspension, solution, suppository, nasal spray, etc.

The dosage of the cardiovascular agent can be determined from published material, and may range from 1 to 1000 mg per dose.

In this specification, the term"at least one compound of Formula I"means that one to three different compounds of Formula I may be used in a pharmaceutical composition or method of treatment. Preferably one compound of Formula I is used.

Similarly, the term"one or more additional cardiovascular agents"means that one to three additional drugs may be administered in combination with a compound of Formula I ; preferably, one additional compound is administered in combination with a compound of Formula I. The additional cardiovascular agents can be administered sequentially or simultaneously with reference to the compound of Formula I.

When separate compounds of Formula I and the other cardiovascular agents are to be administered as separate compositions, they can be provided in a kit comprising in a single package, one container comprising a compound of Formula I in a pharmaceutical acceptable carrier, and a separate container comprising another cardiovascular agent in a pharmaceutical acceptable carrier, with the compound of Formula I and the other cardiovascular agent being present in amounts such that the combination is therapeutically effective. A kit is advantageous for administering a

combination when, for example, the components must be administered at different time intervals or when they are in different dosage forms.

The following formulations'exemplify some of the dosage forms of this invention. In each, the term"active compound"designates a compound of formula 1.

EXAMPLE A-Tablets No. Ingredient mq/tablet mg/tablet 1 Active Compound 100 500 2 Lactose USP 122 113 3 Corn Starch, Food Grade, as a 10% 30 40 paste in Purified Water 4 Corn Starch, Food Grade 45 40 5 Magnesium Stearate 3 7 Total 300 700 Method of Manufacture Mix Item Nos. 1 and 2 in suitable mixer for 10-15 minutes. Granulate the mixture with Item No. 3. Mill the damp granules through a coarse screen (e. g., 1/4", 0.63 cm) if necessary. Dry the damp granules. Screen the dried granules if necessary and mix with Item No. 4 and mix for 10-15 minutes. Add Item No. 5 and mix for 1-3 minutes. Compress the mixture to appropriate size and weight on a suitable tablet machine.

EXAMPLE B-Capsules No. Ingredient mg/tablet mg/tablet 1 Active Compound 100 500 2 Lactose USP 106 123 3 Corn Starch, Food Grade 40 70 4 Magnesium Stearate NF 4 7 Total 250 700 Method of Manufacture Mix Item Nos. 1,2 and 3 in a suitable blender for 10-15 minutes. Add Item No.

4 and mix for 1-3 minutes. Fill the mixture into suitable two-piece hard gelatin capsules on a suitable encapsulating machine.

The activity of the compounds of formula I can be determined by the fol lowing procedures.

In Vitro Testing Procedure for Thrombin Receptor Antagonists : Preparation of F3HlhaTRAP A (pF-F) R (ChA) (hR) (12-Y)-NH2 (1.03 mg) and 10% Pd/C (5.07 mg) were

suspended in DMF (250/il) and diisopropylethylamine (10, ul). The vessel was attached to the tritium line, frozen in liquid nitrogen and evacuated. Tritium gas (342

mCi) was then added to the flask, which was stirred at room temperature for 2 hours.

At the completion of the reaction, the excess tritium was removed and the reacted peptide solution was diluted with DMF (0.5 ml) and filtered to remove the catalyst.

The collected DMF solution of the crude peptide was diluted with water and freeze dried to remove the labile tritium. The solid peptide was redissolved in water and the freeze drying process repeated. The tritiated peptide ([3H] haTRAP) was dissolved in 0.5 ml of 0. 1 % aqueous TFA and purified by HPLC using the following conditions: column, VydacT C18, 25 cm x 9.4 mm I. D.; mobile phase, (A) 0. 1% TFA in water, (B) 0. 1% TFA in CH3CN ; gradient, (A/B) from 100/0 to 40/60 over 30 min; flow rate, 5 mi /min ; detection, UV at 215 nm. The radiochemical purity of [3H] haTRAP was 99% as analyzed by HPLC. A batch of 14.9 mCi at a specific activity of 18.4 Ci/mmol was obtained.

Preparation of platelet membranes Platelet membranes were prepared using a modification of the method of Natarajan et al. (Natarajan et al, Int. J. Peptide Protein Res. 45: 145-151 (1995) ) from 20 units of platelet concentrates obtained from the North Jersey Blood Center (East Orange, NJ) within 48 hours of collection. All steps were carried out at 4° C under approved biohazard safety conditions. Platelets were centrifuged at 100 x g for 20 minutes at 4° C to remove red cells. The supernatants were decanted and centrifuged at 3000 x g for 15 minutes to pellet platelets. Platelets were resuspended in 10 mM Tris-HCI, pH 7.5, 150 mM NaCI, 5 mM EDTA, to a total volume of 200 mi and centrifuged at 4400 x g for 10 minutes. This step was repeated two additional times.

Platelets were resuspended in 5 mM Tris-HCI, pH 7.5, 5 mM EDTA to a final volume of approximately 30 ml and were homogenized with 20 strokes in a DounCOTM homogenizer. Membranes were pelleted at 41,000 x g, resuspended in 40-50 ml 20 mM Tris-HCI, pH 7.5, 1 mM EDTA, 0.1 mM dithiothreitol, and 1 O ml aliquots were frozen in liquid N2 and stored at-80° C. To complete membrane preparation, aliquots were thawed, pooled, and homogenized with 5 strokes of a Dounce homogenizer.

Membranes were pelleted and washed 3 times in 10 mM triethanolamine-HCI, pH 7.4, 5 mM EDTA, and resuspended in 20-25 mi 50 mM Tris-HCI, pH 7.5, 10 mM MgCl2, 1 mM EGTA, and 1% DMSO. Aliquots of membranes were frozen in liquid N2 and stored at-80° C. Membranes were stable for at least 3 months. 20 units of platelet concentrates typically yielded 250 mg of membrane protein. Protein concentration was determined by a Lowry assay (Lowry et al., J. Biol. Chem., 193: 265-275 (1951)).

High Throughput Thrombin Receptor tor Binding Assay Thrombin receptor antagonists were screened using a modification of the thrombin receptor radioligand binding assay of Ahn et al. (Ahn et al., Mol. Pharmacol., 51: 350-356 (1997) ). The assay was performed in 96 well Nunc plates (Cat. No.

269620) at a final assay volume of 200 NI. Platelet membranes and [3H] haTRAP were diluted to 0.4 mg/ml and 22.2 nM, respectively, in binding buffer (50 mM Tris-HCI, pH 7.5, 10 mM MgCi2, 1 mM EGTA, 0. 1% BSA). Stock solutions (10 mM in 100% DMSO) of test compounds were further diluted in 100% DMSO. Unless otherwise indicated, 10 @ ul of diluted compound solutions and 90 nul of radioligand (a final concentration of 10 nM in 5% DMSO) were added to each well, and the reaction was started by the addition of 100 pl of membranes (40, ug protein/well). The binding was not significantly inhibited by 5% DMSO. Compounds were tested at three concentrations (0.1, 1 and 10µM). The plates were covered and vortex-mixed gently on a Lab-Linew Titer Plate Shaker for 1 hour at room temperature. Packard UniFilter GF/C filter plates were soaked for at least 1 hour in 0. 1% polyethyleneimine. The incubated membranes were harvested using a Packard FilterMateT Universal Harvester and were rapidly washed four times with 300, cul ice cold 50 mM Tris-HCI, pH 7. 5,10 mM MgCl2, 1 mM EGTA. MicroScint 20 scintillation cocktail (25, ul) was added to each well, and the plates were counted in a Packard TopCount Microplate Scintillation Counter. The specific binding was defined as the total binding minus the nonspecific binding observed in the presence of excess (50, uM) unlabeled haTRAP. The % inhibition by a compound of [3H] haTRAP binding to thrombin receptors was calculated from the following relationship : % Inhibition = Total binding-Binding in the presence of a test compound x 100 Total binding-Nonspecific binding Materials A (pF-F) R (ChA) (hR) Y-NH2 and A (pF-F) R (ChA) (hR) (12-Y)-NH2, were custom synthesized by AnaSpec Inc. (San Jose, CA). The purity of these peptides was >95%. Tritium gas (97%) was purchased from EG&G Mound, Miamisburg, Ohio. The gas was subsequently loaded and stored on an IN/US Systems Inc. Trisorber.

MicroScintz 20 scintillation cocktail was obtained from Packard Instrument Co.

Protocol For Ex-Vivo Platelet Aggregation In Cvnomolqus Whole Blood Drua Administration and Blood Collection : Conscious chaired cynomolgus monkeys are allowed to equilibrate for 30 min.

A needle catheter is inserted into a brachial vein for infusion of test drugs. Another needle catheter is inserted into the other brachial or saphenous vein and used for blood sampling. In those experiments where the compound is administered orally only one catheter is used. A baseline blood sample (1-2 ml) is collected in vacutainer tubes containing a thrombin inhibitor CVS 2139 (100, ug/0. 1 ml saline) as an anticoagulant. The drug is then infused intravenously over a period of 30 min. Blood

sampies (1 ml) are collected at 5,10, 20,30 min during and 30,60, 90 min after termination of the drug infusion. In PO experiments the animals are dosed with the drug using a gavage canula. Blood samples are collected at 0,30, 60,90, 120,180, 240,300, 360 min after dosing. 0.5 ml of the blood is used for whole blood aggregation and the other 0.5 ml is used for determining the plasma concentration of the drug or its metabolites. Aggregation is performed immediately after collection of the blood sample as described below.

Whole Blood Aggregation : A 0.5 mi blood sample is added to 0.5 ml of saline and warmed to 37°C in a Chronolog whole blood aggregometer. Simultaneously, the impedance electrode is warmed in saline to 37°C. The blood sample with a stir bar is placed in the heating block well, the impedance electrode is placed in the blood sample and the collection software is started. The software is allowed to run until the baseline is stabilized and then a 20 fi calibration check is performed. 20 Q is equal to 4 blocks on the graphic produced by the computer software. The agonist (haTRAP) is added by an adjustable volume pipette (5-25 pI) and the aggregation curve is recorded for 10 minutes. Maximum aggregation in 6 minutes following agonist is the value recorded.

In vitro Platelet Aggregation Procedure: Platelet aggregation studies were performed according to the method of Bednar et aL (Bednar, B. , Condra, C., Gould, R. J. , and Connolly, T. M. , Throm. Res., 77: 453-463 (1995)). Blood was obtained from healthy human subjects who were aspirin free for at least 7 days by venipuncture using ACD as anticoagulant. Platelet rich plasma was prepared by centrifugation at 1 O0xg for 15 minutes at 15 deg C.

Platelets were pelleted at 3000xg and washed twice in buffered saline containing 1 mM EGTA and 20, ugiml apyrase to inhibit aggregation. Aggregation was performed at room temperature in buffered saline supplemented with 0.2 mglml human fibrinogen. Test compound and platelets were preincubated in 96-well flat-bottom plates for 60 minutes. Aggregation was initiated by adding 0. 3, uM haTRAP or 0.1 Ulml thrombin and rapidly vortexing the mixture using a Lab LineT" Titer Plate Shaker (speed 7). Percent aggregation was monitored as increasing light transmittance at 405 nm in a Spectromax Plate Reader.

In vivo Antitumor Procedure: Tests in the human breast carcinoma model in nude mouse are conducted according to the procedure reported in S. Even-Ram et a/., Nature Medicine, 4,8 (1988), p. 909-914.

Cannabinoid CB Receptor Binding Assay

Binding to the human cannabinoid CB2 receptor was carried out using the procedure of Showalter, et aL (1996, J. Pharmacol Exp Ther. 278 (3), 989-99), with minor modifications. All assays were carried out in a final volume of 100 ul. Test compounds were resuspended to 10 mM in DMSO, then serially diluted in 50 mM Tris, pH 7.1, 3 mM MgCI2, 1 mM EDTA, 50% DMSO. Aliquots (10 ul) of each diluted sample were then transferred into individual wells of a 96-well microtiter plate.

Membranes from human CB2 transfected CHO/Ki cells (Receptor Biology, Inc) were resuspended in binding buffer (50 mM Tris, pH 7.1, 3 mM MgCI2, 1 mM EDTA, 0.1 % fatty acid free bovine serum albumin), then added to the binding reaction (-15 ug in 50 ul per assay). The reactions were initiated with the addition of [3H] CP-55,940 diluted in binding buffer (specific activity = 180 Ci/mmol ; New England Nuclear, Boston, Mass. ). The final ligand concentration in the binding reaction was 0.48 nM.

Following incubation at room temperature for 2 hours, membranes were harvested by filtration through pretreated (0.5% polyethylenimine ; Sigma P-3143) GF-C filter plates (Unifilter-96, Packard) using a TomTecT Mach 3U 96-well cell harvester (Hamden, Ct). Plates were washed 10 times in 100 ul binding buffer, and the membranes allowed to air dry. Radioactivity on membranes was quantitated following addition of Packard OmniscintT 20 scintillation fluid using a TopCountT NXT Microplate Scintillation and Luminescence Counter (Packard, Meriden, Ct). Non-linear regression analysis was performed using Prism 20b. (GraphPad Software, San Diego, Ca).

Using the test procedures described above, representative compounds of formula I were found to have thrombin receptor ICs0 values (i. e., the concentration at which a 50% inhibition of thrombin receptor was observed) of 1 to 1000 nM, preferably 1-100 nM, more preferably 1-20 nM. CB2 Ki values range from 1 to 1000 nM, preferably 1-200 nM, more preferably 1-100 nM. For example, iCso values of Example Nos. 8BU, 8CA, 8CB, 8CL, 17H, 20E, 20F, 20G and 20H range from 1-100 nM.