Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
INHIBITORS OF FLAVIVIRIDAE VIRUSES
Document Type and Number:
WIPO Patent Application WO/2011/011303
Kind Code:
A1
Abstract:
Provided are compounds of Formula I: (I) and pharmaceutically acceptable salts and esters thereof. The compounds, compositions, and methods provided are useful for the treatment of Flaviviridae virus infections, particularly hepatitis C infections.

Inventors:
CANALES EDA (US)
CHONG LEE S (US)
CLARKE MICHAEL O'NEIL HANRAHAN (US)
DOERFFLER EDWARD (US)
LAZERWITH SCOTT E (US)
LEW WILLARD (US)
LIU QI (US)
MERTZMAN MICHAEL (US)
MORGANELLI PHILIP A (US)
WATKINS WILLIAM J (US)
YE HONG (US)
Application Number:
PCT/US2010/042394
Publication Date:
January 27, 2011
Filing Date:
July 19, 2010
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
GILEAD SCIENCES INC (US)
CANALES EDA (US)
CHONG LEE S (US)
CLARKE MICHAEL O'NEIL HANRAHAN (US)
DOERFFLER EDWARD (US)
LAZERWITH SCOTT E (US)
LEW WILLARD (US)
LIU QI (US)
MERTZMAN MICHAEL (US)
MORGANELLI PHILIP A (US)
WATKINS WILLIAM J (US)
YE HONG (US)
International Classes:
A61K31/381; C07D409/12; C07D333/68; C07D409/14; C07D413/12; C07D417/12; C07D487/04; C07D493/04; A61P31/12
Domestic Patent References:
WO2008058393A12008-05-22
WO2005095386A12005-10-13
WO2007093365A22007-08-23
WO2008058393A12008-05-22
WO2006072347A22006-07-13
WO2002100851A22002-12-19
Foreign References:
US5861421A1999-01-19
Other References:
K. J. HERLIHY ET AL.: "Development of intergenotypic chimeric replicons to determine the broad-spectrum antiviral activities of hepatitis C virus polymerase inhibitors", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, vol. 52, no. 10, 11 August 2008 (2008-08-11), pages 3523 - 3531, XP002601089
CALISHER ET AL., J. GEN. VIROL., vol. 70, 1993, pages 37 - 43
MOENNIG, V. ET AL., ADV. VIR. RES., vol. 48, 1992, pages 53 - 98
BOYER, N. ET AL., J HEPATOL., vol. 32, 2000, pages 98 - 112
DI BESCEGLIE, A.M.; BACON, B. R., SCIENTIFIC AMERICAN, October 1999 (1999-10-01), pages 80 - 85
GORDON, C. P. ET AL., J. MED. CHEM., vol. 48, 2005, pages 1 - 20
MARADPOUR, D. ET AL., NAT. REV. MICRO., vol. 5, no. 6, 2007, pages 453 - 463
DYMOCK ET AL., ANTIVIRAL CHEMISTRY & CHEMOTHERAPY, vol. 11, no. 2, 2000, pages 79 - 95
NEUMANN ET AL., SCIENCE, vol. 282, 1998, pages 103 - 7
FUKIMOTO ET AL., HEPATOLOGY, vol. 24, 1996, pages 1351 - 4
DOMINGO ET AL., GENE, vol. 40, 1985, pages 1 - 8
MARTELL ET AL., J. VIROL., vol. 66, 1992, pages 3225 - 9
SCOTT, L. J. ET AL., DRUGS, vol. 62, 2002, pages 507 - 556
J. AM. CHEM. SOC., vol. 82, 1960, pages 5566
J. ORG. CHEM., vol. 65, 2000, pages 1158 - 1174
J. AM. CHEM. SOC., vol. 124, 2002, pages 7421 - 7428
J. AM. CHEM. SOC., vol. 110, no. 4, 1988, pages 1238 - 1256
SCHUL ET AL., J. INFECTIOUS DIS., vol. 195, 2007, pages 665 - 74
HILL, A. V.: "The Possible Effects of the Aggregation of the Molecules of Hæmoglobin on its Dissociation Curves", J. PHYSIOL., vol. 40, 1910, pages IV - VII
Attorney, Agent or Firm:
WARD, John et al. (Inc.333 Lakeside Driv, Foster City CA, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A compound of Formula i:

Formula (I),

or a pharmaceutically acceptable salt or ester thereof, wherein:

R1 is selected from the group consisting of optionally substituted C^12 afkyl, optionally substituted C2_i2 aikenyl, optionally substituted C2-I2 aikynyl, optionally substituted C3^12 cycloalkyl, optionally substituted C6-U aryi, optionally substituted 3 - 14 membered heteroaryi, optionally substituted 3 -12 membered heterocyciyi, optionally substituted 3-18 membered heteroaryialkyl, optionally substituted 3-18 membered heterocyclylalkyl and optionally substituted C6-ie arylalkyl,

wherein, each substituted R1 is substituted with one or more Q1;

each Q1 is independently selected from the group consisting of

halogen, oxo, oxide, -NO2, -N{=0), -SR10, -S(O)R10, -S(O)2R10, -S(O)2 NR10R11, -NR10C(O)R11, -NR10C(O)NR11R12, -NR10S(O)R11, -NR10S(O)2R11, -OP(O)R11R12, -P(O)R11R12, -P(O)OR11R12, -P(O)(OR11)OR12, -C(O)NR11R12, optionally substituted C1-6 alkyl, optionally substituted C2.6 alkenyi, optionally substituted C2-6 alkynyj, optionally substituted C3.6 cycioalkyl, optionally substituted C6.12 arylalkyl, optionally substituted C6^2 aryl, optionally substituted 3 - 14 membered heteroaryi, optionally substituted C^ alkyloxy, optionally substituted C2.6 alkenyloxy, optionally substituted C2^6 alkynyloxy, optionally substituted C3-6 cycloalkyloxy, optionally substituted C6_i2 aryloxy, optionally substituted 3 - 14 membered heteroaryioxy, optionally substituted 4-12 membered heterocyclyloxy, optionally substituted -C(O)Ci-6 aSkyl, optionally substituted -C(O)C2-6 aikenyl, optionally substituted -C(O)C2-6 alkynyl, optionally substituted -C(O)C3-6 cycioalkyl, optionally substituted -C(O)C6-I2 aryl, optionally substituted -C(0)-3 - 14 membered heteroaryi, optionally substituted -C(O)C6-I2 arylaikyl, optionally substituted -3-10 membered

heterocyclyl, -OH, -NR11R12, -C(O)OR10, -CN, -N3, -C(=NR13)NR11R12,

-C(=NR13)OR10, -NR10C(=NR13)NR11R12, -NR11C(O)OR10, and -OC(O)NR11R12;

each R10, R11, and R1Z, iπdependentfy, is selected from the group consisting of H, optionally substituted C1-12 alkyl, optionaliy substituted C2-12 aikenyl, optionally substituted C2-12 alkynyl, optionally substituted C3-12 cycloaikyl, optionally substituted C6-i4 aryl, optionally substituted 3 - 14 membered heteroaryl, optionally substituted 3- 12 membered heterocyclyl, optionally substituted 3-18 membered heteroarylalkyl, and optionally substituted C6-18 arylaikyl;

or R11 and R12 taken together with the atoms to which they are attached form a 3 to 10 membered heterocyclyl;

each R13, independently, is selected from the group consisting of H, optionally substituted CM 2 alkyl, optionally substituted C2-i2 aikenyl, optionally substituted C2-12 alkynyi, optionally substituted C3-12 cycloaikyl, optionally substituted C6-14 aryl, optionally substituted 3 - 14 membered heteroaryl, optionally substituted 3-12 membered heterocyclyl, optionally substituted 3-18 membered heteroarylalkyi, optionally substituted C6-18 arylaikyl, -CN, -C(O)R14, -CHO and -S(O)2R14;

each R14, independently, is optionally substituted C1-12 alkyl;

wherein, each substituted Q1, substituted R10, substituted R11, substituted R12, substituted R13, or substituted R14 is independently substituted with one or more Q6;

R2 is selected from the group consisting of optionally substituted C1-12 alkyl, optionally substituted C2--I2 aikenyl, optionally substituted C2-12 aikynyl, optionally substituted C3-12 cycloaikyl, optionally substituted C6-14 aryl, optionally substituted 3 - 14 membered heteroaryl, optionally substituted 3 -12 membered heterocyclyl, optionaliy substituted 3-18 membered heteroarylalkyl, and optionally substituted C6-18 arylaikyl;

wherein, each substituted R2 is substituted with one or more Q2;

each Q2, independently, is selected from the group consisting of

halogen, oxo, oxide, -NO2, -N(=0), -SR20, -S(O)R20, -S(O)2R20, -S(O)2 NR20R21, -NR20C(O)R21, -NR20C(O)NR21R22, -NR20S(O)R21, -NR20S(O)2R21, -OP(O)R21R22, -P(O)R21R22, -P(O)OR21R22, -P(O)(OR21 )OR22, -C(O)NR21R22, optionally substituted C1-6 alkyl, optionally substituted C2-6 aikenyl, optionally substituted C2^ alkynyl, optionally substituted C3-6 cycloaikyl, optionally substituted C6-I2 arylalkyl, optionally substituted C6-12 aryl, optionally substituted 3 - 14 membered heteroaryl, optionally substituted C1-6alkyloxy, optionally substituted C2-6 alkenyloxy, optionaliy substituted C2.6 alkynyloxy, optionally substituted C3-6 cycloafkyioxy, optionaliy substituted C6-i2 aryloxy, optionally substituted 3 - 14 membered heteroaryloxy, optionaily substituted 4-12 membered heterocyclyloxy, optionally substituted -C(O)C1-6 alkyl, optionally substituted -C(O)C2-6 alkenyl, optionally substituted -C(O)C2-6 alkynyl, optionally substituted -C(O)C3-6 cycloaikyl, optionally substituted -C(O)C6-I2 aryl, optionaily substituted -C(0)-3 -14 membered heteroaryl, optionaily substituted -C(O)C6-12 arylalkyl, optionaily substituted 3-10 membered

heterocyclyl, -OH, -NR21R22, -C(O)OR20, -CN, -N3, -C(=NR23)NR21R22,

-C(=NR23)OR20, -NR20C(=NR23)N R21R22, -NR21C(O)OR20, and -OC(O)NR21R22;

each R20, R21, and R22, independently, is selected from the group consisting of H, optionaliy substituted C1-12 alkyl, optionally substituted C2-12 alkenyl, optionally substituted C2-12 alkynyl, optionally substituted C3-12 cycloaikyl, optionally substituted C6-14 aryl, optionally substituted 3 - 14 membered heteroaryl, optionaliy substituted 3- 12 membered heterocyclyl, optionally substituted 3-18 membered heteroarylalkyl, and optionaliy substituted C6^e aryialkyl;

or R21 and R22 taken together with the atoms to which they are attached form a 3 to 10 membered heterocyclyl;

each R23, independently, is selected from the group consisting of H, optionally substituted C1-I2 alkyl, optionaliy substituted C2-12 alkenyl, optionally substituted C2-12 alkynyl, optionally substituted C3.12 cycloalkyi, optionally substituted C6-14 aryl, optionally substituted 3 - 14 membered heteroaryi, optionaily substituted 3-12 membered heterocyclyl, optionaliy substituted 3-18 membered heteroarylaikyl, optionally substituted C6-18 aryiaikyl, -CN, -C(O)R24, -CHO and -S(O)2R24;

each R24 individually is optionally substituted C1-12 alkyl;

wherein, each substituted Q2, substituted R20, substituted R21, substituted R22, substituted R23, or substituted R24 is independently substituted with one or more Q6;

R3 is selected from the group consisting of optionally substituted C1-12 alkylene, C2-12 alkenylene, substituted C2-12 aikenylene, C2-12 alkynylene, substituted C2-12 alkynylene, C3-12 cycloalkylene, substituted C3-12 cycioalkylene, optionally substituted C6-14 aryiene, optionally substituted 3 - 14 membered heteroarylene, optionally substituted 3 -12 membered heterocyclylene, optionally substituted 3-18 membered heteroarylaikylene, and optionally substituted C6.i8 arylaikylene;

wherein each substituted R3 is substituted with one or more Q3; each Q3, independently, is selected from the group consisting of

halogen, oxo, oxide, -NO2, -N(=O), -SR30, -S(O)R30, -S(O)2R30, -S(O)2 NR30R31, -NR30C(O)R31, -NR30C(O)NR31R32, -NR30S(O)R31, -NR30S(O)2R31, -OP(O)R31R32, -P(O)R31R32, -P(O)OR31R32, -P(O)(OR31 )OR32, -C(O)NR31R32, optionally substituted C1-6 alkyS, optionally substituted C2.6 alkenyl, optionally substituted C2_6 alkynyl, optionally substituted C3-6 cycioalkyi, optionally substituted C6-i2 aryiaikyl, optionally substituted C6-I2 aryl, optionally substituted 3 - 14 membered heteroaryl, optionally substituted C1-6 alkyioxy, optionally substituted C2-6 alkenyloxy, optionally substituted C2-6 alkynyloxy, optionally substituted C3.6 cycloaSkyloxy, optionally substituted C6-i2 aryioxy, optionally substituted 3 - 14 membered heteroaryloxy, optionally substituted 4-12 membered heterocyclyloxy, optionally substituted -C(O)C1-6 aikyl, optionally substituted -C(O)C2-6 alkenyl, optionally substituted -C(O)C2-6 aikynyl, optionally substituted -C(O)C3-6 cycioalkyi, optionally substituted -C(O)C6.12 aryl, optionally substituted -C(O)- 3 -14 membered heteroaryl, optionally substituted -C(O)C6-I2 aryiaikyl, optionally substituted 3-10 membered

heterocyclyl, -OH, -NR31R32, -C(O)OR30, -CN, -N3, -C(= N R33JNR31R32,

-C(=NR33)OR30, -NR30C(=NR33)N R31R32, -NR31C(O)OR30, and -OC(O)NR31R32;

each R30, R31, and R32, independently is selected from the group consisting of H, optionally substituted C1-12 alkyl, optionally substituted C2-12 alkenyi, optionally substituted C2-I2 aikynyl, optionally substituted C3-12 cycioalkyi, optionally substituted C6-14 aryl, optionally substituted 3 - 14 membered heteroaryl, optionally substituted 3- 12 membered heterocyciyl, optionally substituted 3-18 membered heteroarylalkyf, and optionally substituted C6.18 aryiaikyl;

or R31 and R32 taken together with the atoms to which they are attached form a 3 to 10 membered heterocyclyl;

each R33 independently is selected from the group consisting of H, optionally substituted Ci-12 alkyl, optionally substituted C2-12 alkenyl, optionally substituted C2-I2 alkynyl, optionally substituted C3-12 cycioalkyi, optionally substituted C6-i4 aryl, optionally substituted 3 - 14 membered heteroaryl, optionally substituted 3-12 membered heterocyclyl, optionally substituted 3-18 membered heteroarylalkyl, optionally substituted C6-18 aryiaikyl, -CN, -C(O)R34, -CHO and -S(O)2R34;

each R34 individually is optionally substituted C1-12 alkyl;

wherein, each substituted Q3, substituted R30, substituted R31, substituted R32, substituted R33, or substituted R34 is independently substituted with one or more Q6; L is selected from the group consisting of -O-, -S-, -S(O)-, and -S(O)2-;

Het is an optionally substituted 3-12 membered heterocyclyl or optionally substituted 3-14 membered heteroaryl;

wherein, each substituted Het is substituted with one or more Q4;

each Q4, independently, is selected from the group consisting of

halogen, oxo, oxide, -NO2, -N(=0), -SR40, -S(O)R40, -S(O)2R40, -S(O)2NR40R41, -NR40C(O)R41, -NR40C(O)NR41R42, -NR40S(O)R41, -NR40S(O)2R41, -OP(O)R41R42, -P(O)R41R42, -P(O)OR41R42, -P(O)(OR41)OR42, -C(O)NR41R42, optionally substituted C1-6 alkyi, optionally substituted C2.6 aikenyi, optionally substituted C2-6 alkynyϊ, optionally substituted C3.6 cycioalkyl, optionally substituted C6-12 arylalkyl, optionally substituted C6-12 aryl, optionally substituted 3 - 14 membered heteroaryl, optionally substituted C^ alkyloxy, optionally substituted C2-6 alkenyloxy, optionally substituted C2-6 alkynyioxy, optionally substituted C3-6 cycloalkyloxy, optionally substituted C6-I2 aryioxy, optionally substituted 3 - 14 membered heteroaryloxy, optionally substituted 4-12 membered heterocyclyloxy, optionally substituted -C(O)C1^ alkyl, optionally substituted -C(O)C2-6 alkeny!, optionally substituted -C(O)C2-6 aikynyl, optionally substituted -C(O)C3-6 cycioalkyl, optionally substituted -C(O)C6-I2 aryl, optionally substituted -C(O)- 3 -14 membered heteroaryl, optionally substituted -C(O)C6--I2 arylalkyi, optionally substituted 3-10 membered

heterocyclyl, -OH, -NR41R42, -C(O)OR40, -CN, -N3, -C(=NR43)NR41R42,

-C(=NR43)OR40, -NR40C(=NR43)N R41R42, -NR41C(O)OR40, and -OC(O)NR41R42; each R40, R41, and R42, independently is selected from the group consisting of H, optionally substituted C1-12 aikyl, optionally substituted C2-12 alkeny!, optionally substituted C2-I2 aikynyl, optionally substituted C3-I2 cycioalkyl, optionally substituted C6-U aryl, optionally substituted 3 - 14 membered heteroaryl, optionally substituted 3- 12 membered heterocyclyl, optionally substituted 3-18 membered heteroarylalkyl, and optionally substituted C6--J8 arylalkyl;

or R41 and R42 taken together with the atoms to which they are attached form a 3 to 10 membered heterocyclyl;

each R43, independently, is selected from the group consisting of H, optionally substituted CM2 alkyl, optionally substituted C2-12 alkenyl, optionally substituted C2_i2 aikynyl, optionally substituted C3.12 cycioalkyl, optionally substituted C6-I4 aryl, optionally substituted 3 - 14 membered heteroaryl, optionally substituted 3-12 membered heterocyclyl, optionally substituted 3-18 membered heteroarylalkyl, optioπaily substituted C6.18 arylalkyl, -CN1 -C(O)R44, -CHO and -S(O)2R44;

each R44 individually is optionally substituted Cv12 alkyl;

wherein, each substituted Q4, substituted R40, substituted R41, substituted R42, substituted R43, or substituted R44 is independently substituted with one or more Q5; each Q5, individually, is selected from the group consisting of

halogen, oxo, oxide, -NO2, -N(=O), -SR50, -S(O)R50, -S(O)2R50, -S(O)2 NR50R51, -NR50C(O)R51, -NR50C(O)NR51R52, -NR50S(O)R51, -NR50S(O)2R51, -OP(O)R51R52, -P(O)R51R52, -P(O)OR51R52, -P(O)(OR51 )0R5Z, -C(O)NR51R52, optionally substituted C1-6 alkyl, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3-6 cycloalkyl, optionally substituted C6-I2 arylalkyl, optionally substituted C6-12 aryl, optionally substituted 3 - 14 membered heteroaryi, optionally substituted C^ alkyloxy, optionally substituted C2-6 alkenyloxy, optionally substituted C2-6 alkynyloxy, optionally substituted C3-6 cycloaSkyloxy, optionally substituted C6-12 aryioxy, optionally substituted 3 - 14 membered heteroaryloxy, optionally substituted 4-12 membered heterocyclyloxy, optionally substituted -C(O)C1-6 alkyl, optionally substituted -C(O)C2-B alkenyl, optionally substituted -C(O)C2-6 atkynyl, optionally substituted -C(O)C3-6 cycloaikyl, optionally substituted -C(O)C6-12 aryl, optionally substituted -C(O)- 3 -14 membered heteroaryl, optionally substituted -C(O)C6-I2 arylalkyl, optionally substituted 3-10 membered

heterocyclyl, -OH1 -NR51R52, -C(O)OR50, -CN, -N3, -C(=NR53)NR51R52,

-C(=NR53)OR50, -NR50C(=NR53)N R51R52, -NR51C(O)OR50, and -OC(O)NR51R52;

each R50, R51, and R52, independently is selected from the group consisting of H, optionally substituted C1-12 alkyl, optionally substituted C2-I2 alkenyl, optionally substituted C2-i2 alkynyl, optionally substituted C3-12 cycloalkyl, optionally substituted C6-14 aryl, optionally substituted 3 - 14 membered heteroaryl, optionally substituted 3- 12 membered heterocyclyl, optionally substituted 3-18 membered heteroarylalkyl, and optionally substituted C6_i8 arylalkyl;

or R51 and R52 taken together with the atoms to which they are attached form a 3 to 10 membered heterocyclyl;

each R53, independently, is selected from the group consisting of H, optionally substituted CM2 alkyl, optionally substituted C2-12 alkenyj, optionally substituted C2.12 alkynyl, optionally substituted C3-12 cycloalkyl, optionally substituted C6-14 aryl, optionally substituted 3 - 14 membered heteroary!, optionally substituted 3-12 membered heterocyclyl, optionally substituted 3-18 membered heteroaryialkyl, optionally substituted C6-18 aryla!kyi, -CN, -C(O)R54, -CHO and -S(O)2R54;

each R54, independently, is optionally substituted C1--I2 alky!;

wherein, each substituted Q5, substituted R50, substituted R51, substituted R52, substituted R53, or substituted R54 is independently substituted with one or more Q6; each Q6, independently, is selected from the group consisting of halogen, oxo, oxide, -NO2, -N{=0), -SR60, -S(O)R60, -S(O)2R60, -S(O)2 NR60 R61 , -NR60C(O)R61, -NR60C(O)NR61R62, -NR60S(O)R61 ,-NR60S(O)2R61, -OP(O)R61R62,

-P(O)R61R62, -P(O)OR61R62, -P(O)(OR61 )OR62, -C(O)NR61R62, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-6 cycloalkyl, C6-i2 arylalkyl, C6^2 a ry I, 3 - 14 membered heteroaryl, Ci-6 alkyloxy, C2-6 alkenyloxy, C2-6 alkynyioxy, C3-6 cycloalkyloxy, C6-i2 aryloxy, 3 - 14 membered heteroaryloxy, 4-12 membered heterocyctyloxy, -C(O)C1-6 aikyl, - C(O)C2-6 alkenyi, -C(O)C2-6 aikynyl, -C(O)C3-6 cycloalkyl, -C(O)C1-6

haioalkyi, -C(O)C6^12 aryl, -C(O)- 3-14 membered heteroaryl, -C(O)C6-12 arylalkyl, 3-10 membered heterocyciyl, -OH, -NR61R62, -C(O)OR60, -CN, -N3, -C(=NR63)NR61R62, -C(=NR63)OR60, -NR60C(=NR63)N R61R62, -NR61C(O)OR60, and -OC(O)NR61R62;

each R60, R61, and R62, independently, is selected from the group consisting of H, C1-12 alkyl, C2-12 alkenyl, C2.12 alkynyl, C3-12 cycloaikyl, d.12 haioalkyi, C6-i4 aryl, 3 - 14 membered heteroaryl, 3-12 membered heterocyclyl, 3-18 membered heteroaryialkyl, and C6-18 arylalkyl;

or R61 and R62 taken together with the atoms to which they are attached form a 3 to 10 membered heterocyciyl ;

each R63 independently is selected from the group consisting of H, C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl, C3-12 cycloaSkyl, C6-14 aryl, 3 - 14 membered heteroaryl, 3- 12 membered heterocyclyS, 3-18 membered heteroarySalkyl, C6-i8 arylalkyl, -CN, - C(O)R64, -CHO and -S(O)2R64; and

each R64 individually is C1-12 alkyl.

2. The compound of claim 1 , wherein R1 is optionally substituted C1-12 alkyl or optionally substituted C3-12 cycloalkyl.

3, The compound of claim 1 or 2, wherein R1 is optionally substituted C3-C7 secondary or tertiary alky! or optionally substituted C3-C5 cycloaikyl.

4. The compound of any one of claims 1-3, wherein R2 is optionally substituted methylcyclohexyl or optionally substituted methyicyciohexenyl.

5. The compound of any one of claims 1-4, wherein R3 is optionally substituted C1-12 alkylene, C3-12 cycloalkylene, substituted C3-12 cycloaikylene, optionally substituted C6-M arylene or optionally substituted 3 -12 membered heterocyclylene.

6. The compound of any one of claims 1-5, wherein Het is an optionally substituted 3-12 membered heterocyclyl or optionally substituted 3-14 membered heteroaryl wherein said optionally substituted 3-12 membered heterocyclyl or optionally substituted 3-14 membered heteroaryl comprises one to four heteroatomε selected from O, S, or N.

7. The compound of any one of claims 1-6, wherein Het is optionaliy substituted pyridinyl, optionally substituted pyridazinyl, optionally substituted tetrahydro-2H- pyranyl, optionally substituted piperidinyl, optionally substituted pyrroiidϊnyl, optionally substituted tetrahydrothiophenyi, optionally substituted pyrazinyl, optionally substituted 1H-tetrazolyl, optionally substituted azetidinyl, optionally substituted tetrahydrofuranyl, optionally substituted tetrahydro-2H-furo[2,3-b]furanyl, optionally substituted thiazoy!, optionally substituted IH-imidazolyl, optionally substituted 4H- 1,2,4-triazolyl, optionaliy substituted 1 H-pyrazolyi, optionally substituted 1 ,3,4- thiadiazolyl, optionally substituted quinolinyi, optionally substituted [1 ,2,4]triazolo[4,3- a]pyridinyl, optionaliy substituted thiophenyl, optionally substituted 1 ,2,4-thiadiazolyi, optionally substituted pyrimidinyl, optionally substituted 1 H-1 ,2,3-triazoiyl, optionaliy substituted 1 ,3,4-oxadiazolyl or optionally substituted tmidazo[1 ,2-b]pyridazinyl.

8. The compound of any one of claims 1-7 represented by Formula II:

Formula Il or pharmaceutically acceptable salts and esters thereof, wherein:

R2 is optionally substituted 4-methylcyclohexyi or optionally substituted

methylcyciohexenyi.

9. The compound of any one of claims 1-8, wherein R2 is

10. The compound of any one of claims 1-8, wherein R is

1 1. The compound of any one of claims 1-10, wherein R3 is optionally substituted cyclohexylene.

12. The compound of claim 1 that is

or ester thereof

13 A pharmaceutical composition compπsing a therapeutically effective amount of a compound of any one of claims 1-12 and a pharmaceutically acceptable carrier or excipient

14 The pharmaceutical composition of claim 13 further comprising at least one additional therapeutic agent selected from the group consisting of interferons, ribavirin or its analogs, HCV NS3 protease inhibitors, NS5a inhibitors, alpha- giucosidase 1 inhibitors, hepatoprotectants, mevalonate decarboxylase antagonists, antagonists of the renin-angiotensin system, endothelin antagonists, other anti- fibrotic agents, nucleoside or nucleotide inhibitors of HCV NS5B polymerase, non- nucleoside inhibitors of HCV NS5B polymerase, HCV NS5A inhibitors, TLR-7 agonists, cyclophillin inhibitors, HCV IRES inhibitors, pharmacokinetic enhancers and other drugs for treating HCV; or mixtures thereof.

15. A method for treating a Flaviviridae viral infection comprising administering a therapeutically effective amount of a compound or pharmaceutical composition of any one of claims 1-13 to a mammal in need thereof.

16. The method of claim 15 wherein the viral infection is caused by a Hepatitis C virus.

17. The method of claim 15 or 16 further comprising administering at least one additional therapeutic agent selected from the group consisting of interferons, ribavirin or its analogs, HCV NS3 protease inhibitors, NS5a inhibitors, alpha- glucosidase 1 inhibitors, hepatoprotectants, mevalonate decarboxylase antagonists, antagonists of the renin-angiotensin system, endothelin antagonists, other anti- fibrotic agents, nucleoside or nucleotide inhibitors of HCV NS5B polymerase, non- nucleoside inhibitors of HCV NS5B polymerase, HCV NS5A inhibitors, TLR-7 agonists, cyclophillin inhibitors, HCV IRES inhibitors, pharmacokinetic enhancers and other drugs for treating HCV; or mixtures thereof.

18. Use of a compound of any one of claims 1 -12 for the treatment of a

Flaviviridae viral infection or a Hepatitis C virus infection.

Description:
INHIBITORS OF FLA VIVIRIDAE VIRUSES

FIELD OF THE INVENTION

The present application includes novel inhibitors of Flaviviridae viruses, compositions containing such compounds, therapeutic methods that include the administration of such compounds.

BACKGROUND OF THE INVENTION

Viruses comprising the Flaviviridae family include at least three

distinguishable genera including pestiviruses, flaviviruses, and hepaciviruses

(Calisher, et al., J. Gen. Virol., 1993, 70, 37-43). While pestiviruses cause many economically important animal diseases such as bovine viral diarrhea virus (BVDV), classical swine fever virus (CSFV, hog cholera) and border disease of sheep (BDV), their importance in human disease is iess well characterized (Moennig, V., et al., Adv. VIr. Res. 1992, 48, 53-98). Flaviviruses are responsible for important human diseases such as dengue fever and yellow fever while hepaciviruses cause hepatitis C virus infections in humans. Other important virai infections caused by the

Flaviviridae family include West Ni!e virus (WNV) Japanese encephalitis virus (JEV), tick-borne encephalitis virus, Junjin virus, Murray Valley encephalitis, St Louis enchaplitis, Omsk hemorrhagic fever virus and Zika virus.

The hepatitis C virus (HCV) is the leading cause of chronic liver disease worldwide (Boyer, N. et al. J Hepatol. 32:98-1 12, 2000) so a significant focus of current antiviral research is directed toward the development of improved methods of treatment of chronic HCV infections in humans (Di Besceglie, A.M. and Bacon, B. R., Scientific American, Oct.: 80-85, (1999); Gordon, C. P., et al., J. Med. Chem. 2005, 48, 1-20; Maradpour, D.; et al., Nat. Rev. Micro, 2007, 5(6), 453-463). A number of HCV treatments are reviewed by Dymock et al. in Antiviral Chemistry &

Chemotherapy, 11 :2; 79-95 (2000). Virologic cures of patients with chronic HCV infection are difficult to achieve because of the prodigious amount of daily virus production in chronically infected patients and the high spontaneous mutability of HCV virus (Neumann, et al., Science 1998, 282, 103-7; Fukimoto, et al., Hepatology, 1996, 24, 1351-4; Domingo, et al., Gene, 1985, 40, 1-8; Martell, et al., J. Virol. 1992, 66, 3225-9.

Currently, there are primarily two antiviral compounds, ribavirin, a nucleoside analog, and interferon-aipha (α) (IFN), that are used for the treatment of chronic HCV infections in humans. Ribavirin alone is not effective in reducing viral RNA levels, has significant toxicity, and is known to induce anemia. The combination of IFN and ribavirin has been reported to be effective in the management of chronic hepatitis C (Scott, L. J., et al. Drugs 2002, 62, 507-556) but less than half the patients given this treatment show a persistent benefit.

Combined, infections from the Flaviviridae virus family cause significant mortality, morbidity and economic losses throughout the world. Alkynyl substituted thiophenes with virus activity have been disclosed by Chan, et al., WO 2008058393; Wunberg, et al., WO 2006072347; and Chan, et al., WO

2002100851 ; but none of these are currently clinically approved antiviral therapeutics. Therefore, there remains a need to develop effective treatments for Flaviviridae virus infections.

SUMMARY OF THE INVENTION

Provided are compounds of Formula i:

Formula (I),

or a pharmaceutically acceptable salt or ester thereof, wherein:

R 1 is selected from the group consisting of optionally substituted C 1-12 alkyl, optionally substituted C 2-I2 alkenyl, optionally substituted C 2 -i 2 alkynyi, optionally substituted C 3 ^ 12 cycloalkyl, optionally substituted C 6 -^ aryl, optionally substituted 3

14 membered heteroaryl, optionally substituted 3 -12 membered heterocyclyl, optionally substituted 3-18 membered heteroarylalkyi, optionaily substituted 3-18 membered heterocyciylafkyl and optionally substituted C 6 .-| 8 arylalkyj,

wherein, each substituted R 1 is substituted with one or more Q 1 ;

each Q 1 is independently selected from the group consisting of

halogen, oxo, oxide, -NO 2 , -N{=0), -SR 10 , -S(O)R 10 , -S(O) 2 R 10 , -S(O) 2 NR 10 R 11 , -NR 10 C(O)R 11 , -NR 10 C(O)NR 11 R 12 , -NR 10 S(O)R 11 , -NR 10 S(O) 2 R 11 , -OP(O)R 11 R 12 , -P(O)R 11 R 12 , -P(O)OR 11 R 12 , -P(O)(OR 11 )OR 12 , -C(O)NR 11 R 12 , optionally substituted C 1-6 aikyl, optionally substituted C 2-6 aikenyi, optionalfy substituted C 2- e alkyny!, optionally substituted C 3-6 cycloaikyl, optionally substituted C 6 - I2 arylalkyl, optionaily substituted C 6 - I2 aryl, optionally substituted 3 - 14 membered heteroaryl, optionally substituted Ci -6 alkyioxy, optionally substituted C 2-6 alkenyloxy, optionally substituted C 2-6 aikynyloxy, optionaily substituted C 3 . 6 cycloalkyioxy, optionally substituted C 6-12 aryloxy, optionally substituted 3 - 14 membered heteroaryloxy, optionally substituted 4-12 membered heterocyclyloxy, optionally substituted -C(O)C 1 - B alkyl, optionally substituted -C(O)C 2 -e aikenyi, optionally substituted -C(O)C 2-6 aikynyl, optionaily substituted -C(O)C 3^6 cycSoalkyi, optionaily substituted -C(O)C 6-12 aryl, optionally substituted -C(0)-3 - 14 membered heteroaryl, optionaily substituted -C(O)C 6-I2 arylalkyl, optionaliy substituted -3-10 membered

heterocyciyl, -OH, -NR 11 R 12 , -C(O)OR 10 , -CN, -N 3 , -C(=NR 13 )NR 11 R 12 ,

-C(=NR 13 )OR 10 , -NR 10 C(=NR 13 )NR 11 R 12 , -NR 11 C(O)OR 10 , and -OC(O)NR 11 R 12 ;

each R 10 , R 11 , and R 12 , independently, is selected from the group consisting of H, optionally substituted C 1-12 alkyl, optionaliy substituted C 2 . 12 aikenyi, optionally substituted C 2 _i 2 aikynyl, optionally substituted C 3-12 cycSoalkyi, optionaily substituted C 6-14 aryl, optionaily substituted 3 - 14 membered heteroaryl, optionaily substituted 3- 12 membered heterocyclyi, optionally substituted 3-18 membered heteroarytalkyl, and optionally substituted C 6-16 arylalkyl;

or R 11 and R 12 taken together with the atoms to which they are attached form a 3 to 10 membered heterocyciyl;

each R 13 , independently, is selected from the group consisting of H, optionally substituted C 1-12 aikyi, optionally substituted C 2-12 aikenyi, optionally substituted C 2-12 aikynyl, optionally substituted C 3 . 12 cycioalkyl, optionaily substituted C 6-I4 aryl, optionally substituted 3 - 14 membered heteroaryl, optionaily substituted 3-12 membered heterocyclyi, optionaliy substituted 3-18 membered heteroarylalkyl, optionaliy substituted C 6-18 aryialkyi, -CN, -C(O)R 14 , -CHO and -S(O) 2 R 14 ; each R 14 , independently, is optionally substituted C M2 alkyl;

wherein, each substituted Q 1 , substituted R 10 , substituted R 11 , substituted R 12 , substituted R 13 , or substituted R 14 is independently substituted with one or more Q 6 ;

R 2 is selected from the group consisting of optionally substituted C 1-12 alkyl, optionally substituted C 2 - 12 aikenyl, optionally substituted C 2 - I2 alkynyl, optionally substituted C 3-12 cycloalkyl, optionally substituted C 6- i 4 aryl, optionally substituted 3 - 14 membered heteroaryl, optionally substituted 3 -12 membered heterocyclyl, optionally substituted 3-18 membered heteroarylalkyl, and optionally substituted C 6- is arylalkyl;

wherein, each substituted R 2 is substituted with one or more Q 2 ;

each Q 2 , independently, is selected from the group consisting of

halogen, oxo, oxide, -NO 2 , -N(=O), -SR 20 , -S(O)R 20 , -S(O) 2 R 20 , -S(O) 2 NR 20 R 21 , -NR 20 C(O)R 21 , -NR 20 C(O)NR 21 R 22 , -NR 20 S(O)R 21 , -NR 20 S(O) 2 R 21 , -OP(O)R 21 R 22 , -P(O)R 21 R 22 , -P(O)OR 21 R 22 , -P(O)(OR 21 )OR 22 , -C(O)NR 21 R 22 , optionally substituted C 1-6 alkyi, optionally substituted C 2-6 aikenyl, optionally substituted C 2 - 6 aSkynyl, optionally substituted C 3-6 cycloaikyi, optionally substituted C 6- i 2 arylalkyl, optionally substituted C 6-12 aryl, optionally substituted 3 - 14 membered heteroaryl, optionally substituted C 1-e alkyloxy, optionally substituted C 2-6 alkenyloxy, optionally substituted C 2 ^ 6 alkynyloxy, optionally substituted C 3-6 cycloalkyloxy, optionally substituted C 6-12 aryloxy, optionally substituted 3 - 14 membered heteroaryloxy, optionally substituted 4-12 membered heterocyclyioxy, optionally substituted -C(O)C 1-6 alkyl, optionally substituted -C(O)C 2 ^ aikenyl, optionally substituted -C(O)C 2-6 alkynyl, optionally substituted -C(O)C 3-6 cycioalkyl, optionally substituted -C(O)C 6-I2 aryl, optionally substituted -C(0)-3 -14 membered heteroaryi, optionally substituted -C(O)C 6 - I2 arylalkyl, optionally substituted 3-10 membered

heterocyclyl, -OH, -NR 21 R 22 , -C(O)OR 20 , -CN, -N 3 , -C(=NR 23 )NR 21 R 22 ,

-C(=NR 23 )OR 20 , -NR 20 C(=NR 23 )N R 21 R 22 , -NR 21 C(O)OR 20 , and -OC(O)NR 21 R 22 ;

each R 20 , R Z1 , and R 22 , independently, is selected from the group consisting of H, optionally substituted Ci -12 alkyl, optionally substituted C 2-12 aikenyl, optionally substituted C 2-12 alkynyl, optionally substituted C 3 ^ 2 cycloalkyl, optionally substituted C 6-1 4 aryl, optionally substituted 3 - 14 membered heteroaryl, optionally substituted 3- 12 membered heterocyclyl, optionally substituted 3-18 membered heteroarylalkyl, and optionally substituted C 6- - I8 arylalkyl; or R 21 and R 22 taken together with the atoms to which they are attached form a 3 to 10 membered heterocyclyl;

each R 23 , independently, is selected from the group consisting of H, optionally substituted C 1 ^ 2 alkyl, optionally substituted C 2- I 2 alkenyl, optionally substituted C 2 -i 2 aikyπyl, optionally substituted C 3 . 12 cycloalkyl, optionally substituted C 6-14 aryl, optionally substituted 3 - 14 membered heteroaryl, optionally substituted 3-12 membered heterocyclyi, optionally substituted 3-18 membered heteroarylalkyl, optionally substituted C 6-18 arylalkyl, -CN, -C(O)R 24 , -CHO and -S(O) 2 R 24 ;

each R 24 individually is optionally substituted C 1-I2 alky!;

wherein, each substituted Q 2 , substituted R 20 , substituted R 21 , substituted R 22 , substituted R 23 , or substituted R 24 is independently substituted with one or more Q 6 ;

R 3 is selected from the group consisting of optionally substituted C 1- - I2 alkylene, C 2 - I2 alkenylene, substituted C 2- - I2 alkenySene, C 2 - I2 alkynylene, substituted C 2-12 alkynylene, C 3-12 cycloalkylene, substituted C 3- - I2 cycloaikylene, optionaiiy substituted C 6- - U arylene, optionally substituted 3 - 14 membered heteroarylene, optionally substituted 3 -12 membered heterocyclylene, optionally substituted 3-18 membered heteroarylaSkylene, and optionally substituted C 6-1S arylalkylene;

wherein each substituted R 3 is substituted with one or more Q 3 ;

each Q 3 , independently, is selected from the group consisting of

halogen, oxo, oxide, -NO 2 , -N(O), -SR 30 , -S(O)R 30 , -S(O) 2 R 30 , -S(O) 2 NR 30 R 31 , -NR 30 C(O)R 31 , -NR 30 C(O)NR 31 R 32 , -NR 30 S(O)R 31 , -NR 30 S(O) 2 R 31 , -OP(O)R 31 R 32 , -P(O)R 31 R 32 , -P(O)OR 31 R 32 , -P(O)(OR 31 )OR 32 , -C(O)NR 31 R 32 , optionally substituted C 1-6 alkyl, optionally substituted C 2 ^ alkeny!, optionally substituted C 2 . 6 alkynyl, optionally substituted C 3-6 cycloalkyl, optionally substituted C 6 - I2 arylaikyl, optionally substituted C 6- - I2 aryl, optionally substituted 3 - 14 membered heteroary!, optionaiiy substituted C 1-6 a!kyloxy, optionally substituted C 2-6 alkenyloxy, optionaiiy substituted C 2-6 alkynyioxy, optionally substituted C 3 . 6 cycloalkyioxy, optionally substituted C 6 . 12 aryloxy, optionaiiy substituted 3 - 14 membered heteroaryloxy, optionaiiy substituted 4-12 membered heterocyclyloxy, optionally substituted -C(O)C 1-6 alkyl, optionally substituted -C(O)C 2-6 alkenyl, optionally substituted -C(O)C 2-6 alkynyl, optionally substituted -C(O)C 3-6 cycloaikyl, optionally substituted -C(O)C 6-I2 aryl, optionally substituted -C(O)- 3 -14 membered heteroaryl, optionally substituted -C(O)C 6-I 2 arylalkyl, optionally substituted 3-10 membered

heterocyclyi, -OH, -NR 31 R 32 , -C(O)OR 30 , -CN, -N 3 , -C(=NR 33 )NR 31 R 32 , -C(=NR 33 )OR 30 , -NR 30 C(=NR 33 )N R 31 R 32 , -NR 31 C(O)OR 30 , and -OC(O)NR 31 R 32 ;

each R 30 , R 31 , and R 32 , independently is selected from the group consisting of H, optionaily substituted C 1-12 alky!, optionally substituted C 2-I2 alkenyl, optionally substituted C 2- i 2 alkyny!, optionally substituted C 3-12 cycloalkyl, optionally substituted C 6-14 aryl, optionally substituted 3 - 14 membered heteroaryf, optionally substituted 3- 12 membered heterocyclyl, optionally substituted 3-18 membered heteroarylalkyl, and optionally substituted C 6-1S aryialkyl;

or R 31 and R 32 taken together with the atoms to which they are attached form a 3 to 10 membered heterocyclyl;

each R 33 independently is selected from the group consisting of H, optionally substituted C 1^12 alkyi, optionally substituted C 2-I2 alkenyl, optionally substituted C 2 - 12 alkynyl, optionally substituted C 3-12 cycloalkyl, optionally substituted C 6-14 aryl, optionally substituted 3 - 14 membered heteroaryi, optionally substituted 3-12 membered heterocyclyl, optionally substituted 3-18 membered heteroarylalkyl, optionally substituted C 6 . aryialkyl, -CN, -C(O)R 34 , -CHO and -S(O) 2 R 34 ;

each R 34 individually is optionally substituted C M2 alkyl;

wherein, each substituted Q 3 , substituted R 30 , substituted R 31 , substituted R 32 , substituted R 33 , or substituted R 34 is independently substituted with one or more Q 6 ;

L is selected from the group consisting of -0-, -S-, -S(O)-, and -S(O) 2 -;

Het is an optionally substituted 3-12 membered heterocyclyl or optionally substituted 3-14 membered heteroaryi;

wherein, each substituted Het is substituted with one or more Q 4 ;

each Q 4 , independently, is selected from the group consisting of

halogen, oxo, oxide, -NO 2 , -N(=0), -SR 40 , -S(O)R 40 , -S(O) 2 R 40 , -S(O) 2 NR 40 R 41 , -NR 40 C(O)R 41 , -NR 40 C(O)NR 41 R 42 , -NR 40 S(O)R 41 , -NR 40 S(O) 2 R 41 , -OP(O)R 41 R 42 , -P(O)R 41 R 42 , -P(O)OR 41 R 42 , -P(O)(OR 41 )OR 42 , -C(O)NR 41 R 42 , optionally substituted C-i-e alkyl- optionally substituted C 2 . 6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted C 3-6 cycloalkyl, optionaily substituted C 6-12 aryialkyl, optionaily substituted C 6-I 2 aryl, optionally substituted 3 - 14 membered heteroaryi, optionally substituted C-i-ealkyloxy, optionally substituted C 2 - 6 alkenyloxy, optionally substituted C 2 -6 alkynyioxy, optionally substituted C 3 . 6 cycloalkyloxy, optionally substituted C 6 - I2 aryloxy, optionally substituted 3 - 14 membered heteroaryloxy, optionally substituted 4-12 membered heterocyclyloxy, optionally substituted -C(O)Ci -6 alkyl, optionally substituted -C(O)C 2-6 alkenyl, optionally substituted -C(O)C 2 ^ alkynyi, optionally substituted -C(O)C 3-6 cycioalkyl, optionally substituted -C(O)C 6-12 aryl, optionally substituted -C(O)- 3 -14 membered heteroaryi, optionally substituted -C(O)C 6-I2 aryialkyl, optionally substituted 3-10 membered

heterocyclyl, -OH, -NR 41 R 42 , -C(O)OR 40 , -CN, -N 3 , -C(=NR 43 )NR 41 R 42 ,

-C(=NR 43 )OR 40 , -NR 40 C(=NR 43 )N R 41 R 42 , -NR 41 C(O)OR 40 , and -OC(O)NR 41 R 42 ;

each R 40 , R 41 , and R 42 , independently is selected from the group consisting of H, optionally substituted d. 12 alky!, optionally substituted C 2-I 2 alkenyl, optionally substituted C 2 . 12 alkynyl, optionally substituted C 3^12 cycloalkyl, optionally substituted C 6-14 aryl, optionally substituted 3 - 14 membered heteroaryi, optionally substituted 3- 12 membered heterocyciyi, optionally substituted 3-18 membered heteroaryialkyl, and optionally substituted C 6-I8 aryialkyl;

or R 41 and R 42 taken together with the atoms to which they are attached form a 3 to 10 membered heterocyciyi;

each R 43 , independently, is selected from the group consisting of H, optionally substituted C 1-12 alkyl, optionally substituted C 2- i 2 alkenyl, optionally substituted C 2-I2 alkynyl, optionaliy substituted C 3 . 12 cycloalkyl, optionally substituted C 6-14 aryl, optionally substituted 3 - 14 membered heteroaryi, optionally substituted 3-12 membered heterocyciyi, optionally substituted 3-18 membered heteroaryialkyl, optionally substituted C 6-18 arylaikyS, -CN 1 -C(O)R 44 , -CHO and -S(O) 2 R 44 ;

each R 44 individually is optionally substituted C t- i 2 alky!;

wherein, each substituted Q 4 , substituted R 40 , substituted R 41 , substituted R 42 , substituted R 43 , or substituted R 44 is independently substituted with one or more Q 5 ; each Q 5 , individually, is selected from the group consisting of

halogen, oxo, oxide, -NO 2 , -N(=0), -SR 50 , -S(O)R 50 , -S(O) 2 R 50 , -S(O) 2 NR 50 R 51 , -NR 50 C(O)R 51 , -NR 50 C(O)NR 51 R 52 , -NR 50 S(O)R 51 , -NR 50 S(O) 2 R 51 , -OP(O)R 51 R 52 , -P(O)R 51 R 52 , -P(O)OR 51 R 52 , -P(O)(OR 51 )OR 52 , -C(O)NR 51 R 52 , optionally substituted C 1-6 atkyl, optionaliy substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted C 3-6 cycloalkyl, optionally substituted C 6-12 aryialkyl, optionally substituted Ce -I2 aryl, optionally substituted 3 - 14 membered heteroaryi, optionally substituted Ci -6 alkyloxy, optionally substituted C 2 - 6 alkenyloxy, optionally substituted C 2-6 alkynyloxy, optionally substituted C 3-6 cycloaikyloxy, optionally substituted C 6 . 12 aryloxy, optionally substituted 3 - 14 membered heteroaryloxy, optionally substituted 4-12 membered heterocyclyioxy, optionally substituted -C(O)Cv 6 alkyl, optionally substituted -C(O)C 2-6 alkenyl, optionally substituted -C(O)C 2-6 alkynyl, optionally substituted -C(O)C 3-6 cycloalkyi, optionally substituted -C(O)C 6-I2 aryi, optionaliy substituted -C(O)- 3 -14 membered heteroaryf, optionally substituted -C(O)C 6-I2 arylalkyl, optionally substituted 3-10 membered

heterocyclyl, -OH, -NR 51 R 52 , -C(O)OR 50 , -CN, -N 3 , -C(=NR 53 )NR 51 R 52 ,

-C(=NR 53 )OR 50 , -NR 50 C(=NR 53 )N R 51 R 52 , -NR 51 C(O)OR 50 , and -OC(O)NR 51 R 52 ;

each R 50 , R 51 , and R 52 , independently is selected from the group consisting of H, optionaliy substituted C 1-12 alkyl, optionally substituted C 2-I 2 alkenyl, optionally substituted C 2-I2 alkynyl, optionally substituted C 3-12 cycloalkyi, optionally substituted C 6 -i 4 aryl, optionally substituted 3 - 14 membered heteroaryl, optionally substituted 3- 12 membered heterocyclyl, optionally substituted 3-18 membered heteroarylalky!, and optionally substituted C 6 .i 8 arylalkyl;

or R 51 and R 52 taken together with the atoms to which they are attached form a 3 to 10 membered heterocyclyl;

each R 53 , independently, is selected from the group consisting of H, optionally substituted Cv 12 alkyl, optionally substituted C 2-12 alkenyl, optionally substituted C 2-12 alkynyl, optionally substituted C 3 - 12 cycloalkyi, optionally substituted C 6-14 aryl, optionally substituted 3 - 14 membered heteroaryl, optionally substituted 3-12 membered heterocyclyl, optionally substituted 3-18 membered heteroarylalkyl, optionaily substituted C 6 . 18 arylalkyl, -CN, -C(O)R 54 , -CHO and -S(O) 2 R 54 ;

each R 54 , independently, is optionally substituted C 1-12 alkyl;

wherein, each substituted Q 5 , substituted R 50 , substituted R 51 , substituted R 52 , substituted R 53 , or substituted R 54 is independently substituted with one or more Q 6 ; each Q 6 , independently, is selected from the group consisting of halogen, oxo, oxide, -NO 2 , -N(=O), -SR 60 , -S(O)R 60 , -S(O) 2 R 60 , -S(O) 2 NR 60 R 61 , -NR 60 C(O)R 61 , -NR 60 C(O)NR 61 R 62 , -NR 60 S(O)R 6 VNR 60 S(O) 2 R 61 , -OP(O)R 61 R 62 ,

-P(O)R 61 R 62 , -P(O)OR 61 R 62 , -P(O)(OR 61 )OR 62 , -C(O)NR 61 R 62 , C 1 * alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyi, C 6- I 2 arylalkyl, C 6-12 aryl, 3 - 14 membered heteroaryl, Ci -6 alkyloxy, C 2 * alkenySoxy, C 2 - 6 alkynyloxy, C 3 . 6 cycloalkyloxy, C 6 . 12 aryioxy, 3 - 14 membered heteroaryloxy, 4-12 membered heterocyclyloxy, -C(O)C 1-6 alkyl, - C(O)C 2-6 alkenyS, -C(O)C 2-6 alkynyl, -C(O)C 3-6 cycloalkyi, -C(O)C 1-6

haloalkyl, -C(O)C 6-12 aryl, -C(O)- 3-14 membered heteroaryi, -C(O)C 6 . 12 aryiaikyl, 3-10 membered heterocyclyl, -OH, -NR 61 R 62 , -C(O)OR 60 , -CN, -N 3 , -C(=NR 63 )NR 61 R 62 , -C(=NR 63 )OR 60 , -NR 60 C(=NR 63 )N R 61 R 62 , -NR 61 C(O)OR 60 , and -OC(O)NR 61 R 62 ; each R 60 , R 81 , and R 62 r independently, is selected from the group consisting of H, Ci -12 alkyl, C2-12 alkenyl, C 2 -i 2 alkynyl, C 3-12 cycfoaikyl, C 1-I2 haioalkyl, C 6-H aryl, 3 - 14 membered heteroaryl, 3-12 membered heterocycfyl, 3-18 membered heteroarylalkyl, and C 6 .^ arylalkyl;

or R 61 and R 62 taken together with the atoms to which they are attached form a 3 to 10 membered heterocyclyl;

each R 63 independently is selected from the group consisting of H, C 1-12 alkyl, C 2-12 alkenyl, C 2-12 alkynyl, C 3-12 cycioalkyl, C 6-I4 aryl, 3 - 14 membered heteroaryl, 3- 12 membered heterocyclyi, 3-18 membered heteroarylalkyl, C 6-18 arylalkyl, -CN, - C(O)R 64 , -CHO and -S(O) 2 R 64 ; and

each R 64 individually is C 1-12 alkyl.

In another aspect, a method for treating Flaviviridae viral infections is provided comprising administering a therapeutically effective amount of a compound of Formula I to a mammal in need thereof. The compound of Formula I is

administered to a human subject in need thereof, such as a human being who is infected with viruses of the Flaviviridae family. In another embodiment, the compound of Formula I is administered to a human subject in need thereof, such as a human being who is infected with a HCV virus. In one embodiment, the treatment results in the reduction of one or more of the in viral loads or clearance of viral RNA in a patient.

In another embodiment, provided is a method of treating and/or preventing a disease caused by a viral infection wherein the viral infection is caused by a virus selected from the group consisting of dengue virus, yellow fever virus, West Nile virus, Japanese encephalitis virus, tick-borne encephalitis virus, Junjin virus, Murray Valley encephalitis virus, St Louis encephalitis virus, Omsk hemorrhagic fever virus, bovine viral disarrhea virus, Zika virus and Hepatitis C virus; by administering to a subject in need thereof a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt or ester thereof.

In another aspect, provided is the use of a compound of Formula I for the manufacture of a medicament for the treatment of Flaviviridae viral infections. In another aspect, provided is a compound of Formula I for use in treating a Flaviviridae viral infection. In one embodiment, the Flaviviridae viral infection is acute or chronic HCV infection. In one embodiment of each aspect of use and compound, the treatment results in the reduction of one or more of the viral loads or clearance of RNA in the patient.

In another aspect, provided is a method for treating or preventing HCV comprising administering an effective amount of a compound of Formula I to a patient in need thereof. In another aspect, provided is the use of a compound of the present invention for the manufacture of a medicament for the treatment or prevention of HCV.

In another aspect, provided is a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt or ester thereof and one or more pharmaceutically acceptable carriers or excipients. The pharmaceutical composition of Formula I may further comprise one or more additional therapeutic agents. The one or more additional therapeutic agent may be, without limitation, selected from: interferons, ribavirin or its analogs, HCV NS3 protease inhibitors, NS5a inhibitors, alpha-glucosidase 1 inhibitors, hepatoprotectants, mevaionate decarboxylase antagonists, antagonists of the renin-angiotensin system, other anti- fibrotic agents, endothelin antagonists, nucleoside or nucleotide inhibitors of HCV NS5B polymerase, non-nucleoside inhibitors of HCV NS5B polymerase, HCV NS5A inhibitors, TLR-7 agonists, cyclophillin inhibitors, HCV IRES inhibitors,

pharmacokinetic enhancers and other drugs for treating HCV; or mixtures thereof. in another aspect, provided is a method for the treatment or prevention of the symptoms or effects of an HCV infection in an infected animal which comprises administering to, i.e. treating, said animal with a pharmaceutical combination composition or formulation comprising an effective amount of a Formula I compound, and a second compound having anti-HCV properties.

In another embodiment, provided are compounds of Formula I and

pharmaceutically acceptable salts and esters thereof and all racemates, enantiomers, diastereomers, tautomers, polymorphs, pseudopolymorphs and amorphous forms thereof.

In another aspect, provided are processes and novel intermediates disclosed herein which are useful for preparing Formula I compounds.

In other aspects, novel methods for synthesis, analysts, separation, isolation, purification, characterization, and testing of the compounds of Formula I are provided.

The present invention includes combinations of aspects and embodiments, as weil as preferences, as herein described throughout the present specification. DETAILED DESCRIPTION

Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated in the accompanying structures and formulas. While the invention will be described in conjunction with the enumerated

embodiments, it will be understood that they are not intended to limit the invention to those embodiments. On the contrary, the invention is intended to cover all alternatives, modifications, and equivalents, which may be included within the scope of the present invention as defined herein.

Each document referenced herein is incorporated by reference in its entirety for all purposes.

In one embodiment of Formula i, R 1 is optionally substituted C 1 ^ 2 alkyl, optionally substituted C 2 - 12 afkenyl, optionally substituted C 2-12 alkynyl, or optionally substituted C 3-12 cycloalkyl. In another aspect of this embodiment, R 1 is optionally substituted C 1 -C 12 alkyl. In another aspect of this embodiment, R 1 is optionally substituted C 3 -C 7 secondary or tertiary alkyl. in another aspect of this embodiment, R 1 is optionally substituted C 3 -C 5 cycloalkyl. in another aspect of this embodiment, R 1 is prop-2-yl (isopropyl) or 2-methylprop-2-yl (/--butyl).

In another embodiment of Formula I, R 2 is optionally substituted C^ 12 alkyi, optionally substituted C 2-I2 alkenyl, optionally substituted C 2^12 alkynyl, optionally substituted C 3 . 12 cycloalkyl, or optionally substituted C 6-16 arylalkyl. In another aspect of this embodiment, R 2 is optionally substituted C 3-12 cycloalkyl. In another aspect of this embodiment, R 2 is optionally substituted methylcyclohexyl. In another aspect of this embodiment, R 2 is optionally substituted methylcyclohexenyi. In another aspect of this embodiment, R 2 is optionally substituted 4-methylcyclohexyi. In a preferred aspect of this embodiment, R 2 is

In another preferred aspect of this embodiment, R is Sn another preferred aspect of this embodiment, R 2 is

In another preferred aspect of this embodiment, R 2 is

in another embodiment of Formuia I, R 3 is optionaliy substituted C 1-12 aikylene, C3- 12 cycloalkylene, substituted C 3- I 2 cycloalkylene, optionally substituted C 6-14 arylene or optionally substituted 3 -12 membered heterocyclylene. In another aspect of this embodiment, R 3 is optionally substituted C 2 -C 6 aikylene. In another aspect of this embodiment, R 3 is optionally substituted C 3 -C 6 cycloalkylene. In another aspect of this embodiment, R 3 is optionally substituted 4 -6 membered heterocyciylene. In another aspect of this embodiment, R 3 is an optionally substituted 4-6 membered nitrogen-containing heterocyclylene.

In one embodiment of Formula I, L is— O-. In another embodiment of Formula I, L is -S-. In another embodiment of Formula I, L is -S(O)-. In another embodiment of Formula I, L is -S(O) 2 -.

in another embodiment of Formula I, Het is an optionally substituted 3-12 membered heterocyclyl or optionally substituted 3-14 membered heteroaryl wherein said optionally substituted 3-12 membered heterocyciyl or optionally substituted 3-14 membered heteroaryi comprises one to four heteroatoms selected from O 1 S, or N. In another aspect of this embodiment, Het is an optionally substituted 3-12

membered heterocyclyi comprising one or two heteroatoms selected from O 5 S, or N. In another aspect of this embodiment, Het is an optionally substituted 5-10

membered heteroaryl comprising one to four hetroatoms selected from O, S, or N. In another aspect of this embodiment, Het is optionally substituted pyridinyl. In another aspect of this embodiment, Het is optionaliy substituted pyridazinyl. In another aspect of this embodiment, Het is optionally substituted tetrahydro-2H-pyranyl. In another aspect of this embodiment, Het is optionally substituted piperidinyl. In another aspect of this embodiment, Het is optionally substituted pyrrolidinyi. In another aspect of this embodiment, Het is optionally substituted tetrahydrothiophenyl. In another aspect of this embodiment, Het is optionally substituted pyrazinyl. In another aspect of this embodiment, Het is optionally substituted 1H-tetrazolyl. In another aspect of this embodiment, Het is optionally substituted azetidinyl. In another aspect of this embodiment, Het is optionally substituted tetrahydrofuranyl. in another aspect of this embodiment, Het is optionally substituted tetrahydro-2/-/- furo[2,3-b]furanyl. In another aspect of this embodiment, Het is optionally substituted thiazoyl. In another aspect of this embodiment, Het is optionally substituted 1H- imidazolyl. In another aspect of this embodiment, Het is optionally substituted AH- 1 ,2,4-triazolyl. In another aspect of this embodiment, Het is optionally substituted 1 tf-pyrazolyl. in another aspect of this embodiment, Het is optionally substituted

1 ,3,4-thiadiazolyl. In another aspect of this embodiment, Het is optionally substituted quinolinyl. In another aspect of this embodiment, Het is optionally substituted

[1 ,2,4]triazolo[4,3-a]pyridinyi. In another aspect of this embodiment, Het is optionally substituted thiophenyl. in another aspect of this embodiment, Het is optionally substituted 1 ,2,4-thiadiazolyl. In another aspect of this embodiment, Het is optionally substituted pyrimidinyl. In another aspect of this embodiment, Het is optionally substituted 1 H- 1 ,2,3-triazolyl. In another aspect of this embodiment, Het is optionally substituted 1 ,3,4-oxadiazolyl. In another aspect of this embodiment, Het is optionally substituted imidazo[1 ,2-b]pyridazinyi.

In another embodiment of Formula I, R 1 is optionally substituted C 1 -C 12 alkyl,

In another aspect of this embodiment, R 2 is optionally substituted methylcyclohexyl or optionally substituted methylcyclohexenyl. In another aspect of this embodiment, R 3 is optionally substituted C 1-12 alkylene, C 3-12 cycloalkylene, substituted C 3- - I2 cycloalkylene, optionally substituted C 6 - I4 arylene or optionally substituted 3 -12 membered heterocyclylene. In another aspect of this embodiment, Het is an optionally substituted 3-12 membered heterocyclyl or optionally substituted 3-14 membered heteroaryl wherein said optionally substituted 3-12 membered

heterocyclyl or optionally substituted 3-14 membered heteroaryl comprises one to four heteroatoms selected from O, S 1 or N. In another aspect of this embodiment, Het is optionally substituted pyridinyl, optionally substituted pyridazinyl, optionally substituted tetrahydro-2/-/-pyranyl, optionally substituted piperidinyl, optionally substituted pyrrolidinyl, optionally substituted tetrahydrothiophenyi, optionally substituted pyrazinyl, optionally substituted 1H-tetrazolyl, optionally substituted azetidinyl, optionally substituted tetrahydrofuranyl, optionally substituted tetrahydro- 2H-furo[2,3-b]furanyS, optionally substituted thiazoyl, optionally substituted 1H- imidazolyl, optionally substituted 4H-1 ,2,4-triazolyf, optionally substituted 1H- pyrazolyi, optionally substituted 1 ,3,4-thiadiazolyl, optionally substituted quinolinyl, optionally substituted [1 ,2,4]triazoio[4,3-a]pyridinyl, optionally substituted thiophenyl, optionally substituted 1 ,2,4-thiadiazolyl, optionally substituted pyrimidinyl, optionally substituted 1H-1 ,2,3-triazolyl, optionally substituted 1 ,3,4-oxadiazoly! or optionally substituted imidazo[1 ,2-b]pyridazinyi. In another aspect of this embodiment, L is -O. In another aspect of this embodiment, L is -S-. In another aspect of this embodiment, L is -S(O)-. In another aspect of this embodiment, L is -S(O) 2 -.

In another embodiment of Formula I 1 R 1 is optionally substituted C 3-12 cycloaikyl. in another aspect of this embodiment, R z is optionally substituted methylcyclohexy! or optionally substituted methylcyclohexenyl. In another aspect of this embodiment, R 3 is optionally substituted C 1-12 alkylene, C 3-12 cycloalkylene, substituted C 3 .-^ cycioalkylene, optionally substituted C 6 -i 4 arylene or optionally substituted 3 -12 membered heterσcyclylene. In another aspect of this embodiment, Het is an optionally substituted 3-12 membered heterocyclyl or optionally substituted 3-14 membered heteroaryl wherein said optionally substituted 3-12 membered heterocyclyl or optionally substituted 3-14 membered heteroaryl comprises one to four heteroatoms selected from O, S, or N. in another aspect of this embodiment, Het is optionally substituted pyrtdtnyl, optionally substituted pyridazinyl, optionally substituted tetrahydro-2H-pyranyl, optionally substituted piperidinyl, optionally substituted pyrrolidinyl, optionally substituted tetrahydrothiophenyl, optionally substituted pyrazinyl, optionally substituted 1H-tetrazolyl, optionally substituted azetidinyl, optionally substituted tetrahydrofuranyl, optionally substituted tetrahydro- 2/-/-furo[2,3-b]furanyl, optionally substituted thiazoyl, optionally substituted 1 H- imidazolyl, optionally substituted 4H-1 ,2,4-triazolyl, optionally substituted 1H- pyrazolyl, optionally substituted 1 ,3,4-thiadiazolyl, optionally substituted quinolinyl, optionally substituted [1 ,2,43triazolo[4,3-a]pyridinyl, optionally substituted thiophenyl, optionally substituted 1 ,2,4-thiadiazolyl, optionally substituted pyrimidinyl, optionally substituted 1H-1 ,2,3-triazoiyi, optionally substituted 1 ,3,4-oxadiazolyl or optionally substituted imidazo[1 ,2-b]pyridazinyl. in another aspect of this embodiment, L is -0-. In another aspect of this embodiment, L is -S-. In another aspect of this embodiment, L is -S(O)-. In another aspect of this embodiment, L is -S(O) 2 -. in another embodiment of Formula !, R 1 is optionally substituted C 1 -C 12 alkyl and R 2 is optionally substituted C 3-12 cycloalkyl. In another aspect of this embodiment, R 2 is optionally substituted methylcyclohexyl or optionally substituted

methylcydohexenyl. in another aspect of this embodiment, R 3 is optionally substituted C M2 alkylene, C 2 _i 2 alkenylene, substituted C 2 -u alkenylene, C 2-I2 alkynylene, substituted C 2 - I2 alkynylene, C 3-12 cycloaikylene, substituted C 3-12 cycloalkyiene, optionally substituted C 6 . 14 arylene, optionally substituted 3-14 membered heteroarylene, optionally substituted 3 -12 membered heterocyclylene, optionally substituted 3-18 membered heteroarylalkylene, or optionally substituted C 6 . 18 arylalkylene. In another aspect of this embodiment, R 3 is optionally substituted C 1 . - I2 alkylene, C 3 ^ 12 cycloaikylene, substituted C 3 .i 2 cycloalkyiene, optionally substituted C 6- i 4 arylene, or optionally substituted 3 -12 membered heterocyclylene. In another aspect of this embodiment, L is -O-, -S-, -S(O)- or -S(O) 2 -. In another aspect of this embodiment, L is -O-. In another aspect of this embodiment, L is -S-. In another aspect of this embodiment, L is -S(O)-. In another aspect of this embodiment, L is - S(O) 2 -.

In another embodiment of Formula I, R 1 is optionally substituted C 3 -Ci 2 cycloaikyl and R 2 is optionally substituted C 3 . 12 cycloalkyl. In another aspect of this embodiment, R 2 is optionally substituted methylcyclohexyl or optionally substituted methylcydohexenyl. In another aspect of this embodiment, R 3 is optionally

substituted C M2 alkylene, C 2- - I2 alkenylene, substituted C 2-12 alkenylene, C 2-12 alkynylene, substituted C 2-12 alkynylene, C 3 . 12 cycloalkyiene, substituted C 3-12 cycloaikylene, optionally substituted C 6 ^ arylene, optionally substituted 3 - 14 membered heteroarylene, optionally substituted 3 -12 membered heterocyclylene, optionally substituted 3-18 membered heteroaryialkySene, or optionally substituted C 6 - 1 8 aryialkylene. in another aspect of this embodiment, R 3 is optionally substituted C 1 - 12 alkyiene, C 3-12 cycloaikylene, substituted C 3 . 12 cycloaikylene, optionally substituted C 6-14 arylene, or optionally substituted 3 -12 membered heterocyclylene. in another aspect of this embodiment, L is -0-, -S-, -S(O)- or -S(O) 2 -. in another aspect of this embodiment, L is -0-. In another aspect of this embodiment, L is -S-. In another aspect of this embodiment, L is -S(O)-. In another aspect of this embodiment, L is - S(O) 2 -.

In another embodiment of Formula I, R 1 is optionally substituted C 1 -C 12 alkyl, R 2 is optionally substituted C 3-12 cycioalkyi and R 3 is optionally substituted C 1-12 alkyienθ, C 3-12 cycloaikylene, substituted C 3-12 cycloalkyiene, optionally substituted C 6~ i 4 arylene, or optioπaily substituted 3 -12 membered heterocyclyiene. in another aspect of this embodiment, R 3 is optionally substituted Ci_ 6 alkyiene. In another aspect of this embodiment, R 3 is C 4 ^ cycloalkySene or substituted C 4-6 cycioalkylene. In another aspect of this embodiment, R 3 is an optionally substituted 5-6 membered heterocyclyiene. In another aspect of this embodiment, L is -O-, -S-, -S(O)- or - S(O) 2 -. In another aspect of this embodiment, L is -0-. In another aspect of this embodiment, L is -S-. In another aspect of this embodiment, L is -S(O)-. In another aspect of this embodiment, L is -S(O) 2 -. In another aspect of this embodiment, Het is an optionally substituted 3-12 membered heterocyclyl or optionally substituted 3-14 membered heteroaryl wherein said optionally substituted 3-12 membered

heterocyclyl or optionally substituted 3-14 membered heteroaryl comprises one to four heteroatoms selected from O, S, or N. In another aspect of this embodiment, Het is an optionally substituted 3-12 membered heterocyclyl comprising one or two heteroatoms selected from O, S, or N. In another aspect of this embodiment, Het is an optionally substituted 5-10 membered heteroaryi comprising one to four hetroatoms selected from O 1 S, or N.

In another embodiment of Formula I, R 1 is optionally substituted C 3 -C 7 secondary or tertiary alkyl and R 2 is optionally substituted methylcyclohexyl or optionally substituted methylcyclohexenyl. In another aspect of this embodiment, R 1 is prop-2-yl (isopropyl) or 2-methyiprop-2-yl (/-butyl). In another aspect of this embodiment, R 2 is optionally substituted 4-methylcyclohexyl. In a preferred aspect of this embodiment, R 2 is

In another preferred aspect of this embodiment, R 2 is

In another preferred aspect of this embodiment, R 2 is

In another preferred aspect of this embodiment, R is

In another embodiment of Formula I, R 1 is optionally substituted C 3 -C 5 cycloalkyl and R 2 is optionally substituted methylcyciohexy! or optionally substituted methylcyclohexenyl. In another aspect of this embodiment, R 1 is prop-2-yl (isopropyl) or 2-methyiprop-2-yl (f-butyl). In another aspect of this embodiment, R 2 is optionally substituted 4-methylcydohexyl, In a preferred aspect of this embodiment, R 2 is

In another preferred aspect of this embodiment, R is

In another preferred aspect of this embodiment, R 2 is

In another preferred aspect of this embodiment, R 2 is

In another embodiment of Formula I, R 1 is optionally substituted C 3 -C 7 secondary or tertiary alky! and R 2 is optionally substituted methylcyclohexyl or optionally substituted methylcyciohexenyl. In another aspect of this embodiment, R 3 is optionally substituted C 1-6 alkylene. In another aspect of this embodiment, R 3 is C 4 . 6 cycioaikylene or substituted C 4-6 cycloalkylene. In another aspect of this

embodiment, R 3 is an optionally substituted 4-6 membered nitrogen-containing heterocyclylene. In another aspect of this embodiment, L is -O-, -S-, -S(O)- or - S(O) 2 -. In another aspect of this embodiment, L is -O-. in another aspect of this embodiment, L is -S-. In another aspect of this embodiment, L is -S(O)-. !n another aspect of this embodiment, L is -S(O) 2 -. In another aspect of this embodiment, Het is an optionally substituted 3-12 membered heterocyclyl or optionally substituted 3-14 membered heteroaryl wherein said optionally substituted 3-12 membered

heterocycly! or optionally substituted 3-14 membered heteroaryl comprises one to four heteroatoms selected from O, S, or N. In another aspect of this embodiment, Het is an optionally substituted 3-12 membered heterocyciyl comprising one or two heteroatoms selected from O, S, or N. In aspect of this embodiment, Het is an optionally substituted 5-10 membered heteroaryl comprising one to four hetroatoms selected from O, S, or N.

In another embodiment of Formula I, L is -O-, R 1 is optionally substituted C 3 - C 7 secondary or tertiary alkyl and R 2 is optionally substituted methylcyclohexyl or optionally substituted methylcyciohexenyl. In another aspect of this embodiment, R 3 is optionally substituted C 1-6 alkylene. In another aspect of this embodiment, R 3 is C 4 . 6 cycloalkylene or substituted C 4 . 6 cycloalkylene. In another aspect of this

embodiment, R 3 is an optionally substituted 5-6 membered heterocyclylene. In another aspect of this embodiment, Het is an optionally substituted 3-12 membered heterocyclyl or optionally substituted 3-14 membered heteroaryl wherein said optionally substituted 3-12 membered heterocycly! or optionally substituted 3-14 membered heteroaryl comprises one to four heteroatoms selected from O, S 1 or N. In another aspect of this embodiment, Het is an optionally substituted 3-12

membered heterocyclyl comprising one or two heteroatoms selected from O, S, or N. In aspect of this embodiment, Het is an optionally substituted 5-10 membered heteroaryl comprising one to four hetroatoms selected from O, S, or N.

in another embodiment, compounds of Formula I are represented by compounds of Formula Ii: or pharmaceutically acceptable salts and esters thereof, wherein:

R 2 is optionally substituted 4-methylcyclohexyI or optionally substituted

methylcyciohexenyl and the remaining variables are defined as for Formula

In one embodiment of Formula II, R 2 is:

In one embodiment of Formula II, R 2 : i,s

in another embodiment of Formula II, R 2 is

in another embodiment of Formula If, R is

In one embodiment of Formula It, R 3 is optionally substituted C 1-6 alkylene. In another embodiment of Formula II, R 3 is C 4 . 6 cycloalkyiene or substituted C 4-6 cycloalkylene. in one embodiment of Formula II, R 3 is an optionally substituted 5-6 membered heterocyclylene. In another embodiment of Formula It, L is -O- , -S-, - S(O)- or -S(O) 2 -. In another embodiment of Formuia I!, L is -O-. In another embodiment of Formula Ii, L is -S-. In another embodiment of Formuia II, L is -S(O)-. In another embodiment of Formuia II, L is -S(O) 2 -. In another embodiment of

Formula Ii, Het is an optionally substituted 3-12 membered heterocyciyl or optionally substituted 3-14 membered heteroaryl wherein said optionally substituted 3-12 membered heterocyciyl or optionally substituted 3-14 membered heteroaryl comprises one to four heteroatoms selected from O, S, or N. in another embodiment of Formula II, Het is an optionally substituted 3-12 membered heterocyciyl comprising one or two heteroatoms selected from O, S, or N. In another embodiment of Formuia II, Het is an optionally substituted 5-10 membered heteroaryl comprising one to four hetroatomε selected from O, S, or N.

In another embodiment of Formula Ii, Het is an optionally substituted 3-12 membered heterocyciyl or optionally substituted 3-14 membered heteroaryl wherein said optionally substituted 3-12 membered heterocyciyl or optionally substituted 3-14 membered heteroaryi comprises one to four heteroatoms selected from O, S, or N. In another aspect of this embodiment, Het is optionally substituted pyridinyl. In another aspect of this embodiment, Het is optionally substituted pyridine-2-yl. In another aspect of this embodiment, Het is optionally substituted pyridaziπyl. In another aspect of this embodiment, Het is optionally substituted tetrahydro-2H~ pyranyl. In another aspect of this embodiment, Het is optionally substituted piperidinyl. In another aspect of this embodiment, Het is optionally substituted pyrrolidinyl. In another aspect of this embodiment, Het is optionally substituted tetrahydrothiophenyl. In another aspect of this embodiment, Het is optionally substituted pyrazinyi. In another aspect of this embodiment, Het is optionally substituted 1 H-tetrazo!yi. In another aspect of this embodiment, Het is optionally substituted azetidinyl. In another aspect of this embodiment, Het is optionally substituted tetrahydrofuranyi. In another aspect of this embodiment, Het is optionally substituted tetrahydrofuran-3-y!. In another aspect of this embodiment, Het is optionally substituted tetrahydro-2/-/-furo[2,3-b]furanyl. In another aspect of this embodiment, Het is optionally substituted thiazoyl. In another aspect of this embodiment, Het is optionally substituted 1H-tmidazolyl. In another aspect of this embodiment, Het is optionally substituted 4H-1,2,4-triazoiyl. In another aspect of this embodiment, Het is optionally substituted 1 H-pyrazolyl. In another aspect of this embodiment, Het is optionally substituted 1 ,3,4-thiadiazoiyi. In another aspect of this embodiment, Het is optionally substituted quiπolinyl. In another aspect of this embodiment, Het is optionally substituted [1 ,2,4]triazolo[4,3-a]pyridinyl. in another aspect of this embodiment, Het is optionally substituted thiophenyl. In another aspect of this embodiment, Het is optionally substituted 1 ,2,4-thiadiazolyl. In another aspect of this embodiment, Het is optionally substituted pyrimidinyl. In another aspect of this embodiment, Het is optionally substituted 1 H- 1 ,2,3-triazolyl. In another aspect of this embodiment, Het is optionally substituted 1 ,3,4-oxadiazolyl. In another aspect of this embodiment, Het is optionally substituted imidazo[1 ,2-b]pyridazinyl. In another aspect of this embodiment, L is -O-. In another aspect of this embodiment, L is -S-. in another aspect of this embodiment, L is -S(O)-. in another aspect of this embodiment, L is -S(O) 2 -. In another aspect of this embodiment, R 2 is:

in another aspect of this embodiment, R is:

In another preferred aspect of this embodiment, R 2 is

In another preferred aspect of this embodiment, R is !n one embodiment of Formula II, L is -O-. In another aspect of this embodiment, R 3 is optionally substituted C 1 ^ alkylene. In another aspect of this embodiment, R 3 is C 4 , 6 cyctoalkylene or substituted C 4 . 6 cycloaikylene. In another aspect of this embodiment, R 3 is an optionally substituted 5-6 membered

heterocyciylene. in another aspect of this embodiment, Het is an optionaliy substituted 3-12 membered heterocyclyl or optionally substituted 3-14 membered heteroaryl wherein said optionally substituted 3-12 membered heterocyciy! or optionally substituted 3-14 membered heteroaryl comprises one to four heteroatoms selected from O, S, or N. In another aspect of this embodiment, Het is an optionally substituted 3-12 membered heterocyclyl comprising one or two heteroatoms selected from O, S, or N. In another aspect of this embodiment, Het is an optionally substituted 5-10 membered heteroaryl comprising one to four hetroatoms selected from O, S, or N. In another aspect of this embodiment, Het is optionally substituted pyridinyl. In another aspect of this embodiment, Het is optionally substituted pyridazinyl. In another aspect of this embodiment, Het is optionally substituted tetrahydro-2/-/-ρyranyl. in another aspect of this embodiment, Het is optionally substituted piperidinyi. In another aspect of this embodiment, Het is optionally substituted pyrrofidinyl. In another aspect of this embodiment, Het is optionally substituted tetrahydrothiopheny!. in another aspect of this embodiment, Het is optionally substituted pyrazinyl. In another aspect of this embodiment, Het is optionally substituted 1 H-tetrazolyl. In another aspect of this embodiment, Het is optionally substituted azetidinyl. In another aspect of this embodiment, Het is optionally substituted tetrahydrofuranyl. In another aspect of this embodiment, Het is optionally substituted tetrahydro-2/-/-furo[2,3-b]furanyl. In another aspect of this embodiment, Het is optionally substituted thiazoyl. In another aspect of this embodiment, Het is optionally substituted 1 H-imidazolyl. in another aspect of this embodiment, Het is optionally substituted 4H- 1 ,2,4-triazolyi. In another aspect of this embodiment, Het is optionally substituted 1 H-pyrazolyl. In another aspect of this embodiment, Het is optionally substituted 1 ,3,4-thiadiazolyl. in another aspect of this embodiment, Het is optionally substituted quinolinyl. In another aspect of this embodiment, Het is optionally substituted [1 ,2,4]triazolo[4,3-a]pyridinyl. In another aspect of this embodiment, Het is optionally substituted thiophenyl. in another aspect of this embodiment, Het is optionally substituted 1,2,4-thiadiazolyl. In another aspect of this embodiment, Het is optionally substituted pyrimidinyl. In another aspect of this embodiment, Het is optionally substituted 1H-1 ,2,3-triazo!yl. In another aspect of this embodiment, Het is optionally substituted 1,3,4-oxadiazolyl. In another aspect of this embodiment, Het is optionally substituted imidazo[1 ,2-b]pyridazinyl. In another aspect of this embodiment, R 2 is:

In another aspect of this embodiment, R is:

In another preferred aspect of this embodiment, R 2 is

in another preferred aspect of this embodiment, R is

In one embodiment of Formula Ii, L is -O- and R 3 is C 4-6 cycloaikylene or substituted C 4 . 6 cycioalkylene. In another aspect of this embodiment, Het is an optionally substituted 3-12 membered heterocyciy! or optionaliy substituted 3-14 membered heteroaryi wherein said optionaliy substituted 3-12 membered heterocyciyl or optionally substituted 3-14 membered heteroaryi comprises one to four heteroatoms selected from O, S, or N. in another aspect of this embodiment, Het is an optionally substituted 3-12 membered heterocyciyl comprising one or two heteroatoms selected from O, S, or N. In another aspect of this embodiment, Het is an optionally substituted 5-10 membered heteroaryi comprising one to four hetroatoms selected from O, S, or N. In another aspect of this embodiment, Het is optionally substituted pyridinyl. In another aspect of this embodiment, Het is optionally substituted pyridazinyl. In another aspect of this embodiment, Het is optionally substituted tetrahydro-2H-pyranyl. In another aspect of this embodiment, Het is optionally substituted piperidinyi. In another aspect of this embodiment, Het is optionally substituted pyrrolidinyl. in another aspect of this embodiment, Het is optionally substituted tetrahydrothiophenyl. In another aspect of this embodiment, Het is optionally substituted pyrazinyl. In another aspect of this embodiment, Het is optionally substituted 1 /-/-tetrazoiyl. in another aspect of this embodiment, Het is optionally substituted azetidinyi. In another aspect of this embodiment, Het is optionally substituted tetrahydrofurany!. !n another aspect of this embodiment, Het is optionally substituted tetrahydro-2H-furo[2,3-b]furanyi. In another aspect of this embodiment, Het Ss optionally substituted thiazoyl. In another aspect of this embodiment, Het is optionally substituted 1 H-imidazolyl. In another aspect of this embodiment, Het is optionally substituted 4/-/-1,2,4-triazolyl. In another aspect of this embodiment, Het is optionally substituted 1 H-pyrazolyl. In another aspect of this embodiment, Het is optionally substituted 1 ,3,4-thiadiazolyl. In another aspect of this embodiment, Het is optionally substituted quinolinyl. In another aspect of this embodiment, Het is optionally substituted [1 ,2,4]triazofo[4,3-a]pyridinyl. In another aspect of this embodiment, Het is optionally substituted thiophenyl. In another aspect of this embodiment, Het is optionally substituted 1,2,4-thiadiazolyl. In another aspect of this embodiment, Het is optionally substituted pyrimidinyl. In another aspect of this embodiment, Het is optionally substituted 1H-1 ,2,3-triazolyl. In another aspect of this embodiment, Het is optionally substituted 1 ,3,4-oxadiazolyi. In another aspect of this embodiment, Het is optionally substituted imidazo[1 ,2-b]pyhdazinyj. In another aspect of this embodiment, R 2 is:

In another aspect of this embodiment, R is:

In another preferred aspect of this embodiment, R 2 is

In another preferred aspect of this embodiment, R is

In one embodiment of Formula II, L is -O-, R 3 is C 4 . 6 cycloalkyiene or substituted C 4 . 6 cycloalkylene and R 2 is optionally substituted methylcyclohexyS optionally substituted methylcyclohexenyl. In another aspect of this embodiment, Het is an optionally substituted 3-12 membered heterocyclyl or optionally substituted 3-14 membered heteroaryl wherein said optionally substituted 3-12 membered

heterocyclyl or optionally substituted 3-14 membered heteroaryl comprises one to four heteroatoms selected from O, S, or N. In another aspect of this embodiment, Het is an optionally substituted 3-12 membered heterocyclyl comprising one or two heteroatoms selected from O, S, or N. In another aspect of this embodiment, Het is an optionally substituted 5-10 membered heteroaryl comprising one to four hetroatoms selected from O, S, or N. In another aspect of this embodiment, Het is optionally substituted pyridinyi. In another aspect of this embodiment, Het is optionally substituted pyridazinyl. In another aspect of this embodiment, Het is optionally substituted tetrahydro-2H-pyranyi. In another aspect of this embodiment, Het is optionally substituted piperidinyl. in another aspect of this embodiment, Het is optionally substituted pyrrolidinyl. In another aspect of this embodiment, Het is optionally substituted tetrahydrothiophenyi. In another aspect of this embodiment, Het is optionally substituted pyrazinyl. In another aspect of this embodiment, Het is optionally substituted 1H-tetrazolyl. In another aspect of this embodiment, Het is optionally substituted azetidinyl. In another aspect of this embodiment, Het is optionally substituted tetrahydrofuranyl. In another aspect of this embodiment, Het is optionally substituted tetrahydro-2/-/-furo[2,3-b]furanyl. In another aspect of this embodiment, Het is optionally substituted thiazoyl. In another aspect of this embodiment, Het is optionally substituted 1 /-/-imidazolyl. In another aspect of this embodiment, Het is optionally substituted 4H-1,2,4-triazolyl. In another aspect of this embodiment, Het is optionally substituted 1H-pyrazolyl. in another aspect of this embodiment, Het is optionally substituted 1 ,3,4-thiadiazolyl. In another aspect of this embodiment, Het is optionally substituted quinolinyl. In another aspect of this embodiment, Het is optionally substituted [1 ,2,4]triazolo[4,3-ajpyridiπyl. In another aspect of this embodiment, Het is optionally substituted thiophenyl. In another aspect of this embodiment, Het is optionally substituted 1 ,2,4-thιadιazoiyl In another aspect of this embodiment, Het is optionally substituted pyπmidinyl In another aspect of thts embodiment, Het is optionally substituted 1H-1 ,2,3-tπazolyl in another aspect of thts embodiment, Het is optionally substituted 1 ,3,4-oxadiazoiyl In another aspect of this embodiment, Het is optionally substituted smιdazo[1 ,2-b]pyrιdazιnyl In another aspect of this embodiment, R 2 ιs-

In another aspect of this embodiment, R is

In another preferred aspect of this embodiment, R 2 is

In another preferred aspect of this embodiment, R is in another embodiment, the compound of Formula I or Formuia Il is

or a pharmaceutically acceptable salt or ester thereof.

Definitions

Unless stated otherwise, the following terms and phrases as used herein are intended to have the following meanings. The fact that a particular term or phrase is not specificaily defined should not be correlated to indefiniteness or lacking clarity, but rather terms herein are used within their ordinary meaning. When trade names are used herein, applicants intend to independently include the tradename product and the active pharmaceutical ingredient(s) of the tradename product. The term "treating", and grammatical equivalents thereof, when used in the context of treating a disease, means slowing or stopping the progression of a disease, or ameliorating at least one symptom of a disease, more preferably ameSiorating more than one symptom of a disease. For example, treatment of a hepatitis C virus infection can include reducing the HCV viral load in an HCV infected human being, and/or reducing the severity of jaundice present in an HCV infected human being.

"Alkyf" is hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms. For example, an alkyl group can have 1 to 20 carbon atoms (i.e, C 1 -C 2O alkyl), 1 to 10 carbon atoms (i.e., C 1 -C 10 alkyl}, or 1 to 6 carbon atoms (i.e., C 1 -C 6 alkyi). Examples of suitable alkyl groups include, but are not limited to, methyi (Me 1 -CH 3 ), ethyi (Et, -CH 2 CH 3 ), 1 -propyl (n-Pr, n-propyl, -CH 2 CH 2 CH 3 ), 2-propyl Q-Pr,

i-propyl, -CH(CH 3 J 2 ), 1 -butyl (n-Bu, n-butyl, -CH 2 CH 2 CH 2 CH 3 ), 2-methyl-1 -propyl Q-Bu, l-butyl, -CH 2 CH(CH 3 J 2 ), 2-butyl (s-Bu, s-butyl, -CH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propyl (t-Bu, t-butyl, -C(CHg) 3 ), 1 -pentyl (n-pentyl, -CH 2 CH 2 CH 2 CH 2 CH 3 ), 2-pentyl

(-CH(CH 3 )CH 2 CH 2 CH 3 ), 3-pentyl (-CH(CH 2 CH 3 J 2 ), 2-methy(-2-butyϊ

(-C(CH 3 J 2 CH 2 CH 3 ), 3-methyl-2-butyl (-CH(CH 3 )CH(CH 3 ) 2 ), 3-methyl-1 -butyl

(-CH 2 CH 2 CH(CH 3 ) 2 ), 2-methyl-1 -butyl (-CH 2 CH(CH 3 )CH 2 CH 3 ), 1 -hexyi

(-CH 2 CH 2 CH 2 CH 2 CH 2 CH 3 ), 2-hexyl (-CH(CH 3 )CH 2 CH 2 CH 2 CH 3 ), 3-hexyl (- CH(CH 2 CH 3 )(CH 2 CH 2 CH 3 )), 2-methyi-2-pentyl (-C(CHa) 2 CH 2 CH 2 CH 3 ), 3-methyi-2- pentyl (-CH(CH 3 )CH(CH 3 )CH 2 CH 3 ), 4-methyi-2-penty! (-CH(CH 3 )CH 2 CH(CH 3 ) 2 ), 3- methyl-3-pentyl (-C(CH 3 )(CH 2 CH 3 ) 2 ), 2-methyl-3-pentyl (-CH(CH 2 CH 3 )CH(CH 3 ) 2 ), 2,3- dimethyl-2-butyl (-C(CH 3 ) 2 CH(CH 3 ) 2 ), 3,3-dimethyl-2-butyi (-CH(CH 3 JC(CH 3 J 3 , and octy! (-(CH 2 J 7 CH 3 ).

"Alkoxy" means a group having the formula— O-alkyl, in which an alkyl group, as defined above, is attached to the parent molecule via an oxygen atom. The alkyl portion of an aikoxy group can have 1 to 20 carbon atoms (i.e., C 1 -C 20 alkoxy), 1 to 12 carbon atoms (i.e., C 1 -C 12 atkoxyj, or 1 to 6 carbon atoms(i.e., C 1 -C 6 alkoxy).

Examples of suitable alkoxy groups include, but are not limited to, methoxy (-0-CH 3 or -OMe), ethoxy (-OCH 2 CH 3 or -OEt), t-butoxy (-0-C(CH 3 J 3 or -OtBu), and the like.

"Haloalkyl" is an alkyl group, as defined above, in which one or more hydrogen atoms of the alkyl group is replaced with a halogen atom. The alkyl portion of a haloalkyl group can have 1 to 20 carbon atoms (i.e., C 1 -C 20 haloaSkyl), 1 to 12 carbon atoms(i.e., C 1 -C 12 haloalkyl), or 1 to 6 carbon atoms (i.e., C 1 -C 6 alkyl).

Examples of suitable haloalkyl groups include, but are not limited to, -CF 3 , -CHF 2 , -CFH 2 , -CH 2 CF 3 , and the like.

"Alkenyi" is a hydrocarbon containing normal, secondary, tertiary, or cyclic carbon atoms with at least one site of unsaturation, i.e. a carbon-carbon, sp2 double bond. For example, an alkeny! group can have 2 to 20 carbon atoms (i.e., C 2 -C 2O alkenyi}, 2 to 12 carbon atoms (i.e., C 2 -C 12 alkenyi), or 2 to 6 carbon atoms (i.e., C 2 - C 6 aSkenyi). Examples of suitable alkeny! groups include, but are not limited to, vinyl (-CH=CH 2 ), ally! (-CH 2 CH-CH 2 ), cyclopentenyi (-C 5 H 7 ), and 5-hexenyl

(-CH 2 Cn 2 CH 2 CH 2 CH=CH 2 ).

"Alkynyi" is a hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms with at least one site of unsaturation, i.e. a carbon-carbon, sp triple bond. For example, an aikynyl group can have 2 to 20 carbon atoms (i.e., C 2 -C 20 alkyny!), 2 to 12 carbon atoms (i.e., C 2 -Ci 2 alkyne,), or 2 to 6 carbon atoms (i.e., C 2 - C 6 alkynyi). Examples of suitable alkynyi groups include, but are not limited to, acetyienic (-CsCH), propargyl (-CH 2 C=CH), and the like.

"Aikylene" refers to a saturated, branched or straight chain radical or or cyclic hydrocarbon radical having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane. For example, an aikylene group can have 1 to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms. Typical aikylene radicals include, but are not limited to, methylene (-CH 2 -), 1 ,1 -ethylene (-CH(CH 3 )-), 1 ,2-ethylene (-CH 2 CH 2 -), 1 ,1-propylene (-CH(CH 2 CH 3 )-), 1 ,2-propyiene (-CH 2 CH(CH 3 )-), 1,3-ρropylene (-CH 2 CH 2 CH 2 -), 1 ,4- butyiene (-CH 2 CH 2 CH 2 CH 2 -), and the like.

"Aikenylene" refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent aikene. For example, and aikenyiene group can have 1 to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms. Typical aikenylene radicals include, but are not limited to, 1,2-ethylene (-CH=CH-).

"Afkynylene" refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent aikyne. For example, an alkynylene group can have 1 to 20 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms. Typical alkynyiene radicals include, but are not limited to, acetylene (-C=C-), propargyl (-CH 2 C=C-), and 4-pentynyl (-CH 2 CH 2 CH 2 C=C-). "Aikylyne" refers to a saturated, branched or straight chain radicai having two radical centers derived by the removal of three hydrogen atoms from two carbon atoms of a parent afkane. For example, an aikylyne group can have 2 to 20 carbon atoms, 2 to 10 carbon atoms, or 2 to 6 carbon atoms. Typical aikyfyne radicals include, but are not limited to, 1 ,2-ethyiyne (-CH 2 CH=), 1 ,2-proρylyne (-CH 2 C(CH 3 )=), 1 ,3-propyiyne (-CH 2 CH 2 CH=), 1 ,4-butylyne (-CH 2 CH 2 CH 2 CH=), and the like.

"Aryi" means a monovalent aromatic hydrocarbon radical derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system. For example, an aryi group can have 6 to 20 carbon atoms, 6 to 14 carbon atoms, or 6 to 12 carbon atoms. Typical aryi groups include, but are not limited to, radicals derived from benzene (e.g., phenyl), substituted benzene, naphthalene, anthracene, btphenyi, and the like.

"Arylene" refers to an aryi as defined above having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent aryi. Typical arylene radicals include, but are not limited to, phenylene.

"ArylalkyP refers to an acyclic alky! radicai in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with an aryi radical. Typical arylalkyl groups include, but are not limited to, benzyl, 2- ρheny!ethan-1-yi, naphthylmethyi, 2-naρhthylethan-1-yi, naphthobenzyl,

2-naphthophenySethan-1-yl and the like. The arylalkyl group can comprise 6 to 20 carbon atoms, e.g., the alky! moiety is 1 to 6 carbon atoms and the aryi moiety is 6 to 14 carbon atoms.

"Cycloalkyl" refers to a saturated or partially unsaturated ring having 3 to 7 carbon atoms as a monocycle, 7 to 12 carbon atoms as a bicycle, and up to about 20 carbon atoms as a poiycycle. Monocyclic cycloalkyl groups have 3 to 6 ring atoms, still more typically 5 or 6 ring atoms. Bicyclic cycloalkyl groups have 7 to 12 ring atoms, e.g., arranged as a bicyclo (4,5), (5,5), (5,6) or (6,6) system, or 9 or 10 ring atoms arranged as a bicycio (5,6) or (6,6) system. Cycloalkyl groups include hydrocarbon mono-, bi-, and poly-cyclic rings, whether fused, bridged, or spiro. Non- limiting examples of monocyclic cycloalkyls include cyciopropyl, cyclobutyl, cyclopentyl, 1 -cyclopent-i-enyl, 1-cyc!opent-2-enyl, 1-cyclopent-3-enyi, cyclohexyl, 1- cyclohex-1-enyl, 1 -cyclohex-2-enyl and 1-cyclohex-3-enyi. Non-limiting examples of bicyclo cycloalkyls includes naphthyl, tetrahydronapthalene, decaliπe and

bicyclo[3.1.0]hex-6-yl and the like.

"Cycloalkylene" refers to a cycioalkyl as defined above having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent cycioalkyl. Typical cycloalkylene radicals include, but are not limited to, cyclopropylene, cyclobutylene, cyclopentylene and cyclohexylene.

"Halogen" refers to F, Ci 1 Br, or I.

As used herein the term "haioalkyl" refers to an alky! group, as defined herein, that is substituted with at least one halogen. Examples of branched or straight chained "haioalkyl" groups as used herein include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, and t-butyl substituted independently with one or more halogens, for example, fluoro, chbro, bromo, and iodo. The term "haioalkyl" should be interpreted to include such substituents as perfluoroalkyi groups such as -CF 3 .

As used herein, the term "haioalkoxy" refers to a group -OR a , where R a is a haioalkyl group as herein defined. As non-limiting examples, haioalkoxy groups include -0(CH 2 )F, -0(CH)F 2 , and -OCF 3 .

Ηeterocycle" or "heterocyclyl" refers to a saturated or partially saturated cyclic group having from 1 to 14 carbon atoms and from 1 to 6 heteroatoms selected from N, S, P, or O, and includes single ring and multiple ring systems including, fused, bridged, and spiro ring systems. "Heterocycle" or "heterocyclyl" as used herein includes by way of example and not limitation those heterocycles described in

Paquette, Leo A.; Principles of Modern Heterocyclic Chemistry (W. A. Benjamin, New York, 1968), particularly Chapters 1 , 3, 4, 6, 7, and 9; The Chemistry of Heterocyclic Compounds, A Series of Monographs" (John Wiley & Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and 28; and J. Am. Chem. Soc. (1960) 82:5566. In one embodiment, the carbon, nitrogen, phosphorous, or sulfur atom(s) of the heterocyclic group may be oxidized to provide for C(=O), N-oxide, phosphinane oxide, sulfinyl, or sulfonyl moieties.

As one example, substituted heterocyciyls include, for example, heterocyclic rings substituted with any of the substituents disclosed herein including oxo groups. A non-limiting example of a carbonyl substituted heterocyclyl is:

Examples of heterocycles include by way of example and not limitation dihydroypyridyl, tetrahydropyrtdyl (piperidyl), tetrahydrothiophenyl, sulfur oxidized tetrahydrothiophenyl, piperidinyl, 4-piperidonyl, pyrrolidinyl, azetidinyl, 2-pyrrolidonyf, tetrahydrofuranyi, decahydroquinolinyl, octahydroisoquinolinyl, pyranyl, morpholinyl, and bis-tetrahydrofuranyl:

"Heterocyclene" or "heterocyclyiene" refers to a "heterocycie" or "heterocyciyi" as defined above having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent heterocycie, the removal of two hydrogen atoms from two nitrogen atoms of a parent heterocycie, or the removal of a hydrogen atom from a nitrogen and the removal of a hydrogen atom from a carbon atome of a parent heterocycie. Non-limiting examples of heterocyclene or heterocyclylenes are:

Ηeteroaryl" refers to a monovalent aromatic heterocyclyl having at least one heteroatom in the ring. Thus, "heteroaryl" refers to an aromatic group of from 1 to 14 carbon atoms and 1 to 6 heteroatoms selected from oxygen, nitrogen, sulfur, or phosphorous. For multiple ring systems, by way of example, the term "heteroaryl" includes fused, bridged, and spiro ring systems having aromatic and non-aromatic rings. . In one embodiment, the carbon, nitrogen, or sulfur ring atom(s) of the heteroaryl group may be oxidized to provide for C(=O), N-oxide, suifinyl, or sulfonyl moieties.

Examples of heteroaryls include by way of example and not limitation pyridyl, thiazoiyl, pyrimidinyl, furanyl, thienyl, pyrroiyl, pyrazoiyl, imidazolyl, tetrazolyi, benzofuranyl, thianaphthaienyl, indolyl, quinolinyl, isoquinolinyl, benzimidazoiyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, 6H-1 ,2,5-thiadiazinyl, 2H,6H-1 ,5,2- dithiaziny!, thienyl, thianthrenyl, isobenzofuranyl, chromenyl, xanthenyl,

phenoxathinyl, 2H-pyrrolyl, isothiazolyl, isoxazolyi, pyrazinyl, pyridazinyl, indolizinyl, isoindolyl, 3H-indolyl, 1 H-indazoly, purinyl, 4H-quinoϋzinyi, naphthyridiny!, quinoxalinyl, quinazolinyi, cinnolinyl, pteridiny!, 4aH-carbazolyl, carbazolyl, β- carbolinyl, phenanthridinyl, acridinyl, pyrimidinyl, phenanthroliny!, phenazinyl, phenothiazinyl, furazanyl, phenoxazinyl, isochromanyl, chromanyl, imidazolidinyl, imidazolinyi, pyrazoϋdinyi, pyrazolinyl, piperazinyl, indofinyl, isoindolinyl, quinuclidinyl, morpholinyl, oxazoϋdinyl, benzotriazolyl, benzisoxazolyl, oxindolyl, benzoxazoiinyl, and isatinoyl. "Heterocyclylene" refers to a heterocyclyl, as defined herein, derived by replacing a hydrogen atom from a carbon atom or heteroatom of a heterocyclyl, with an open valence. Similarly, "heteroarylene" refers to an aromatic heterocycfylene.

"Heterocyciyialkyl" refers to an acyclic aikyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with a heterocyclyl radical (i.e., a heterocyciyl-alkyiene- moiety). Typical heterocyclyl alkyl groups include, but are not limited to heterocyclyl-Ch^-, 2- (heterocyclyl)ethan-i -yl, and the like, wherein the "heterocyclyl" portion includes any of the heterocyclyl groups described above, including those described in Principles of Modern Heterocyclic Chemistry. One skilled in the art will also understand that the heterocyclyl group can be attached to the afky! portion of the heterocyclyl alkyi by means of a carbon-carbon bond or a carbon-heteroatom bond, with the proviso that the resulting group is chemicalSy stable. The heterocyclylalky! group comprises 2 to 20 carbon atoms and 1-6 heteroatoms, e.g., the alkyl portion of the heterocyclylalkyl group comprises 1 to 6 carbon atoms and the heterocyciyi moiety comprises 1 to 14 carbon atoms. Examples of heterocyciylalkyls include by way of example and not limitation 5-membered sulfur, oxygen, phosphorus, and/or nitrogen containing heterocycles such as pyrrolidiylmethyl, 2-tetrahydrofuranylylethan-1-yl, and the like, 6-membered sulfur, oxygen, and/or nitrogen containing heterocycles such as piperidinylmethyl, morpholinylmethyi, piperidinylethyi, teterahydropyranylethyl, and the like.

"Heteroarylalkyf" refers to an alkyl group, as defined herein, in which a hydrogen atom has been replaced with a heteroaryl group as defined herein. Non- limiting examples of heteroaryl alkyl include -CH 2 -pyridinyl, -CH 2 -pyrroiyi, -CH 2 - oxazolyl, -CH 2 -indoiyl, -CH 2 -isoindotyl, -CH 2 -purinyl, -CH 2 -furanyl, -CH 2 -thienyl, -CH 2 - benzofuranyl, -CH 2 -benzothiophenyl, -CH 2 -carbazolyl, -CH 2 -imidazoiyi, -CH 2 -thiazolyl, -CHHsoxazolyl, -CH 2 -pyrazoiyl, -CH 2 -isothiazolyi, -CH 2 -quinoiyl, -CH 2 -isoquino!yl, - CH 2 -pyridazyl, -CH 2 -pyrimidyl, -CH 2 -pyrazyl, -CH(CH 3 )-pyridinyl, -CH{CH 3 )-pyrro!yl, - CH(CH 3 )-oxazolyi,

-CH(CH 3 )-indolyl, -CH<CH 3 )-isoindolyl, -CH(CH 3 )-purinyl, -CH(CH 3 )-furanyl, - CH(CH 3 )-thienyl, -CH{CH 3 )-benzofuranyl, ~CH(CH 3 )-benzothiσphenyi, -CH(CH 3 )- carbazolyl,

-CH(CH 3 )-imidazolyl, -CH(CH 3 )-thiazolyl, -CH(CH 3 )-isoxazolyl, -CH(CH 3 )-pyrazolyl, -CH(CH 3 )-isothiazolyl, -CH(CH 3 )-quinolyl, -CH(CH 3 )-isoqυinolyl, -CH(CH 3 )-pyridazyl, -CH(CH 3 )-pyrimidyt, -CH(CH 3 )-pyrazyl, and the like.

The term "heterocyciyloxy" represents a heterocyclyl group attached to the adjacent atom by an oxygen.

When there is a sulfur atom present, the sulfur atom can be at different oxidation levels, namely, S, SO, SO 2 , or SO 3 . All such oxidation levels are within the scope of the present invention.

When there is a phosphorous atom present, the phosphorous atom can be at different oxidation levels, namely, POR a R b R c , PO 2 R a R b , or PO 3 R a R b , where R a , R b , and R c each independently is chosen from H, C M2 alky!, C 2-12 alkenyl, C 2-I2 alkynyl, C 6-14 aryl, 3-12 membered heterocycle, 3-18 membered heteroarylalkyl, C 6 ^e aryialky!; or two taken together (with or without oxygens) form a 5 to 10 membered heterocycle. All such oxidation levels are within the scope of the present invention

The term "optionally substituted" in reference to a particular moiety of the compound of Formula i-ll (e.g., an optionally substituted aryl group) refers to a moiety wherein all substiutents are hydrogen or wherein one or more of the hydrogens of the moiety may be replaced by substituents such as those listed under the definition of "substituted" or as otherwise specified.

The term "substituted" in reference to aikyl, aikylene, aryl, arylalkyl, alkoxy, heterocyclyl, heteroaryi, carbocyclyl, etc. , for example, "substituted aikyl",

"substituted aikylene", "substituted aryl", "substituted arylalkyl", "substituted heterocyciyf", and "substituted carbocycly!" means aikyl, aikylene, aryl, arylalkyl, heterocyciyl, carbocyclyl respectively, in which one or more hydrogen atoms are each independently replaced with a non-hydrogen substituent. Divalent groups may also be similarly substituted. Unless otherwise indicated, typical substituents include, but are not limited to, -X, -R b , -O " , =O, -OR b , -SR b , -S " , -NR b 2 , -fsTR b 3 ,

=NR b , -CX 3 , -CN, -OCN, -SCN, -N=C=O, -NCS, -NO, -NO 2 ,

=N 2 , -N 3 , -NHC(=O)R b , -OC(=O)R b , -NHC(=0)NR b 2 , -S(=O) 2 -, -S(=O) 2 OH, -S(=O) 2 R b , - OSf=O) 2 OR', -S(=O) 2 NR b 2 , -S(=O)R b , -OP(=O)(OR & ) 2 , -P(=O)(OR b ) 2 , -P(=O)(O " ) 2 , -P(= O)(OH) 2 , -P(O)(OR b )(O-), -C(=O)R b , -C(=O)X, -C(S)R b , -C(O)OR b , -C(O)O ' , -C(S)OR b , -C(O)SR b , -C(S)SR b , -C(O)NR b 2 , -C(S)NR b 2 , -C(=NR b )NR b 2 , where each X is independently a halogen: F, Cl, Br, or I; and each R b is independently H, aikyl, aryl, arylalkyl, a heterocycle, or a protecting group or prodrug moiety. Alkyiene, alkenylene, and aikynyiene groups may also be similarly substituted. Unless otherwise indicated, when the term "substituted" is used in conjunction with groups such as arylalkyl, which have two or more moieties capable of substitution, the substituents can be attached to the aryl moiety, the alky! moiety, or both.

Those skilled in the art will recognize that when moieties such as "aikyl", "aryl",

"heterocyclyl", etc. are substituted with one or more substituents, they could alternatively be referred to as "aikylene", "arylene", "heterocyclylene", etc. moieties (i.e., indicating that at least one of the hydrogen atoms of the parent "aikyl", "aryl", "heterocyclyl" moieties has been replaced with the indicated substituent(s)). When moieties such as "alkyl", "aryj", "heterocyciyl", etc. are referred to herein as "substituted" or are shown diagrammatically to be substituted (or optionally substituted, e.g., when the number of substituents ranges from zero to a positive integer), then the terms "alkyl", "aryS", "heterocyclyf, etc. are understood to be interchangeabie with "aikylene", "arylene", "heterocyciylene", etc.

As will be appreciated by those skilled in the art, the compounds of the present invention may exist in soivated or hydrated form. The scope of the present invention includes such forms. Again, as will be appreciated by those skilled in the art, the compounds may be capable of esterification. The scope of the present invention includes esters and other physiologically functional derivatives. The scope of the present invention includes prodrug forms of the compound herein described.

"Ester" means any ester of a compound in which any of the -COOH functions of the molecule is replaced by a -C(O)OR function, or in which any of the -OH functions of the molecuie are replaced with a -OC(O)R function, in which the R moiety of the ester is any carbon-containing group which forms a stable ester moiety, including but not limited to alky!, alkenyl, alkynyl, cycioalkyl, cycloalkylalkyl, aryl, aryialkyl, heterocyciyl, heterocyclylalky! and substituted derivatives thereof.

The term "prodrug" as used herein refers to any compound that when

administered to a biological system generates the drug substance, i.e., active ingredient, as a result of spontaneous chemical reaction(s), enzyme catalyzed chemicai reaction(s), photolysis, and/or metabolic chemical reactioπ(s). A prodrug is thus a covalently modified analog or latent form of a therapeutically active compound. Non-limiting examples of prodrugs include ester moieties, quaternary ammonium moieties, glycol moieties, and the like.

One skilled in the art will recognize that substituents and other moieties of the compounds of Formula I or Il should be selected in order to provide a compound which is sufficiently stable to provide a pharmaceutically useful compound which can be formulated into an acceptably stable pharmaceutical composition. Compounds of Formula I or Il which have such stability are contempiated as falling within the scope of the present invention.

As will be appreciated by those skilled in the art, the compounds of the present invention may contain one or more chiral centers. The scope of the present invention includes such forms. Again, as will be appreciated by those skilled in the art, the compound is capable of esterification. The scope of the present invention includes esters and other physiologically functional derivatives. In addition, the scope of the present invention includes prodrug forms of the compound herein described.

A compound of Formula 1-11 and its pharmaceutically acceptable salts may exist as different polymorphs or pseudopoiymorphs. As used herein, crystalline polymorphism means the ability of a crystalline compound to exist in different crystal structures. Polymorphism generally can occur as a response to changes in temperature, pressure, or both. Polymorphism can also result from variations in the crystallization process. Polymorphs can be distinguished by various physical characteristics known in the art such as x-ray diffraction patterns, solubility, and melting point. The crystalline polymorphism may result from differences in crystal packing (packing polymorphism) or differences in packing between different conformers of the same molecule (conformational polymorphism). As used herein, crystalline pseudopolymorphtsm means the ability of a hydrate or solvate of a compound to exist in different crystal structures. The pseudopoiymorphs of the instant invention may exist due to differences in crystal packing (packing

pseudopolymorphism) or due to differences in packing between different conformers of the same molecule (conformational pseudopolymorphism). The instant invention comprises all polymorphs and pseudopoiymorphs of the compounds of Formula I-II and their pharmaceutically acceptable salts.

A compound of Formula l-ll and its pharmaceutically acceptable salts may also exist as an amorphous solid. As used herein, an amorphous solid is a solid in which there is no long-range order of the positions of the atoms in the solid. This definition applies as well when the crystal size is two nanometers or less. Additives, including solvents, may be used to create the amorphous forms of the instant invention. The instant invention comprises all amorphous forms of the compounds of Formula l-l! and their pharmaceutically acceptable salts.

Certain of the compounds described herein contain one or more chiral centers, or may otherwise be capable of existing as multiple stereoisomers. The scope of the present invention includes mixtures of stereoisomers as well as purified enantiomers or enantiomeπcally/diastereomericaily enriched mixtures. Also included within the scope of the invention are the individual isomers of the compounds represented by the formulae of the present invention, as well as any wholly or partially equilibrated mixtures thereof. The present invention also includes the individual isomers of the compounds represented by the formuias above as mixtures with isomers thereof in which one or more chira! centers are inverted.

The term "chiral" refers to molecules which have the property of non- superimposability of the mirror image partner, whiie the term "achiral" refers to molecules which are superimposable on their mirror image partner.

The term "stereoisomers" refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space.

"Diastereomer" refers to a stereoisomer with two or more centers of chirafity and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g., melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography.

"Enantiomers" refer to stereoisomers of a compound which are non- superimposable mirror images of one another.

"Atropisomers" refer to stereoisomers of a compound resulting from hindered rotation about single bonds where the steric strain barrier to rotation is high enough to allow for the isolation of the individual conformer. Atropisomers display axial chtrality. Atropisomers may be equilibrated thermally and the interconversion barrier may be measured kinetically. Atropisomerism may occur apart from the presence of other forms of chiral isomerism. Thus, as illustrated, the depicted nitrogen atom is planar and compounds of Formula I are capable of existing as atropisomers:

ln one embodiment of the present invention, the compounds exist in a conformeric form of Formula Ia:

° * \ R 2 (Ia). Stereochemical definitions and conventions used herein generally follow S. P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and ESiel, E. and Wiien, S., Stereochemistry of Organic Compounds (1994) John Wiley & Sons, inc., New York.

Many organic compounds exist in optically active forms, i.e., they have the ability to rotate the plane of plane-polarized light. !n describing an optically active compound, the prefixes D and L or R and S are used to denote the absolute configuration of the molecule about its chirai center(s). The prefixes d and I or (+ ) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or 1 meaning that the compound is levorotatory. A compound prefixed with (+) or d is dextrorotatory.

A specific stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture. A 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereoεpecificity in a chemical reaction or process. The terms "racemic mixture" and "racemate" refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.

The present invention includes a salt or solvate of the compounds herein described, including combinations thereof such as a solvate of a salt. The

compounds of the present invention may exist in solvated, for example hydrated, as well as unsolvated forms, and the present invention encompasses all such forms.

Typically, but not absolutely, the salts of the present invention are

pharmaceutically acceptable salts. Salts encom passed within the term "pharmaceutically acceptable salts" refer to non-toxic salts of the compounds of this invention.

Examples of suitable pharmaceutically acceptable salts include inorganic acid addition salts such as chloride, bromide, sulfate, phosphate, and nitrate; organic acid addition salts such as acetate, galactarate, propionate, succinate, lactate, giycolate, malate, tartrate, citrate, maieate, fumarate, methanesulfonate, p-to!uenesulfonate, and ascorbate; salts with acidic amino acid such as aspartate and glutamate; alkali metal salts such as sodium salt and potassium salt; alkaline earth metal salts such as magnesium salt and calcium salt; ammonium salt; organic basic salts such as trimethylamine salt, triethylamine salt, pyridine salt, picoiine salt, dicyclohexylamine salt, and N.N'-dibenzylethylenediamine salt; and salts with basic amino acid such as iysine salt and arginine sail The salts may be in some cases hydrates or ethanol solvates.

The modifier "about" used in connection with a quantity is inclusive of the stated value and has the meaning dictated by the context (e.g., inciudes the degree of error associated with measurement of the particular quantity).

Whenever a compound described herein is substituted with more than one of the same designated group, e.g., "R" or "R 1 ", then it wili be understood that the groups may be the same or different, i.e., each group is independently selected. Wavy lines,

J_

^ ,

indicate the site of covalent bond attachments to the adjoining substructures, groups, moieties, or atoms.

The compounds of the invention can also exist as tautomeric isomers in certain cases. Although only one delocaSized resonance structure may be depicted, all such forms are contemplated within the scope of the invention. For example, ene- amine tautomers can exist for purine, pyrimidine, imidazole, guanidine, amidine, and tetrazoie systems and ali their possible tautomeric forms are within the scope of the invention.

Selected substituents comprising the compounds of Formula l-ll may be present to a recursive degree. In this context, "recursive substituent" means that a substituent may recite another instance of itself. The multiple recitations may be direct or indirect through a sequence of other substituents. Because of the recursive nature of such substituents, theoretically, a large number of compounds may be present in any given embodiment. One of ordinary skill in the art of medicinal chemistry understands that the total number of such substituents is reasonably limited by the desired properties of the compound intended. Such properties include, by way of example and not limitation, physical properties such as molecular weight, solubility or log P, application properties such as activity against the intended target, and practical properties such as ease of synthesis. Recursive substituents may be an intended aspect of the invention. One of ordinary skill in the art of medicinai chemistry understands the versatility of such substituents. To the degree that recursive substituents are present in an embodiment of the invention, they may recite another instance of themselves, 0, 1 , 2, 3, or 4 times. The compounds of Formula i-ll also include molecules that incorporate isotopes of the atoms specified in the particular molecules. Non-limiting examples of these isotopes include D, T, 14 C, 13 C and 15 N.

Protecting Groups

In the context of the present invention, protecting groups include prodrug moieties and chemical protecting groups.

Protecting groups are available, commonly known and used, and are optionally used to prevent side reactions with the protected group during synthetic procedures, i.e. routes or methods to prepare the compounds of the invention. For the most part the decision as to which groups to protect, when to do so, and the nature of the chemical protecting group "PG" will be dependent upon the chemistry of the reaction to be protected against (e.g., acidic, basic, oxidative, reductive or other conditions) and the intended direction of the synthesis. The PG groups do not need to be, and generally are not, the same if the compound is substituted with multiple PG. In general, PG will be used to protect functional groups such as carboxyl, hydroxyl, thio, or amino groups and to thus prevent side reactions or to otherwise facilitate the synthetic efficiency. The order of deprotection to yield free, deprotected groups is dependent upon the intended direction of the synthesis and the reaction conditions to be encountered, and may occur in any order as determined by the artisan.

Various functional groups of the compounds of the invention may be protected. For example, protecting groups for -OH groups (whether hydroxyl, carboxylic acid, phosphonic acid, or other functions) include "ether- or ester-forming groups". Ether- or ester-forming groups are capable of functioning as chemical protecting groups in the synthetic schemes set forth herein. However, some hydroxyl and thio protecting groups are neither ether- nor ester-forming groups, as will be understood by those skilled in the art, and are included with amides, discussed below.

A very large number of hydroxy! protecting groups and amide-forming groups and corresponding chemical cleavage reactions are described in Protective Groups in Organic Synthesis, Theodora W. Greene and Peter G. M. Wuts (John Wiley & Sons, Inc., New York, 1999, ISBN 0-471-16019-9) ("Greene"). See also Kocienski, Philip J.; Protecting Groups (Georg Thieme Veriag Stuttgart, New York, 1994), which is incorporated by reference in its entirety herein. In particular Chapter 1 , Protecting Groups: An Overview, pages 1-20, Chapter 2, Hydroxy! Protecting Groups, pages 21-94, Chapter 3, Diol Protecting Groups, pages 95-1 17, Chapter 4, Carboxyl Protecting Groups, pages 118-154, Chapter 5, Carbonyl Protecting Groups, pages 155-184. For protecting groups for carboxylic acid, phosphonic acid, phosphonate, sulfonic acid and other protecting groups for acids see Greene as set forth beiow. Such groups include by way of example and not limitation, esters, amides, hydrazides, and the like.

Ether- and Ester-forming protecting groups

Ester-forming groups inciude: (1 ) phosphonate ester-forming groups, such as phosphonamidate esters, phosphorothioate esters, phosphonate esters, and phosphon-bis-amidates; (2) carboxy! ester-forming groups, and (3) sulphur ester- forming groups, such as sulphonate, sulfate, and sulfinate. Metabolites of the Compounds of the Invention

Also falling within the scope of this invention are the in vivo metabolic products of the compounds described herein. Such products may result for example from the oxidation, reduction, hydrolysis, amidation, esterification and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the invention includes compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yield a metabolic product thereof. Such products typically are identified by preparing a radiolabeled {e.g., C 14 or H 3 ) compound of the invention, administering it parenterally in a detectable dose (e.g., greater than about 0.5 mg/kg) to an animal such as rat, mouse, guinea pig, monkey, or to man, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours) and isolating its conversion products from the urine, blood or other biological samples. These products are easily isolated since they are labeled (others are isolated by the use of antibodies capable of binding epitopes surviving in the metabolite). The metabolite structures are determined in conventional fashion, e.g., by MS or NMR analysis. In general, analysis of metabolites is done in the same way as conventional drug metabolism studies well-known to those skilled in the art. The conversion products, so long as they are not otherwise found in vivo, are useful in diagnostic assays for therapeutic dosing of the compounds of the invention even if they possess no anti-infective activity of their own.

The definitions and substituents for various genus and subgenus of the present compounds are described and illustrated herein. It should be understood by one skilled in the art that any combination of the definitions and substituents described above should not result in an inoperable species or compound,

"Inoperable species or compounds" means compound structures that violates relevant scientific principles (such as, for example, a carbon atom connecting to more than four covalent bonds) or compounds too unstable to permit isolation and formulation into pharmaceutically acceptable dosage forms.

Pharmaceutical Formulations

The compounds of this invention are formulated with conventional carriers and excipients, which will be selected in accord with ordinary practice. Tablets will contain excipients, giidants, fillers, binders and the like. Aqueous formulations are prepared in sterile form, and when intended for delivery by other than oral

administration generally will be isotonic. All formulations will optionally contain excipients such as those set forth in the Handbook of Pharmaceutical Excipients (1986), herein incorporated by reference in its entirety. Excipients include ascorbic acid and other antioxidants, chelating agents such as EDTA, carbohydrates such as dextrin, hydroxyalkylcellulose, hydroxyalkylmethylcellulose, stearic acid and the like. The pH of the formulations ranges from about 3 to about 1 1 , but is ordinarily about 7 to 10.

Whϋe it is possible for the active ingredients to be administered alone it may be preferable to present them as pharmaceutical formulations. The formulations of the invention, both for veterinary and for human use, comprise at least one active ingredient, together with one or more acceptable carriers and optionally other therapeutic ingredients. The carrier(s) must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and physiologically

innocuous to the recipient thereof.

The formulations include those suitable for the foregoing administration routes. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations generally are found in Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, Pa.), herein incorporated by reference in its entirety. Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.

Formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oi!-in-water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient may aiso be administered as a bolus, electuary or paste.

A tablet is made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered active ingredient moistened with an inert liquid diluent. The tablets may optionally be coated or scored and optionally are formulated so as to provide slow or controlled release of the active ingredient.

For administration to the eye or other external tissues e.g., mouth and skin, the formulations are preferably applied as a topical ointment or cream containing the active ingredient(s) in an amount of, for example, 0.075 to 20% w/w (including active ingredient(s) in a range between 0.1 % and 20% in increments of 0.1 % w/w such as 0.6% w/w, 0.7% w/w, etc.), preferably 0.2 to 15% w/w and most preferably 0.5 to 10% w/w. When formulated in an ointment, the active ingredients may be employed with either a paraffinic or a water-miscible ointment base. Alternatively, the active ingredients may be formulated in a cream with an oil-in-water cream base.

If desired, the aqueous phase of the cream base may include, for example, at least 30% w/w of a polyhydric alcohol, i.e. an alcohol having two or more hydroxyl groups such as propylene glycol, butane 1,3-diol, mannitol, sorbitol, glycerol and polyethylene glycol (including PEG 400) and mixtures thereof. The topical formulations may desirably include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas. Examples of such dermal penetration enhancers include dimethyl sulphoxide and related analogs.

The oily phase of the emulsions of this invention may be constituted from known ingredients in a known manner. Whiie the phase may comprise merely an emulsifier (otherwise known as an emuigent), it desirably comprises a mixture of at ieast one emulsifier with a fat or an oil or with both a fat and an oil. Preferably, a hydrophiiic emulsifier is included together with a lipophilic emulsifier which acts as a stabilizer. It is also preferred to include both an oil and a fat. Together, the

emulsifier(s) with or without stabilizer(s) make up the so-called emulsifying wax, and the wax together with the oil and fat make up the so-called emulsifying ointment base which forms the oily dispersed phase of the cream formulations.

Emulgents and emulsion stabilizers suitable for use in the formulation of the invention include Tween® 60, Span® 80, cetostearyl alcohol, benzyl alcohol, myristyl alcohol, glyceryl mono-stearate and sodium lauryl sulfate.

The choice of suitable oils or fats for the formulation is based on achieving the desired cosmetic properties. The cream should preferably be a non-greasy, non- staining and washable product with suitable consistency to avoid leakage from tubes or other containers. Straight or branched chain, mono- or dibasic alkyl esters such as di-isoadipate, isocetyl stearate, propylene glycol diester of coconut fatty acids, isopropyl myristate, decyl oleate, isopropyl palmitate, butyl stearate, 2-ethylhexyl palmitate or a blend of branched chain esters known as Crodamol CAP may be used, the last three being preferred esters. These may be used alone or in combination depending on the properties required. Alternatively, high melting point lipids such as white soft paraffin and/or liquid paraffin or other mineral oils are used.

Pharmaceutical formulations according to the present invention comprise one or more compounds of the invention together with one or more pharmaceutically acceptable carriers or excipients and optionally other therapeutic agents.

Pharmaceutical formulations containing the active ingredient may be in any form suitable for the intended method of administration. When used for oral use for example, tablets, troches, iozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs may be prepared. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation. Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable. These excipientε may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, lactose monohydrate, croscarmellose sodium, povidone, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or aiginic acid; binding agents, such as cellulose, microcrystaliine cellulose, starch, gelatin or acacia; and iubricating agents, such as magnesium stearate, stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.

Formulations for oral use may be also presented as hard gelatin capsules where the active ingredient is mixed with an inert solid diluent, for example calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, such as peanut oil, liquid paraffin or olive oil.

Aqueous suspensions of the invention contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.

Such excipients include a suspending agent, such as sodium carboxymethylceliulose, methyicellulose, hydroxypropyl methylcelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g.,

heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexito! anhydride {e.g., polyoxyethylene sorbitaπ monooleate). The aqueous suspension may also contain one or more preservatives such as ethyl or n-propyi p-hydroxy-benzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin.

Oil suspensions may be formulated by suspending the active ingredient in a vegetable oil, such as arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oral suspensions may contain a thickening agent, such as beeswax, hard paraffin or cetyt alcohol. Sweetening agents, such as those set forth herein, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an antioxidant such as ascorbic acid.

Dispersible powders and granules of the invention suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent, and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those disclosed above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present.

The pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, such as olive oil or arachis oil, a mineral oil, such as liquid paraffin, or a mixture of these. Suitable emulsifying agents include naturally-occurring gums, such as gum acacia and gum tragacanth, naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan monooieate, and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan monoofeate. The emulsion may also contain sweetening and flavoring agents. Syrups and elixirs may be formulated with sweetening agents, such as glycerol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent.

The pharmaceutical compositions of the invention may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned herein. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenteraliy acceptable diluent or solvent, such as a solution in 1 ,3-butane-diol or prepared as a lyophiiized powder. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils may

conventionally be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid may likewise be used in the preparation of injectables. The amount of active ingredient that may be combined with the carrier material to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. For example, a time-reiease formulation intended for oral administration to humans may contain approximately 1 to 1000 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions (weight:weight). The pharmaceutica! composition can be prepared to provide easily measurable amounts for administration. For example, an aqueous solution intended for intravenous infusion may contain from about 3 to 500 μg of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.

Formulations suitable for administration to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent for the active ingredient. The active ingredient is preferably present in such formulations in a concentration of 0.5 to 20%, advantageously 0.5 to 10% particularly about 1.5% w/w.

Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as geϊatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.

Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.

Formulations suitable for intrapuimoπary or nasal administration have a particle size for example in the range of 0.1 to 500 μm (including particle sizes in a range between 0.1 and 500 μm in increments such as 0.5 μm, 1 μm, 30 μm, 35 μm, etc.), which is administered by rapid inhalation through the nasal passage or by inhalation through the mouth so as to reach the alveolar sacs. Suitable formulations include aqueous or oily solutions of the active ingredient. Formulations suitable for aerosol or dry powder administration may be prepared according to conventional methods and may be delivered with other therapeutic agents such as compounds heretofore used in the treatment or prophylaxis of infections as described herein.

Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.

Formulations suitable for parenteral administration include aqueous and nonaqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterϋe suspensions which may include suspending agents and thickening agents.

The formulations are presented in unit-dose or multi-dose containers, for example sealed ampouies and vials, and may be stored in a freeze-dried

(lyophitized) condition requiring oniy the addition of the sterile liquid carrier, for example water for injection, immediately prior to use. Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets of the kind previously described. Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, of the active ingredient.

It should be understood that in addition to the ingredients particularly mentioned above the formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.

Compounds of the invention can also be formulated to provide controlled release of the active ingredient to allow less frequent dosing or to improve the pharmacokinetic or toxicity profile of the active ingredient. Accordingly, the invention also provided compositions comprising one or more compounds of the invention formulated for sustained or controlled release.

The effective dose of an active ingredient depends at least on the nature of the condition being treated, toxicity, whether the compound is being used

prophyiactically (lower doses) or against an active viral infection, the method of delivery, and the pharmaceutical formulation, and will be determined by the clinician using conventional dose escalation studies. The effective dose can be expected to be from about 0.0001 to about 100 mg/kg body weight per day; typically, from about 0.01 to about 10 mg/kg body weight per day; more typically, from about .01 to about 5 mg/kg body weight per day; most typically, from about .05 to about 0.5 mg/kg body weight per day. For example, the daily candidate dose for an adult human of approximately 70 kg body weight will range from 1 mg to 1000 mg, preferably between 5 mg and 500 mg, and may take the form of single or multiple doses.

In yet another embodiment, the present application discloses pharmaceutical compositions comprising a compound of Formula I or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or exipient.

Routes of Administration

One or more compounds of the invention (herein referred to as the active ingredients) are administered by any route appropriate to the condition to be treated. Suitable routes include oral, rectal, nasal, topical (including buccal and sublingual), vagina! and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural), and the like. It will be appreciated that the preferred route may vary with for example the condition of the recipient. An advantage of the compounds of this invention is that they are orally bioavailable and can be dosed orally.

Combination Therapy, Including HCV Combination Therapy

In another embodiment, the compounds of the present invention may be combined with one or more active agent. Non-limiting examples of suitable combinations include combinations of one or more compounds of the present invention with one or more interferons, ribavirin or its analogs, HCV NS3 protease inhibitors, NS5a inhibitors, alpha-glucosidase 1 inhibitors, hepatoprotectants, mevalonate decarboxylase antagonists, antagonists of the renin-angiotensin system, other anti-fibrotic agents, endothelin antagonists, nucleoside or nucleotide inhibitors of HCV NS5B polymerase, non-nucleoside inhibitors of HCV NS58 polymerase, HCV NS5A inhibitors, TLR-7 agonists, cyclophillin inhibitors, HCV IRES inhibitors, pharmacokinetic enhancers and other drugs for treating HCV; or mixtures thereof.

More specifically, one or more compounds of the present invention may be combined with one or more compounds selected from the group consisting of

1 ) interferons, e.g., pegylated rIFN-alpha 2b (PEG-lntron), pegylated rlFN- alpha 2a (Pegasys), rIFN-alpha 2b (Intron A), rIFN-alpha 2a (Roferon-A), interferon alpha (MOR-22, OPC-18, Aifaferone, Alfanative, Multiferon, subalin), interferon alfacon-1 (Infergen), interferon alpha~n1 (Weiiferon), interferon alpha-n3 (Alferon), interferon-beta (Avonex, DL-8234), interferon-omega (omega DUROS, Biomed 510), albinterferon alpha-2b (Albuferon), IFN alpha XL 1 BLX-883 (Locteron), DA-3021 , glycosylated interferon alpha-2b (AVI-005), PEG-!nfergen, PEGylated interferon iambda {PEGylated IL-29), and belerofon,

2) ribavirin and its analogs, e.g., ribavirin (Rebetot, Copegus), and taribavirin (Viramidine),

3) HCV NS3 protease inhibitors, e.g., boceprevir (SCH-503034 , SCH-7), teiaprevir (VX-950), VX-813, TMC-435 (TMC435350), ABT-450, BI-201335, BI-1230, MK-7009, SCH-900518, VBY-376, VX-500, GS-9256, GS-9451 , BMS-790052, BMS- 605339, PHX-1766, AS-101 , YH-5258, YH5530, YH5531 , and ITMN-191 (R-7227), 4) alpha-glucosidase 1 inhibitors, e.g., celgosivir (MX-3253), Miglitol, and UT-

231 B,

5) hepatoprotectants, e.g., emericasan (IDN-6556), ME-3738, GS-9450 (LB- 84451 ), silibilin, and MitoQ,

6) nucleoside or nucleotide inhibitors of HCV NS5B polymerase, e.g., R1626, R7128 (R4048), IDX 184, IDX-102, PSi-7851, BCX-4678, valopicitabine (NM-283), and MK-0608,

7) non-nucleoside inhibitors of HCV NS5B polymerase, e.g., fϋibuvir (PF- 868554), ABT-333, ABT-072, BI-207127, VCH-759, VCH-916, JTK-652, MK-3281 , VBY-708, VCH-222, A848837, ANA-598, GL60667, GL59728, A-63890, A-48773, A- 48547, BC-2329, VCH-796 (nesbuvir), GSK625433, BILN-1941 , XTL-2125, and GS- 9190,

8) HCV NS5A inhibitors, e.g., AZD-2836 (A-831 ), AZD-7295 (A-689), and BMS-790052,

9) TLR-7 agonists, e.g., imiquimod, 852A, GS-9524, ANA-773, ANA-975, AZD-8848 (DSP-3025), PF-04878691 , and SM-360320,

10) cyclophillin inhibitors, e.g., DEBIO-025, SCY-635, and NIM811 ,

1 1 ) HCV IRES inhibitors, e.g., MCI-067,

12) pharmacokinetic enhancers, e.g., BAS-100, SPi-452, PF-4194477, TMC- 41629, GS-9350, GS-9585, and roxythromycin,

13) other drugs for treating HCV, e.g., thymosin alpha 1 (Zadaxin), nitazoxanide (Alinea, NTZ), BIVN-401 (virostat), PYN-17 (aftirex), KPE02003002, acttlon (CPG-10101 ), GS-9525, KRN-7000, civacir, GI-5005, XTL-6865, B1T225, PTX-111, ITX2865, TT-033i, ANA 971 , NOV-205, tarvacin, EHC-18, VGX-410C, EMZ-7G2, AVi 4065, BMS-650032, BMS-791325, Bavituximab, MDX-1 106 (ONO- 4538), Ogiufanide, FK-788, and VX-497 (merimepodib)

14) mevalonate decarboxylase antagonists, e.g., statins, HMGCoA synthase inhibitors (e.g., hymeglusin), squalene synthesis inhibitors (e.g., zaragozic acid);

15) angiotensin Ii receptor antagonists, e.g., losartan, irbesartan, olmesartan, candesartan, valsartan, telmisartan, eprosartan;

16) angiotensin-converting enzyme inhibitors, e.g., captopril, zofenopril, eπalapri!, ramipril, quinapril, perindopriϊ, iisinopril, benazepril, fosinopril;

17) other anti-fibrotic agents, e.g., amiloride and

18) endotheiin antagonists, e.g. bosentan and ambrisentan.

In yet another embodiment, the present application discloses pharmaceutical compositions comprising a compound of the present invention, or a pharmaceutically acceptable salt thereof, in combination with at least one additional active agent, and a pharmaceutically acceptable carrier or excipient. In yet another embodiment, the present application provides a combination pharmaceutical agent with two or more therapeutic agents in a unitary dosage form. Thus, it is also possible to combine any compound of the invention with one or more other active agents in a unitary dosage form.

The combination therapy may be administered as a simultaneous or sequential regimen. When administered sequentially, the combination may be administered in two or more administrations.

Co-administration of a compound of the invention with one or more other active agents generally refers to simultaneous or sequential administration of a compound of the invention and one or more other active agents, such that therapeutically effective amounts of the compound of the invention and one or more other active agents are both present in the body of the patient.

Co-administration includes administration of unit dosages of the compounds of the invention before or after administration of unit dosages of one or more other active agents, for example, administration of the compounds of the invention within seconds, minutes, or hours of the administration of one or more other active agents. For example, a unit dose of a compound of the invention can be administered first, followed within seconds or minutes by administration of a unit dose of one or more other active agents. Alternatively, a unit dose of one or more other active agents can be administered first, followed by administration of a unit dose of a compound of the invention within seconds or minutes. In some cases, it may be desirable to administer a unit dose of a compound of the invention first, followed, after a period of hours (e.g., 1-12 hours), by administration of a unit dose of one or more other active agents. In other cases, it may be desirable to administer a unit dose of one or more other active agents first, followed, after a period of hours (e.g., 1-12 hours), by administration of a unit dose of a compound of the invention.

The combination therapy may provide "synergy" and "synergistic effect", i.e. the effect achieved when the active ingredients used together is greater than the sum of the effects that results from using the compounds separately. A synergistic effect may be attained when the active ingredients are: (1 ) co-formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen. When delivered in alternation therapy, a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g., in separate tablets, pills or capsules, or by different injections in separate syringes. In general, during alternation therapy, an effective dosage of each active ingredient is administered sequentially, i.e. serially, whereas in combination therapy, effective dosages of two or more active ingredients are administered together.

As will be appreciated by those skilled in the art, when treating a viral infection such as HCV, such treatment may be characterized in a variety of ways and measured by a variety of endpoints. The scope of the present invention is intended to encompass all such characterizations.

Synthetic Examples

Certain abbreviations and acronyms are used in describing the experimental details. Although most of these would be understood by one skilled in the art, Table 1 contains a list of many of these abbreviations and acronyms.

Table 1. List of abbreviations and acronyms.

Abbreviation Meaning

Ac 2 O acetic anhydride

AIBN 2,2'-azobis(2-methylpropionitrile)

Bn benzyl BnBr benzylbromide

BSA bis(trimethylsilyi)acetamide

BzCI benzoyl chloride

CDI carbonyl dϋmidazoie

DABCO 1 ,4-diazabicycio[2.2.2]octane

DBN 1 ,5-diazabicycio[4.3.0]non-5-ene

DDQ 2,3-dichloro-5,6-dicyano-1 ,4-benzoquinone

DBU 1 ,5-diazabicyclo[5.4.0]undec-5-ene

DCA dichloroacetamide

DCC dicydohexylcarbodiimide

DCM dichloromethane

DMAP 4-dimethylaminopyridine

DME 1 ,2-dimethoxyethane

DMTCI dimethoxytrityi chloride

DMSO dimethylsuSfoxide

DMTr 4, 4'-dimethoxytrityi

DMF dimethyiformamide

EtOAc ethyl acetate

ESI eiectrospray ionization

HMDS hexamethyldisilazane

HPLC High pressure liquid chromatography

LDA lithium diisopropylamide

LRMS low resolution mass spectrum

MCPBA meta-chloroperbenzoic acid

MeCN acetonitrsle

MeOH methanol

MMTC mono methoxytrityl chloride

m/z or m/e mass to charge ratio

MH + mass plus 1

MH " mass minus 1

MsOH methanesulfonic acid

MS or mε mass spectrum

NBS N-bromosuccinsmide

Ph phenyl rt or r.t. room temperature

TBAF tetrabutylammonium fluoride

TMSC! chlorotrimethylsilane

TMSBr bromotrimethyisilane

TMSI iodotrimethyisilane

TMSOTf (trimethylsilyl)trifluoromethy!sulfonate

TEA triethylamine

TBA trSbutylamine

TBAP tributylammonium pyrophosphate

TBSCI t-butyldimethylsilyl chloride

TEAB triethylammonium bicarbonate

TFA trifluoroacetic acid

TLC or tic thin layer chromatography

Tr triphenyimethyl

To! 4-methyfbenzoyi

Turbo Grignard 1 :1 mixture of isopropyimagπesium chloride and lithium chloride δ parts per million down field from tetramethylsiiane

General Schemes

The compounds of this invention may be synthesized by several routes with key bond-forming steps as indicated in Schemes A-C, in which the carboxylate substituent R indicates either a protecting group such as an alkyi ester (where necessary), or the free acid itself. Alkyl ester protecting groups are conveniently removed by saponification with an alkali metal hydroxide in a protic solvent such as water or an alcohol, and may be facilitated by use of ethereal solvent mixtures and/or heating. Alternatively they may be removed by dealkySation through heating with an alkali metal halide in an aprotic solvent. As will be appreciated, substituentε on Het may be modified subsequent to other bond-forming steps by, for exampSe, N- oxidation with a typical oxidant such as metachioroperbenzoic acid in a solvent such as dichloromethane, O-dealkylation through treatment with a reagent such as boron tribromide in a solvent such as dichioromethane, or hydrolysis.

Scheme A

The bond between L and Het may be formed by displacement of X on Het, where X is a leaving group such as a halide, suifinate, sulfonate or phosphate moiety. The reaction is conveniently performed by deprotonation of L-H with a base such as sodium hydride or potassium hexamethyldisiiazide, or is facilitated by the presence of a tertiary amine; it can be carried out in a variety of solvents such as THF, dioxane, dichloromethane, NMP, DMF or DMSO and may be accelerated by heating.

Scheme B

The bond between R 3 and L may be formed by nucleophilic displacement of a leaving group X on R 3 . The leaving group may vary widely and includes, but is not limited to, haiide, carboxylate, suifinate, sulfonate or phosphate moieties, and it may be generated from the corresponding alcohol in situ through treatment with reagents such as dialky! azodicarboxylates. The reaction may also be facilitated by deprotonation of Het-L-H with a base such as sodium hydride or potassium hexamethyldisiiazide, or is facilitated by the presence of a tertiary amine; it can be carried out in a variety of solvents such as THF, dioxane, dichloromethane, NMP, DMF or DMSO and may be accelerated by heating.

Scheme C

The starting materia! in Scheme A may be synthesized as depicted in Scheme C. Substituted 3-aminothiophenes Il may be generated by reductive amination of Y-R 3 -L-R (where Y indicates an aldehyde or ketone and R and R a depict optional protecting groups), or by direct alkylation (where Y indicates a leaving group such as a halide, sulfinate, sulfonate or phosphate moiety) of the 3-aminothiophene I (see patent application WO2008/58393). In the latter case the aikylation may be facilitated by deprotonation of the amine with a base such as sodium hydride or potassium hexamethyldisilazide, and can be carried out in a variety of solvents such as THF, dioxane, dichloromethane, NMP, DMF or DMSO and may be accelerated by heating. In cases where R 3 is aromatic, the reaction may be catalyzed by Pd (J. Org. Chem., 2000, 65, 1158-1174). Alternatively H may be generated by coupling of an amine with a 3-iodothiophene IV catalyzed by Pd (J. Org. Chem., 2000, 65, 1158- 1 174). The amine Ii is converted to the amide III by acyiation with a carboxylic acid derivative such as an acyl chloride or anhydride in the presence of a base such as pyridine or a tertiary amine in an inert solvent such as dichloromethane. Alternatively IV may be converted to IM directly by amidation catalyzed by Cu (J. Am. Chem. Soc, 2002, 124, 7421-7428).

The starting material for Scheme B may be generated in an analogous fashion, with the leaving group X being generated in a final step by standard methods from the precursor alcohol. The synthesis of iodothiophene IV is illustrated beiow for the case where R 1 tBu, and other variants may be synthesized in analogous fashion:

Scheme D nBuLi (2.2 equiv)

To a solution of 5-(3,3-Dimethy!-but-1-yny!)-thiophene-2-carboxySic acid (6.2 g, 30 mmol; see patent application US5861421 ) in THF (100 mL) was added a solution of nBuLi (2.0 M in pentane, 33 mL, 66 mmol) via an addition funnel at -78 0 C. After addition, the reaction was stirred at -78 0 C for 1 h. A solution of I 2 (7.7 g, 30 mmoi) in THF (100 mL) was added slowly (ca. 15 min) to the flask. After a further 10 mins, the reaction was quenched with 1 N HCI (50 mL) and warmed to room temperature. The volatiles were removed in vacuo and the residue was dissolved in ether (500 mL). The organic solution was washed with 1 M Na 2 S 2 O 3 (100 mL x 2), brine (100 mL) and dried over Na 2 SO 4 . After concentrated in vacuo, the residue was purified by silica gel chromatography (EtOAc/hexanes) to give 5-(3,3-Dimethy!-buM-yny!)-3-iodo- thiophene-2-carboxylic acid (5.9 g, 65%) as a white solid. Scheme E

To a solution of 5-(3,3-Dimethyl-but-1-ynyl)-3-iodo-thiophene-2-carboxyiic acid (1.0 g, 3.0 mmol) and DMF (20 μL) in dry dichloromethane (10 mL) was added oxalyl chloride (508 μL, 6.0 mmol) at room temperature. After stirring at room temperature for 90 min, the reaction was concentrated in vacuo to remove volatiles. The residue was dissoived in pyridine (5 ml_) and methanol (5 mL) and stirred for 2 h. The voiatites were removed in vacuo and the residue was participated between ether (150 mL) and saturated NH 4 CI solution (50 mL). The organic layer was washed with saturated NH 4 CI solution (50 mL) and dried over Na 2 SO 4 . After concentration in vacuo, the residue was purified by siiica gel chromatography (EtOAc/hexanes) to give the desired product (835 mg, 80%).

Experimentais

Example 1 - Compound 1 : 5-f3,3-Dimethyi-but-1-vnyl)-3-{(fr'ans-4-methyl- cvclohexanecarbonvi)-rfrans-4-(ρyridin-2-yioxy)-cvclohexyl] -aminol-thiophene-2- carboxyiic acid Scheme 1

ompound 1

A mixture of 5-(3,3-Dimethyl-but-1-ynyl)-3-[(£ran$-4-hydroxy-cyclohexyi) - (frans-4-methyl-cyclohexanecarbonyl)-amino]-thsophene-2-carb oxyiic acid (78 mg,

0.175 mmol) and 2-fluoro-pyridine (145 μL, 1.68 mmol) in DMF (0.6 mL) was treated with sodium hydride (67 mg, 1.68 mmol, 60% oil dispersion) in two or three portions.

The mixture was stirred until the bubbling slowed, and was sealed and heated by microwave at 100 deg C for 30 min. After cooling, ethyl acetate (2-3 mL) was added and the mixture was carefully quenched with citric acid (10% aqueous soiution, 2-3 mL). Water was added and the mixture was extracted with ethyl acetate (2x30 mL).

The combined organic layers were dried over sodium sulfate, filtered and concentrated. The residue was purified by HPLC (Gemini column, 35% acetonitrile:water, 2 min, 35-50% acetonitrile:water, 2 min, 50-95% acetonitrile: water 13 min, both solvents containing 0.1% trifluoroacetic acid). This resulted in 65 mg (58% yield) of the title compound as a white powder (TFA salt): MS (m/z): 520.9 [M- H] " ; HPLC retention time: 4.61 min (2-98% acetonitrile:water with 0.05% trifluoroacetic acid).

Example 2 - Compound 2: 5-(3,3-Dimethvi-but-1-vnyl)-3-{(trans-4-methγl- cyciohexa ι neca ι rbgnvl)-[frans-4-fpyrazin-2-yloxy)-cvclohexyπ-arninoH hiophene-2- carboxylic acid

Compound 2

The title compound was synthesized in a manner analogous to Example 1 , using 2-chloro-pyrazine in place of 2-fluoro-pyridine. Also, siiica gei chromatography (1 % EtOH:dichloromethane 2min, 1-8% EtOH:dichloromethane, 12 min, 30 mL/min, 12 g siiica column) was utilized instead of HPLC for purification: MS (m/z): 522.1 [M- H] " ; HPLC retention time: 5.04 min (2-98% acetonitri!e:water with 0.05% tifluoroacetic acid).

Example 3 - Compound 3: 5-(3,3-Dimethyl-but-1-vnyi)-3-((trans-4-methyl- cyclohexanecarbonγl)-j " frans-4-(6-methyl-pyridazin-3-yloxy)-cyciohexyll-amino }- thiophene-2-carboxyiic acid

Compounod The title compound was synthesized in a manner analogous to Example 1 , using 3-chloro-6-methyS-pyridazine in place of 2-fluoro-pyridine: MS (m/z): 536.1 [M- H] " ; HPLC retention time: 3.73 min (2-98% acetonitrile:water with 0.05% tifluoroacetic acid).

Example 4 - Compound 4: 5-f3.3-Dimethyi-but-1 -vnylV3-((trans-4-fnethyl- cvclohexanecarfaonyl)-| ' fra/7S-4-(pyrimidin-2-yloxy)-cyc[ohexyn-aminoHhtophene -2- carboxyiic acid

Compound 4 The title compound was synthesized in a manner analogous to Example 1 , using 2-chloro-pyrimidine in place of 2-fiuoro-pyridine: MS (m/z): 522.0 [M-H] " ; HPLC retention time: 4.82 min (2-98% acetonitrile:water with 0.05% tifluoroacetic acid).

Example 5 - Compound 5: 5-f3,3-Dimethyl"but-1-ynylV3-{(frans-4-methyl- cyciohexanecarbonyl)-ffraπS"4-(pyridin-3-yloxy)-cvclohexyπ -aminoHhiophene-2- carboxyiic acid

CompOound 5-

The title compound was synthesized in a manner analogous to Example 1 , using 3-fluoro-pyridine in piace of 2-fluoro-pyridine: MS (m/z): 520.9 [M-H] " ; HPLC retention time: 3.54 min (2-98% acetonitrile:water with 0.05% tifiuoroacetic acid).

Example 6 - Compound 6: 5-f3,3-Dimethyl-but-1-ynylV3-rf^ans-4-fβ- methanesulfonyl-pyridin-2-yloxy)-cyc [ ohexylHfrans-4-methyi-cyclohexanecarbonyl)- aminoHhiophene-2-carboxylic acid

Compound 6

The title compound was synthesized in a manner analogous to Example 1 , using 2-chioro-6-methanesulfonyl-pyridine in place of 2-fluoro-pyridine: MS (m/z): 598.9 [M-HP; HPLC retention time: 4.79 min (2-98% acetonitrϊle:water with 0.05% tifluoroacetic acid).

Example 7 - Compound 7: 5-(3,3-Pimethyl-but-1-γnvh-3-{«τaπs-4-methyl- cvdohexanecarbonyiHfran5-4-(6-methyl-pyridin-3-vioxy)-cycJoh exyi1-amino}- thiophβne-2-carboxylic acid

Compound 7

The title compound was synthesized in a manner analogous to Example 1 , using 5-fluoro-2-methyl-pyridine in place of 2-fluoro-pyridine: MS (m/z): 534.9 [M-H] " ; HPLC retention time: 4.40 min (2-98% acetonitriie:water with 0.05% tifluoroacetic acid).

Example 8 - Comgound 8: 5-(3,3-Dimethyl-but-1-ynyl)-3-{ffrans-4-methyl- cvclohexanecarbonylHfrans-4-(6-methyl-pyridin-2-yloxy)-cvcio hexyl]-amino}- thiophene-2-carboxylic acid

Compound 8

The title compound was synthesized in a manner analogous to Exampie 1 , using 2-chloro-6-methyl-pyridine in place of 2-fIuoro-pyridine: MS (m/z): 534.8 [M-H] " ; HPLC retention time: 4.05 min {2-98% acetonitrile:water with 0.05% tifluoroacetic acid).

Exampie 9 - Compound 9: 3-fl ' fr ι ans-4-f5-Cvano-pyridin-2-yloxy)-cvclohexyπ-(fr " ans-4- methyi-cvclohexanecarbonvπ-aminol-S-fS^-dimethyl.-bMMiyJI^O -thiophβrie-S- carboxylic acid

Compound 9

The title compound was synthesized in a manner analogous to Example 1 , using 6-chloro-nicotinonitriSe in place of 2-fluoro-pyridine: MS (m/z): 545.9 [M-H] " ; HPLC retention time: 5.17 min (2-98% acetonitrile:water with 0.05% tifluoroacetic acid).

Example 10 - Compound 10: 5-(3.3-Dimethyl-but-1-ynyl)-3-f(frans-4-methyl- cyciohexanecarbonyl)-rfrans-4-(5-trifiuoromethyi-pyridin-2-v ioxy)-cycJohexyn-amino)- thiophene-2-carboxylic acid

Compound 10 The title compound was synthesized in a manner analogous to Example 1 , using 2-chloro-5-trifluoromethy!-ρyridine in place of 2-fluoro-pyridine: MS (m/z): 588.8 [M-H] ' ; HPLC retention time: 5.55 min (2-98% acetonitriie: water with 0.05% tifluoroacetic acid).

Example 11 - Compound 11 : 5-(3,3-Dimethyl-but-1-ynvO-3~{(frans-4-rnethyl- cvcSohexanecarbonγl)-ffran5-4-(5-methyl-pyridin-2-vioxy)-cv c[ohexyi1-amino}- thiophene-2-carboxylic acid

Compound 11 The title compound was synthesized in a manner analogous to Example 1 , using 2-chloro-5-methy!-pyridine in place of 2-fluoro-pyridine: MS (m/z): 534.8 [M-H] " ; HPLC retention time: 4.44 min (2-98% acetonitrile:water with 0.05% tifluoroacetic acid). Example 12 - Compound 12: 5-f3.3-Dimethyl-but-1-vnyl)-3-fffrans-4-methyl- cvciohexanecarbonyl)-rfrans-4-(3-methyl-pyridin-2-yloxy)-cvc Johexyl1-amino)- thiophene-2-carboχylic acid

Compound 12

The title compound was synthesized in a manner analogous to Example 1 , using 2-chloro-3-methyi-pyridine in place of 2-fluoro-pyridine: MS (m/z): 534.9 [M-H] " ; HPLC retention time: 5.28 min (2-98% acetonitrile:water with 0.05% tifluoroacetic acid). Example 13 - Compound. JJ3: 3-[rfra/7s-4-f6-Cyano-pyridin-2-vioxy)-cvclohexyπ- ffrans-4-methv!-cvclohexanecarbonvi)-arninol-5-f3 1 3-ciirnethv[-but--1-vnviHhiophene- 2-carboxylic acid

Compoound 13

The title compound was synthesized in a manner analogous to Exampie 1 , using δ-chioro-pyridine^-carbonitrile in place of 2-fiuoro-pyridine: MS (m/z): 545.9 [M-H] " ; HPLC retention time: 5.17 min (2-98% acetonitrile:water with 0.05% tifluoroacetic acid).

Example 14 - Compound 14: 5-f3,3-Dimethyl-but-1-vnyl)-3-((frans-4-methyi- cyciohexanecarbonyl)-^f'an5-4-(6-oxo-1 ,6-dihvdropyridazin-3-yloxy)-cyclohexyll- amino}-thiophene-2-carboxyiic acid

Scheme 2

Compound 14

A mixture of 5-(3,3-Dimethyi-but-1 -ynyt)-3-[(frans-4-hydroxy-cycfohexyl)- (fra/7s-4-methyl-cyclohexanecarbonyl)-amino]-thiophene-2-car boxy]ic acid (53 mg,

0.12 mmol) and 3,6-dichloropyridazine (86 mg, 0.58 mmol) in DMF (0.5 mL) was treated with sodium hydride (58 mg, 1.4 mmol, 60% oil dispersion) in two portions.

The mixture was stirred until the bubbling slowed, and was sealed and heated by microwave at 100 deg C for 30 min. After cooling, ethyl acetate (2-3 mL) was added and the mixture was carefuNy quenched with citric acid (10% aqueous solution, 2-3 mL). Water was added and the mixture was extracted with ethyt acetate (2x30 mL).

The combined organic layers were dried over magnesium sulfate, filtered and concentrated. The residue was passed through a plug of silica (10% MeOH: DCM eluent) and concentrated to yield 69 mg of 5-(3,3-Dimethyl-but-1-ynyl)-3-{(frans-4- methyl~cyclohexanecarbony!)-[fΛa/7s-4-(6-chloro-pyridazin-3 ~yioxy)-cyclohexyl]- amino}-thiophene-2-carboxyiic acid as a black oil which was carried on without further purification.

A crude sample of (3,3-Dimethyl-but-1-ynyl)-3-{(frans-4-methyl- cyclohexanecarbonyI)-[frans-4-(6-chloro-pyridazin-3-ySoxy)-c yclohexyl]-amino}- thiophene-2-carboxylic acid (69 mg) was dissolved in acetic acid (1 mL) and treated with sodium acetate (36 mg). The mixture was heated at 100 deg C for 4 hours and then partitioned between water and ethyl acetate. The aqueous phase was neutralized by addition of aqueous sodium hydroxide and extracted with ethyl acetate (3X30 mL). The combined organic layers were dried over magnesium sulfate, filtered and concentrated. The residue was purified by HPLC (Gemini column; 25% acetonitrile:water, 2 min; 25-100% acetonitrile:water, 16 min; 100% acetonitrile, 3 min; both solvents containing 0.1% trifluoroacetic acid). This resulted in 14 mg (19% yield over 2 steps) of the title compound as a white powder (TFA salt): MS (m/z): 537.9 [M-Hf; HPLC retention time: 4.31 min (2-98% acetonitrile:water with 0.05% trifluoroacetic acid).

Example 15 - Compound 15: 5-(3,3-Dimethyl-but-1 -vnyl)-3-f(frans-4-methyl- cyclohexanecarbon ι γ ι lj ι: rfrans-4-(thiazol-2-yioxy)-cvclohexyπ-amino)-thiophen e-2- carboxyiic acid

Compound 15

The title compound was synthesized in a manner analogous to Example 1 , using 2-chioro-thtazole in place of 2-fluoro-pyridine: MS (m/z): 526.9 [M-H] " ; HPLC retention time: 5.08 min (2-98% acetonitrile:water with 0.05% tifiuoroacetic acid).

Example 16 - Compound 16: 5-(3,3-Dimethyl-but-1-vnyl)-3-{(frans-4-methyl- cvclohexanecarbonvO-rfrans^-fS-trifluoromethyl-pyridin-Σ-yl ox^jh^y^lghexyll-arnino}- thiophene-2-carboxylic acid

The title compound was synthesized in a manner analogous to Example 1 , using 2-bromo-3-trifluoromethyl-pyridine in place of 2-fluoro-ρyridine: MS (m/z): 588.8 [M-H] " ; HPLC retention time: 5.47 min (2-98% acetonitrile:water with 0.05% tifluoroacetic acid).

Example 17 - Compound 17: 5-(3,3-Dimethyl-but-1-vnyi)-3-{ffrans-4-methγi- cvciohexanecarbonvi)-r^ran5-4-f6-trifluoromethyl-pyridin-2-y loxy)-cvciohexyil-amino)- thiophene-2-carboxylic acid

Compound 17

The title compound was synthesized in a manner anaiogous to Example 1 , using 2-chloro-6-trifluoromethyl-pyridine in place of 2-fluoro-pyridine: MS (m/z): 588.9 [M-H] " ; HPLC retention time: 5.45 min (2-98% acetonitrile:water with 0.05% tiffuoroacetic acid).

Example 18 - Compound 18: 5-(3,3-Dimethyl-but-1-ynyl)-3-fjϊraf75-4-(6-methoxy- pyridin-2-vioxyVcvclohexylV(fraπs-4-methyl-cvclohexa

2-carboxyiic acid

Compound 18

The title compound was synthesized in a manner analogous to Example 1 , using 2-chloro-6-methoxy-pyridinein place of 2-fluoro~pyridine: MS (m/z): 550.9 [M- H] " ; HPLC retention time: 5.29 min (2-98% acetonitrile:water with 0.05% tifluoroacetic acid).

Example 19 - Compound 19: 5-f3.3-Dimethyl-but-1 -vny[)-3-frfrans-4-(2-methoxy- βyπdiπ:,4:Vlo χ v)-cvciohexyl1-(fra/?s-4-methvi-cyclohexanecarbonyl)-a mino1-thiophene- 2-carboxyiic acid

CompSou,nd 19 The title compound was synthesized in a manner analogous to Example 1 , using 4-chloro-2-methoxy-pyridinein place of 2-fluoro-pyridine: MS (m/z): 550.9 [M- H] " ; HPLC retention time: 3.75 min (2-98% acetonitrile:water with 0.05% tifluoroacetic acid). Example 20 - Compound 20: 5-f3,3-Dimethyl-but-1-vnyl)-3-f(-rans-4-methv8- cvclohexanecarbonyl)-fc/s-3-(pyridin-3-yloxy)-cyclobutyl1-am ino)-thiophene-2- carboxylic acid

Compound 20

Scheme 3

A mixture of 3-amino-5-(3,3-dimethyl-but-1-ynyl)-thiophene-2-carboxyIic acid methyl ester, hydrochloride salt (750 mg, 2.74 mmol) and 3-benzyloxy-cyclobutanone (2.5 g, 14.3 mmo)) in DCM (40 mL) was treated with sodium triacetoxyborohydride (3.34 g, 15.8 mmol) portioπwise. The mixture was stirred at room temperature for 3 hours. The solution was diluted with ethyl acetate, washed sequentially with saturated aqueous sodium bicarbonate, water, and brine. The organic phase was dried over magnesium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (0-10% ethyl acetate:hexanes). This resulted in 1.26 g (quant, yield) of 3-(3-benzyloxy-cycSobutyiamino)-5-(3,3-dimethyl-but-1-ynyl)- thiophene-2-carboxylic acid methyl ester as a mixture of cis- and tranε-isomers.

A mixture of 3-(3-Benzyloxy-cyc!obutylamino)-5-(3,3-dimethyl-but-1-ynyi)- thiophene-2-carboxylic acid methyl ester (1.26 g, 3.17 mmol) and N, N- diisopropySethylamine (2.4 mL, 13.8 mmol) in DCE (8 mL) was treated with trans-4- methyl-cyclohexanecarbonyl chloride and heated to 100 deg C for 5 hours. The reaction mixture was cooled to room temperature, adsorbed onto silica, and purified by silica gel chromatography (0-30% ethyl acetate:hexanes). This resulted in 590 mg (36% yield) of c/s-cyclobutyl isomer (c/s-S-^S-benzySoxy-cyclobutyO-tΦmethyl- cyclohexanecarbonyl)-amino]-5-(3,3-dimethyl-but-1 -ynyl)-thiophene-2-carboxylic acid methy! ester) and 230 mg (14% yield) of the frans-cyclobuty! isomer (trans-3-[(3- benzySoxy-cycJobutylH4-methyl-cyclohexanecarbonyf)-amino]-5- (3,3-dimethyl-but-1- yny!)-thiophene-2-carboxylic acid methyl ester).

3-[(c/s-3-benzyloxy-cyciobutyl)-(fraπs-4-methyl-cyclohexane carboπyi)-amino]- 5-(3,3-dimethyI-but-1-ynyi)-thtophene-2-carboxylic acid methyl ester (290 mg, 0.55 mmol) in DCM (2 mL) at 0 deg C was treated with boron tribromide (0.85 mL, 1.0 M solution in DCM). The mixture was stirred at 0 deg C for 15 minutes and quenched with the addition of silica gel. Voiatiles were evaporated under reduced pressure and the reaction mixture was purified by siϋca gel chromatography (0-100% ethyl acetate: hexanes). This resulted in 210 mg (88% yield) of 5-{3,3-Dimethyl-but-1-ynyI)- 3-[(c/s-3-hydroxy-cydobutyl)-(frans-4-methy j -cyclohexanecarbonyl)-amino]- thϊophene-2-carboxylic acid methyl ester.

5-(3,3-Dimethyl-but-1 -ynyl)-3-[(c/s-3-hydroxy-cyclobutyl)-(frans-4-methyl- cyclohexanecarbonyl)-amino]-thiophene-2-carboxylic acid methyl ester (400 mg, 0.93 mmol) in a 3:2:1 mixture of THF:MeOH:water (20 mL) was treated with lithium hydroxide (4.5 mL, 1.0 M aqueous solution) and heated to 60 deg C for 2 hours. The organic voiatiies were evaporated under reduced pressure and remaining solution was acidified with 10% HCI (aq) . A white precipitate was collected by vacuum filtration, washed with water, and dried to afford 290 mg (74% yield) of 5-(3,3-Dimethyl-but-1 - ynyl)-3-[(c/s » 3-hydroxy-cyclobutyl)-(4-methyi-cyclohexanecarbonyl)-a mino]- thiophene-2-carboxyiic acid as a white powder. MS (m/z): 415.8 [M-H] " ; HPLC retention time: 4.10 min (2-98% acetonitπ!e:water with 0.05% trifluoroacetic acid).

The title compound was synthesized in a manner analogous to Example 1 , using 5-(3,3-dimethyl-but-1-ynyl)-3-[(c/s-3-hydroxy-cyclobuty!)-(4 -methyl- cyclohexanecarbonyl)-amtno]-thiophene-2-carboxylic acid in place of 5-(3,3- Dimethyi-but-1-ynyl)-3-[(/rans-4-hydroxy-cyclohexyl)-(f/ * ans-4-methyl- cyclohexanecarbonyl)-amino]-thiophene-2-carboxy)ic acid and 3-fluoro-pyridine in place of 2-fluoro-pyridiπe: MS (m/z): 492.8 [M-H] " ; HPLC retention time: 3.44 min (2- 98% acetonitrile: water with 0.05% tifluoroacetic acid). Example 21 - Compound 21 : 5-(3,3-Dimethyl-but-1-vnyl)-3-{(frans-4-rnethyl- cyclohexanecarbonylHfr'ans-3-(pyridin-3-yloxy)-cyclobutvπ-a mino)-thioρhene-2- carboxylic acid

Compound 21

The title compound was synthesized in a manner analogous to Example 20, using 3-[(frans-3-benzyloxy-cyclobutyl)-(frans-4-methyl-cyc!ohexan ecarbonyI)- amino]-5-(3,3-dimethyl-but-1-ynyl)-thiophene-2-carboxylfC acid methyl ester in piace of 3-[(c/s-3-benzyloxy-cyclobutyl)-(fr-ans-4-methyi-cyclohexane carbonyl)-amino]-5- (3,3-dimethyl-but-1-ynyl)-thiophene-2-carboxylic acid methyl ester: MS (m/z): 492.8 [M-H] " ; HPLC retention time: 3.45 min (2-98% acetonitrile:water with 0.05%

tifluoroacetic acid). Example 22 - Compound 22: 5-f3,3-Dimethyl-but-1-vnvπ-3-f(frans-4-methvi- cvclohexanecarbonviHc/s-3-(pyhmidin-2-yloxy)-cyclobutyi]-ami noHhiophene-2- carboxyiic acid

Compound 22

The title compound was synthesized in a manner analogous to Example 20, using 2-chioro-pyrimidine in place of 3-fluoro-pyridine: MS (m/z): 493.8 [M-H] " ; HPLC retention time: 3.52 min (2-98% acetonitrile:water with 0.05% tifluoroacetic acid). Example 23 - Compound 23: 5-(3,3-Dimethyl-but-1 -vnylV3-{(frans-4-methyt- cvclohexanecarbonv!)-rf/'ans-3-(pyrimidin-2-yloxy)-cvclobutv []-aminoHhiophene-2- carboxylic acid

Compound 23

The title compound was synthesized in a manner analogous to Example 20, using 3-[(frans-3-benzyloxy-cyclobutyl)-(frans-4-methyl-cyciohexan ecarbonyl)- amino]-5-(3,3-dimethyl-but-1-ynyl)-thiophene-2-carboxylic acid methyl ester in place of 3-[(c/s-3-benzyloxy-cyclobutyl)-(frans-4-methyl-cyciohexanec arbonyl)-amino]-5- (3,3-dimethyl-but-1-yny!)-thiophene-2-carboxylic acid methyl ester and 2-chioro- pyrimidine in place of 3-fluoro-pyridine: MS (m/z): 493.8 [M-H] " ; HPLC retention time: 3.53 min (2-98% acetonitrite:water with 0.05% tifiuoroacetic acid). Example 24 - Compound 24: S-fS^Dimethyl-but-i-vnvD-S-^frans-Φmethyl- cycSohexanecarbonyl)-[frans-4-(1 -oxy-pyridin-3-yloxy)-cvciohexyπ-amino)-thiophene- 2-carboxylic acid

Compound 24

The title compound was synthesized in a manner analogous to Example 1 , except 3-fluoro-pyridine 1 -oxide was used in piace of 2-fluoro-pyridine and the reaction was run at 60 deg C for 1 h instead of 100 deg C for 30 min: MS (m/z):

536.8 [M-H] " ; HPLC retention time: 4.16 min (2-98% acetonitrile:water with 0.05% tifiuoroacetic acid). Example 25 - Compound 25: 5-(3,3-Dimethvi -but-1 -vnvO-3-{(4-methyl- cvclohexanecarbonvi)-F4-(Quinoliπ-4-vioxy)-cvclohexyll-amin oHhiophene-2- carboxyiic acid

Compound 25

The title compound was synthesized in a manner analogous to Example 1 , using 4-chloroquinoiine in piace of 2-fiuoro-pyridine: MS (m/z):572.8 [M+H]; HPLC retention time 3.66 min (2-98% acetonitrile: water with 0.05% trifiuoroacetic acid)

Example 26 - Compound 26: 3-f[4-(2,6-diethyl-pyridin-4-vSoxy)-cvclohexyi]-(4- methyl-cvclohexanecarbony])-amino ' l- 5-f 3,3-Dimethyl -but-1 -ynyl)-thiophene-2- carboxyiic acid

The title compound was synthesized in a manner analogous to Example 1 , using 4-chloroquinoline in place of 2-fiuoro-pyridine: MS (m/z): 578.0 [M+H]+; HPLC retention time 3.72 min (2-98% acetonitrile: water with 0.05% trifiuoroacetic acid)

Example 27 - Compound 27: 5-(3,3-DimethyJ -but-1 -ynyi)-3-(ftrans-4-methyl- cvclohexanecarbonyl)-[trans-4-(4-pyrrolidin-1-ylmethyl-pyrid in-2-vioxy)-cyclohexyll- aminoHhiophene-2-carboxylic acid

Scheme 4

Compound 27

The mixture of 2-chloro-pyridine-4-carbaldehyde and (300mg, 2.12mmoi) and pyrrolidine (0.19ml, 2.12mmol) in DCM (10 ml) was treated with acetic acid (0.25m!, 4.24mmol). The mixture was stirred at room temperature for 10 minutes and

NaBH(OAc) 3 ( 899 mg, 4.24mmol) was added. After overnight stirring, the reaction was washed with sat. NaHCO 3 and extracted twice with DCM. The organic layers were combined and washed with water, brine, dried over sodium sulfate, filtrated and concentrated. The crude material was purified by column chromatography with MeOH/DCM (0-10%) and concentrated to give 120mg of 2-chloro-4-pyrrolidin-1- yimethyl-pyridine as a clear oil.

The title compound was synthesized in a manner analogous to Example 1 , using 2-chloro-4-pyrroIidin-1-ylmethyi-pyridine in place of 2-fiuoro-ρyridine: MS (m/z); 606.1[M+H]+; HPLC retention time 3.64 min (2-98% acetonitriie: water with 0.05% trifluoroacetic acid).

Example 28 - Compound 28: 5-(3,3-Dimethyl -but-1-vnyl)-3-(f4-methyl- cyclohexanecarbony ι j ι )-[4-(4-morphol ι in : 4-yJ ι m

thiophene-2-carboxylic acid

The title compound was synthesized in a manner analogous to Example 27, using 4-{2-chloro-pyπdin-4-yimethy!)morpholιne (prepared from 2-chloro-pyridine-4- carbaldehyde and morpholine) in place of 2-ch!oro-4-pyrroiidin-1-ylmethyl-pyridtne: MS (m/z): 622,0 [M+H]+; HPLC retention time 3.59 min (2-98% acetonitriie: water with 0.05% tπ ' fluoroacetic acid). Example 29 - Compound 29. 3-r[-4-(4-dimethylamιnomehtvi-pyndιn-2-yloxy)- cycJohexyπ-(4-methyl-cyclohexanecarbonyl)-aminol-5-(3,3-Dι methyl-but-1-ynyl)- thιophene-2-carboxylic acid

The title compound was synthesized in a manner analogous to Example 27, using (2-chioro-pyrιdιn-4-ylmethyl)dimethyl-amine (prepared from 2-chioro-ρyrιdine-4- carbaidehyde and dimethyi amine) in place of 2-chloro-4-pyrrolιdin-1-ylmethyl- pyridine: MS (m/z):580.1 [M+H]+; HPLC retention time 3.57 mm (2-98% acetonitriie: water with 0 05% trifiuoro acetic acid).

Example 30 - Compound 30. 5-{3,3-Dιmethγl-but-1-vnvJ)-3-f(4-rnethyl- cyclohexanecarbonylH4-(pyridin-2-yloxy)-phenyn-aminoHhιophe ne-2-carboxylic acid Scheme 5

Compound 30

A mixture of 3-Amiπo-5-(3,3-dimethyl-but-1 "ynyl)-thiophene-2-carboxylic acid methyl ester (0.200 g, 0.843 mmol), palladium acetate (0.019 g, 0.084 mmol), xantphos (0.058 g, 0.101 mmol), cesium carbonate (0.767 g, 2.36 mmol} and 2-(4- Bromo-phenoxy)-pyridine (0.252 g, 1.01 mmol) in toluene (3 ml) was heated by microwave to 110 0 C for 45 minutes. The reaction was diluted with ethyl acetate filtered through a Ceiite pad and purified by silica gel chromatography to give 5-(3,3- Dimethyl-but-1 -ynyl)-3-[4-(pyridin-2-yloxy)-phenylamino]-thiophene-2-carbo xy!ic acid methyl ester in 66% yield.

To a cooled (0 0 C) THF (5 mL) solution of 5-(3,3-Dimethyi-but-1-ynyl)-3-[4- (pyridin-2-yloxy)-phenylamino]-thiophene~2-carboxyiic acid methyl ester (0.225 g : 0.55 mmol) was first added KHMDS (0.66 mmol, 0.5 M in toluene), followed by neat fr-ans-4-methyi-cyclohexanecarbonyl chloride (0.132 g, 0.825 mmol). The reaction was warmed slowly to room temperature and quenched with saturated ammonium chloride solution. The reaction mixture was diluted with EtOAc, washed with water, brine, dried over sodium sulfate, filtrated and concentrated. The crude material was purified by silica gel column chromatography to give 5-(3,3-Dimethyl-but-1-ynyi)-3- {(4-methyt-cycSohexanecarbonyI)-[4-(pyrtdin-2-yioxy)-phenyl] -amino}4hiophene-2- carboxylic acid methyl ester in 60% yield.

S-fS^-Dimethyl-but-i-ynyO-S-^^methyl-cyclohexanecarbonyiJ-^p yridin^- yloxy)-phenyi]-amino}-thioρhene-2-carboxylic acid methyl ester (174 mg, 0.33 mmol) in a 3:2:1 mixture of THF:MeOH:water (5 ml_) was treated with Sithium hydroxide monohydrate (0.69 g, 1.65 mmol) and heated to 60 0 C for 1 hour. The organic volatiies were evaporated under reduced pressure and the crude material was purified by reverse-phase HPLC to afford the title compound in 70% yield. MS (m/z): 517.0 [M+H] " ; HPLC retention time: 4.90 min (2-98% acetonitrile:water with 0.05% trifluoroacetic acid).

The hpic/mass spectrometer equipment used to analyze Example 31-77, unless otherwise indicated, were: LC: Thermo Electron Surveyor HPLC

MS: Finnigan LCQ Advantage MAX Mass Spectrometer

Column: Phenomenex Polar RP 30 mm X 4.6 mm

Solvents: Acetonitrϋe with 0.1% formic acid, Water with 0.1% formic acid. Method A

Scheme 6

^[[S^S^-dimethyi-but-i -ynyO^-methoxycarbonyl-thiophen-S-ylH^trans- methy!-cyclohexanecarbonyi)-amino]-piperidine-1-carboxylic acid tert-butyl ester (1.5g, 2.73mmo!) was dissolved in ACN (1OmL). To the solution was added a solution of lithium hydroxide (253mg, 11.01mmoi) in water (1 OmL). The reaction was stirred at room temperature for 6 hours. The reaction was complete as determined by LC/MS. The pH was adjusted to 5 with 1N HCI in water. The product was extracted with ethyl acetate (3x1 OmL). The combined organics were dried with sodium sulfate, filtered and were concentrated under reduced pressure and 4~[[2- carboxy-5-(3,3-dimethyl-but-1-ynyl)-thiophen-3-yl]-(4-trans- methyl- cyc(ohexanecarbonyl)-amino]-piperidine-1-carboxylic acid tert-butyl ester (1.35g, 93%) was recovered as a white solid.

4-[[2-carboxy-5-(3,3-dimethyl-but-1-ynyi)-thiophen-3-y!]-(4- trans-methyl- cyclohexanecarbony!)-amino]-piperidine-1-carboxylic acid tert-butyl ester (1.35g, 2.53mmo!) was dissolved in 4N HCI in dioxane (6mL, 24mmol). The reaction was stirred at room temperature for 0.5 hours and found to be complete as determined by LC/MS, The reaction was concentrated under reduced pressure to give the HCI salt of S-fS.S-dimethyl-but-i-ynyO-S-^-trans-methyl-cyclohexanecarbo nyO-piperidin^-yl- amino]-thiophene-2-carboxylic acid. Method B

δ-fS.S-Dimethyl-but-i-vnviVS-ffΦtrans-methyl-cvclohexan ecarbonvD-pyrrolidiπ-S^SV vi-aminoHhiophene-2-carboxylic acid was synthesized as foSlows: a) Synthesis of S-O.S-dimethyi-but-i-vnyD-S-iodo-thiophene^-carboxySic acid methyi ester

Scheme 8

oxalyl chloride, MeOH

A mixture of 5-Bromo-thiopheπe-2-carboxylic acid ethyl ester (7g, 30 mmot), copper iodide (1.2g, 6 mmol), triethylamine (20 mL) in DMF (100 mL) was degassed in a 35OmL pressure bottle. Then tris{dibenzyiideneacetone)dipalladium(0) (2.1 g, 3 mmoi) and 3,3-dimethyl-but-1-yne (18.3 mL, 150 mmoi) were added and heated at 80 degree for 3 hours. The reaction mixture was filtered on ceiite and washed with ethyl acetate. The solution was diluted with water and extracted twice with ethyl acetate. The organic phases were combined and washed with water. After drying and concentration, the crude residue was purified by flash chromatography to yield 6.9g (95%) of 5-(3,3-Dimethy!-but-1-ynyl)-thiophene-2-carboxylic acid ethyl ester as a yeilow oil.

A solution of 5-(3,3-dimethyl-but-1-ynyl)-thiophene-2-carboxylic acid ethyl ester (6.9g) in THF (100 mL) was added LiOH (1.5N, 10OmL). The mixture was stirred at room temperature for 4 hours. Acidified reaction with HCi to pH =2, then remove voiatiies under vacuo. The resulting beige color solid was collected by filtration, washed with water then dried overnight to give 6.2 g of product which was used without further puriftcation.

To a solution of 5-{3 s 3-Dimethyl-but-1-ynyi)-thioρhene-2-carboxyiic acid (6.2 g, 30 mmol; see patent application US5861421 ) in THF (100 mL) was added a solution of nBuU (2.0 M in pentane, 33 mL, 66 mmol) via an addition funnel at -78 0 C. After addition, the reaction was stirred at -78 0 C for 1 h. A solution of I 2 (7.7 g, 30 mmol) in THF (100 mL) was added siowly (ca. 15 min) to the flask. After a further 10 mins, the reaction was quenched with 1 N HCI (50 mL) and warmed to room temperature. The voiatiies were removed in vacuo and the residue was dissolved in ether (500 mL).

The organic solution was washed with 1 M Na 2 S 2 O 3 (100 mL x 2), brine (100 mL) and dried over Na 2 SO 4 . After concentrated in vacuo, the residue was purified by silica gel chromatography (EtOAc/hexanes) to give 5-(3,3-Dimethyl-but-1-ynyl)-3-iodo- thiophene-2-carboxylic acid (5.9 g, 65%) as a white solid.

To a soiution of 5-(3,3-dimethyl-but-1-ynyl)-3-iodo-thiophene-2-carboχylic acid

(1.0 g, 3.0 mmol) and DMF (20 μL) in dry dichloromethane (10 mL) was added oxalyl chloride (508 μL, 6.0 mmol) at room temperature. After stirring at room temperature for 90 min, the reaction was concentrated in vacuo to remove volatites. The residue was dissolved in pyridine (5 mL) and methanol (5 mL) and stirred for 2 h. The voiatiies were removed in vacuo and the residue was participated between ether

(150 mL) and saturated NH 4 C! solution (50 mL). The organic layer was washed with saturated NH 4 C! soiution (50 mL) and dried over Na 2 SO 4 . After concentration in vacuo, the residue was purified by silica gel chromatography (EtOAc/hexanes) to give the desired product (835 mg, 80%). b) Synthesis of δ-O.S-dimethvi-but-i -vnvπ-S-ffΦmethyl-cvclohexanecarbonyl)- pyrrolidin-3$-vi-amino " |-thioρhene-2-carboxylic acid methyl ester

Scheme 9

A mixture of of 5-(3,3~dιmethyl-but-1-yπyl)-3-ιodo-thιophene-2-carboxyl c acid methyl ester_(0 5 g, 1 5 mmol), palladium acetate (0.015 g, 0 32 mmoi), BlNAP (0 009 g, 0 15 mmol), cesium carbonate {1 2 g, 4 5 mmol) and (3S)-Amtno- pyrroiιdιne-1-carboxyiιc acid tert-butyl ester (0.252 g, 1 01 mmol) in toluene (8 mL) was heated to 110 0 C for 8 h The reaction was diluted with ethyi acetate filtered through a Celite pad and purified by silica gel chromatography to give 3-[5-(3,3- dιmethyl-but-1-yny!)-2-methoxycarbonyl-thιophen-3S-ylammo] -pyrrolιdιne-1- carboxylic acid tert-butyl ester in 70% yield.

To a cooled (0 0 C) THF (3 mL) solution of 3-[5-(3,3-dιmethyl-but-1-ynyl)-2- methoxycarbonyl-thiophen-SS-ylaminoJ-pyrrolidine-i-carboxyli c acid tert-buty! ester was first added KHMDS (1 0 mmol, 0 5 M in toluene), followed by neat trans-4- methyi-cyciohexanecarbonyl chloride (0 2 mL, 1 24 mmol) The reaction was warmed slowly to room temperature and quenched with saturated ammonium chloride solution The reaction mixture was diluted with EtOAc, washed with water, brine, dried over sodium sulfate, filtrated and concentrated. The crude was then diluted with EtOAc (10 mL), treated with 4M HCI in dioxane (0 5 mL) and heated to 50 0 C for 30 mm The reaction mixture was cooled to room temperature

concentrated and purified by silica gel column chromatography to give the title compound in 70% yield

δ-tS.S-dimethyl-but-i-ynylJ-S-K^methyl-cyclohexanecarbonylJ -pyrrolidin-SS- yl-amιno]-thιophene-2-carboxylic acid methyl ester (0 3 g, 0.7 mmol) in a 3 2 1 mixture of THF.MeOH water (5 mL) was treated with lithium hydroxide monohydrate (0 69 g, 1 65 mmol) and heated to 60 0 C for 1 hour The organic volatiies were evaporated under reduced pressure and the crude material was purified by reverse- phase HPLC to afford 5-{3,3-dimethy[-but-1-yny])-3-[(4-trans-methy!- cyclohexanecarbonyl)-pyrrolidin-3-(S)-yl-amino|-thiophene-2- carboxyiic acid in 60% yield. Method C.

Example 58 - Compound 58: 5-(3 : 3-Dimethyi-but-1-vnyl)-3-(ftrans-4-methyl- cyclohexanecarbonviHI-fSVmethyl-S-pyridin-S-yloxyVpropyii-am inoHhiophene^- carboxylic acid

Compound 58

L-Alaninemethyl ester (10.Og, 71.6mmol) was taken up in CH 2 Ci 2 (60OmL). The solution was cooled to 0 0 C with an ice bath and benzylchloroformate (1 1.09mL, 78.8mmol) was added followed by a slow addition of diisopropylethyl amine (28.2mL, 157.5mmol). The reaction was allowed to warm to rt and stirred. The reaction was monitored and determined to be complete at 1h by TLC. The reaction was quenched with VT. sat. NaHCO 3(aq) . The layers were separated and the aqueous was washed with CH 2 CI 2 (SOOmL). The organics were combined and dried over Na 2 SO 4 . The solids were filtered and solvent removed under reduced pressure. S-2- benzyloxycarbonylamino-propionicacid methyl ester (14.22g, 84%) was isolated by siiica gei chromatography as a colorless oil.

To an Argon purged flask was added S-2-benzyloxycarbony!amino- propionicacid methyl ester (14.1Og, 59.4mmol) and CH 2 CI 2 (50OmL). The solution was cooled to -78 0 C internal using dry ice and methanol. DIBAL-H 1.0M in Toluene (119mL, 119mmol) was added over a period of 1h ensuring the internal temperature does not exceed -68 °C. The solution was allowed to stir at -78 0 C for a period of 1.5h. The reaction was quenched with a slow addition of CH 3 OH (2OmL) followed by the addition of sat NH 4 Ci (aq) (15OmL). The solution was then allowed to warm to rt. The solids were filtered through Celite. The layers were separated and the aqueous was washed with CH 2 CI 2 (50OmL). The organics were combined and washed with brine and dried over Na 2 SO 4 . Solids were removed by filtration and solvent removed under reduced pressure. (1-(S)-methy!-2-oxo-ethyl)-carbamic acid benzy! ester (9.78g, 79%) was isolated by silica gel chromatography as a colorless oil,

Methyltriphenylphosphonium bromide (16.92g, 47.30mmol) was taken up in toluene (35OmL). The heterogeneous solution was cooled to 0 0 C then KHMDS 0.5M in toluene (9OmL, 45mmol) was added. The reaction was stirred at 0 0 C for 30 min, then cooled to -78 °C and (1-(S)-methyl-2-oxo-ethyl)-carbamic acid benzyl ester (6.11g, 29.56mmol) that was dissolved in toluene (45mL) was added. The solution was warm to rt, stirred at rt for 30 min, then quenched with sat NH 4 CI (aq} (15OmL). The layers were separated and the aqueous was washed with EtOAc. The organics were combined and dried over Na 2 SO 4 . Solids were removed by filtration and solvent removed under reduced pressure. (1-(S)-methyl-allyl)-carbamic acid benzyl ester (4.9Og, 81%) was isolated by silica gel chromatography as a colorless oil.

(1-(S)-Methy!-allyl)-carbamic acid benzyl ester (1.96g, 9.56mmol) was taken up in THF (20OmL). 9-BBN 0.5M in THF (38mL, 19.0mmol) was added slowly to the reaction mixture. The solution was allowed to stir at rt for 16h. Quenched with H 2 O (65mL), followed by 1.0M NaOH (aq) (2OmL). The solution was cooled to 0 0 C in an ice bath and H 2 O 2 30% in H 2 O (7.OmL) was slowly added. The reaction was partitioned between Et 2 O and H 2 O. The layers were separated and the aqueous washed with Et 2 O. The combine organics were dried over Na 2 SO 4 . Solids were filtered and the solvent removed under reduced pressure. (3-hydroxy-1-(S)-methyi- propyi)-carbamic acid benzyl ester (1.85g, 87%) was isolated by silica ge! chromatography as a white solid.

(3-Hydroxy-1-(S)-methyl-propyl)-carbamic acid benzyl ester (8.02g,

35.9mmol) was taken up in DMF (75mL). terf-Butyl-dimethyisily! chloride (8.12g, 53.9mmol) was added to the solution followed by imidazole (4.15g, 61.0mmol). The reaction was stirred at rt and monitored by TLC. The reaction was determined to be complete at 1h by TLC. The reaction was taken up in EtOAc (20OmL) and partitioned between 5% LiCI (aq ) (20OmL). The aqueous was back extracted with EtOAc. The organics were combined and washed with 3 X 20OmL 5% LiCi (aq) , and dried over

Na 2 SO 4 . Soiids were filtered and solvent removed under reduced pressure. [3-(tert- Butyl-silanoxy)-1-(S)-methyl-propyi]-carbamic acid benzyl ester (12.73g, 100%) was isolated by silica gel chromatography as a white solid.

3-(fert-Butyl-siSanoxy)-1-(S)-methyl-propyl]-carbamic acid benzyl ester (12.37g, 36.7mmoi) was taken up in EtOH (20OmL). The solution was purged with argon for 15min, then 10% Pd/C (1.24g) was added. The vessel was evacuated and backfilled with H 2(g) three times. Stir under a H 2(g) atmosphere monitoring by TLC. The reaction was determined to be complete after 2h. The reaction was put through a PTFE filter and solids washed with CH 2 CI 2 . Solvent was removed under reduced pressure and the colorless oil 3-(terf-butyl-dimethyJ-silanoxy)-1-(S)- methylpropylamine (8.34g, 100%) was used without any purification.

An argon purged flask was charged with 3-(ferf-butyl-dimethyl-silanoxy)-1-(S)- methylpropylamine (3.5Og, 17.24mmmol), 5-(3,3-dimethyl-but-1ynyl)-3-iodo- thiophene-2-carboxylic acid methyl ester (3.Og, 8.62mmol), Pd(OAc) 2 (288mg, 1.29mmol), and ±-BINAP 801 mg, 1.29mmol). Argon degassed toluene (9OmL) was add to the reaction mixture and the reaction was placed in a preheated 120 0 C oil bath and stirred for 16h. The solvent was removed under reduced pressure and taken up in hexanes. 3-[3-(te/t-butly-dimethy!-silanoxy)-1-(S)-methyl-propylamino ]-5- (3,3-dimethyl-but-1-ynyϊ)thiophene-2-carboxylic acid methyl ester was isolated by silica gel chromatography (2.87g, 79%) as a yellow solid.

3-[3-(tert-Butyl-dimethyl-silanoxy)-1-(S)-methyl-propylamino ]-5-(3,3-dimethyl- but-1-yny!)thiophene-2-carboxylic acid methyl ester (8.2g, 18.24mmol) was taken up in THF (20OmL). TBAF 1 .0M in THF (88.15mL, 88.15mmol) was added in 20 mL increments at rt. The reaction was monitored by LC/MS. The reaction was determined to be complete at 30 mm and was quenched with sat NH 4 C!^ ) (20OmL) After stirring at rt for 1h the organicε were removed under reduced pressure and treated with EtOAc The solution was dried over Na 2 SO 4 , solids were filtered and the solvent removed under reduced pressure 5-(3,3-dιmethyl-but-1-ynyi)3-(3-hydroxy-1- (S)-methyl~propylamino)-thιophene-2-carboxy!ιc acid methyl ester (4 Og, 73%) was isolated by silica gel chromatography as a yellow solid

5-(3,3-Dιmethyl-but-1-ynyi)3-(3-hydroxy-1-(S)~methyl-propyl amιno)- thιophene-2-carboxyiιc acid methyi ester (4 Og, 12 94mmo!) was taken up in pyridine (10OmL) Trans^-methyi-cyclohexanecarbonyl chloride (12 4Og, 77 66mmol) was added and the reaction was placed in a 100 0 C preheated oil bath and stirred for 16h After cooling to rt the solvent was removed under reduced pressure The residue was partitioned between EtOAc and 2N HCl (aq) , the organics dried over Na 2 SO 4 , and the solids filtered Solvent was removed under reduced pressure and 5-(3,3- dιmethyi-but-1-ynyi)-3~[(trans-4-methyicyclohexanecarbonyl) - [1-(S)-methyl-3-(trans- 4-methyi-cyc!ohexanecarbonyloxy)-propyl]-amino]-thιophene-2 - carboxyhc acid methyl ester (4 15g, 58%) was isolated by silica gel chromatography as a yellow solid A mixture of THF (2OmL) and CH 3 OH (1 OmL) and 5-(3,3-dιmethyi-but-1-ynyJ)- 3-[(trans-4-methylcyclohexanecarbony!)- [1-(S)-methyl-3-(trans-4-methyl- cyclohexanecarbonyloxy)-propyl]-amino]-thιophene-2- carboxyhc acid methyl ester (4 15g, 7 45mmoi) was treated with dissolved LiOH H 2 O (1 56g, 37 20mmol) in H 2 O (1OmL) After stirring at rt for 3h , the reaction was determined to be complete by LC/MS After adjusting pH - 2 with 2N HCI( aQ ) the organics were removed under reduced pressure and the residue partitioned with EtOAc The organics were separated, dried and the solvent removed under reduced pressure 5-(3,3-Dιmethyi- but-1 -ynyi)-3-[(3-hydroxy-1 -(S)-methyl-propyl)-(trans-4-methyl-cyclohexanecarbonyl)- amιno]-thιophene-2-carboxylιc acid TFA salt (3 96g, 95%) was isolated by reverse phase HPLC as a white solid

A mixture of 5-(3,3-dιmethyl-but-1-ynyl)-3-[(3-hydroxy-1-(S)-methyl-prop yl)- (trans-4-methyl-cyclohexanecarbonyl)-amino3-thiophene-2-carb oxyhc acid TFA salt (0 10Og, 0 238mmol) and 3-fluoropyrιdsne (0 046g, 0 476mmo!) in DMSO (3 OmL) was treated with KOtBu (0 106g, 0 952mmo!) in one portion The reaction was determined to be complete by LC/MS in 30 mm After quenching the reaction with 20% AcOH in H 2 O (2mL), 5-(3,3-d)methyl-but-1-ynyl)-3-[(trans-4-methyl- cyclohexanecarbonylJ-EI-tSJ-methyl-S-pyπdin-S-yloxyJ-propyl l-aminoJ-thiophene^- carboxyiic acid TFA salt (0.029g, 26%) was isolated by reverse phase HPLC as an off-white solid.

LC/MS = 496.84 (M + +1 )

Retention time: 4.14 min

Gradient: 0 min-0.2min 5% ACN, 0.2 min~~3.95 min 5%-100% ACN, 3.95 min-5.20 min 100% ACN, 5,20 min-5.5 min 100%-5% ACN, 5.5 min-6 min 5% ACN.

Example 59 - Compound 59: 5-(3,3-dimethyi-but-1-vnyl)-3-r(trans-4-methyl- cvclohexanecarbonyD-fi-fSVmethyi-S-pyrimidin^-yloxyVpropyll- aminoHhjophene^" carboxyiic acid

Compound 59

A mixture of 5-(3,3-dimethyl-but-1-ynyl)-3-[(3-hydroxy-1-(S)-methyi-propy I)- (trans-4-methyl-cyclohexanecarbonyl)-amino]-thiophene-2-carb oxylic acid TFA salt (0.100g, 0.238mmol) and 2-chioropyrimidine (0.029g, 0.250mmoi) in DMSO (3.OmL) was treated with KOtBu (0.087g, 0.776mmol) in one portion. The reaction was determined to be complete by LC/MS in 30 min. After quenching the reaction with 20% AcOH in H 2 O (2mL), 5-(3,3-dimethyl-but-1-ynyl)-3-[(trans-4-methyl- cycSohexanecarbonyO-EI-fSJ-methyl-S-pyrimidin^-yloxyJ-propyl J-aminoj-thiophene^- carboxylic acid TFA salt (0.015g, 13%) was isolated by reverse phase HPLC as an off-white solid.

LC/MS = 520.18 (M + +Na)

Retention time: 3.79 min

Gradient: 0 min-0.2min 5% ACN, 0.2 min-3.95 min 5%-100% ACN, 3.95 min-5.20 min 100% ACN, 5.20 min-5.5 min 100%-5% ACN, 5.5 min-6 min 5% ACN.

Example 60 - Compound 60: 5-(3,3-dimethyl-but-1-ynvO-3-F(trans-4-methyi- cyclohexanecarbonylHI-fSl-methyl-S-pyridin^-vioxyVpropyll-am inoHhiophene^- carboxyiic acid

A mixture of 5-(3,3-dimethyl-but-1-ynyl)-3-[(3-hydroxy-1 (S)-methyi-propyl)- (trans-4-methyl-cyclohexanecarbonyl)-amino]-thiopheπe-2-car boxylic acid TFA salt (0.182g, 0.340mmo!) and 2-fluoropyridine (164μL, 1.70mmol) in DMSO (5.OmL) was treated with KOtBu (0.152g, 1.36mmol) in one portion. The reaction was determined to be complete by LC/MS in 30 min. After quenching the reaction with 20% AcOH in H 2 O (2ml_), 5-(3,3-dimethyl-but-1 -ynylJ-S-Ktrans^-methyl-cyclohexanecarbonylHI - (S)-methy!-3-pyridin-2-yloxy)-propyi]-amino]-thiophene-2-car boxyiic acid TFA salt (0.195g, 94%) was isolated by reverse phase HPLC as an off-white solid.

LC/MS = 496.95 (M + +1 )

Retention time: 2.33 min

Gradient: 0 min-0.1min 5% ACN, 0.1 min-1.95 min 5%-100% ACN, 1.95 min-3.50 min 100% ACN, 3.50 min-3.55 min 100%-5% ACN, 3.55 min-4.0 min 5% ACN.

Example 61 - Compound 61: 3-rf3-(4-dimethylaminomethy[-pyridin-2-yloxy)-1-fS)- methyl-propylHtrans^-methyf-cyciohexanecarbonyD-aminol-S-fS^ -dimethyl-but-i - ynyl)-thioρhene-2-carboxyiic acid

Compound 61

A mixture of 5-(3,3-dimethyl-but-1-ynyl)-3-[(3-hydroxy-1-(S)-methyl-propy l)- (trans-4-methyl-cyclohexanecarbonyl)-amino]-thiophene-2-carb oxylic acid TFA salt (0.21 δg, 0.598mmol) and (2-fiuoropyridin-4-yl-methyl)-dimethyiamine (0.40Og, 2.59mmol) in DMSO (5.OmL) was treated with KOtBu (0.267g, 2.39mmol) in one portion. The reaction was determined to be complete by LC/MS in 30 min. After quenching the reaction with 20% AcOH in H 2 O (5m L), 3-[[3-{4-dimethylaminomethyl- pyridin^-yioxyJ-I ^SJ-methyl-propylHtrans-^methyl-cyciohexanecarbonylJ-aminoj- δ- (3,3-dimethyl-but-1 -ynyl)-thiophene-2-carboxylic acid TFA salt (0.272g, 68%) was isolated by reverse phase HPLC as an off-white solid.

LC/MS = 553.94 (M + +1)

Retention time: 3.30 min

Gradient: 0 min-0.2min 5% ACN, 0.2 min-3.95 min 5%-100% ACN, 3.95 min-5.20 min 100% ACN, 5.20 min-5.5 min 100%-5% ACN, 5.5 min-6 min 5% ACN.

Example 62 - Compound 62: 5-(3,3-dimethyl-but-1-vnyl)-3-rftrans-4-methyl- cvclohexanecarbonyl)-[1-(S)-methyl-3-f4-pyrroiidin-1-vi-meth yl-pyridin-2-yloxy)- propyn-amino]-thiophene-2-carboxyNc acid

A mixture of 5-(3,3-dimethyi-but-1-ynyi)-3-[(3-hydroxy-1-(S)-methyi-propy l)- (trans-4-methyl-cyc!ohexanecarbonyi)-amino]-thiophene-2-carb oxy!ic acid TFA salt (0.05Og, 0.1 19mmol) and 2-fluoro-4-pyrrolidin-1-yl-methyl-pyridine (0.10Og,

0.555mmol) in DMSO (1.5mL) was treated with KOtBu (0.053g, 0.476mmol) in one portion. The reaction was determined to be complete by LC/MS in 30 min. After quenching the reaction with 20% AcOH in H 2 O (1 mL), 5-(3,3-dimethyl-but~1-ynyl)-3- [(trans-4-methyl-cyclohexanecarbonyl)-[1-(S)-methyl-3-(4-pyr rolidin-1 -yl-methyl- pyridin-2-yioxy)-propyl]-amino]-thiophene-2-carboxylic acid TFA salt (0.03Og, 37%) was isolated by reverse phase HPLC as an off-white solid.

LC/MS = 580.00 (M + +1 )

Retention time: 3.25 min

Gradient: 0 min-0.2min 5% ACN, 0.2 min-3.95 min 5%-100% ACN, 3.95 min-5.20 min 100% ACN 1 5.20 min-5.5 min 100%-5% ACN, 5.5 min-6 min 5% ACN. Example 63 - Compound 63: 5-(3,3-dimethy[-but-1-vnyl)-3-ff3-(6-hydro^

v!oxy)-1-(S)-methyl-propylHtrans-4-methy^

2-carboxyiJc acid

Compound 63

A mixture of 5-(3,3-dimethy[-but-1-ynyl}-3-[(3-hydroxy"1~(S}-methyi-propy l)- (trans-4-methyl-cyclohexanecarboπyl)-amSno]-thiophene-2-car boxylic acid TFA salt (0.10Og, 0.238mmol) and 2,6-dichloropyridazine (0.176g, 1.19mmol) in DMSO (3mL) was treated with KOtBu (0.106g, 0.952mmol) in one portion. The reaction was determined to be complete by LC/MS in 30 min. After quenching the reaction with 20% AcOH in H 2 O (5mL), 3-[[3-(6-chloro-pyridazin-3-yloxy)-1-(S)-methyi-propy!]- (trans^-methyi-cyclohexanecarbonyl^aminol-S-fS^-dimethyl-but -i-ynylJ-thiophene- 2-carboxyiic acid TFA sait (0.056g, 36%) was isolated by reverse phase HPLC as an off-white soϋd.

A mixture of 3-[[3-(6-chloro-pyrsdazin-3-yloxy)-1 (S)-methyl-propyl]-(trans-4- methyl-cyciohexanecarbonyO-aminoJ-δ-fS^-dimethyl-but-i-yny J-thiophene^- carboxylfc acid TFA salt (0.56g, 0.087mmo!) in AcOH (2.OmL) and NaOAc (0.37g, 0.434mmoi) was placed in a preheated 100 0 C oil bath and stirred for 3h. The reaction was determined to be complete by LC/MS. After cooling, the reaction was diluted with CH 3 OH (5mL). 5-(3,3-Dimethyl-but-1-ynyl)-3-[[3-(6-hydroxy-pyridazin-3- yioxyJ-i-tSJ-methyl-propylJ^trans^-methyl-cyclohexanecarbony O-aminoJ-thiophene- 2-carboxylic acid TFA salt (0.07g, 13%) was isolated by reverse phase HPLC as an off-white solid,

LC/MS = 536.12 (M + +Na)

Retention time: 3.50 min

Gradient: 0 min-0.2min 5% ACN, 0.2 min-3.95 min 5%-100% ACN, 3.95 min-5.20 min 100% ACN, 5.20 min-5.5 min 100%-5% ACN, 5.5 min-6 min 5% ACN. Example 84- Compound 84: 5-(3, 3-dimethylbut-1 -vnyl)-3-(4-methyl-N-

(4-(4-methyJ-4H-1 ,2,4-triazol-3- ylthio)cvclohexyncyclohexanecarboxamido)thiophene-2-carboxyl ic acid

Compound 84

3-[(4-Bromo-cyciohexy!)-(4-methyl-cyciohexanecarbony!)-am ino]-5-(3,3- dimethyl-but-1-ynyl)-thiophene-2-carboxyiic acid methyl ester (75 mg, 0.143 mmol, 1.0 eq.) was weighed into a round-bottomed flask containing 1 ,4-dioxane (2 ml_) and purged with N 2 . 4-Methyl-4H-[1 ,2,4]triazo!e-3-thiol (83 mg, 0.718 mmol, 5.0 eq.) and DBU (107 μl_, 0.718 mmol, 5 eq.) were then added to the solution. The reaction was stirred at 80 0 C until the complete consumption of bromide. The solution was then cooied to room temperature and 50% KOH (1.0 ml_) was added to the fSask. The reaction was heated at 40 0 C for 2 h. The reaction was cooled to 0 0 C and acidified with 2 N HCI. After removing volatiles, the residue was purified by reverse phase preparative HPLC to afford Example 1 (5-(3,3-Dimethyl-buM-yny!)-3-{(4-methyt- cyclohexanecarbonyl)-[4-{4-methyl-4H-[1 ,2,4]triazol-3-yIsulfanyl)-cyclohexyl]~amino}- thiophene-2-carboxylic acid, 32 mg, 41%).

MS (m/z) 543.2 [M+H] + HPLC retention time: 4.40 min (5-95% acetonitrile with 0.05% TFA: water with 0.05% TFA).

Example 85 - Compou nd 85: 5-(3 , 3-d imethylbut-1 -vnvO-3-(4-methyl-N- (4-(4-methyl-4H-1 .2.4-triazol-3- vlthio)cvclohexvl )cycJohexanecarboxamido )thiophene-2-carboxylic acid

5-(3,3-Dimethyi-but-1-yny!)-3-[(4-hydroxy-cydohexyl)-(4-m ethy!- cyclohexanecarboπyl)-amino]-thiophene-2-carboxyiic acid methy! ester (92 mg, 0.2 mmol, 1.0 eq.), 4-methyl-4H-[1 ,2,4]tπazo(e-3-thiol (46 mg, 0.4 rmmo!, 2.0 eq.), triphenylphosphine (105 mg, 0.4 mmoi, 2.0 eq.), DiAD (77 μl_, 0.4 mmol, 2.0 eq.) and triethyl amine (42 μL, 0.3 mmol, 1.5 eq.) were weighed into a round-bottomed flask containing THF (2.0 ml_). The solution was purged with N 2 and stirred at room temperature for 12 h. The solution was concentrated to an oil and then purified by siiica gel chromatography (0-100% EtOAc in hexanes) to afford 5-(3,3-Dimethyl-but- 1-ynyl)-3-{(4-methyl-cyclohexanecarbonyi)-[4-(4-methyl-4H-[1 ,2,4]triazol-3- ylsuifanyl)-cyclohexyl]-amino}-thiophene-2-carboxylic acid methyi ester (63 mg, 57%).

Compound 85

5-(3,3-DimethyS-but-1-ynyl)-3-{(4-methy!~cyciohexanecarbonyl )~[4-(4-methyl~ 4H-[1 ^^JtriazoS-S-yisulfanyO-cyclohexy^-aminoJ-thiophene^-carboxy lic acid methyl ester (63 mg, 0.113 eq.) was dissolved in THF (2.0 ml_) in a round-bottomed flask. A solution of LiOH {14 mg, 0.34 mmol, 3.0 eq.) in water (1.0 mL) was added. The reaction was stirred at room temperature overnight. The reaction was then neutralized with 0.2 mL 2 N HCI, concentrated, and purified by reverse phase preparative HPLC to afford Exampie 2 (5-(3,3-Dimethyl-but-1-ynyi)-3-{(4-methyi- cyclohexanecarbonylHΦf^methyl^H-Ii ^.^triazoi-S-yisuifanylJ-cyciohexyO-amino}- thiophene-2-carboxyIic acid, 27 mg, 44%).

MS (m/z) 543.2 [M+H] + HPLC retention time: 4.92 min (5-95% acetonitrile with 0.05% TFA: water with 0.05% TFA).

Examples 86-108 were synthesized by the same method as Examples 84 or 85 using the appropriate heterocyclic thiol as substrate.

Example 86 - Compound 86: 3-(N-(4-f1 ,3,4-Thiadiazol-2-yJthio)cyclphexyi)-4- methylcvciohexanecarboxamido)-5-(3,3-dimethvibut-1-vnyl)thio phene-2-carboxylic acid

Compound 86

MS (m/z) 546 [M+H] + HPLC retention lime: 3.3 min (50-98% acetonitri!e:water with 0.05% tifluoroacetic acid; 6 min run).

Example 87 - Compound 87: 5-(3,3-DimethyJbut-1-vnyl)-3-(4-methyl-N-(4-(pyridin-4- ylthio)cvclohexyi)cvclohexanecarpoxamido)thiophene-2-carboxy lic acid

Compound 87

MS (m/z) 539 [M+H] + HPLC retention time: 4.08 min (6min HPLC method 2- 98% acetonitrile:water with 0.05% tifluoroacetic acid).

Example 88 - Compound 88: 5-f3,3-Dimethylbut-1-vnvJ)-3-(4-methyl-N-4-fpyridin-2- ylthio-N-oxide)cvciohexyl)cvclohexanecarboxamido)thiophene-2 -carboxylic acid

Compound 88

MS (m/z) 555 [M+H] + HPLC retention time: 2.43 min (50-98%

acetonitriie:water with 0.05% tifiuoroacetic acid; 6 min run). Example 89 - Compound 89: 5-(3,3-Dimethγ!but-1-ynvQ-3-(N-(4-(1-ethγH H^m^ ^^ ^ ^ 2-v!thio)cvclohexyi)-4-methvicvclohexanecarboxamido)thiophen e-2-carboxyiic acid

Compound 89

MS (m/z) 556 [M+H] + HPLC retention time: 4.19 min (2-98% acetonitrile:water with 0.05% tifluoroacetic acid; 6 min run).

Example 90 - Compound 90: 3-(N-(4-(4~Cvclopropyl-4H-1 ,2.4-triazol-3- v]thio)cvciohexyl)-4-methylcvciohexanecarboxamido)-5-(3,3-di methylbut-1- ynyl)thiophene-2-carboxylic acid

Compound 90

MS (m/z) 569 [M+H] + HPLC retention time: 4.66 min (2-98% acetonitrile:water with 0.05% tifluoroacetic acid; 6 min run).

Example 91 - Compound 91 : 5-f3.3-Dimethylbut-1 -vnyl)-3-(4-methyl-N-f4-fαuinolin-2- ylthio)cvclohexyi)cyclohexanecarboxamido)thiophene-2-carboxy iic acid

Compound 91

MS (m/z) 589 [M+H] + HPLC retention time: 4.37 min (50-98%

acetonitri!e:water with 0.05% tifluoroacetic acid; 6 mSn run).

Example 92 - Compound 92: 3-fN-(4-(π ,2,41-Triazolor4,3-a1pyπdin-3- ylthio)cvclohexyi)-4-methylcvclohexanecarboxamtdo)-5-(3,3-di methylbut-1- ynyl)thiophene-2-carboxylic acid

Compound 92

MS (m/z) 579 [M+H] + HPLC retention time: 2.44 min (50-98%

acetonitriie:water with 0.05% tifluoroacetic acid; 6 min run).

Example 93 - Compound 93: 5-f3.3-Dimethylbut-1-vnyl)-3-f4-methy[-N-(4-fthiophen- 2-yithio)cvclohexyi)cycloheχanecarboxamido)thiophene-2-carb oxylic acid

Compound 93

MS (m/z) 544 [M+H] + HPLC retention time: 4.36 min (50-98%

acetonitrile:water with 0.05% tifSuoroacetic acid; 6 min run).

Example 94 - Compound 94: 3-(N-(4-(1 H-)midazol-2-ylthio)cvciohexyl)-4- methvicvciohexanecarboxamido)-5-(3,3-dimethvibut-1-vnyi)thio phene-2-carboxylic acid

Compound 94

MS (m/z) 528 [M+H] + HPLC retention time: 3.92 min (2-98% acetonitrile:water with 0.05% tifSuoroacetic acid; 6 min run).

Exampie 95 - Compound 95: 3-(N-(4-(5-Amino-2H-1.2,4-triazol-3-vithio)cvclohexyl)- 4-methvicvciohexanecarboxamido)-5-(3,3-dimethylbut-1-vnyl)th iophene-2-carboxyiic acid

Compound 95

MS (m/z) 544 [M+H] + HPLC retention time: 3.99 min (2-98% acetonitriie:water with 0.05% tifluoroacetic acid; 6 min run).

Example 96 - Compound 96: 3-fN-(4-f3H-1.2,3-Triazoi-4-ylthio)cvciohexyi)-4- methylcvclohexanecarboxamido)-5-(3,3-dJmethyibut-1-vnv0thiop hene-2-carfaoxylic acid

Compound 96

MS (m/z) 529 [M+H] + HPLC retention time: 4.85 min (2-98% acetonitrile:water with 0.05% tifluoroacetic acid; 6 min run).

Example 97 - Compound 97: 3-fN-(4-(1 H-1 ,2,4-Thazoi-3-ylthio)cvciohexyl)-4- methylcvciohexanecarboxamido)-5-(3 : 3-dimethy[but-1-vnyl)thiophene-2-carboxylic acid

Compound 97

MS (m/z) 529 PvRH] + HPLC retention time: 4.59 min (2-98% acetonitriie:water with 0.05% tifluoroacetfc acid; 6 min run).

Example 98 - Compound 98: 5-(3,3-Dimethylbut-1 -vnvn-3-(4-methyl-N-(4-(5-methyl- 1 ,3,4-oxadiazo[-2-ylthio)cvclohexyl)cyclohexanecarboxamido)th iophene-2-carboxy(ic acid

Compound 98

MS (m/z) 540 [M+Hj + HPLC retention time: 5.21 min (2-98% acetonttriie:water with 0.05% tifiuoroacetic acid; 6 min run).

Example 99 - Compound, 99: δ^g^Dimethylbut-i -ynyl)-3-(4-methyl-N-(4-f pyridin-2- ylthio)cyciohexyi)cvciohexanecarboxamido)thiophene-2-carboxy lic acid

Compound 99

MS (m/z) 539 [M+H] + HPLC retention time: 5.23 min (2-98% acetonitrile:water with 0.05% tifiuoroacetic acid; 6 min run).

Exampie 100 - Compound 100: 5-(3,3-Dimethylbut-1 -vnyl)-3-(4-methy[-N-(4- fpyrimidin-2-ylthio)cvclohexyl)cvclohexanecarboxamido)thioph ene-2-carboxylic acid

Compound 100

MS (m/z) 540 [M+H| + HPLC retention time: 5.58 min (2-98% acetonitrile:water with 0.05% tifluoroacetic acid; 6 min run).

Example 101 - Compound 101 : 5-f3,3-Dimethvibut-1-vnyl>-3-(4-methv8-N-f4-f1- methvJ-1 H-tetrazol-5-ylthio)cvclohexy[)cyclohexanecarboxamido)thioph ene-2- carboxyiic acid

Compound 101

MS (m/z) 543 [M+Hf HPLC retention time: 4.42 min (2-98% acetonitrile:water with 0.05% tifluoroacetic acid; 6 min run).

Example 102 - Compound 102: 5-(3.3-Dimethylbut-1-vnvi)-3-f4-methv1-N-(4-f5- methv[-1 ,3,4-thJadiazoi-2-ylthio)cvclohexyl)cγc[ohexanecarboxamido) thiophene-2- carboxylic acid

Compound 102

MS (m/z) 560 [M+H] + HPLC retention time: 5.42 min (2-98% acetonitriie:water with 0.05% tifluoroacetic acid; 6 min run).

Example 103 - Compound 103: 5-f3.3-Dimethylbut-1-vnylV3-f4-methyl-N-(4-f5- methyl-1 ,3,4-thiadiazol-2-ylthio)cyclohexyl)cvctohexanecarboxamido)t hiophene-2- carboxyiic acid

Compound 103

MS (m/z) 560 [MH-H] + HPLC retention time: 5.33 min (2-98% acetonitriie:water with 0.05% tiftuoroacetic acid; 6 min run).

Exampie 104 - Compound 104: 5-(3,3-Dimethylbut-1-vnvO-3-(4-methyl-N-(4-f1- .Ω]M , byl:J.H , :^ razo l~5-ylthio)cyc[ohexγ[)cvciohexanecarboxarnido)thiophe ne-2- carboxyiic acid

Compound 104

MS (m/z} 544 [M+Hf HPLC retention time: 4.99 min (2-98% acetonitriie:water with 0.05% tifluoroacetic acid; 6 min run).

Exampie 105 - Compound 105: 3-(N-(4-f1,3,4-Thiadiazoi-2-ylthio)cvclohexyi)-4- methvJcvclohexanecarboxamido)-5-f3,3-dimethylbut-1-vnylHhiop hene-2-carboxylic acid

Compound 105

MS (m/z) 546.2 [M+H] + HPLC retention time: 2.61 min (2-98%

acetonitπ!e:water with 0.05% tifluoroacetic acid; 6 min run).

Example 106 - Compound 106: 5-f3,3-Dimethylfaut-1-vnyl)-3-(4-methyi-N-(4-(thiazol- 2-ylthio)cvclohexyi)cvciohexanecarboxamido)thiophene-2-carbo xyiic acid

Compound 106

MS (m/z) 545.2 [M+H] + HPLC retention time: 3.2 min (2-98%

acetonitrileiwater with 0.05% tifluoroacetic acid; 6 min run).

Exampie 107 - Compound 107: 5-f3,3-Dimethylbut-1-vnyl)-3-(4-methyi-N-f4-(thiazol- 2-ylthio)cvclohexyl)cyciohexanecarboxamido)thiophene-2-carbo xyiic acid

Compound 107

MS (m/z) 545.1 [M+H] + HPLC retention time: 3.15 min (2-98%

acetonitrileiwater with 0.05% tifluoroacetic acid; 6 min run).

Example 108 - Compound 108: 5-(3,3-Dimethvlbut-1-vnvlV3-(4-methvJ-N-f4- (pyrimidin-2-vSthio)cvclohexy[)cvclohexanecarboxamido)thioph ene-2-carboxy[ic acid

Compound 108

MS (m/z) 540.2 [M+H] + HPLC retention time: 2.89 min (2-98%

acetonitriie:water with 0.05% tifluoroacetic acid; 6 min run).

Examples 109-1 1 1 were synthesized from the corresponding thio compounds by oxidation with peracetic acid.

Example 109 - Compound 109: 5-(3,3-Dimβthylbut-1-vnyl)-3-f4-methyl-N-(4- (tetrahvdro-2H-pyran-4-ylsϋifonyl)cvclohexyl)cyclohexanecar boxamido)thiophene-2- carboxylic acid

Compound 109 MS (m/z) 558 [M+H] + HPLC retention time: 4.09 min (50-98%

acetonitrϊle:water with 0.05% tifluoroacetic acid; 6 min run).

Example 110 - Compound 110: 5-(3,3-Dimethylbut-1 -vnyl)-3-f4-methyl-N-(4-f1- methvi-1 H-tetrazol-5-visulfonyl)cvclohexyl)cvclohexanecarboxamido)th iophene-2- carboxylic acid

Compound 110 MS (m/z) 576 [M +H] * HPLC retention time: 4.95 min (2-98% acetonitrile:water with 0.05% tifluoroacetic acid; 6 min run).

Exampie 111 - Compound 111: 5-(3,3-Dimethvibut-1-vnyl)-3-ff1 r 1 4S>-4-methvl-N-(4- f(R)-1-methvJ-1 H-tetrazol-5-ylsuifiny[)cyclohexyl)cyclohexanecarboxamido)th iophene- 2-carboxylic acid

Compound 111

MS (m/z) 560 [M+H] + HPLC retention time: 4.73 min (2-98% acetonitriie:water with 0.05% tifluoroacetic acid; 6 min run).

Exampie 138 - Compound 138: 5-f3,3-dimethyl-but-1-vnvO-3-{(4-methy[- cyclohexanecarbonylH3-(pyridin-3-yloxy)-ethyl1-amino)-thioph ene-2-carboxyJic acid

Compound 138

A mixture of 3~Amino-5-(3,3-dimethy!-but-1-ynyt)-thiophene-2-carboxylic acid methyl ester (100 mg, 0.42 mmol), (pyridin-3-yloxy)-acetaIdehyde (96 mg, 0.7 mmol), AcOH (300 mg, 5 mmol) in DCE (2 mL) was treated with NaBH(OAc) 3 (179 mg, 0.9 mmoi) for 16 h. The reaction was quenched with water (10 mL) and extracted with EtOAc. The crude material was then dissolved in pyridine (5 mL) and treated with neat frans-4-methyl-cyclohexanecarbonyl chloride (0.132 g, 0.825 mmol). The reaction was heated for 16 h at 85 0 C and quenched with saturated ammonium chloride solution. The reaction mixture was diluted with EtOAc, washed with water, brine, dried over sodium suifate, filtered and concentrated. The crude material was then dissolved in a 3:2:1 mixture of THF:MeOH:water (5 ml_), treated with iithium hydroxide monohydrate (0.69 g, 1.65 mmol) and heated to 60 0 C for 1 hour. The organic volatiles were evaporated under reduced pressure and the crude material was purified by HPLC to afford the title compound. MS (m/z): 470.0 [M+H] " ; HPLC retention time: 3.36 min (2-98% acetonitri!e:water with 0.05% trifluoroacetic acid).

Example 139 - Compound 139: 5-(3,3-dimethyl-but-1-vnylV34(4-methyl- cvclohexanecarbonvl)-r3-(pvridin-2-yloxy)-ethyl]-aminoHhioph ene-2-carboxylic acid

Compound 139

The title compound was synthesized in a manner analogous to Example 138, using (pyridin-2-yloxy)-acetaldehyde in place of (pyridin-3-yloxy)-acetaldehyde: MS (m/z): 468.9 [M-H] + ; HPLC retention time: 4.47 min (2-98% acetonitriie:water with 0.05% trifluoroacetic acid) 30 min run.

Exampie 143 - Compound 143: 5-(3,3-dimethyl-but-1-vnyl)-3-|ftrans-4-methvi- cvclohexanecarbonvO-n-fSVmethvi-S-fpyrimidin-S-yloxyVpropyn- aminoHhiophene- 2-carboxylic acid

Compound 143 The 5-(3,3-dimethyl-but-1 -ynyi)-3-[(3-hydroxy-1 -(S)-methyf-propyl)-(trans-4- methyl-cyclohexanecarbonyl)-amino]-thiophene-2-carboxylic acid TFA salt (100mg, 0.239mmol) and 5-chloro-pyrimidine (137mg, 1.194mmol) were dissolved in DMSO (2mL). To this solution tBuOK {107mg, 0.956mmol) was added in one portion at rt. The reaction was allowed to stir at rt for 1 h and then heated to 6O 0 C for 1.5h.

Additional tBuOK (50mg) and 5-chloro-pyrimidine (69mg) were added and the reaction was run at 6O 0 C for another 1 h. The reaction was then heated to 100°C for another 1 h. Additional tBuOK (50mg) and 5-chloro-pyrimidine (69mg) were added and the reaction was run at 100 0 C overnight. The reaction was cooled to rt and quenched with a 20% (v/v) solution of acetic acid in water. The reaction was diluted with MeOH and δ-fS^-dimethyl-but-i -ynyO-S-^trans-^methyl-cydohexanecarbonyl)- [1 -(S^methyi-S-fpyrimidin-S-yioxyJ-propylj-aminoHhiophene^-car boxylic acid (4mg, 2.7%) was isolated by reverse phase HPLC as the TFA salt.

LC/MS (m/z): 497.88 [M-M]

Retention time: 2.56min

LC: Thermo Electron Surveyor HPLC

MS: Finnigan LCQ Advantage MAX Mass Spectrometer

Column: Phenomenex Polar RP 30 mm X 4.6 mm

Solvents: Acetonitriie with 0.1 % formic acid, Water with 0.1 % formic acid

Gradient: 0 min-0.1 min 5% ACN, 0.1 min-1.95 min 5%-100% ACN, 1.95 min-3.5 min 100% ACN, 3.5 min-3.55 min 100%-5% ACN, 3.55 min-4 min 5% ACN

Example 144 - Compound 144: 5-(3,3-dimethyl-but-1-vnyl)-3-[T3-(2-ethoxy-7- methoxy-8-methy[-quinolin-4-yloxy)-1-(S)-methyl-propyiH4-met hyl- cyclohexanecarbonyl)-amino1-thiophene-2-carboxylic acid

The 5-(3,3-dimethyl-but-1 -ynyl)-3-[(3-hydroxy-1 -(S)-methyl-propyl)-(trans-4- methyl-cycioheχanecarbonyl)-amino]-thiophene-2-carboxylic acid TFA salt (lOOrng, 0.239mmol) and 4-chloro-7-methoxy-2-ethoxy-8-methyt-quinoline (301 mg,

1.195mmol) were dissolved in DMSO (3mL). To this solution tBuOK (107mg, 0.956mmoi) was added in one portion at rt. The reaction was run at rt for 15min and then cooled in an ice bath. The reaction was quenched with a 20% (v/v) solution of acetic acid in water. The reaction was partitioned between water and EtOAc. The phases were separated and the organic phase was extracted with brine. The organic phase was extracted with brine and then concentrated. The residue was suspended in MeOH and remaining solids were removed by filtration. The filtrate was concentrated, suspended in MeOH and filtered again. The filtrate was clarified with a small amount of EtOAc and 5-(3,3-dimethyl-but-1 -ynyl)-3-[[3-(2-ethoxy-7-methoxy-8- methyl-quinolin-Φ-yloxyJ-i-fSJ-methyl-propyiJ^Φ-methyl-cyc lohexanecarbony!)- amino]-thiophene-2-carboxylic acid (29mg, 16%) was isolated as the TFA salt by reverse phase HPLC.

LC/MS (m/z): 635.00 [M+1]

Retention time: 2.73min

LC: Thermo Electron Surveyor HPLC

MS: Finnigan LCQ Advantage MAX Mass Spectrometer

Column: Phenomenex Polar RP 30 mm X 4.6 mm

Solvents: Acetonitrile with 0.1% formic acid, Water with 0.1% formic acid

Gradient: 0 min-0.1 min 5% ACN, 0.1 min-1.95 min 5%-100% ACN, 1.95 min-3.5 min 100% ACN, 3.5 miπ-3.55 min 100%-5% ACN 5 3.55 min-4 min 5% ACN Example 145 - Compound 145: 5-(3,3-Dimethyl-but-1-vnylV3-f(frans-4-methyi- cycSohexanecarbonyl)-fc/s-3-fpyridin-3-yloxy-N-oxide)-cvclob utySVamino>-thiophene- 2-carboxylic acid

Prepared in the same manner as Example 20 by repiacing 3-ftuoropyridine with 3-chloropyridine-1 -oxide. MS (m/z): 51 1.1 [M+H] + ; HPLC retention time: 4.00 min.

Example 146 - Compound 146: 5-(3,3-Dimethyibut-1-ynγl)-3-(4-methyl-N-(3-fpyridin- 2-yloxy)cvclobutvi)cyclohexanecarboxamido)thiophene-2-carbox y[ic acid

Compound 146

Prepared in the same manner as Example 20 by replacing 3-fluoropyridine with 2-fluoropyridine. MS (m/z): 495.1 [M+Hf; HPLC retention time: 4.57 min.

Example 147 : , Compound 147: 5-(3,3-Dimethyl-but-1-vnyl)-3-{(frans-4-methyi- cvclohexanecarbonyl)-[frans-4-(2-oxo-1 : 2-dihvdro-pyrimidin-4-yloxy)-cvciohexy!l- aminoHhiophene-2-carboxylic acid

Compound 147

5-(3,3-Dimethyl-but-1~yny!)-3-[{frans-4-hydroxy-cycIohexyl)- (ifraπs-4-methyl- cyclohexanecarbonyl)-amino]-thiophene-2-carboxy!ic acid (211 mg, 0.46 mmol) in DMF (2 mL) at 0 deg C was treated with sodium hydride (110 mg, 2.8 mmol, 60% oil dispersion) in portions. The mixture was stirred for 30 minutes at 0 deg C and a soiution of 2,4-dichloropyrimidine (210 mg, 1.4 mmol) in DMF (1 mL) was added dropwise. The reaction mixture was allowed to warm and stir at room temperature. After completion, ethyl acetate (2-3 mL) was added and the mixture was carefuliy quenched with citric acid (10% aqueous solution, 2-3 mL). Water was added and the mixture was extracted with ethyl acetate (2x30 mL). The combined organic layers were dried over magnesium sulfate, filtered and concentrated. The residue was passed through a plug of silica (0-5% MeOH:DCM eluent) and concentrated to yield 162 mg of 5-(3,3-Dimethyl-but-1-ynyl)-3-{(fraπs-4-methyl-cyclohexanec arbonyl)-

[frans-4-(2-chloro-pyrimidin-4-yloxy)-cyc!ohexyl]-amino}- thiophene-2-carboxyiic actd as an impure black oil which was carried on without further purification.

A crude sample of (3,3~Dimethyl-but-1-ynyl)-3-{(frans-4-methyl- cydohexanecarbonyl)-[frans-4-(2-chloro-pyπmidin-4-yloxy)-cy clohexy!]-amino}- thiophene-2-carboxylic acid (162 mg, 0.29 mmol) was dissolved in a 1 : 1 mixture of dioxane:water (8 mL) and treated with potassium carbonate (120 mg, 0.87 mmol) and DABCO (27 mg, 0.24 mmol). The mixture was heated at 70 deg C for 4 hours and then partitioned between water and ethyl acetate. The aqueous phase was neutralized by addition of aqueous citric acid and extracted with ethyl acetate (3X30 ml_). The combined organic layers were dried over magnesium sulfate, filtered and concentrated. The residue was purified by HPLC (Gemini column; 25%

acetonitrile:water, 2 min; 25-100% acetonitrile:water, 16 min; 100% acetonitriie, 3 min; both solvents containing 0.1 % trifluoroacetic acid). This resulted in 22.6 mg (8% yield over 2 steps) of the title compound as a white powder (TFA salt): MS (m/z): 537.9 [M-H] " ; HPLC retention time: 4.14 min (2-98% acetoπttrile:water with 0.05% trifluoroacetic acid).

Example 148 - Compound 148: 5-(3,3-Dimethylbut-1-vnyl)-3-fN-(4-(5-ethylpyhmidin- 2-yioxy)cyctohexyl)-4-methvicyclohexanecarboxamido)thiophene -2-carboxylic acid

Compound 148

Prepared in the same manner as Example 1 by replacing 2-fluoropyridine with 2-chloro-5-ethyipyrimidine. MS (m/z): 552.1 [M+H] + ; HPLC retention time: 5.07 min.

Example 149 - Compound 149: 5-(3,3-Dimethylbut-1-vnyl)-3-(4-methyl-N-(4-f2- (p_y_rroli ι dJn-1-yl)pyridin-3-yloxy)cvclohexyl)cvclohexanecarboxa mido)thiophene-2- carboxylic acid

Compound 149 Prepared in the same manner as Example 1 by replacing 2-fIuoropyridine with 3-fluoro-2-pyrrolidSn-1-yi-pyridine. MS (m/z): 592.2 [M+H] + ; HPLC retention time: 3.76 min.

Example 150 - Compound 150: 5-f3.3-Dimethylbut-1-vnyl)-3-(4-methvi-N-f4-(6- fρyrroiidlin-1 -vπpyhmidin-4-yloxy)cvciohexyi)cyciohexanecarboxamido)thiop hene-2- carboxylic acid

Compound 150 Prepared in the same manner as Example 1 by replacing 2-fluoropyridine with

4-chloro-6-pyrroiidin-1-yl-pyrimidine. MS (m/z): 593.0 [M+H] + ; HPLC retention time: 3.73 min.

Example 151 - Compound 151 : 3-(N-(4-(4-(Dimethviamino)-5-fluoropyπmidin-2- vioxy)cyciohexyl)-4-methyJcvciohexanecarboxamido)-5-(3,3-dim ethylbut-1 - ynyl)thiophene-2-carboxylic acid

Compound 151 Prepared in the same manner as Example 1 by replacing 2-fluoropyridine with

(2-chloro-5-fiuoro-pyrimidin-4-yl)-dimethylamine. MS (m/z): 585.0 [MH-H] + ; HPLC retention time: 3.70 min.

Example 152 - Compound 152: 5-(3,3-Dimethylbut-1-vnylV3-(4-methy[-N-f4-(6- (pyrrolidin-1-yl)pyridin-2-yloxy)cyclohexyl)cyciohexanecarbo xamido)thiophene-2- carboxyiic acid

Compound 152

Prepared in the same manner as Example 1 by replacing 2-fIuoropyridine with 2-bromo-6-pyrroiidin-1-yl-pyridtne. MS (m/z): 592.1 [M+Hf; HPLC retention time: 4.04 min.

Example 153 - Compound 153: 5-(3,3-Dimethylbut-1-vnyl)-3-(4-methv!-N-f4-(6- fpyrroiidin-1-yl)pyrazin-2-vioxy)cvciohexyl)cyclohexanecarbo xamido)thiophene-2- carboxylic acid

Compo 153

Prepared in the same manner as Example 1 by replacing 2-fluoropyridine with 2-chloro-6-pyrrolidin-1-yl-pyrazine. MS (m/z): 593.0 [M+H] + ; HPLC retention time: 4.40 min.

Example 154 - Compound 154: 5-f3,3-Dimethylbut-1-vnyl)-3-(4-methyl-N-(4-f3- methyl-1 ,2,4-thiadiazol-5-yloxy)cyclohexyl)cvclohexanecarboxamido)th iophene-2- carboxylic acid

Compound 154 Prepared in the same manner as Example 1 by replacing 2-fluoropyridiπe with 5-chioro-3-methyi-[1 ,2,4]-thiadiazole. MS (m/z): 541.8 [M-H] ' ; HPLC retention time: 5.10 min.

Synthesis of 2-Chioro-4-(pyrrolidin-1-ylmethvO-thiazole

2-Chloro-4-chloromethyl-thiazole (610 mg, 3.6 mmol), pyrrolidine (0.4 mL, 4.8 mmoi), and potassium carbonate (1.05 g, 7.6 mmoi) were stirred in DMF (6 mL) for 5 hours. After completion, the reaction mixture was partitioned between ethyl acetate and water. The aqueous phase was thrice extracted with ethyl acetate and the combined organic phases were washed with brine, dried over magnesium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (0- 5% [8:1 EtOH:NH 4 OH (aQ) ]/DCM). This resulted in 457 mg (63% yield) of the titie compound as a white solid. Exampie 155 - Compound 155: 5-f3,3-Dimethylbut-1-ynvi)-3-f4-methy[-N-(4-f4- fpyrrolidin-1-ylmethyl)thiazoJ-2-yloxy)cvclohexyl)cyclohexan ecarboxamido)thiophene- 2-carboxylic acid

Compound 155 Prepared in the same manner as Example 1 by replacing 2-fluoropyridine with 2-ch!oro-4-(pyrrolidin-1 -ylmethyl)-thiazo!e. MS (m/z): 612.1 [M+H] + ; HPLC retention time: 3.66 min.

Exampie 156 - Compound 156: 3-(N-(4-(4-((Dimethyiamino)methvπthiazol-2- yioxy)cyclohexyl)-4-methvicvclohexanecarboxamido)-5-(3,3-dim ethylbut-1- vnvPthiophene-2-carboxyiic acid

Compound 156

Prepared in the same manner as Exampie 1 by replacing 2-fluoropyridine with 2-chloro~4-(dimethylaminomethyl)-thiazole (prepared in the same manner as 2- chloro-4-(pyrrolidin-1-ylmethyl)-thiazote by substituting dimethylamine for pyrrolidine). MS (m/z): 586.1 [M+H] + ; HPLC retention time: 3.59 min.

Example 157 - Compound 157: 5-(3,3-Dimethylbut-1 -vnyl)-3-(4-methyl-N-(4-(pyridin- 4-yloxy-N-oxide)cvclohexyl)cvclohexanecarboxamido)thiophene- 2-carboxyiic acid

Compound 157

Prepared in the same manner as Example 1 by replacing 2-fluoropyridine with 4-nitro-pyridtne-1 -oxide. MS (m/z): 539.0 [M+H] + ; HPLC retention time: 3.73 min.

Exampie 158 - Compound 158: 3-(N-f4-(6-(Dimethylamino)pyrimidin-4- vJoxy)cvcJohexyl)-4-methylcyclohexanecarboxamido)-5-(3,3-dim ethylbut-1- ynyl)thiophene-2-carboxylic acid

Compound 158

Prepared in the same manner as Example 1 by replacing 2-fiuoropyridine with (6-chloro-pyrimidin-4-yi)-dimethyiamine. MS (m/z): 566.9 [M+H] + ; HPLC retention time: 3.67 min.

Example 159 - Compound 159: 5-f3,3-Dimethylbut-1-vnvπ-3-f4-methyl-N-f4-f6- (pyrrolidin-1-yl)pyrida2in-3-vioxy)cvclohexyπcyclohexanecar boxamido)thiophene-2- carboxylic acid

Compound 159

Prepared in the same manner as Example 1 by replacing 2-fluoropyridine with S-chloro-θ-pyrrolidin-i -yi-pyridazine. MS (m/z): 592.9 [M+H] + ; HPLC retention time: 3.59 min.

Example 160 - Compound 160: 3-(N-(4-(6-(Dimethy[amino)pyridazin-3- yloxy)cvciohexyl)-4-methylcyciohexanecarboxamido)-5-(3,3-dim ethylbut-1 - ynvQthiophene-2-carboxyiic acid

Compound 160 Prepared in the same manner as Example 1 by replacing 2-fiuoropyridine with (6-chioro-pyridazin-3-y!)-dimethy!amine. MS (m/z): 564.9 [M-H] " ; HPLC retention time: 3.51 min.

Exampie 161 - Compound 161 : 5-(3,3-Dimethvibut-1-vnylV3-f4-methyl-N-(4-(3- ( pyrroiidin-1 -vimethylH ,2.4-thiadiazol-5- yloxy)cvclohexyl)cvclohexanecarboxamido)thiophene-2-carboxyl ic acid

Compound 161

Prepared in the same manner as Example 1 by replacing 2-fiuoroρyridine with 5-chloro-3-(pyrroiidin-1-ylmethyS)-[1 ,2,4]-thiadiazole MS (rn/z): 613.1 [M+H] + ; HPLC retention time: 4.88 min.

Exampie 162 - Compound 162: 5-f3,3-Dimethylbut-1-vnyi)-3-f4-methyl-N-(4- (pvrimidin-5-yioxy)cyciohexyl)cyciohexanecarboxamido)thiophe ne-2-carboxylic acid

Compound 162

Prepared in the same manner as Example 1 by replacing 2-f!uoropyridine with 5-chloro-pyrimidine. MS (m/z): 524.0 [M+H] + ; HPLC retention time: 4.58 min.

Example 163 - Compound 163: 5-(3,3-Dimethylbut-1-ynyl)-3-(4-methvJ-N-(4- (pyrimidin-4-ytoxy)cvciohexyl)cycb ^

Compound 163

Prepared in the same manner as Example 1 by replacing 2-fluoropyridine with 4-chioro-pyrimidine. MS (m/z): 529.1 [M-H] " ; HPLC retention time: 4.14 min.

Example 166 - Compound 166: Sodium 5-(3,3-dimethylbut-1-vnylV3-f4-methvi-N-f4-

(4-(pyrrolidine-1 -carbonvOpyridin-2- yloxy)cyclohexyl)cyclohexanecarboxamido)thiophene-2-carboxyi ate.

Compound 166

Neat 2-chioro-tsonicotinoyl chloride (500 mg, 2.84 mmol) was added dropwise to a solution of pyrrolidine (1.18 ml_, 14.2 mmol) in dichloromethane (20 ml_) at 0 °C. After 30 min the ice bath was removed and the reaction was allowed to warm to room temperature. The reaction mixture was evaporated to dryness and the residue was partitioned between ethyl ether and sat. NaHCO 3 The aqueous layer was separated, extracted with ethyl ether and the combined organic layers were washed with brine, dried over Na 2 SO 4 and concentrated to provide the product as a viscous oil (614 mg, quantitative). Use in the next step without further purification,

NaH (50 mg of a 60% oil dispersion, 1.14 mmof) was added in portions to a solution of 5-(3 1 3-dimethyl-but-1-ynyl)-3~[(4-hydroxy-cyclohexyl)-(4-me thyl- cyclohexanecarbony!)-amino]-thiophene-2-carboxylic acid (128 mg, 0.287 mmol) and (2-chloro-pyridin-4-yl)-pyrrolidin-1-yl-methanone from the previous step (300 mg, 1.42 mmol) in THF (5.0 mL) at 0 0 C. After 5 min the reaction was warmed to room temperature and then heated at 85 0 C overnight. The reaction was cooled, aqueous NaOH (0.5 mL of a 1.0 N solution) was added and the reaction evaporated to dryness to give a solid residue. The solid was suspended in ethyl ether and sonicated for several minutes. The ether iayer was removed to give an orange solid that was dried under vacuum and purified by column chromatography on reverse phase Ci 8 silica gel (100% water to 20% acetonitrile/water). Fractions containing product were pooled and lyophilized to give the desired product as a colorless solid (30 mg, 16%). MS (m/z): 618.0 [M-H] " ; HPLC retention time: 6.43 min (2-98% acetonitrile:water with 0.05% tifSuoroacetic acid).

Example 167 - Compound 167: Sodium 3-(N-(4-(4-((1-methylpiperidin-4- yl)carbamoyl)pyridin-2-yloxy)cyclohexyl)-4-methvicyclohexane carboxamido)-5-(3.3- dimethylbut-1-vnyl)thiophene-2-carboxylate.

Compound 167

Neat 2-ch!oro-isonicotinoyl chloride (1.0 g, 5.69 mmol) was added dropwise to a soiution of 1-methyl-piperidin-4-ylamine (1.0 g, 8.75 mmoi) and triethyiamine (2.4 ml_, 17.22 mmol) in dichloromethane (25 m!_) at 0 °C. After 1 h the reaction was evaporated to dryness and partitioned between ethyl ether and sat, NaHCO 3 , The aqueous layer was separated, extracted with ethyl ether and the combined organic layers were washed with brine, dried over Na 2 SO 4 and concentrated to give the product as a pale yellow solid (190 mg, 13%). Use in the next step without further purification.

The title compound (23 mg, 20%) was synthesized in a manner analogous to Example 166 using 2-chloro-N-(1-methyl-piperidin-4-yl)-isonicotinamide from the previous step in place of (2-chioro-pyridin-4-yi)-pyrrolidin-1 -yl-methanone: MS (m/z):663.1 [M+H]+; HPLC retention time 5.59 min (2-98% acetonitrile: water with 0.05% trifiuoroacetic acid).

Example 168 - Compound 168: Sodium 3-(N-(4-(4-(azetidine-1-carbonyl)pyridin-2- yloxy^vclohexyD^-methylcvclohexanecarboxamidoVS-O^-dimethylb ut-i- ynyi)thiophene-2-carboxylate.

Compound 168

Azetidine (800 mg, 14 01 mmol) was added dropwise to a solution of 2- chloro-isonicotinoyl chloπde (500 mg, 2 84 mmol} in dichloromethane (20 ml_) at 0 0 C The reaction was allowed to slowly warm to room temperature over a 90 mm period and then evaporated to dryness The residue was partitioned between ethyl ether and sat NaHCO 3 The aqueous layer was separated, extracted with ethyl ether and the combined organic layers were washed with brine, dried over Na 2 SO 4 and concentrated to give the product as a colorless solid (291 mg, 55%) Use in the next step without further purification

NaH (60 mg of a 60% oii dispersion, 1 50 mmoi) was added in portions to a solution of 5-{3,3-dιmethy!-but-1 -yπyl)-3-[(4-hydroxy-cyclohexyl)-(4-methyi- cycJohexanecarbonyl)-amtno]-th[ophene-2-carboxylιc acid (130 mg, 0 291 mmoi) and azetidιn-1-yl-{2-ch!oro-pyrιdιn-4-yl)-methanone from the previous step (290 mg, 1 47 mmol) in DMF (3 0 mL) at 0 0 C After 30 msn the reaction was warmed to room temperature and then heated at 85 0 C for 3 5 h The reaction was cooled, aqueous NaOH (0 5 mL of a 1 0 N solution) was added and the reaction evaporated to dryness to give a solid residue The solid was purified by column chromatography on reverse phase C 18 silica gel (100% water to 20% acetonitrile/water) Fractions containing product were pooled and lyophilized to give the desired product as a coioriess solid (140 mg, 77%). HPLC retention time: 6.41 min (2-98% acetonitri!e:water with 0.05% tifluoroacetic acid).

Example 169 - Compound 169: Sodium 3-(N-(4-(4-(3-(dimethylamino)pyrroiidine-1- carbonyl)pyridiπ-2-γi ι o ι xy)cycjphexyl)-4-methylcvclohexanecarboxamiφj ι -5 : (3 J 3 : dimethylbut-1-vnyl)thiophene-2-carboxylate.

Compound 169 The title compound (87 mg, 38%) was synthesized in a manner analogous to

Exampie 168, using dimethyl-pyrrolidin-3-yl-amine in place of azetidine and in the second step the reaction was heated at 65 0 C. MS (m/z): 663.1 [M+H]+; HPLC retention time 5.18 min (2-98% acetonitrile: water with 0.05% trifluoroacetic acid). Example 170 - Compound 170: Sodium 5-(3,3-dimethyibut-1-ynyl)-3-(4-methyl-N-(4- (5-((2-oxopyrrolidin-1-yl)methvi)pyridin-2- yioxy)cyciohexyi)cyciohexanecarboxamido)thiophene-2-carboxyl ate.

Compound 170

NaH (520 mg of a 60% oil dispersion, 13.00 mrnol) was added in portions to a solution of 2-pyrrolidinone (1.1 g, 13.00 mmol) in DMF/THF (60 mL, 1 :10) at O 0 C. After stirring at this temperature for 1.5 h, solid 2-chioro-5-chioromethyl-pyridine (2.0 g, 12.34 mmol) was added along with a catalytic amount of TBAl. The reaction was allowed to warm to room temperature and stirred overnight. The reaction was hydrolyzed with the addition of satd. NH 4 CI, concentrated to near dryness and partitioned between ethyl acetate and water. The organic layer was separated, washed with 5% aqueous LiCI, brine, dried over Na 2 SO 4 and concentrated to give a dark orange residue. Purification by flash column chromatography on silica gel with ethyl acetate provided the desired product as a pale yellow solid (942 mg, 36%).

The title compound (22 mg, 10%) was synthesized in a manner analogous to Example 168, using 1-(6-chloro-pyridin-3-ylmethyl)-pyrrolidin-2-one from the previous step in place of azetidine. MS (m/z): 618.0 [M-H] ' ; HPLC retention time: 6.00 min (2- 98% acetonitrile:water with 0.05% tifluoroacetic acid).

Example 171 - Compound 171 : Sodium 5-(3.3-dimethvibut-1-vnyl)-3-(4-methyl-N- ff1 R)-4-(pyridin-2-yloxy)-3-(pyrroJidin-1- ylmethv0cyclohexyi)cyclohexanecarboxamido)thiophene-2-carbox viate.

Compound 171 5-(3,3-Dimethyl-but-1~ynyi)~3-[(4-methyl-cyclohexanecarbonyl )-(4-oxo- cyclohexyl)-amino]-thiophene-2-carboxylic acid methyl ester (350 mg, 0.76 mmol), pyrrolidine HCI (160 mg, 1.48) and paraformaldehyde (100 mg) were combined in ethanoi (2.5 ml_). A catalytic drop of concentrated HCI was added and the reaction was heated at 85 0 C for 24 h. The reaction was cooled, partitioned between ethyl acetate and sat. NaHCO 3 /water. The aqueous layer was extracted and the combined organic layers were washed with brine, dried over Na 2 SO 4 and concentrated to give a foam that was purified by flash column chromatography on silica gel with 10% methanol in dichloromethane to provide the desired product (262 mg, 63%) as a colorless foam.

To a solution of the ketone from the previous step (262 mg, 0.484 mmol) in

MeOH (5.0 ml_) cooled to 0 0 C was added NaBH 4 (40 mg, 1.05 mmol) in several portions. After stirring for 2 h, dilute HCI was added and the reaction was

concentrated to dryness to give a residue that was partitioned between ethyl acetate and sat. NaHCO 3 /bπne. The organic layer was separated, washed with brine, dried over Na 2 SO 4 and concentrated to give the product as a pale foam (275 mg, quantitative). NaH (10 mg of a 60% oil dispersion, 0.25 mmo!) was added in one portion to a solution of the alcohol from the previous step (43 mg, 0.08 mmol) in THF (1.0 mL) at 0 0 C. After stirring for 5 min, 2-fluoropyridine (0.07 mL, 0.81 mmo!) was added and the reaction was warmed to room temperature and then heated at 80 0 C overnight. After cooling to room temperature, 1.0 N NaOH (0.10 mL) was added and the reaction was concentrated to dryness to give an orange solid. The solid was purified by column chromatography on reverse phase C 18 silica gel (100% water to 50% acetonitrile/water) to provide the desired product as a colorless solid (15 mg, 30%) after iyophilization. MS (m/z): 606.2 [M+H]; HPLC retention time: 5.90 min (2-98% acetonitrile:water with 0.05% tifluoroacetic acid).

Example 173 - Compound 173: Sodium 5-(3,3-dimethylbut-1-vnyl)-3-(4-methvi-N-(4- (1-methγl-6-oxo-1 ,6-dihvdropyridazin-3- yloxy)cvcJohexyl)cγclohexanecarboxamido)thiophene-2-carboxy late.

To a solution of 5-(3,3-dimethyl-but-1 -ynyJ)-3-[(4-hydroxy-cyclohexylH4- methyl-cyclohexanecarbonyl)-amino]-thiophene-2-carboxylic acid methyl ester (250 mg, 0.54 mmol) and 6-hydroxy-2-methyi-2H-pyridazin-3-one (86 mg, 0.68 mmol) in THF (4.O mL) was added PPh 3 (157 mg, 0.60 mmol) at 0 0 C. DIAD (0.12 mL, 0.60 mmol) was added dropwise and the reaction was allowed to slowly warm to room temperature and stirred overnight. Sat. NH 4 CI was added and the reaction mixture was extracted with ethyl ether. The combined organic layers were washed with water, brine, dried over Na 2 SO 4 and concentrated to give a pale yellow foam. Purification by flash column chromatography on silica gel with 3% methanol in dichloromethane provided 335 mg of the desired product as a colorless foam that was contaminated with triphenylphosphine oxide.

To a solution of the methyl ester from the previous step (335 mg, assume 0.50 mmol) in methanol (0.75 ml_) and THF (0.75 mL) was added aqueous NaOH (0.75 mL of a 1.0 N solution) at room temperature. After stirring for 90 min the reaction was concentrated to dryness to give a pale yellow solid. Purification by column chromatography on reverse phase C 18 silica gel (100% water to 5%

acetonithle/water) provided the desired product as a colorless solid (28 mg, 9% for two steps) after lyophilization. HPLC retention time: 6.00 min (2-98%

acetonitrile:water with 0.05% tifluoroacetic acid).

Example 174 - Compound 174: 5-(3,3-Dimethylbut-1 -vnyl)-3-(4-methyl-N-(4-(1- methyi-6-oxo-1 ,6-dihydropyridazin-3- vioxy)cyciohexyi)cyclohexanecarboxamido)thiophene-2-carboxyl ic acid.

Compound 174

Cs 2 CO 3 (90 mg, 0.28 mmoϊ) was added to a solution of (5-{3,3-dimethyl-but-1- ynyO-S-^Φmethyl-cyclohexanecarbonylH^Θ-oxo-i .θ-dihydro-pyndazin-S-yloxy)- cyclohexyi]-amino}-thioρhene-2-carboxylic acid methyi ester (50 mg, 0.09 mmol) in DMF (2.0 mL) at room temperature. After stirring for 5 min, neat methyl iodide (40 mg, 0.28 mmol) was added and the reaction was stirred for 2 h. The reaction was filtered, concentrated and partitioned between ethyl acetate and 5% aqueous LiCI, The organic layer was separated, washed with brine, dried over Na 2 SO 4 and concentrated to give an orange foam that was used in the next step without further purification.

To a solution of the crude methyl ester from the previous step (assume 0.09 mmol) in methanol (0.50 mL) and THF (0.50 mL) was added aqueous NaOH (0.10 mL of a 1.0 N solution) at 0 0 C. The reaction was warmed to room temperature and after 90 min the reaction is evaporated to dryness to give a colorless solid. .

Purification by column chromatography on reverse phase C 18 silica gel (100% water to 20% acetonitrile/water) provided a colorless solid that was taken up in a minimum amount of water and acidified with 1.0 N HCI to give a colorless precipitate that was collected by filtration and dried under vacuum to provide the desired product (19 mg, 37% for two steps). MS (m/z): 553.9 [M+]; HPLC retention time: 4.50 min (2-98% acetonitrile:water with 0.05% trifluoroacetic acid).

Example 175 - Compound 175: Sodium 3-(N-(4-(4-((dimethyiamino)methyl)pyridin-2- yloxy)cyclohexyl)-4-methylcvclohexanecarboxamido)-5-(3,3-dim ethylbut-1- ynyl)thiophene-2-carboxvJate.

Compound 175 NaH {65 mg of a 60% oil dispersion, 1.62 mmol) was added to soiution 5-(3,3- dimethyi-but-1-ynyl)-3-[(4-hydroxy-cyclohexyl)-(4-methyl-cyc lohexanecarbonyl)- amino]-thiophene-2-carboxylic acid methy! ester (155 mg, 0.34 mmol) and (2-fluoro- pyπdin-4-y(methyl)-dimethyl-amiπe (130 mg, 0.84 mmo!) in DMF (4.0 mL) at 0 0 C. After 5 min the reaction was warmed to room temperature and then heated at 65 0 C for 2 h. The reaction was cooled, aqueous NaOH (0.5 mL of a 1.0 N solution) was added and the reaction mixture evaporated to dryness. Purification by column chromatography on reverse phase C 18 silica ge! (100% water to 20%

acetonitrile/water) provided the desired product as a colorless solid (62 mg, 31 %) after lyophtlization. MS (rn/z): 580.2 [M+H]; HPLC retention time: 5.05 min (2-98% acetonitrile:water with 0.05% trifluoroacetic acid).

Example 176 - Compound 176: Sodium 5-f3.3-dimethyibut-1.-ynγll-MN,:.(4 :

(imidazof1 ,2-b]pyridazin-6-yloxy)cvclohexyi)-4- methvicvclohexanecarboxamido)thiophene-2-carboxylate.

Compound 176

NaH (30 mg of a 60% oil dispersion, 0.75 mmol) was added to a solution of 5- (3,3-dimethyS-but-1-ynyl)-3-[(4-hydroxy-cyclohexy!)-(4-methy l-cyclohexanecarbonyl)- amino]-thiophene-2-carboxylic acid methyl ester (100 mg, 0.22 mmol) and 6-chloro- imidazo[1 ,2-b]ρyridazine (167 mg, 1.08 mmol) in THF (4.0 mL) at room temperature. The reaction was heated at 80 0 C overnight, cooled, aqueous NaOH (1.0 mL of a 1.0 N solution) was added and the reaction evaporated to dryness to give a brown solid. Purification by column chromatography on reverse phase C 18 silica gel (100% water to 30% acetonitrile/water) provided the desired product as a solid (17 mg, 14%) after iyophilization. MS (m/z): 563.0 [M+H]; HPLC retention time: 5.55 min (2-98% acetonitrile:water with 0.05% trifluoroacetic acid). Examples 177-183 were prepared by the same methods described for either Example 173 or 175 by substituting the appropriate heterocycie.

Example 177 - Compound 177: 3-(N-f4-f6-Chloropyridazin-3-yloxy)cvclohexyl)-4- methylcvclohexaπecarboxamido)-5-f3,3-dimethylbut-1-ynyl)thi opheπe-2-carboxylic acid

Compound 177 MS (m/z): 558.1 [M-HH]; HPLC retention time: 3.16 mtn (2-98%

acetonitrife:water with 0.05% trifluoroacetic acid).

Example 178 - Compound 178: 5-(3,3-Dimethylbut-1 -vnyl)-3-(N-f4-f6- hvdroxypyridazin-3-vioxy)cvclohexyS)-4-methylcyciohexanecarb oxamido)thiophene-2- carboxylic acid

Compound 178

MS (m/z): 540.2 [M+H]; HPLC retention time: 2.96 min (2-98%

acetonitriie:water with 0.05% trifluoroacetic acid). Example 179 - Compound 179: 5-(3.3-Dimethylbut-1-vnylV3-(4-rnethyl-N-(4- (pvrimidin-5-vloxv)cvclohexvi)cvciohexanecarboxamido)thiophe ne-2-carboxvlic acid

Compound 179

MS (m/z): 524.2 [M+H]; HPLC retention time: 2.46 min (2-98%

acetonitrile:water with 0.05% trifluoroacetic acid).

Exampie 180 - Compound 180: 5-(3,3-Dimethvibut-1-vnyl)-3-f4-methvi-N-{4-fpyridin- 3-yloxy)cvclohexyl)cvclohexanecarboxamido)thiophene-2-carbox yiic acid

Compound 180

MS (m/z): 523.2 [M+H]; HPLC retention time: 2.14 min (2-98%

acetonitriie:water with 0.05% trifiuoroacetic acid).

Exampie 181 - Compound 181 : 5-{3.3-Dimethylbut-1-vnvO-3-f4-methvl-N-f4- (pyrimidin-2-yloxy)cvciohexyl)cvclohexanecarboxamido)thiophe ne-2-carboxyIic acid

Compound 181

MS (m/z): 524.2 [M+H]; HPLC retention time: 2.49 min {2-98%

acetonitrile:water with 0.05% trifluoroacetic acid).

Example 182 - Compound 182: 3-(N-(4-(5-Ch[oropyrazin-2-yloxy)cvclohexyi)-4- methylcvclohexanecarboxamido)-5-f3,3-dimethylbut-1-vnvi)thio phene-2-carboxyiJc acid

Compound 182

MS (m/z): 558.1 [M+H]; HPLC retention time: 4.09 min (2-98%

acetonitrile:water with 0.05% trifluoroacetic acid).

Example 183 - Compound 183: 5-(3.3-Dimethylbut-1 -vnyl)-3-f4-methyl-N-f4- fpvridazin-4-vloxv)cvciohexvl)cvclohexanecarboxamido)thiophe πe-2-carboxyiic acid

Compound 183

MS (m/z): 524.2 [M+H]; HPLC retention time: 4.02 min (2-98%

acetonitri!e:water with 0.05% trifluoro acetic acid).

Examples 184 and 185 were prepared by the same method described for Example 1 by substituting the appropriate heterocycie. Example 184 - Compound 184: 3-(N-(4-(5-Chioroρyra2in-2-yloxy)cvciohexyl)-4-

acid

Compound 184

MS (m/z): 558.1 [M+H]; HPLC retention time: 4.32 min (2-98%

acetonitri!e:water with 0.05% trifluoroacetic acid).

Example 185 - Compound 185: 5-(3,3-Dimethylbut-1 -vnylV3-(4-methyl-N-f4- (pyridazin-4-yloxy)cvclohexyl)cyclohexanecarboxamido)thiophe ne-2-carboxyijc acid

Compound 185

MS (m/z): 524.1 [M+H]; HPLC retention time: 3,71 min (2-98%

acetonitriie:water with 0.05% trifluoroacetic acid).

Exampie 187 - Compound 187: 3-Fr-4-f6-pyrrolidin-1-ylmehtyl-pyridin-2-yloxy)- cvciohexylH4-methyl-cvclohexanecarbonvi)-aminol-5-(3,3-Dimet hy[-but-1-vnyl)- thiophene-2-carboxylic acid

The titie compound was synthesized in a manner analogous to Exampie 27, using (2-fluoro-pyridin-6-yimethyl)pyrrolidtne {prepared from 2-fluoro-pyridine-6- carbaidehyde and pyrrolidine in a manner analogous to Example 27) in place of 2- chloro-4~pyrro!idin-1-ylmethyl-pyridine: MS (m/z):606.2 [M+H]+; HPLC retention time 3.65 min (2-98% acetonitrile: water with 0.05% trifluoroacetic acid). Exampie 188 - Compound 188: 3-ff-4-f6-morpholin-1-yimehtyl-pyridin-2-yloxy)- cyclohexyl]-(4-methyi-cyclohexanecarbonyl)-amino1-5-(3,3-Dim ethyl-but-1-ynyl)- thiophene-2-carboxyiic acid

The title compound was synthesized in a manner analogous to Example 27, using 4-(2-chloro-pyridin-6-ylmethyl)morphoiine (prepared from 2-chloro-pyridine-6- carbaldehyde and morphoiine) in place of 2-chloro~4-pyrrolidin-1-yimethyl-pyridine: MS (m/z): 622.0 [M+H]+; HPLC retention time 3.58 min (2-98% acetonitriie: water with 0.05% trifluoroacetic acid). Exampie 189 - Compound 189: 3-[f-4-(4-pyrroSidin-2-one -1-ylmethyl-pyridin-2-yioxy)- cyclohexyl1-(4-methyl-cvciohexanecarbonyl)-amino]-5-(3,3-Dim ethyl-but-1-vnyl)- thiophene-2-carboxyiic acid

The title compound was synthesized in a manner analogous to Example 27, using 4-(2-chSoro-pyridin-4-ylmethy!)ρyrrolidin-2-one (prepared from 4-bromomethyl- 2-chioro-pyridine and pyrroiidin-2-one) in place of 2-chloro-4-pyrrolidin-1 -ylmethyl- pyridine: MS (m/z): 618.3 [M+H]+; HPLC retention time 4.20min (2-98% acetonitriie: water with 0.05% trifluoroacetic acid).

Example 190 - Compound 190: 3-[[-4-[4-(3-hvdroxy-p-yrrpjidin-l : ylmehMkByddjn:.2 ι : ι yloxy]-cyclohexyl]-(4-methyl-cyclohexanecarbonyl)-aminol-5-( 3,3-Dimethyl-but-1- ynyi)-thiophene-2-carboxylic acid

Compound 190

The title compound was synthesized in a manner analogous to Example 27, using 4-(2-fiuoro-pyridin-4-ylππethyl)pyrrolidin-3-ol (prepared from 2-fluoro-pyridine-4- carbaldehyde and pyrrrolidin-3-oi) in place of 2-chloro-4-pyrroiidin-1-ylmethyl- pyridine: MS (m/z): 622.1 [M+H]+; HPLC retention time 3.55min (2-98% acetonitrsle: water with 0.05% trifiuoroacetic acsd). Example 191 - Compound 191 : 3-[r-4-[4-ffl2-hvdroxy-ethvn-methyl-aminol-1- vimethyl)-ρyridin-2-yloxy1-cvclohexyi1-(4-methyi-cvciohexan ecarbonyl)-amino]-5-(3,3- Dimethyl-but-1 -vnyl)-thiophene-2-carboxyJic acid

Compound 191

The title compound was synthesized in a manner analogous to Example 27, using 2-[(2-fluoro-pyridin-4-ylmethyl)-methyl-amino]-ethanol (prepared from 2-fluoro- pyridine-4-carbaidehyde and 2-methylamino-ethano!) in place of 2-chloro-4- pyrrolidin-1 -yimethyl-pyridine: MS (m/z): 610.1 [M+H]+; HPLC retention time 3.54min (2-98% acetonitrile: water with 0.05% trifluoroacetic acid). Example 192 - Compound 192: 3-[[-4-f5-dimethylaminomethyl-pyridin-3-vioxy)- cvclohexyπ-f4-methvS-cvciohexanecarbonyl)-aminol-5-(3,3-dim ethyl-but-1 -ynγS)- thiophene-2-carboxyiic acid

Compound 192

The title compound was synthesized in a manner analogous to Example 27, using (5-fluoro-pyridin-3-ylmethyl)-dimethyl-amine (prepared from 5-fluoro-pyridine-3- carbaldehyde and dimethylamine) in place of 2-chloro-4-pyrrolidin-1-yimethyl- pyridine: MS (m/z): 580.2 [M+H]+; HPLC retention time 4.30min (2-98% acetonitrile: water with 0.05% trifiuoroacetic acid).

Example 193 - Compound 193: 3-ff-4-f5-pyrrolidn-1 -ylmethyl-pyridin-3-yloxy)- cvclohexyJH4-methyl-cyclohexanecarbonyπ-aminol-5-f3,3-dimet hyi~but-1-ynyl)- thiophene-2-carboxylic acid

The title compound was synthesized in a manner analogous to Example 27, using (3-fluoro-5-pyrroiidin-1-ylmethyl)-pyridine (prepared from 5-f!uoro-pyridine-3- carbaldehyde and pyrrolidine) in place of 2~ch!oro-4-pyrrolidin-1-ylmethyl-pyridine: MS (m/z): 606.2 [M+H]+; HPLC retention time 3.38min (2-98% acetonitrile: water with 0.05% trifiuoroacetic acid). Example 194 - Compound 194: 3-f[-4-f5-morpholin-4-ylmethyl-pyridin-3-yloxy)- cvclohexy[]-(4-methyl-cvc[ohexanecarbonvπ-amino]-5-(3,3-dim ethyl-but-1-vnvi)- thiophene-2-carboxyiic acid

Compound 194

The title compound was synthesized in a manner analogous to Example 27, using (5-fluoro-pyridin-3-ylmethyl)-morpholine (prepared from 5-f!uoro-pyridine-3- carbaSdehyde and morpholtne) in place of 2-chSoro-4-pyrrolidin-1 -ylmethyl-pyridine: MS (m/z): 622.2 [M+HJ+; HPLC retention time 3.39 min (2-98% acetonitrile: water with 0.05% trifluoroacetic acid).

Example 195 - Compound 195: 3-ff-4-(3-dimethylamino-pyrrolidn-1 -ylmethyl-pyridin- 3-yloxy)-cvclohexylH4-methyl-cvciohexanecarbonyl)-aminol-5-( 3,3-dimethyl-but-1 - vnylHhiophene-2-carboxylic acid

Compound 195

The title compound was synthesized in a manner analogous to Example 27, using [1-(3-fluoro-pyridin-4-ylmethyl)-pyrrolidin-3-yl]-dimethyl-a mine (prepared from 3-fluoro-pyridine-4-carbaldehyde and dimethyi-pyrroiidine-3-yl-amine) in place of 2- chioro-4-pyrrolidin-1-ylmethyi-pyridine: MS (m/z): 649.3 [M+H]+; HPLC retention time 2.98min (2-98% acetonitrile: water with 0.05% trifluoroacetic acid). Example 196 - Compound 196: 3-IT-4-(4- pyrrolidn-1 -ylmethyl-pyridin-3-ylox\0- cycjoJiexylH4-me^^

thiophene-2-carboxylic acid

Compound 196

The title compound was synthesized in a manner anafogous to Example 27, using 3-fluoro-4-pyrrolidine-1 -ylmethyl-pyridine (prepared from 3-fluoro-pyridine-4- carbaldehyde and pyrrolidine) in place of 2-chloro-4-pyrroiidin-1-y!methyi-pyridine: MS (m/z): 606.2 [M+H]+; HPLC retention time 3.23min (2-98% acetonitriie: water with 0.05% trifiuoroacetic acid).

Example 197 - Compound 197: 3-[[-4-(4-azetidin -1 -ylmethyi-pyridin-3-yioxy)- cvciohexyll-(4-methyl-cyciohexanecarbonyl)-aminol-5-f3,3-dim ethyl-"but-1 -vnyl)- thiophene-2-carboxylic acid

The title compound was synthesized in a manner analogous to Example 27, using 4-azetidine-1-ylmethy!-3-fiuoro-pyridine (prepared from 3-fluoro-pyridine-4- carbaldehyde and azetidine) in place of 2-ch!oro-4-pyrroiidin-1-ylmethyI-pyridine: MS (m/z): 592,3 [M+H]+; HPLC retention time 3.18min (2-98% acetonitriie: water with 0.05% trifiuoroacetic acid). Example 198 - Compound 198: 3-[[-4-f4-dimethylaminomethyl-pyridin-3-yloxyV cvclohexyi1-(4-methvi-cvciohexanecarbonyl)-amino1-5-(3,3-dim ethvi-but-1-vnyl)- thiophene-2-carboxyJic acid

Compound 198

The title compound was synthesized in a manner analogous to Example 27, using (3-f!uoro-pyridin-4-ySmethyl)-dimethyl-amine (prepared from 3-fiuoro-pyridine-4- carbaldehyde and dimethyiamine) in place of 2-chloro-4-pyrroiidin-1-ylmethyi- pyridine: MS (m/z): 580.2 [M+H]+; HPLC retention time 3.18min (2-98% acetoπitriie: water with 0.05% trifluoroacetic acid).

Exampie 199 - Compound 199: 3-Ff-4-(2-pyrrølidin-1-ylmethyl -pyridin-4-yloxy)- cvclohexyil-f^methyhcyclohexanecarbonvO-aminoi-S-fS^-dimethy i-but-i-ynyl)- thiophene-2-carboxylic acid

Compound 199

The title compound was synthesized in a manner analogous to Exampie 27, using 4-chloro-2-pyrrolidin-1-yimethyl-pyridine (prepared from 4-chloro-pyridiπe-2- carbaldehyde and pyrrolidine) in place of 2-chloro-4-pyrrolidin-1-yimethyl-pyridine: MS (m/z): 606.1 [M+H]+; HPLC retention time 3.46min (2-98% acetonitrile: water with 0.05% trifluoroacetic acid).

Example 200 - Compound 200: 3-Ff-4-(2-dimethylaminomethyl -pyridin-4-yloxy)- cvclohexyll-(4-methy!-cyclohexanecarbonyi)-amino1-5-(3,3-dim ethyJ-but-1-ynyi)- thiophene-2-carboxylic acid

Compound 200

The title compound was synthesized in a manner analogous to Example 27, using (4-chloro-pyridin-2-yimethyl)-dimethyl-amine (prepared from 4-chloro-pyridine- 2-carbaldehyde and dimethylamine) in place of 2-chloro-4-pyrrolidin-1-ylmethyl- pyridine: MS (m/z): 580.1 [M+H]+; HPLC retention time 3.43mtn (2-98% acetonitrile: water with 0.05% trifiuoroacetic acid). Example 201 - Compound 201 : 5-(3,3-Dimethyl-but-1-vπvlV3-(f4-methvS- cvclohexanecarbonviHI-d -methyl-I H-imidazole^-sulfonySVpiperidin^-vn-amino)- thiophene-2-carboxyiic acid

Compound 201

The title compound was synthesized in a manner analogous to Example 140, using 1-methyi-1 H-imidazoie-4-su!fonyi chloride in place of pyridine-2-carbonyl chloride: MS (m/z): 575.1 [M+H]; HPLC retention time: 4.34 min (2-98%

acetonitrile:water with 0.05% trifiuoroacetic acid; 6 min run).

Example 202 - Compound 202: 5-(3,3-DimethyJ-but-1-vnvi)-3-((4-methyl- cvclohexanecarbonyn-F1 -(4H-f1 ,2,41tna2oie-3-sulfonvn-piperidin-4-vn-amino)- thiophene-2-carboxylic acid

Compound 202

The title compound was synthesized in a manner analogous to Example 140, using 4H-[1,2,4]triazoie-3-suJfonyl chloride in place of pyridine-2-carbonyl chloride: MS (m/z): 561.7 [M+H]; HPLC retention time: 4.34 min (2-98% acetonitriSe:water with 0.05% trifluoroacetic acid) 6 min run.

Example 203 - Compound 203: 5-f3,3-Dimethyl-but-1-vnyl)-3-{(4-methyl- cycSohexanecarbonvD-fi-fΣ-methyS^H-pyrazole-S-sulfonyD-pipe ridin^-vil-aminoV thiophene-2-carboxylic acid

Compound 203

The title compound was synthesized in a manner analogous to Example 140, using 2-methy!-2H-pyrazole-3-su!fonyl chloride in place of pyridine-2-carbonyl chloride: MS (m/z): 575.1 [M+H]; HPLC retention time: 4.85 min (2-98%

acetonitrile:water with 0.05% trifluoroacetic acid) 6 min run.

Example 204 - Compound 204: 3-[ri-(2-DimethyJamino-pyrimidine-5-sulfonyl)- piperidin-4-yn-(4-methyl-cvclohexanecarbonylVamino]-5-(3,3-d imethyl-but-1-ynyl)- thiophene-2-carboxylic acid

Compound 204

The title compound was synthesized in a manner analogous to Example 140, using 2-dimethylamino-pyrimidine-5-sutfonyl chloride in piace of pyridine-2-carbony! chloride: MS (m/z): 616.1 [M+H]; HPLC retention time: 5.00 miπ (2-98%

acetonitrtle;water with 0.05% trifluoroacetic acid) 6 min run.

Example 205 - Compound 205: 5-(3,3-Dimethyl-but-1-vnyl)-3-(f4-nrιethyl- cyclohexanecarbonyJHlrβhiophθne-S-suifonvπ-piperidin-^yli -arninoHhiophene^- carboxylic acid

Compound 205 The title compound was synthesized in a manner analogous to Example 140, using thiophene-3-sulfonyi chloride in place of pyridine-2-carbonyl chloride: MS (m/z): 576.9 [M+H]; HPLC retention time: 4.94 min (2-98% acetonitrile:water with 0.05% trifluoroacetic acid) 6 min run, Example 206 - Compound 206: 5-f3,3-Dimethyl-but-1-vnylV3-rF1-f1 ,2-dimethyl-1 H- imidazole-4-sulfonyl)-piperidin-4-vn-f4-methyl-cyclohexaneca rbonvi)-amino1- thioρhene-2-carboxylic acid

ompound 206

The title compound was synthesized in a manner analogous to Example 140, using 1 ,2-dimethyi-1 H-imidazole-4-sulfonyl chloride in place of pyridine-2-carbonyi chloride: MS (m/z): 589.1 [M+H]; HPLC retention time: 4.14 min {2-98%

acetonitrile:water with 0.05% trifluoroacetic acid) 6 min run.

Example 207 - Compound 207: 5-(3,3-Dimethyl-but-1-ynyi)-3-{(4-methyi- cvclohexanΘcarbonvD-fi-fthiophene^-suifonvD-pipehdin^-yli-a minol-thiophene-Σ- carboxylic acid

Compound 207 The title compound was synthesized in a manner analogous to Example 140, using Thioρhene-2-sulfony! chloride in place of pyridine-2-carbonyl chloride: MS (m/z): 576.9 [M+H]; HPLC retention time: 5.02 min (2-98% acetonitrile:water with 0.05% trifluoroacetic acid) 6 min run. Example 208 - Compound 208: 5-f3,3-Dimethyl-but-1-ynyl)-3-((4-methyl- cyclohexanecarbonyll-LI- aminoHhiophene-2-carboxylic acid

Compound 208

The title compound was synthesized in a manner analogous to Example 140, using 2-oxo-1 ,2-dihydro-pyrimidine-5-suffonyl chloride in place of pyridine-2-carbonyl chloride: MS (m/z): 589.1 [M+H]; HPLC retention time: 4.22 min (2-98%

acetonitrile:water with 0.05% trifluoroacetic acid) 6 min run.

Example 209 - Compound 209: 5-(3,3-Dimethyl-but-1 -vnyl)-3-{f4-methyl- cYciohexanecarbonylHI-fpy^diπ^S-sulfonvD-piperidin^-viyamin oVthiophene-Σ- carboxylic acid

Compound 209 The title compound was synthesized in a manner analogous to Example 140, using pyridine-2-sulfonyi chloride in place of pyridine-2-carbonyl chloride: MS (m/z): 572.2 [M+H]; HPLC retention time: 4.71 min (2-98% acetonitriie:water with 0.05% trifluoroacetic acid) 6 min run. Example 210 - Compound 210: 5-f3,3-Dimethyl-but-1-vnyl)-3-f(4-methyi- cyclohexanecarbonylHI-fpyridine-S-suifonvD-piperidin^-yn-ami noHhiophene^- carboxylic acid

Compound 210

The title compound was synthesized in a manner analogous to Example 140, using pyridine-3-sulfonyI chloride in place of pyridine-2-carbonyi chloride: MS (m/z): 572.2 [M+HJ; HPLC retention time: 4.67 min (2-98% acetonitrile:water with 0.05% trifluoroacetic acid) 6 min run.

Example 211 - Compound 211 : 5-(3.3-Dimethvi-but-1-vnvO-3-f(4-methyl- cyciohexanecarbonviHI-fthiophene-S-sulfonyO-pyrroiidin-S-vii -aminoHhiophene-Σ- carboxyiic acid

Compound 211 The title compound was synthesized in a manner analogous to Example 142, using thiophene-3-sulfonyl chloride in place of pyridine-2-carbonyl chloride: MS (m/z): 563.0 [M+H]; HPLC retention time: 4.82 min (2-98% acetonitrile:water with 0.05% trifluoroacetic acid) 6 min run. Example 212 - Compound 212: 5-f3,3-Dimethyl-but-1-vnvπ-3-fri-f1 ,2-dimethyl-1 H- imidazoie-4-sulfonyl)-ρyrrolidin-3-yn-(4-methvi-cyclohexane carbonyπ-aminoV thiophene-2-carboxyiic acid

Compound 212

The title compound was synthesized in a manner analogous to Example 142, using 1,2-Dimethyi-1H-imidazo!e-4-sulfonyl chloride in place of pyridine-2-carbonyl chloride: MS (m/z): 575.1 [M+H]; HPLC retention time: 4.13 min (2-98%

acetonitrile:water with 0.05% trifluoroacetic acid) 6 min run.

Example 213 - Compound 213: 5-(3,3-Dimethyl-but-1-vnvi)-3-f(4-methyi- cyclohexanecarbonyiVfi-fthiophene-Σ-sulfonyQ-pyrrolidin-S-y li-aminoHhiophene-Σ- carboxylic acid

The title compound was synthesized in a manner analogous to Example 142, using thiophene-2-sulfonyl chloride in place of pyridine-2-carbony! chloride: MS (m/z): 563.1 [M+H]; HPLC retention time: 4.88 min (2-98% acetonitrϋe:water with 0.05% trifluoroacetic acid) 6 min run. Example 214 - Compound 214: 5-f3,3-Dimethvi-but-1-vnylV3-{(4-methyl- cyclohexanecarbonvi)-M-(2-oxo-1 ,2-dihyciro-ρyrimidine-5-sulfonyl)-ρyrrolidin-3-vπ-

.i.OlJBoHhiophene-g-carbgxylic.acid

Compound 214

The title compound was synthesized in a manner analogous to Example 142, using 2-oxo-1 ( 2-dihydro-pyrimidine-5-sulfonyl chloride in place of pyπ ' dine-2-carbonyi chloride: MS (m/z): 575.1 [M÷H]; HPLC retention time: 4.10 min (2-98%

acetonitrile:water with 0.05% trifiuoroacetic acid) 6 min run.

Example 215 - Compound 215: 5-(3,3-Dimethv[-but-1-vnyl)-34(4-methvi- cvclohexanecarbonyl)-ri-(pyridJne 2 : sylfonyl)-pyrrolidin-3-vπ-amino)-thiophene-2- carboxylic acid

Compound 215 The title compound was synthesized in a manner analogous to Example 142, using pyridine-2-suifonyf chloride in place of pyrtdine-2-carbonyl chloride: MS (m/z): 558.2 [M+H]; HPLC retention time: 4.47 min (2-98% acetonitrile:water with 0.05% trifiuoroacetic acid) 6 min run. Example 216 - Compound 216: 5-f3,3-Dimethy[-but-1-vnylV3-ff4-methyl- cyciohexanβcarbonylHI-fpyridine-S-sulfonvπ-pyrrolidin-S-yl l-aminoHhiophene^- carboxylic acid

Compound 216

The title compound was synthesized in a manner analogous to Example 142, using pyridine-3-sulfonyl chioride in place of pyridine-2-carbonyl chloride: MS (m/z): 558.2 [M+H]; HPLC retention time: 4.56 min (2-98% acetonitrile:water with 0.05% trifluoroacetic acid) 6 min run.

Example 219 and 220 - Compound 219: 5-f3.3-dimethylbut-1-vnyl)-3-ff1 r,4RV4- methyl-N-(hr,4R)-4-qetrahvdro-2H-pvran-4~

yloxy)cyc!ohexyi)cyclohexanecarboxamido)thiophene-2-carbo xylic acid

and Compound 220: 5-f3,3-dtmethylbut-1-vnyl)-3-fM r.4RV4-methyl-N-ff1s,4SV4- ftetrahvdro-2H-pyran-4-yloxy)cyclohexyS)cvclohexanecarboxami do)thiophene-2- carboxylic acid

TMSCl, imidazole

S

220

Tetrahydro-4-pyranol (1 g, 9.8 mmo!) was dissolved in 30 mL of methylene chloride. To it was added imidazole (1 Jg 1 24.5 mmoi) followed by TMSCI (1 ,5 mL, 11 JQ mmol). The reaction was allowed to stir at room temperature overnight. Insoluble material was removed by filtration, and the filtrate was washed with half saturated aqueous sodium bicarbonate and brine, and dried over MgSO 4 , The solids were filtered and solvent removed under reduced pressure to afford 1.6 g of trimethyl(tetrahydro-2H-pyran-4-yloxy)silane as a clear liquid.

To a mixture of Iron (HI) chloride (3.6 mg, 0.022 mmol ) and methyl 5-(3,3- dimethyibut-1 -ynyl)-3-(( 1 r,4R)-4-methyl-N-{4- oxocyciohexyl)cyclohexanecarboxamido)thiophene-2-carboxylate (100 mg,

0.22mmol) in 0.7 mL of nitromethane) were added trimethyi(tetrahydro-2H-pyran-4- yloxy)silane (77 mg, 0.44 mmo!) and triethylsilane (31 μL, 0.264 mmo!) successively, with stirring at 0 0 C under argon. The reaction was allowed to warm to room temperature and stirred at room temperature for 2 hours. The reaction was quenched with a phosphate buffer. The organic materials were extracted with methylene chloride, washed with brine and dried over MgSO 4 , The solids were filtered and solvent removed under reduced pressure. The residue was purified by reverse phase HPLC eluting with acetonitrile /H 2 O with 0.1% TFA to afford 36 mg of methyl 5-(3,3- dimethylbut-1 -ynyl)-3-((1r,4R)-4-methyl-N-((1 r,4R)-4-(tetrahydro-2H-pyran-4- yloxy)cyclohexyI)cyclohexanecarboxamido)thiophene-2-carboxyl ate (front peak) and 20 mg of methyl 5-{3,3-dimethylbut-1-ynyl)-3-{(1 r,4R)-4-methyl-N-((1s,4S)-4- (tetrahydro-2H-pyran-4-yioxy)cyclohexyl)cyciohexanecarboxami do)thsophene-2- carboxylate (back peak).

30 mg of methyl 5-(3,3-dimethylbut-1-ynyl)-3-((1 r,4R)-4-methyi-N-((1r,4R)-4- (tetrahydro-2H-pyran-4-yloxy)cyclohexyl)cyciohexanecarboxami do)thiophene-2- carboxylate was dissolved in 2 mL of THF:H 2 O:MeOH (3:1 :2) and to it was added 0.3 mL of 1N LiOH. The reaction was stirred at room temperature for 5 hours. The solvent was removed and purified by reverse phase HPLC 1 eluting with acetonitrile /H 2 O with 0.1% TFA, to afford 24 mg of 5-(3,3-dimethylbut-1-ynyl)-3-((1r,4RH- methyl-N-((1 r,4R)-4-(tetrahydro-2H-pyran-4- yloxy)cyciohexy!)cyclohexanecarboxamido)thiophene-2-carboχy iic acid as a white solid. MS (m/z): 529.7 [M+H]; HPLC retention time: 4.76 min (2-98%

acetonitrile:water with 0.05% trifluoroacetic acid) 6 min run.

15 mg of methyl 5-(3, 3-dimethylbut-1-ynyl)-3-((1 r, 4R)-4-methyl-N~ ((1 s,4S)-4-(tetrahydro-2H-pyran-4- yloxy)cyclohexy[)cyciohexanecarboxamido)thiophene-2-carboxyl ate was dissolved in 1 mL of THF:H 2 0:MeOH (3:1 :2) and to the solution was added 0.15 mL of 1 N LiOH. The reaction was stirred at room temperature for 5 hours. The solvent was removed and purified by reverse phase HPLC eluting with acetoπitrile /H 2 O with 0.1% TFA to afford 7 mg of 5-(3,3-dimethylbut-1 -ynyl)-3-((1r,4R)-4-methyl-N- {{1 s,4S)-4-(tetrahydro-2H-pyran-4- yloxy)cyciohexy!)cyciohexanecarboxamido)thtophene-2-carboxy! ic acid as a white soiid. MS (m/z): 530.0 [M+H]; HPLC retention time: 4.94 min (2-98%

acetonitrile:water with 0.05% trifluoroacetic acid) 6 min run.

Example 221 - Compound 221 : 5-f3,3-dimethvSbut-1-vnvi)-3-ff 1 r i 4R)-4-methyl-N-f4- ff3S,3aS,6aRHetrahydro-2H-furof2.3-b1furan-3- yloxy)cvclohexyi)cvciohexanecarboxamidoHhiophene-2-carboxyli c acid

The title compound was synthesized in a manner analogous to Example 219, using (3S,3aS,6aR)-tetrahydro-2H-furo[2,3-b]furan-3-ol instead of tetrahydro-4- pyranol: MS (m/z): 558.2 [M+H]; HPLC retention time: 4.67 min (2-98%

acetoni true: water with 0.05% trifluoroacetic acid) 6 min run. Example 222 - Compound 222: 3-(N-(4-(1-ftert-butoxycarbonyl)piperidin-4- yloxy)cvciohexyl)-4-m§thyJcycjohexaj]ecarboxam^

vnvDthSophene-2-carboxylic acid

Compound 222 The title compound was synthesized in a manner analogous to Example 219, using tert-butyl 4-hydroxypiperidine-1-carboxylate instead of tetrahydro-4-pyranol: MS (m/z): 629.3 [M+H]; HPLC retention time; 5.47 min (2-98% acetonitrile:water with 0.05% trifluoroacetic acid) 6 min run.

Example 223 - Compound 223: 5-f3.3-dimethylbut-1-vnvh-3-(4-methyl-N-f4- fp!peridin-4-yloxy)cvclohexyi)cyclohexanecarboxamido)thiophe ne-2-carboxylic acid

Compound 223

The title compound was synthesized from Example 222 by treatment with 20% TFA in methylene chloride at room temperature for 4 hours. MS (m/z): 529.1 [M+H]; HPLC retention time: 3.20 min (2-98% acetonitrileiwater with 0.05% trifluoroacetic acid) 6 min run.

Example 224 - Compound 224: 5-f3.3-dimethylbut-1-vnvO-3-(4-methyl-N-(4-

(tetrahydrofuran-3-yloxy)cvclohexyl)cyclohexanecarboxamid o)thiophene-2-carboxylic acid

Compound 224

The title compound was synthesized in a manner analogous to Example 219, using tetrahydrofuran-3-ol instead of tetrahydro-4-pyranol: MS (m/z): 516.2 [M+H]; HPLC retention time: 4.824 min (2-98% acetonitrile: water with 0.05% trifluoroacetic acid) 6 min run. Example 225 - Compound 225: 3-(N-((1S,4RV4-((SV1-ftert- butoxycarbonvOpyrrolidin-3-ytoxy)cyclohexyl)-4-methyicyciohe xanecarboxamido)-5- (3,3-dimethyibut-1 -ynyl)thiophene-2-carboxylic acid

Compound 225

The title compound was synthesized in a manner analogous to Example 219, using (S)-tert-butyi 3-hydroxypyrroiidine-i -carboxylate instead of tetrahydro-4- pyranol: MS (m/z): 615.3 [M+H]; HPLC retention time: 5.36 min (2-98%

acetonitrile:water with 0.05% trifluoroacetic acid) 6 min run.

Example 226 - Compound 226: 5-(3,3-dimethylbut-1-vnyl)-3-(4-methyi-N-(4-(fS)- pyrroiidin-3-yloxy)cyclohexyl)cyclohexanecarboxamido)thiophe ne-2-carboxylic acid

Compound 226

The title compound was synthesized from Example 225 by treatment with 20% TFA in methylene chloride at room temperature for 4 hours. MS (m/z): 515.2 [M+H]; HPLC retention time: 3.12 min (2-98% acetonitrile:water with 0.05% trifluoroacetic acid) 6 min run. Example 227 - Compound 227: 5-f3.3-dimethvibut-1 -vnyl)-3-f(1 R.2S,4R)-2-hvdroxy- 4-methyl-N-(4-(tetrahydrofuran-3- vioxy)cvclohexyl)cvclohexanecarboxamido)thiophene-2-carboxyl jc acid

Compound 227

The title compound was synthesized in a manner analogous to Exampie 219, using initially methyl 5-(3,3-dimethy!but-1-ynyl)-3-(4-oxocyclohexylamino)thiophene - 2-carboxylate and tetrahydrofuran-3-ol instead of methyl 5-(3,3-dimethylbut-1-ynyl)- 3-(4-methyl-N-(4-oxocyclohexyl)cyclohexanecarboxamido)thioρ hene-2-carboxylate and tetrahydro-4-pyranol; then purified by reverse phase HPLC to afford two products. Upon isolation of the front peak, the initial product was acyiated with (1S,2R,5R)-2-(chSorocarbony!)-5-methylcyclohexyl acetate and then subjected to ester hydrolysis with LiOH: MS (m/z): 532.2 [M+H]; HPLC retention time: 4.530 min (2-98% acetonitrtle: water with 0.05% trifluoroacetic acid)

Exampie 228 - Compound 228: 5-f3,3-dimethylbut-1 -ynylV3-((1 R,2S,4R)-2-hydroxy- 4-methyl-N-(4-(fRHetrahvdrofuran-3- yloxy)cyciohexyl)cvclohexanecarboxamido)thiophene-2-carboxyi ic acid

Compound 228

The title compound was synthesized in a manner analogous to Example 227 but from the second peak isolated in the first reaction therein: MS (m/z): 532.2 [M+H]; HPLC retention time: 4.582 min (2-98% acetonitrile:water with 0.05% trifiuoroacetic acid) 6 min run.

Example 229 - Compound 229: 5-(2-cvciopropylethvnvi)-3-f4-methvi-N-(4-(pyridin-2- v!oxy)cyciohexyl)cyclohexanecarboxamido)thiophene-2-carboxyi ic acid

Compound 229

Compound 229 was synthesized in the same manner as Example 1 using 5- (2-cyc!opropyiethynyl)-3-(N-(4-hydroxycyc)ohexyl)-4- methylcyciohexanecarboxamido)thiophene-2-carboxylic acid as a starting material. [M + H + ] = 507.1. HPLC retention time = 4.65 min (6 minute method 2-98% acetonitrile:water with 0.05% trifluoroacetic acid. Example 230 - Compound 230: 3-(N-f4-f(RV1-ftert-butoxycarbonvl)pyrroiidin-3- yioxy)cvciohexyl)-4-methylcyclohexanecarboxamido)-5-(3,3-dim ethyibut-1- ynvPthiophene-2-carboxylic acid

Compound 230

The title compound was synthesized in a manner analogous to Example 219, using (R)-tert-butyl 3-hydroxypyrrolidine-i-carboxylate instead of tetrahydro-4- pyranol: MS (m/z): 615.3 [M+H]; HPLC retention time: 5.429 min (2-98% acetonitrile:water with 0.05% trifluoroacetic acid) 6 min run.

Exampie 231 - Compound 231 : 5-(3,3-dimethylbut-1-vnvi)-3-(4-methvl-N-(4-((RV pyrrpj id i n-S-yjoxylcyciohexyπcYcSo h exanecarboxa m ido)th io phe ne-2-carboxylic a cid

Compound 231

The title compound was synthesized from Exampie 230 by treatment with 20% TFA in methylene chloride at room temperature for 4 hours. MS (m/z): 515.2 [M+H]; HPLC retention time: 3.418 min (2-98% acetonitrile:water with 0.05% trifluoroacetic acid) 6 min run.

Example 301 - Compound 301 : Synthesis of 5-(3,3-dimethyl-but-1-vnyl)-3-ff4.6S- dimethyl-cvdohex-3-θnecarbonyiH4-(tetrahydro-furan-3S-vioxy Hrans-cyclohexyl " |- aminoHhiophene-2-carboxylic acid (Isomer 1 )

Scheme 1

2 P ridine

A mixture of 3-amino-5-{3,3-dimethyi-but-1 -ynyl)-thiophene-2--carboxylιc acid methyl ester (2 62 g, 1 1.06 mmol), 4-(tetrahydro-furan-3S-yloxy)-cyclohexanone (1.7 g, 9.22 mmol), TFA (2.84 ml_, 36.9 mmol) and triethyisilane (2.94 ml_, 18.44 mmol) in CH 2 Ci 2 (15 ml_) was stirred for 24 h at room temperature. The reaction was concentrated in vacuo to remove volatiles and the crude oil was dissolved in toluene (50 ml_) and concentrated (repeat) to give a yellow solid. The crude solid was purified by column chromatography (hexane/ ethylacetate) to give 5-{3,3-dimethyl- but-1-ynyl)-3-[4-(tetrahydro-furan-3-yloxy)-cycloheχylamιn o]-thiophene-2-carboχylιc acid methyl ester (1.35 g, 3.33 mmol) as a single isomer. 4,6-S-dimethy!-cyclohex-3-ene-1 S-carboxylic acid (944mg, 6.17mmol) was dissolved in CH 2 CI 2 (1OmL) and DMF (20μL) was added. The solution was cooled to 0 0 C and then (COCI) 2 (700μL, 7.38mmol) was slowly added to the solution. The reaction was stirred in the ice bath for 1 hour and then concentrated. The residue was taken up in hexanes and concentrated; this hexanes coevaporatson was repeated once more. To the residue was added 5-(3,3-dimethyl-but-1-ynyl)-3-[4- (tetrahydro-furan-3S-yloxy)-trans-cyclohexylamino3-thiophene -2-carboxylic acid methyl ester (500mg, 1.23mmol) and pyridine (3mL). The solution was heated to 90 0 C overnight. The reaction was cooled to rt, concentrated and taken up in minimal CH 2 CI 2 . The two resulting isomers were purified and separated from each other by sϋica gel column chromatography, eluting with a mixture of EtOAc and hexanes. isomer A yield = 115mg, Isomer B yield = 162mg

Isomer A (105mg, 0.19mmoS) was dissolved in THF (1 mL) and MeOH (0.5mL). To this solution was added a solution of LiOH 1 H 2 O (39.8mg, 0.95mmoi) in H 2 O (0.5mL). The reaction was stirred at rt for 3h and then quenched with TFA. The entire reaction mixture was injected onto a reverse phase HPLC and Compound 301 was isolated (77mg).

LC/MS = 528 (M + +1 )

Retention time: 5,71 min

LC: Thermo Electron Surveyor HPLC

MS: Finnigan LCQ Advantage MAX Mass Spectrometer

Column: Phenomenex Polar RP 30 mm X 4.6 mm

Solvents: (B): Acetonitrile with 0.1% formic acid, (A): Water withθ.1% formic acid

Gradient: 2mL/min, 0 min-0.5min 5% ACN, 0.5 min-6.5 min 5%-100% ACN, 6.5 min-9.0 min 100% ACN, 9.0 min-9.1 min 100%-5% ACN, 9.1 min-9.5 min 5% ACN

Synthesis of (1 S,6S)-4,6-dimethγl-cyclohex-3-enecarboxyiic acid

Scheme 2 30% H 2 O 2 (aq), LiOH-H 2 O 1

THF, H 2 O, then Na 2 SO 3 (aq)

4S-benzyl-3-(4,6S-dimethyl-cyclohex-3-ene-1 S-carbonyl)-oxazofidin-2-one, prepared in a method simitar to that described in J. Am. Chem. Soc. 110(4), 1988, 1238-1256, was dissolved in THF (100OmL) and H 2 O (35OmL). The soSution was cooled in an ice bath and 30% H 2 O 2 (36mL, 354mmol) was slowly added followed by LiOH * H 2 O (S) (9.9Og, 263mmoi) in one portion. The reaction was allowed to slowly warm to rt and was stirred for 16h. The reaction was then cooled in an ice bath. Na 2 SO 3 (6Og, 472mmol) was dissolved H 2 O (40OmL) and very slowly added to the cooled reaction mixture. The solution was stirred for 1 h, then the layers were separated. The organics were removed under reduced pressure. The aqueous was added back to the organics concentrate and was washed with CH 2 CI 2 (2 X 50OmL). Adjust the aqueous pH to 2 with a slow addition of con HCI. Extract the aqueous with EtOAc (4 X 30OmL) and dry over Na 2 SO 4 . Remove organics under reduced pressure and co-evaporate with hexanes to afford (1 S,6S}-4,6-dimethyl-cyclohex-3- enecarboxyiic acid (14.14g, 78%) as a white solid.

Synthesis of 4-(tetrahydro-furan-3S-yloxy)-cycJohexanone Scheme 3

4-(tetrahydro-furan-3S-yloxy)-cyclohexanol (40.86mmol) was dissolved in DCM (20OmL) under an atmosphere of nitrogen. Dess-Martin Periodinane (20.78g, 49.03mmoi) was added in 4 portions. The reaction was stirred for 3 hours at room temperature until complete by TLC. The reaction was cooled to O 0 C and 10OmL of a 1 :1 solution of saturated aqueous NaS 2 O 3 :saturated aqueous NaHCO 3 was added. The reaction was allowed to warm to room temperature and was stirred for 1 hour. The layers were separated. The aqueous layer was extracted with DCM (5OmL x 2). The combined organics were dried with sodium sulfate, filtered and were

concentrated. 4-(Tetrahydro-furan-3S-yloxy)-cyclohexanone was purified by silica gel chromatography to afford 5.2g of a yellow oil (69%).

Example 302 - Compound 302: 5-f3,3-dimethyl-but-1-vnvQ-3-((4,6S-dimethyl- cyclohex-3-enecarbonv[)-f4-ftetrahvdro-furan-3S-yloxy)-trans -cvclohexyJ]-amino)- thiophene-2-carboxylic acid (Isomer 2)

Compound 302 δ^S.S-dimethyl-but-i-ynyO-S-^^βS-climethyl-cyclohex-S-enec arboπy!)-^- (tetrahydro-furan-3S-yloxy)~trans-cyclohexy!]-amino}-thiophe ne-2~carboxyiic acid

(Compound 302) was prepared in a similar fashion as 5-(3,3-dimethyl-but-1-ynyl)-3-

{(4,6S-dimethyl-cyclohex-3-enecarbonyl)-[4-(tetrahydro-fu ran-3S-yloxy)-trans- cyclohexyl]-amino}-thiophene-2-carboxy[ic acid (Isomer 1 ).

LC/MS = 528 (M + +1 )

Retention time: 5.65 mϊn

LC: Thermo Electron Surveyor HPLC

MS: Finnigan LCQ Advantage MAX Mass Spectrometer

Column: Phenomenex PoSar RP 30 mm X 4.6 mm

Solvents: (B): Acetonitrile with 0.1% formic acid, (A): Water withθ.1 % formic acid Gradient: 2mL/min, 0 min-0.5min 5% ACN, 0.5 min-6.5 min 5%-100% ACN, 6.5 min-9.0 min 100% ACN, 9.0 min-9.1 min 100%-5% ACN, 9.1 min-9.5 min 5% ACN. Example 303 - Compound 303: 5-(3,3-dimethyl-but-1-ynvn-3-{(4,6R-dimethyl- cvciohex-3-enecarbonyl)-f4-ftetrahvdro-furan-3S-vioxγ)-tran s-cyclohexvn-amino>- thiophene-2-carboxylic acid (Isomer 1 )

Compound 303

5-(3,3-dimethy!-but-1-ynyi)-3-{(4,6R-dimethyl-cyclohex-3- enecarbonyl)-[4- (tetrahydro-furan-3S-yioxy)-trans-cyciohexyl]-amino}-thtophe ne-2-carboxylic acid (Compound 303) was prepared in a similar fashion as 5-(3,3-dimethyl-but-1-ynyi)-3- {(4,6S-dimethyl-cyclohex-3-enecarbonyl)-[4-(tetrahydro-furan -3S-yloxy)-trans- cyclohexyl]-amino}-thiophene-2-carboxylic acid (Isomer 1 ) using Method A, except that 4,6f?-dimethy!-cyclohex-3-ene-1 R-carboxylic acid (prepared in a similar manner to 4,6S-dimethyi-cyclohex-3-ene-1 S-carboxyiic acid, using the chiral auxiliary AR- benzyl-oxazolidin-2-one) was used instead of 4,6S-dimethyi-cyciohex-3-ene-1 S- carboxyiic acid.

LC/MS = 528 (M + +1 )

Retention time: 5.64 min

LC: Thermo Electron Surveyor HPLC

MS: Finnigan LCQ Advantage MAX Mass Spectrometer

Column: Phenomenex Polar RP 30 mm X 4.6 mm

Solvents: (B): Acetonitrile with 0.1 % formic acid, (A): Water withθ.1 % formic acid Gradient: 2mL/min, 0 min-0.5min 5% ACN, 0.5 min-6.5 min 5%-100% ACN, 6.5 min-9.0 min 100% ACN, 9.0 min-9.1 min 100%-5% ACN, 9.1 min-9.5 min 5% ACN.

Example 304 -Compound 304: 5-(3 l 3-dimethyl-but-1-ynyl)-3-{(4 1 6f?-dimethyl- cyclohex-3-enecarbonyi)-[4-(tetrahydro-furan-3S-yioxy)-trans -cyciohexy[]-amino}- thiophene-2-carboxyiic acid

S-tS.S-Dimethyi-but-i -ynyO-S-^^R-dimethyl-cyclohex-S-enecarbonyl)-^- (tetrahydro-furan-3S-yloxy)-traπs-cyclohexyI]-amino}-thioph ene-2-carboxyltc acid (Compound 304) was prepared in a similar fashion as 5-(3,3-dimethyf-but-1-ynyS)-3- {(4,6S-dimethyl-cyclohex-3-enecarbonyl)-[4-(tetrahydro-furan -3S-yloxy)-trans- cyclohexyl]-amino}-thioρhene-2-carboxylic acid (isomer 1 ), except that 4,6R- dimethyl-cycfohex-3-ene-1R-carboxyiic acid was used instead of 4, 6S-di methyl- cyclohex-3-ene-1 S-carboxylic acid.

LC/MS = 528 (M + +1 )

Retention time: 5.31 min

LC: Thermo Electron Surveyor HPLC

MS: Finnigan LCQ Advantage MAX Mass Spectrometer

Column: Phenomenex Polar RP 30 mm X 4.6 mm

Solvents: (B): Acetonitrile with 0.1% formic acid, (A): Water withθ.1 % formic acid Gradient: 2FnLZmJn, 0 min-0.5min 5% ACN, 0.5 min-6.5 min 5%-100% ACN, 6.5 min-9.0 min 100% ACN, 9.0 min-9.1 min 100%-5% ACN, 9.1 min-9.5 min 5% ACN. Example 305 - Compound 305:5-(3.3-dimethvi-but-1-vnyl)-3-((4,βR-dimethyi- cyclohex-3-enecarbonvl)-[4-(tetrahvdro-furan-3R-yloxy)-trans -cvclohexyl " |-amino)- thiophene-2-carboxyiic acid (Isomer 1)

Compound 305

5-(3,3-Dimethyl-but-1-ynyl)-3-{(4,6R-dimethyl-cyclohex-3- enecarbonyi)-[4- (tetrahydro-furan-3R-yioxy)4rans-cyclohexyl]-amino}-thiophen e-2-carboxylic acid (Compound 305) was prepared in a similar fashion as 5-(3,3-dimethyl-but-1-ynyl)-3- {(4,6S-dimethyl-cycIohex-3-enecarbonyl)-[4-(tetrahydro-furan -3S-yIoxy)-trans- cyclohexy!]-amino}-thiophene-2-carboxyiic acid (isomer 1 ) except that 4-(tetrahydro- furan-3R-yioxy)-cyclohexanone was used instead of 4-(tetrahydro-furan-3S-yloxy)- cyclohexanone and 4,6R-dimethyl-cyciohex-3-ene-1 S-carboxyiic acid was used instead of 4,6S-dimethyl-cyclohex-3-ene-1 S-carboxylic acid.

LC/MS = 528 (M + +1 )

Retention time: 5.59 min

LC: Thermo Electron Surveyor HPLC

MS: Finnigan LCQ Advantage MAX Mass Spectrometer

Coiumn: Phenomenex Poiar RP 30 mm X 4.6 mm

Solvents: (B): Acetomtπle with 0.1 % formic acid, (A): Water with 0.1% formic acid Gradient: 2mL/min, 0 min-0.5min 5% ACN, 0.5 min-6.5 min 5%-100% ACN, 6.5 min-9.0 min 100% ACN, 9.0 min-9.1 min 100%-5% ACN 1 9.1 min-9.5 min 5% ACN. Example 306 - Compound 306: 5-(3,3-Dimethvi-but-1-vnyl)-3-f(4,6R-dimethvS- cyclohex-3-enecarbonyi)-r4-(tetrahvdro-furan-3f?-yloxy)-tran s-cyclohexyl1-amino)- thiophene-2-carboxyiic acid (Isomer 2)

Compound 306

5-(3,3-Dimethyl-but-1-ynyl)-3-{(4,6f?-dimethyl-cyclohex-3 -enecarbonyl)-[4- (tetrahydro-furan-3R-yloxy)-trans-cyclohexyl]-amino}-thiophe ne-2-carboxylic acid (Compound 306) was prepared in a similar fashion as 5-(3,3-dimethyl-but-1-ynyl)-3- {(4,6S-dimethyi-cyc!ohex-3-enecarbony!)-[4-(tetrahydro-furan -3S-yioxy)-trans- cyclohexyi]-amino}-thiophene-2-carboxy!ic acid (Isomer 1 ) except that 4-(tetrahydro- furan-3R-yioxy)-cyclohexanone was used instead of 4~(tetrahydro-furan-3S-yloxy)- cyciohexanone and 4,6R-dimethyl-cyclohex-3-ene-1 S-carboxylic acid was used instead of 4,6S-dimethyl-cyclohex-3-ene-1 S-carboxylic acid.

LC/MS = 528 (M * +1 )

Retention time: 5.66 min

LC: Thermo Electron Surveyor HPLC

MS: Finnigan LCQ Advantage MAX Mass Spectrometer

Column: Phenomenex Polar RP 30 mm X 4.6 mm

Solvents: (B): Acetonitriie with 0.1% formic acid, (A): Water with 0,1 % formic acid Gradient: 2mL/min, 0 min-0.5min 5% ACN, 0.5 min~6.5 min 5%-100% ACN, 6.5 min-9.0 min 100% ACN, 9.0 min-9.1 min 100%-5% ACN, 9.1 min-9.5 min 5% ACN.

Example 7 - Compound 307: 5-f3.3-Dimethyl-but-1-yπvπ-3-{f4,6S-dimethyl- cvciohex-3-enecarbonv!)-F4-(tetrahydro-furan-3ff-vioxy)-tran s-cyciohexyl1-amino}- thiophene-2-carboxylic acid (isomer 1 )

Compound 307

5-(3,3-Dimethyl-but-1-ynyl)-3-{(4,6S-dimethyl-cycfohex-3- enecarbonyl)-[4- (tetrahydro-furan-3R-ySoxy)-trans-cyclohexyl]~amino}-thiophe ne-2-carboxylic acid {Compound 307} was prepared in a similar fashion as 5-{3,3-dimethyl-but-1-ynyl)-3- {(4,6S-dimethyl-cyciohex-3-enecarbonyl)-[4-(tetrahydro-furan -3S-yloxy)-trans- cyclohexy!]-amino}-thiophene-2-carboxylic acid (isomer 1 ) except that 4-(tetrahydro- furan-3R-yloxy)-cyciohexanone was used instead of 4-(tetrahydro-furan-3S-yloxy)- cyclohexanone.

LC/MS = 528 (M" +1)

Retention time: 5.30 min

LC: Thermo Electron Surveyor HPLC MS: Finπigan LCQ Advantage MAX Mass Spectrometer

Column: Phenomenex Polar RP 30 mm X 4.6 mm

Solvents: (B): Acetonitrite with 0.1 % formic acid, (A): Water with 0.1 % formic acid Gradient: 2mL/min, 0 min-0.5min 5% ACN, 0.5 min-6.5 min 5%-100% ACN, 6,5 min-9.0 min 100% ACN, 9.0 min-9.1 min 100%-5% ACN, 9.1 min-9.5 min 5% ACN.

Exampie 308 - Compound 308: 5-f3.3-Dimethyi-faut-1-vnylV3-{f4,6S-dimethyi- cvclohex-3-enecarbonyl)-[4-(tetrarivdro-furan-3R-yloxy)-tran s-cyclohexyn-amino)- thioρhene-2-carboxySic acid (isomer 2)

Compound 308

5-(3,3-Dimethyi-but-1 -yny!)-3~{(4,6S-dimethyl-cyclohex-3-enecarbonyi)-[4- (tetrahydro-furan-3R-yloxy)-trans-cyclohexyl]-amino}-thiophe ne-2-carboxylic acid (Compound 308) was prepared in a similar fashion as 5-(3,3-dimethyl-but-1-yny!)-3- {(4,6S-dimethy(-cyclohex-3-enecarbonyl)-[4-(tetrahydro-furan -3S-yloxy)4rans- cyclohexyl]-amino}-thiophene-2-carboxylic acid (Isomer 1 ) except that 4-(tetrahydro- furan-3R-yioxy)-cyclohexanone was used instead of 4-(tetrahydro-furan-3S-yloxy)- cyclohexanone.

LC/MS = 528 (M + +1 )

Retention time: 5.21 min

LC: Thermo Electron Surveyor HPLC

MS: Finnigan LCQ Advantage MAX Mass Spectrometer

Column: Phenomenex Polar RP 30 mm X 4.6 mm

Soivents: (B): Acetonitrile with 0.1 % formic acid, (A): Water with 0.1 % formic acid Gradient: 2mL/min, 0 min-0.5min 5% ACN, 0.5 min-6.5 min 5%-100% ACN, 6.5 min-9.0 min 100% ACN, 9.0 min-9.1 min 100%-5% ACN, 9.1 min-9.5 min 5% ACN. Example 309 - Compound 309: 5-f3.3-dimethylbut-1-vnyl)-3-((1R2S,4f?V2-hvdroxy- 4-methyl-N-(4-(tetrahvdrofuran-35- yloxy)cvclohexyl)cvclohexanecarboxamido)thiQphene-2-carboxyl ic acid

Compound 309

Compound 309 was synthesized in a manner analogous to Example 301 , acylating with (1S,2R,5R)-2~(chlorocarbonyl)-5-methyicyciohexyl acetate. MS (m/z); 532.2 [M+H]; HPLC retention time: 4.530 min (2-98% acetonitrile:water with 0.05% trifiuoroacetic acid).

Example 310 - Compound 310: 5-f3,3-dimethylbut-1-vnvi)-3-((1R,2SΛff)-2-hydroxy- 4-methyi-N-(4-(tetrahvdrofuran-3f?- yloxy)cyclohexyl)cvclohexanecarboxamido)thiophene-2-carboxyi ic acid

Compound 310 Compound 310 was synthesized in a manner analogous to Example 303, acylating with (1 S,2R,5R)-2-(chlorocarbonyl)-5-methylcyclohexyl acetate. MS (m/z): 532.2 [M+H]; HPLC retention time: 4.530 min (2-98% acetonitrile:water with 0.05% trifiuoroacetic acid). Example 31 1 - Compound 311 : 5-r3,3-Dimethyl-4-(tetrahvdro-pyraπ-2-yloxy)-but-1- vnyl]-3-f(4-methyl-cyciohexanecarbqn^

thiophene-2-carboxylic acid

Scheme 4

Compound 311 To a solution of neopentyl glycol (6.O g, 57.6 mmo!) and 3,4-dihydro-2/-/-pyran

(2.64 mL, 28.94 mmo)) in 2:1 THF/dichloromethane (135 mL) was added p- TsOH-H 2 O (100 mg, 0.53 mmol) and the reaction stirred overnight Soiid NaHCO 3 was then added and after rapid stirring for 15 min the reaction was filtered and concentrated to give a pale liquid. Purification by flash column chromatography on silica gel using 50% ethyl acetate in hexanes provided 2,2-dimethyl-3-(tetrahydro- pyran-2-ySoxy)-propan-1 -ol (3.21 g, 59%) as a pale liquid. DMSO (2.69 mL, 50.97 mmol) was added dropwise to a soiution of oxaiyi chϊoride (2.87 mL, 33.98 mmol) in dicNoromethane (85.0 mL) at -78 0 C. After severai minutes, neat 2,2-dimethyl-3-(tetrahydro-pyran-2-yloxy)-propan-1-ol (3.2 g,

16.99mmoi) was added dropwise and after stirring for 1 h at -78 0 C, triethyiamine (9.5 mL, 68.0 mmol) was added. The reaction was allowed to warm to room temperature and stirred for 30 min. Sat. NH 4 CI was added and the organic layer was separated. The aqueous layer was extracted with ethyl acetate and washed with brine. The organic layers were combined, dried over Na 2 SO 4 and concentrated to give a paie liquid. The crude liquid was passed through a short silica gel column and efuted with 50% ethyl acetate in hexanes to provide 2,2-dimethyl-3-(tetrahydro-ρyran-2-yloxy)- propionaldehyde (3.09 g, 97%) as a pale liquid after removal of solvents under reduced pressure.

(1-Dtazo-2-oxo-propyl)-phosphonic acid dimethyl ester (486 mg, 2.54 mmol) was added to a stirred suspension of 2,2-dimethyi-3-(tetrahydro-pyran-2-yioxy)- propionaldehyde (315 mg, 1.69 mmol) and K 2 CO 3 (700 mg, 5.07 mmol) in methanol (20.0 mL) at room temperature. After overnight stirring, the reaction was diluted with diethyl ether, washed with sat. NaHCO 3 , brine, dried over Na 2 SO 4 and concentrated to provide a crude oil. Purification by flash column chromatography on siϋca gel using 20% ethyl ether in hexanes provided 2-(2,2-dimethyl-but-3-ynySoxy)-tetrahydro-pyran (147 mg, 48%) as a pale liquid.

A solution of 2-(2,2-dimethyi-but-3-yny!oxy)-tetrahydro-pyran (147 mg, 0.806 mmol), 3-[(4-hydroxy-cyclohexyl)-(4-methyl-cyclohexanecarbonyl)-ami no]-5-iodo- thiophene-2-carboxylic acid methyl ester (326 mg, 0.645 mmol) and triethyiamine (1.0 mL s 7.17 mmol) in DMF (1.0 mL) was degassed for 10 minutes with nitrogen. PdCI 2 (PPh 3 ) 2 (20 mg, 0.028 mmol) and CuI (5.0 mg, 0.026 mmol) was added and the resulting mixture was degassed with nitrogen for an additional 5 minutes after which the reaction was in a 60 0 C oil bath and heated for 4 h. The reaction was cooled and partitioned between ethyl acetate and sat. NaHCO 3 . The organic layer was separated, washed with 5% LiCl, brine, dried over Na 2 SO 4 and concentrated to give a dark orange-brown foam. Purification by flash column chromatography on silica gel using 20% hexanes in ethyl acetate provided the desired product (251 mg, 69%) as a pale yellow foam.

NaH (45 mg of a 60% oil dispersion, 1.12 mmol) was added in one portion to a solution of 5-[3,3-dimethyl-4-(tetrahydro-pyran-2-yloxy)-but-1-ynyl]-3-[ (4-hydroxy- cyclohexyl)-(4-methyl-cyclohexanecarbonyl)-amino]-thfophene- 2-carboxylic acid methyl ester (124 mg, 0.221 mmol) and 2-fluoropyridine (0.095 ml_, 1.1 mmo!) in dry DMF (2.0 mL). After several minutes, the reaction was placed in a 85 0 C oil bath and heated for 4 h. The reaction was cooled, water (0.50 mL) was added and the reaction mixture evaporated to dryness to provide a solid residue. The solid was washed with ethyl ether/hexanes then water, collected by centrifugation and washed with ethyl ether followed by water. The solid was collected and dried under vacuum to provide 5-[3 , 3-d i methyl-4-(tetra hyd ro-pyra n-2-yloxy )-but- 1 -ynyl]-3-{(4-methyl- cyclohexanecarbonyl)-[4-(pyridin-2-yloxy)-cyclohexyl]-amiπo }-thiophene-2-carboxylic acid (Compound 31 1 ) (60 mg, 42%) as an off-white solid. MS (m/z): 528.3 [M+H-(2- hydroxypyridine)3+; HPLC retention time: 4.61 min (2-98% acetonitrile:water with 0.05% trifluoroacetic acid).

Example 312 - Compound 312: 5-r3,3-Dimethyl-4-(tetrahvdro-furan-3-vioxy)-but-1- vnyπ-3-f(4-(pyridin-2-vioxy)-cvclohexy!)-(4-methyJ-cvclohex anecarbonvπ-aminol- thiophene-2-carboxyiic acid

Scheme 5

Compound 312

TMSC! (2.30 ml_, 0.018 mo!) was added dropwise to a solution of 3-hydroxy- 2,2-dimethyl-propionic acid methy! ester (2.0 g, 0.015 moi) and triethylamine (3.16 ml_, 0.023 mol) in dichloromethane at 0 0 C. After stirring for 1 h, the reaction was warmed to room temperature and stirred for an additional 1.5 h. Volatϋes were removed under reduced pressure to provide a solid that was slurried in a mixture of ethyi ether and hexanes. The soϋd was removed by filtration and the filtrate was concentrated to provide a yellow liquid that was purified by bulb to bulb distillation under reduced pressure to provide 2,2-dimethyi-3-trimethylsilany!oxy-propionic acid methyl ester (2.8 g, 91 %) as a colorless liquid.

2,2-Dimethyl-3-trimethylsilanyloxy-propionic acid methyl ester (478 mg, 2.34 mmo!) was added to a solution of dihydro-furan-3-one (168 mg, 1.95 mmol) and

FeCI 3 (16 mg, 0.097 mmol) in nitromethane (6.0 ml_) at 0 0 C. Triethylsilane (0.374 ml_, 2,33 mmol) was added and the reaction was warmed to room temperature and stirred for 2 h. Sat. NaHCO 3 was added and the reaction was extracted with dichtoromethane. The organic layer was separated, washed with brine, dried over Na 2 SO 4 and concentrated to give a paie liquid. Purification by flash column

chromatography on silica gel using 30% ethyl acetate in hexanes provided dimethyl- 3-(tetrahydro-furan-3-yloχy)-propionic acid methyl ester (353 mg, 75%) as a colorless liquid.

DiBAL (1.74 ml_ of a 1.0 M solution in hexanes) was added dropwise to a solution of 2,2-dimethyl-3-(tetrahydro-furan-3-yloxy)-propionic acid methyl ester (353 mg, 1.74 mmol) in dichloromethane (9.0 mL) at -78 0 C. After 2 h at this temperature, an additional equivalent of DIBAL was added and after 1 h the reaction was warmed to 0 0 C. After an additional 1 h at 0 0 C, 1.0 N HCI was added dropwise to quench the reaction. The reaction was warmed to room temperature and partitioned between dichloromethane and brine. The iayers were separated and the aqueous layer extracted with dichloromethane. The combined organic layers were dried over

Na 2 SO 4 and concentrated to give an oil. Purification by flash column chromatography on silica gel using 50% ethyl acetate in hexanes provided 2,2-dimethyl-3-(tetrahydro~ furan-3-yloxy)-propan-1-oi (250 mg, 82%) as a colorless oil.

DMSO (0.226 mL, 4.29 mmol) was added dropwise to a solution of oxalyS chloride (0.242 mL, 2.86 mmol) in dichloromethane (6.0 mL) at -78 0 C. After several minutes a solution of 2,2-dimethyl-3-(tetrahydro-furan-3-yloxy)-propan-1-oI (250 mg, 1.43 mmol) in dichioromethane (1.0 mL, with an additional 1.0 mL rinse) was added dropwise and after stirring for 1 h at -78 0 C 1 triethylamine (0.800 mL, 5.72 mmol) was added. The reaction was allowed to warm to room temperature and stirred for 30 min. Sat. NH 4 Ci was added and the reaction mixture was extracted with dichloromethane. The organic iayer was washed with brine, dried over Na 2 SO 4 and concentrated to give a yellow residue. The residue was passed through a short silica gel plug eiuting with 50% ethyl acetate in hexanes to provide 2,2-dimethy!-3-(tetrahydro-furan-3- yloxy)-propionaldehyde (245 mg, 99%) as a greenish-brown oil.

(1-Diazo-2-oxo-propyl)-phosphonic acid dimethyl ester (418 mg, 2.17 mmol) was added to a stirred suspension of 2,2-dimethyl-3-(tetrahydro-furan-3-yloxy)- propionaldehyde (250 mg, 1.45 mmol) and K 2 CO 3 (601 mg, 4.35mmol) in methanol (17.0 mL) at room temperature. After overnight stirring the bulk of the methanol was removed by rotary evaporation and the reaction mixture partitioned between ethyl ether and NaHCO 3 . The organic layer was separated, washed with brine, dried over Na 2 SO 4 and concentrated to provide a crude liquid. Purification by flash column chromatography on silica gel using 30% ethyl acetate in hexanes followed by bulb to bulb distillation under reduced pressure provided 3-(2,2-dimethyl-but-3-ynyioxy)- tetrahydro-furan (169 mg, 69%) as a colorless liquid.

A solution of 3-(2,2-dimethyl-but-3-ynyloxy)-tetrahydro-furan (169 mg, 1.00 mmol), 3-[(4-hydroxy-cyclohexyt)-(4-methyl-cyclohexanecarbonyl)-ami no]-5-iodo- thiophene-2-carboxylic acid methyl ester (404 mg, 0.80 mmol) and triethylamine (1.2 mL, 8.61 mmol) in DMF (1.2 ml.) was degassed for 10 minutes with nitrogen.

PdCl 2 (PPh 3 ) 2 (25 mg, 0.036 mmol) and CuI (6.0 mg, 0.031 mmol) was added and the resulting mixture was degassed with nitrogen for an additional 5 minutes after which the reaction was placed in a 65 0 C oil bath and heated for 2.5 h. The reaction was cooled and partitioned between ethyi acetate and sat. NaHCO 3 . The organic layer was separated, washed with 5% LiCI, brine, dried over Na 2 SO 4 and

concentrated to give a dark brown foam. Purification by flash column

chromatography on silica gel using 50% ethyl acetate in hexanes then 5% methanol in ethyl acetate provided 5-[3,3-dimethyl-4-(tetrahydro-furan-3-y!oxy)-but-1-ynyI]-3- [(4-hydroxy-cyclohexyl)-(4-methyl-cyclohexanecarbonyi)-amino ]-thiophene-2- carboxyiic acid methyl ester (331 mg, 75%) as a pale yellow foam.

Compound 312 may be prepared in the same manner as Compound 31 1 using 5-[3,3-dimethyl-4-(tetrahydro-furan-3-yloxy)-but-1-ynyl]-3-[ (4-hydroxy- cyclohexyl)-(4-methyl-cyclohexanecarbonyl)-amino]-thiophene- 2-carboxyfic acid methyl ester. Example 313 - Compound 313: 5-f3,3-dimethyl-4-(tetrahvdro-pyran-4-yloxyVbut-1- vnyl1-3-r(4-((pyridin-2-v[oxy)-cyclohexyl)-(4-methy[-cyclohe xanecarbonyi)-amino1- thiophene-2-carboxylic acid

Synthesis of 5-r3,3-dimethyi-4-(tetrahvdro-pyran-4-vioxy)-but-1 -vnyl1-3-[(4-ff hydroxy)- cvclohexyl)-(4-methyl-cycSohexanecarbonyl)-amino14hiophene-2 -carboxylic acid methyl ester

This thiophene ester (126 mg, 51%) was synthesized in a manner analogous to 5-[3,3-dimethy!-4-(tetrahydro-furan-3-yloxy)-but-1-ynyl]-3-[ (4-hydroxy-cyc!ohexyl)- (4-methyl-cyclohexanecarbony!)-amino]-thiophene-2-carboxySic acid methyl ester in Example 312, using tetrahydro-pyran-4-one in place of dihydro-furan-3-one.

Compound 313 may be prepared in the same manner as Compound 311 using 5-[3,3-dimethyl-4-(tetrahydro-pyran-4-yloxy)-but-1-ynyl]-3-[ (4-((hydroxy)- cyclohexy!)-(4-methyl-cyclohexanecarbonyl)-amino]-thiophene- 2-carboxylic acid methyl ester.

Example 314 - Compound 314: 5-(5-Hydroxy-3,3-dimethy[-pent-1-ynyl)-3-((4-methyl- cyclohexanecarbonyl)-[4-(tetrahvdro-furan-3-yloxγ)-cyclohex γl1-aminoHhiopheπe-2- carboxyiic acid

Scheme 6

Compound 314

To a solution of 3-hydroxytetrahydrofuran (5.0 ml_, 61.85 mmol) and tπethylamine (13.0 mL, 93.27 mmol) in dichloromethane cooied to 0 0 C was added TMSCI (9.38 mL, 74.17 mmol) dropwise. The reaction was allowed to warm to room temperature and then stirred overnight. Volatiles were removed under reduced pressure to provide a dark red solid that was filtered through a short column of silica gel eluting with ethyl ether. The filtrate was concentrated to afford an orange oii that was purified by bulb to bulb distillation under reduced pressure to provide 5-iodo-3- {(4-methyl-cyciohexanecarbonyl)-[4-(tetrahydro-furan-3-yloxy )-cycfohexyf]-amino}- thiophene-2-carboxyiic acid methyl ester (9.2 g, 93%) as a colorless liquid.

To a suspension of 5-iodo-3~[(4-methyl-cyclohexanecarbonyl)-(4-oxo- cyc!ohexyi)-amino]-thiophene-2-carboxy!ic acid methyl ester (2.5 g, 4.96 mmol) and FeCi 3 (41 mg, 0.252 mmol) in nitromethane (15.0 mL) cooled to 0 0 C was added neat trimethyl-(tetrahydro-furan-3-yloxy)-silane (955 mg, 5.95 mmo!) followed by the dropwise addition of triethylsilane (0.947 mL, 5.92 mmol). The reaction was allowed to slowly warm to room temperature and stirred overnight. Sat. NaHCO 3 was added and the reaction mixture was extracted with dichioromethane. The organic layer was washed with brine, dried over Na 2 SO 4 and concentrated to give a solid. The crude solid was heated in methanol, cooled and collected by filtration to provide 3-[(4- hydroxy~cyclohexyl)-(4-methyl-cyc!ohexanecarbonyl)-amino]-5- iodo-thiophene-2- carboxylic acid methyl ester (1.27 g, 44%) as a colorless solid.

A solution of 3-[(4-hydroxy-cyclohexyl)-(4-methyl-cyclohexanecarbonyl)~ amino]-5-iodo-thiophene-2-carboxylic acid methy! ester (303 mg, 0.53 mmol), 3,3- dtmethy!-pent-4-yn-1-ol (70 mg, 0.63 mmo!) and triethylamine (1.2 ml_, 8.61 mmol) in DMF (1.2 ml_) was degassed for 10 minutes with nitrogen. PdCi 2 (PPh 3 ) 2 (17 mg, 0.024 mmol) and CuI (5.0 mg, 0.026 mmol) was added and the resulting mixture was degassed with nitrogen for an additional 5 minutes after which the reaction was placed in a 65 0 C oil bath and heated for 1.5 h. The reaction was cooled and partitioned between ethyl acetate and sat. NaHCO 3 . The organic layer was separated, washed with 5% LiCI, brine, dried over Na 2 SO 4 and concentrated to give a dark foam. Purification by flash column chromatography on silica gel using 100% ethyl acetate provided 5-(5-hydroxy-3,3-dimethyl-pent-1-ynyl)-3-{(4-methyl-cyclohex anecarbonyl)- [4-(tetrahydro-furan-3-yloxy)-cyclohexyi]-amino}-thiophene-2 -carboxylic acid methyl ester (157 mg, 53%) as a pale yellow foam.

NaOH (0.500 mL of a 1.0 N aqueous solution) was added dropwise to a solution of 5-(5-hydroxy-3,3-dimethyl-pent-1 -ynyl)-3-{(4-methyl- cyc!ohexanecarbonyl)-[4-(tetrahydro-furan-3-yloxy)-cycIohexy !]-amino}4hiophene-2- carboxylic acid methyl ester (157 mg, 0.28 mmol) in methanol (1.5 mL) and THF (1.5 mL) at room temperature. The reaction was stirred until TLC indicated that all of the starting material was consumed. The reaction was evaporated to dryness to give a solid residue that was purified by column chromatography on reverse phase C 18 silica gel (100% water to 5% acetonitrile/water). Fractions containing product were pooled and evaporated to provide 5-(5-hydroxy-3,3-dimethyf-pent-1-ynyl)-3-{(4-methyl- cyclohexanecarbonyl)-[4-(tetrahydro-furan-3-yloxy)-cyclohexy l]-amino}-thiophene-2- carboxylic acid (Compound 314X52 mg, 33%) as a solid. MS (m/z): 546.0 [M+H]+; HPLC retention time 3.75 min (2-98% acetonitriie: water with 0.05% trifluoroacetic acid). Example 317 - Compound 317: 5-(3,3-Dimethv[~but-1-vnyl)-34f4-(4-ds-hvdroxy- tetrahvdro-furan-3-vSoxy)-cvclohexyl]-(4-methyl-cycJohexanec arbonyl)-amino]- thiophene-2-carboxylic acid

Scheme 8 H0 V\)

Compound 317

TMSCi (8.0 mL, 63.32 mmol) was added dropwise to a solution of 1 ,4- anhydroerythritol (3.0 g, 28.81 mmol), triethyiamine (9.2 mL, 66.0 mmol} and DMAP (few crystals) in dichloromethane (200 mL) at 0 0 C. The reaction was allowed to slowly warm to room temperature over 1 h and then stirred for an additional 3 h. The solid was filtered and the filtrate concentrated to give a solid. The solid was then filtered through a short silica gel column eiuting with 33% hexanes in ethyl ether. Removal of solvents under reduced pressure provided 3,4-bis-trimethylsiianyioxy- tetrahydro-furan (7.7 g, -100%) as a colorless liquid.

TMSOTf (0.020 mL, 0.109 mmol) was added to a solution of 3,4-bis- trimethylsiianyloxy-tetrahydro-furan (595 mg, 2.39 mmol) in dichloromethane (5.0 mL) cooled to -78 0 C. A solution of 5-(3,3-dimethyi-but-1-ynyl)-3-[(4-methyl- cyclohexanecarbonyl)-(4-oxo-cyc!ohexyi)-amino]-thiophene-2-c arboxyiic acid methyl ester (1.0 g, 2.18 mmol) in dichloromethane (7.0 mL) was then added dropwise and the reaction allowed to slowly warm to room temperature and then stirred overnight. Sat. NaHCO 3 was added and the reaction mixture was extracted with

dichloromethane. The organic layer was dried over Na 2 SO 4 and concentrated to give a blue-green soiid. The crude solid was triturated with ethyl ether and hexanes to give the desired keta! (525 mg, 44%) as an off-white solid.

TriethyJsilane (0.255 mL, 1.59 mmol) was added to a solution of the ketal from the previous reaction (332 mg, 0.61 mmol) in dichloromethane (3.0 mL) cooled to -78 0 C. TiCI 4 (1.34 mL of a 1.0 M solution in dichioromethane) was then added dropwise over 5 min. The reaction was stirred at -78 0 C 2 h and then warmed to room temperature. After stirring for 2 h, an additional equivalent of triethylsilane and TiCI 4 was added and stirring was continued until the reaction was deemed complete by TLC. Brine was added followed by the cautious dropwise addition of sat. NaHCO 3 . The reaction was diluted with water and extracted with dichloromethane. The organic layer was washed with brine, dried over Na 2 SO 4 and concentrated to give an orange oil. Purification by flash column chromatography on silica gel using 3% methanol in dichloromethane provided 5-{3,3-dimethyi-but-1 -ynyl)-3-[[4-(4-hydroxy-tetrahydro- furan-3-yloxy)-cyclohexyl]-(4-methyl-cydohexanecarbonyl)-ami no]-thiophene-2- carboxylic acid methyl ester 267 mg, 80%) as a colorless foam.

NaOH (0.25 mL of a 1.0 N aqueous solution) was added dropwise to a solution of 5-(3,3-dimethyl-but-1-ynyl)-3-[[4-(4-cis-hydroxy-tetrahydro- furan-3-yloxy)- cyclohexyiH4-methyl-cyclohexanecarbonyi)-amino]-thiophene-2~ carboxyiic acid methyl ester (80 mg, 0.147 mmoi) in methanol (1.0 mL) and THF (1.0 mL) at room temperature. After 2 h, an additional equivalent of NaOH was added and the reaction stirred until all of the starting material was consumed. The reaction was evaporated to dryness to give a residue that was purified by column chromatography on reverse phase C 16 silica gel (100% water to 20% acetonitrile/water). Fractions containing product were pooled and evaporated to provide 5-(3,3-dimethyi-but-1-ynyi)-3-[[4-(4- cis-hydroxy-tetrahydro-furan-3-yloxy)-cyclohexyl]-(4-methyi- cyciohexanecarbonyl)- amino]-thiophene-2-carboxyltc acid (Compound 317) (53 mg, 65%) as a glassy foam. MS (m/z): 532.2 [M+H]+; HPLC retention time 4.24 min (2-98% acetonitrile: water with 0.05% trifluoroacetic acid). Example 318 - Compound 318: 5~(3,3-Dimethγi-but-1-vnyS)-3-f[4-(44rans-hydroxy- tetrahvdro-furan-3-yloxy)-cvclohexyl1-(4-methv[-cvclohexanec arbonyl)-amino]- thiophene-2-carboxylic acid

Scheme 9

Compound 318

To a solution of 5-(3,3~dimethyl-but-1-ynyl)-3-[[4-(4-cis-hydroxy-tetrahydro- furan-3-yloxy)-cyclohexyi]-(4-methyI-cyciohexanecarbonyl)-am ino]-thiophene-2- carboxylic acid methyl ester (267 mg, 0.49 mmoi) in dry THF (9.0 mL) cooled to 0 0 C was added, sequentially, triphenylphosphine (462 mg, 1.76 mrnol) and DIAD (0.29 mL, 1.47 mmol). After 5 min, solid 4-nitrobenzoic acid (295 mg, 1.76 mmol) was added and after stirring for 1 h the reaction was allowed to warm to room temperature. After 3 h the reaction was concentrated to dryness and partitioned between ethyl acetate, sat. NaHCO 3 and water. The layers were separated and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with water, 1.0 N HCl, sat NaHCO 3 , brine, dried over Na 2 SO 4 and concentrated to give a viscous oil. Purification by flash column chromatography on silica gel using 3% methanol in dichloromethane provided 5-(3,3-dimethyl-but-1- ynyl)-3-((4-methyl-cyclohexanecarbonyl)~{4-trans-[4-(4-nitro -benzoyloxy)-tetrahydro- furan-3-y!oxy]-cyclohexyl}-amino)-thioρhene-2-carboxylic acid methyl ester (328 mg, 96%) as a paie yellow foam. Analysis by 1 H NMR indicated a minor impurity due to DIAD.

Solid K 2 CO 3 (100 mg, 0.72 mmol) was added to a solution of 5-(3,3-dimethyl- but-1-ynyl)-3-((4-methyl~cyclohexanecarbonyl)-{4-trans-[4-(4 -nitro-benzoyioxy)- tetrahydro-furan-3-yIoxy]-cyclohexy!}-amino)-thiophene-2-car boxylic acid methyl ester (320 mg, 0.46 mmol) in methanol (3.0 ml_) at room temperature. After 1 h, the reaction was evaporated to dryness and then partitioned between ethyl ether and water. The organic layer was washed with sat. NaHCO 3 , brine, dried over Na 2 SO 4 and concentrated to give a waxy solid. Purification by flash column chromatography on silica gel using 3% methanol in dichloromethane provided 5-(3,3-dimethyl-but-1- yny!)-3-[[4-(4-trans-hydroxy-tetrahydro-furan-3-yloxy)-cyclo hexyl]-(4-methyl- cycloheχanecarbony!)-amino]-thiophene-2-carboxylic acid methyl ester (191 mg, 76%) as a pale foam.

NaOH (0.22 ml_ of a 1.0 N aqueous solution) was added to a solution of 5- (3,3-dimethyl-but-1-ynyl)-3-[[4-(4-hydroxy-tetrahydro-furan- 3-yloxy)-cyclohexyl]-(4- methy!-cyclohexanecarbonyl)-amino]-thiophene-2-carboxyiic acid methyl ester (80 mg, 0.147 mmol) in THF (1.0 mL) and methanol (1 .0 mL) at room temperature. After stirring for 48 h, the reaction was evaporated to dryness to give a residue that was purified by column chromatography on reverse phase Ci 8 silica gel (100% water to 10% acetonitrile/water). Fractions containing product were pooled and evaporated to provide 5-(3,3-dimethyl-but-1 -ynyl)-3-[[4-(4-trans-hydroxy-tetrahydro-furan-3-y!oxy)- cyclohexyl]-(4-methyl-cyclohexanecarbonyl)-amino]-thiophene- 2-carboxylic acid (Compound 318) (53 mg, 43%) as a colorless solid. MS (m/z): 532.2 [M+HJ+; HPLC retention time 4.17 min (2-98% acetonitrile: water with 0.05% trifluoroacetic acid). Example 319 - Compound 319: 5-[3,3-Dimethvi-5-ftetrahγdro-pyran-4-vJoxy)-pent-1- ynyll-3-r(4-(pyridin-2-yloxy)-cvciohexy[)-(4-methyl-cvctohex anecarbonyl)-amino]- thiophene-2-carboxyJic acid

Scheme 10

Compound 319

A solution of the 3,3-dimethyl-pent-4-yr>1-ol (600 mg, 5.36 mmoi), 3-[[4-(tert- Butyl-dimethy!-silanyloxy)-cyclohexyl]-(4-methyl-cyclohexane carbony!)-amino]-5- iodo-thiophene-2-carboxy!ic acid methy! ester (2767 mg, 4.46 mmol) and

triethy!amine (10.8 mL, 77.48 mmol) in DMF (25ml_) was degassed for 10 minutes with nitrogen. PdCI 2 (PPh 3 ) 2 (314 mg, 0.45 mmoi) and Cu! (170 mg, 0.90 mmo!) was added and the resulting mixture was degassed with nitrogen for an additional 5 minutes after which the reaction was placed in a 80 0 C oil bath and heated for 3.5 h. The reaction was cooled and partitioned between ethyl acetate and sat. NaHCO 3 . The organic iayer was separated, washed with 5% LiCI, brine, dried over Na 2 SO 4 and concentrated to give a dark brown foam. Purification by flash column

chromatography on silica gel using 30% ethyl acetate in hexanes then 5% methanol in ethyl acetate provided 3~[[4-(tert-butyl-dimethyl-silanyioxy)-cyciohexyl]-(4~methyl - cyclohexanecarbonyl)-amino]-5-(5-hydroxy-3 > 3-dimethyl-pent-1-ynyl)- thiophene-2- carboxylic acid methyl ester (2.43g, 90%) as a pale yellow foam.

TMSCI (0.08 mL, 0.596 rnmol) was added dropwise to a solution of 3-[[4-(tert- butyl-dimethyl-silany!oxy)-cyclohexyl]-(4-methyl-cyclohexane carbonyl)-amino]-5-(5- hydroxy-3,3-dimethy!-pent-1-ynyl)- thiophene-2-carboxyiic acid methyl ester (300 mg, 0.497m mol) and triethylamine (0.11mL, 0.745 mmoi) in dichloromethane at 0 0 C.

After stirring for 1 h, the reaction was warmed to room temperature and stirred for an additional 1.5 h. The solution was washed with water, brine, dried and concentrated to give crude 3-[[4-(tert-butyl-dimethyl-silanyloxy)-cyciohexyl]~(4-methyi - cyclohexanecarbonylJ-aminoJ-δ^S^-dimethyl-δ-trirπethylsii anyloxy-pent-i-ynyl)- thiophene-2-carboxylic acid methyl ester (330 mg) which was carried to next step without further purification.

3-[[4-(tert-Butyl-dimethyl-silanyioxy)-cyclohexyl]-(4-methy! - cyclohexanecarbonyO-aminol-δ^S.S-dimethyl-δ-trimethylsiian yloxy-pent-i-ynyl)- thiophene-2-carboxylic acid methyl ester (230 mg, 0.34 mmol) was added to a solution of tetrahydro-pyran-4-one (29 mg, 0.29 mmol} and FeCi 3 (2.8 mg, 0.017 mmol) in nitromethane (5.0 mL) at 0 0 C. Triethylsiiane (0.054 mL, 0.34 mmol) was added and the reaction warmed to room temperature and stirred for 2 h. Sat.

NaHCO 3 was added and the reaction extracted with dichloromethane. The organic layer was separated, washed with brine, dried over Na 2 SO 4 and concentrated to give the crude product. Purification by HPLC with CH 3 CN(0.1 % TFA)/H 2 O(0.1 % TFA) provided 5-[3,3-dimethyl-5-(tetrahydro-pyran-4-yloxy)-pent-1-ynyl]-3- [(4-hydroxy- cyclohexyl)-(4-methyl-cyclohexanecarbonyi)-amino]-thiophene- 2-carboxylic acid methyl ester (8 mg, 4% ) as solid.

Compound 319 may be prepared in the same manner as Compound 31 1 using 5-[3,3-dimethyl-5-(tetrahydro-pyran-4-yloxy)~pent-1 ~ynyl]~3-[(4-hydroxy- cyclohexyl)-(4-methyl-cyclohexanecarbonyl)-amino]-thiophene- 2-carboxylic acid methyl ester. Exampie 321 : Compound 321 : 5-f3>Dimethyl-but-1-vnvO-3-f(4-methvi- cvciohexanecarbonyi)-{1 -F2-(pyriciin-2-yloxy)-ethyn-pyrrolidin-3-yl)-amino)-thiophe ne- 2-carboxylic acid

Compound 321

To a solution of 5-(3,3-dimethyl-but-1-ynyl)-3-[(4-methyl- cyclohexanecarbonyl)-pyrrolidin-3S-yl-amino]-thiophene-2-car boxylic acid methyl ester (0.1 g, 0.214 mmol) and acetic acid (37 HL) in dry dichloromethane (1 mL) was added NaBH(OAc) 3 (0.092 g, 0.428 mmol) at room temperature. After stirring at room temperature for 90 min, the reaction was concentrated in vacuo to remove volatiles. The crude oil was dissolved in methanol (2 mL), treated with HCi (1.7 mL, 1 M in methanol) and heated to 60 0 C for 1h. Upon cooling the reaction was concentrated, diluted with ethyl acetate, and neutraiized with NaHCO 3 . The organic layer was washed with saturated NaCi solution (30 mL), dried over Na 2 SO 4 , filtered, concentrated and purified by silica gel column chromatography. A solution of 5-(3,3- DimethyI-but-1-yny!)-3-[[1-(2-hydroxy-ethyl)-pyrrolidin-3-yl ]-(4-methyl- cyclohexanecarbonyl)-amino]-thiophene-2-carboxylic acid methyl ester {Compound 321 ) (0.083 g, 0.175 mmol) in THF (I mL) was treated with KHMDS (0.42 mL, 0.2 mmol, 0.5 M in toluene) at -78 0 C. After 30 min 2-fluro-pyridine (30 r L 1 0.35 mmoi) was added. The reaction was quenched after 1h with saturated NH 4 C! and the organic phase was collected and concentrated. The crude material was purified by reverse-phase HPLC to afford the title compound. MS (m/z): 538.2 [M+H] " ; HPLC retention time: 3.55 min (2-98% acetonitri!e:water with 0.05% trifiuoroacetic acid) 6 min run.

ExampSe 322 - Compound 322: 5-(3,3-Dimethyl-but-1-ynyl)-3-((4-methyl- cyciohexanecarbonyl)-[4-(4-methyl-cyclohexanecarbonyloxγ) : 4 [tet[ahvdro-furan-3" yloxymethy)-cyclohexyπ-aminoHhiophene-2-carboxylic acid

Compound 322

4-Methyi-cyclohexanecarboxylic acid (57 mg, 0.4 mmo!) was dissolved in CH 2 CI 2 (5 mL) and DMF (2 μl_) was added. The solution was cooled to 0 °C and then (COCI) 2 (175 μL, 2 mmoi) was slowly added to the solution. The reaction was stirred in the ice bath for 1 hour and then concentrated. The residue was taken up in hexanes and concentrated; this hexanes co-evaporation was repeated once more. To the residue was added 5-(3,3-dimethyl-but-1-yny!)-3-[4~(tetrahydro-furan-3R- yloxy)-trans-cyclohexylamino]-thiophene-2-carboxy!ic acid methy! ester (84 mg, 0.2 mmol) and pyridine (3m L). The solution was heated to 85 0 C overnight. The reaction was cooled to rt, concentrated and taken up in THF/ MeOH (5:1 ). The solution was then treated with LiOH (1 mmol, 41 mg) and heated to 65 0 C for 1 h. Volatiles were removed under vacuum and the resulting residue purified by HPLC with CH 3 CN (0.1% TFA)/H 2 O(0.1% TFA) to provide Compound 322 as colorless solid. MS (m/z): 516.3 [M+H]+; HPLC retention time 8.53 min (2-98% acetonitriie: water with 0.05% trifluoroacetic acid) 30 min run.

Example 325 - Compound 325: 3-Ff4-(4-dimethylaminomethyl-pyridin-2-yloxy)- cyclohexyl1-(4-methviene-cyclohexanecarbonyl)-amino]-5-(3,3- dimethyl-but-1-vnyl)- thiophene-2-carboxylic acid

Scheme 13

Compound 325 Compound 325 was synthesized in a manner analogous to Example 31 1 :MS (m/z): 579.0 [M+H]+; HPLC retention time 3.4S min (2-98% acetonitriie: water with 0.05% tπ ' fluoroacetic acid).

Example 326 - Compound 326: 5-f3,3-Dimethyi-but-1-vnvi)-3-(f4-methyl- cvciohexanecarbonvO-C^Θ-oxo-i.δ-dihvdro-pyrimidin^-yloxyVc vclohexyii-amino}- thiophene-2-carboxyiic acid

Compound 326

Scheme 14

2,4-Dichloro-pyrimidine (2.00 g, 13.4 mmoi) in THF (30 mL) was treated with sodium hydride (670 mg, 60% oi! dispersion, 16,8 mmof) followed by 2- trimethylsilanyl-ethanol (2.01 mL, 14.1 mmol) in portions over 15 minutes. The reaction mixture was stirred for 16 hours and treated with 1 M HCI (20 mL). Water (20 mL) was added and the mixture was extracted with 1 :1 ethyl acetate:hexanes (100 mL) followed by ethyl acetate (100 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated. Purification by flash

chromatography (10-30% EtOAc: hexanes, 80 g column), afforded 2.45 g (79% yield) of 2-chtoro-4-(2-trimethylsiianyl-ethoxy)-pyrimidine as a mixture of regioisomers.

2-Chloro-4-(2-trimethyisilanyl-ethoxy)-pyrimidine (388 mg, 1.68 mmol) and 5- (3,3-dimethyl-but-1 -ynyi)-3-[(4-hydroxy-cyclohexyI)-(4-methyl-cyclohexanecarbon yl)- amino]-thiophene-2-carboxylic acid (150 mg, 0.337 mmol) were dissolved in DMF (2 mL) and treated with sodium hydride (67 mg, 60% oii dispersion, 1.68 mmol). The mixture was stirred under nitrogen until the bubbling slowed, then was sealed and heated by microwave (90 0 C, 65 min). An additional portion of 2-chioro-4-(2- trimethylsilanyl-ethoxy)-pyrimidine (388 mg, 1.68 mmol) and sodium hydride (67 mg, 60% oil dispersion, 1.68 mrnol) were added and the mixture was stirred under nitrogen unti! the bubbling slowed. The reaction mixture was sealed and heated by microwave (100 0 C, 45 min). After cooling, the mixture was diluted with ethyl acetate (about 5 ml_) and quenched with 10% citric acid (about 2 mL). Water was added and the mixture was extracted twice with

ethyl acetate. After concentration the resulting residue was purified by HPLC with CH 3 CN (0.1% TFA)/H 2 O(0.1 % TFA) to afford 39 mg of 5-(3,3-dimethyl-but-1-ynyl)-3- ((4-methyl-cyclohexanecarbonyl)-{4-[4-(2-trimethylsilanyi-et hoxy)-pyrimidin-2-yloxy]- cyclohexy!}-amino)-thiophene-2-carboxyiic acid.

5-(3,3-Dimethyl-buH-ynyl)-3-((4-methyi-cyclohexanecarbonyl)- {4-[4-(2- trimethyisilanyl-ethoxy)-pyrimidiπ-2-yloxy]-cyclohexyI}~ami no)-thiophene-2-carboxyiic acid (37 mg, 0.058 mmoi) in THF (1 mL) was treated with tetrabutyiammonium fluoride (0.087 mL, 1 M in THF, 0.087 mmol) and stirred at ambient temperature for 1.5 h. An additional portion of tetrabutyiammonium fluoride (0.290 mL) was added and the reaction mixture was stirred at ambient temperature for 3 h. After concentration the residue was purified by HPLC with CH 3 CN (0.1% TFA)/H 2 O(0.1% TFA) to afford 18 mg (58% yield) of Compound 326: MS (m/z): 537.9 [M+H]-; HPLC retention time 4.28 min (2-98% acetonitrile: water with 0.05% trifluoroacetic acid).

Scheme 15

Acrylic acid 4,4-dimethyt-2-oxo-tetrahydro-furan-3-yl ester (R) (2.92 g, 15.9 mmof) in dichloromethane (20 ml_) and hexanes (3 mL) was cooled to -10 °C and treated with titanium tetrachloride (2.4 mL, 2.4 M in dichioromethane, 2.4 mmol). The red solution was stirred for 15 min and treated with isoprene (2.4 mL, 23.8 mmol) dropwise over 5 min. After stirring for 1.5 h, an additional portion of isoprene (2.4 mL, 23.8 mmol) was added and the reaction mixture was stirred at -10 to 0 0 C for 2.5 h. After cooiing to -10 0 C, the reaction mixture was quenched with ammonium chioride (sat. aq.). Water and ethyl acetate: hexanes (1 :1 ) were added. The organic layer was separated and the aqueous layer was extracted again with ethy!

acetate:hexanes (1 :1 ). The combined organic layers were dried over sodium sulfate, filtered and concentrated. The residue was purified by flash chromatography (10- 40% EtOAc:Hex, 80 g column) to afford 3.35 g (84% yield) of 4-methyl-cycSohex-3- (S)-enecarboxyiic acid 4,4-dimethyl-2-oxo-tetrahydro-furan-3-yl ester as a clear oil.

4-MethyI-cyclohex-3-(S)-enecarboxylic acid 4,4-dimethyl-2-oxo~tetrahydro- furan-3-yl ester (3.34 g, 13.2 mmol) in THF (25 mL), water (2.5 mL) and methanol (2.5 mL) was treated with lithium hydroxide monohydrate (2.8 g, 66.2 mmol) and warmed to 50 0 C with stirring. After 1 h, the reaction mixture treated with 1M HCi (about 25 mL). The mixture was extracted with hexanes:ethyl acetate (200 mL: 15 mL), dried over sodium sulfate, filtered and concentrated to 2.4 g of a white semi- solid. The residue was redissolved in hexanes:dichioromethane (100 mL, 95:5), washed with water, dried over sodium sulfate, filtered and concentrated to 1.68 g (91% yield) of (1S)-4-methyl-cyc!ohex-3-enecarboxylic acid as a white powder.

(1 S)-4-Methyt-cyclohex-3-enecarboχy!ic acid (209 mg, 1.5 mmoi),

azeotropically dried by evaporation from toluene, was treated with potassium phosphate tribasic (383 mg, 1.8 mmol), suspended in dichloromethane (4 mL) and treated with dimethylformamide (2 drops). The reaction mixture was cooled to 0 0 C and treated dropwise with oxaiyl chloride (0.3 mL s 3.2 mmol). The reaction mixture was allowed to warm to ambient temperature while stirring for 2 h. After filtering the solids, the solution was concentrated, treated with hexanes and concentrated again to afford 4-methyl-cyciohex-3-enecarbonyl chloride (S) as a light yellow oil which was used immediately in the next step.

Example 327: Compound 331 : 5-(3.3-Dimethyl-but-1-vnviV3-fftrans-4-methyl- cvciohβxanecarbonyl)-[4-(tetrahvdro-furan-3fS)-yloxy)-trans : cyciohexγl]-amino}- thiophene-2-carboxylic acid

Compound 332: 5-f3,3-Dimethv!-but-1-vnvh-3-(ftrans-4-methyl- cvciohexanecarbonyl)-f4-(tetrahvdro-furan-3(S)-yloxy)-cis-cv clohexyl1-amino}- thiophene-2-carboxylic acid

Compound 331 Compound 332

Scheme 17

A mixture of 3-amino-5-(3,3-dimethyl-but-1-yny!)-thiophene-2-carboxyHc acid methyt ester (2.41 g, 10.2 mmoϊ), 4-(tetrahydro-furan-3S-yloxy)-cyclohexanone (1.6 g, 8.5 mmol), TFA (2.6 ml_, 34 mmol) and triethylsilane (2.7 ml, 17 mmol) in CH 2 CI 2 (12 mL) was stirred for 22 h at room temperature. The reaction was concentrated in vacuo and the crude oii was dissolved in toluene (50 mL) and concentrated (repeat) to give a yellow solid. The crude solid was purified by column chromatography (10- 25% ethy! acetate/hexanes) to give 5-(3,3-dimethyl-but-1-ynyl)-3-[4-(tetrahydro-furan- 3S-yloxy)-cyclohexylamino]-thiophene-2-carboxy!ic acid methyl ester (2.28 g, 5.63 mmol) as a mixture of isomers.

S-fS^-dimethyl-but-i-ynyO-S-^tetrahydro-furan-SS-yloxyJ-cycl ohexylamino]- thiophene-2-carboxylic acid methyl ester (105 mg, 0.26 mmol, 6:4 mixture of trans:cis isomers) was dissolved in pyridine (1 mL) and trans-4-methylcyc!ohexanecarbonyl chloride (160 mg, 1.0 mmol) was added. The solution was stirred for 18 h at

90 0 C. 0 C The reaction was cooled to ambient temperature, partitioned between ethyl acetate and water, and the aqueous phase thrice extracted with ethyl acetate. The combined organic layers were sequentially washed with saturated aqueous ammonium chloride, brine, dried over magnesium sulfate, filtered, concentrated, and purified by column chromatography (0-70% ethyl acetate/hexanes) to give 5-(3,3- Dimethyl~but-1-yny!)-3-{(trans-4-methyl-cyciohexanecarbonyl) -[4-(tetrahydro-furan- 3S-yloxy)-cyclohexyl]-amino}-thiophene-2-carboxylSc acid methyl ester (90 mg, 0.17 mmol) as a mixture of isomers.

S-^S-Dimethyl-but-i-ynyO-S-fttrans^-methyl-cyclohexanecarbon yl)-^-

(tetrahydro-furan-3S-yloxy)-cycSohexyi]-amino}-thiophene- 2-carboxylic acid methyl ester (90 mg, 0.17 mmol) was dissolved in THF (1.5 mL), MeOH (0.5 mL), and H 2 O (0.5 mL). To this solution was added LiOH H 2 O (74 mg, 1.8 mmol). The reaction was stirred at 45 0 C for 1 h and then acidified with 10% HCI (aq} . The reaction solution was thrice extracted to ethyl acetate, dried over magnesium sulfate, filtered, concentrated, and purified by reverse phase HPLC to give 5-(3,3-Dimethyl-bui-1-ynyl)-3-{(trans-4- metbyl-cyclohexanecarbonyl)-[4-(tetrahydro-furan-3(S)-yloxy) -trans-cyclohexyi]- amino}-thiophene-2-carboxylic acid (Compound 331 )(39 mg, 0.08 mmol): MS (m/z): 516.0 [M+H]+; HPLC retention time 4.64 min (2-98% acetonitriie: water with 0.05% trifluoroacetic acid), and 5-(3,3-Dimethyl-but-1-yny!)-3-{(trans-4-rnethyl- cyclohexanecarbonyl)-[4-(tetrahydro-furan-3(S)-yloxy)-cis-cy clohexyl]-amino}- thioρhene-2-carboxylic acid (Compound 332)(9 mg, 0.02 mmol): MS (m/z): 515.9 [M+H]+; HPLC retention time 4.70 min (2-98% acetonitriie: water with 0.05% trifluoroacetic acid). Example 333 - Compound 333- 5-f3,3-Dιmethyl-but-1-ynvi)-3-ff 1 SV4-methvi- cvciohex-3-enecarbonvπ-[4-(tetrahvdro-furan-3fS)-yioxy)-tra ns-cyclohexyH-amιno}- thιophene-2-carboxylιc acid

Compound 333

Scheme 18

A mixture of S-amino-S^S^-dimethyi-but-i-ynylJ-thiopheπe^-carboxylic acid methyl ester (2 41 g, 10 2 mmoi), 4-{ietrahydro-furan-3(S)-yloxy)-cyc!ohexanone (1.6 g, 8 5 mmol), TFA (2 6 ml_, 34 mmo!) and tπethylsilane (2.7 ml_, 17 mmol) in CH 2 CI 2 (12 ml_) was stirred for 22 h at room temperature The reaction was concentrated in vacuo and the crude oι! was dissolved in toluene (50 ml_) and concentrated (repeat) to give a yellow solid The crude solid was purified by column chromatography (10- 25% ethyl acetate/hexanes) to give 5-(3,3-dιmethyl-but-1-ynyl)-3-[4-(tetrahydro-furan- 3-yloxy)-trans-cyclohexylamιno]-thιophene-2-carboxyhc acid methyl ester (1 09 g, 2 69 mmol) as a single isomer (ISH-Methyl-cyciohex-S-ene-i-carboxylic acid (130mg, 0.93mmol) was dissolved in CH 2 CI 2 (2mL). K 3 PO 4 (400mg, 1.9mmol) and DMF (20μL) were added. The solution was cooled to 0 0 C and then (COCI) 2 (200μL, 2.1 mmol) was siowly added to the solution. The reaction was warmed to rt and stirred for 2 hours. The reaction solution was decanted from insoluble K 3 PO 4 and concentrated. The residue was taken up in hexanes and concentrated; this hexanes coevaporation was repeated once more. To the residue was added -(3,3-dimethyl-but-1-ynyl)-3-[4- (tetrahydro-furan-3S-yloxy)-trans-cyclohexy!amino]-thiophene -2-carboxylic acid methyl ester (100mg, 0.25mmol), K 3 PO 4 (200mg, 0.98mmol), DCE (1.2mL). The solution was heated to 90 °C overnight. The reaction was cooled to rt, partitioned between ethyl acetate and water, and the aqueous phase thrice extracted with ethyl acetate. The combined organic layers were sequentially washed with saturated aqueous ammonium chloride, brine, dried over magnesium sulfate, filtered, concentrated, and purified by column chromatography (0-70% ethyl acetate/hexanes) to give 5-(3,3-Dimethyl-but-1-ynyl)-3-{(1 S)-4-methyl-cyciohex-3-enecarbonyl)-[4- (tetrahydro-furaπ-3S-yloxy)-trans-cyclohexyl]-amino}-thioph ene-2-carboxylic acid methyl ester (24 mg, 0.05 mmol).

S-fS^-Dimethyl-but-i-ynyO-S-^ISH-methyl-cyclohex-S-enecarbon yl)-^- (tetrahydro-furan-3S-yloxy)-trans-cyclohexyl]-amino}-thiophe ne-2-carboxylic acid methyl ester (19 mg, 0.04 mmol) was dissolved in THF (0.6 ml_), MeOH (0.2 mL), and H 2 O (0.2 mL). To this solution was added LiOH H 2 O (20 mg, 0.5 mmol). The reaction was stirred at rt for 1h, diluted with water, and acidified with 10% HCI (aq) . The reaction solution was thrice extracted to ethyl acetate, dried over magnesium sulfate, filtered, concentrated, and purified by reverse phase HPLC to give 5-(3,3-Dimethyl- but-1-ynyl)-3-{(1 S)-4-methyl-cyclohex-3-enecarbonyl)-[4-(tetrahydro-furan-3S- yloxy)- trans-cyclohexyl]-amino}-thiophene-2-carboxySic acid (Compound 333) (11 mg, 0.02 mmol): MS (m/z): 514.0 [M+HJ+; HPLC retention time 4.53 min (2-98% acetonitrile: water with 0.05% trifluoroacetic acid).

Example 334- Compound 334: 5-(3.3-Dimethyl-but-1-vnyl)-3-{f1 SV4-methyl- cvclohex-3-enecarbonylH4-ftetrahvdro-furan-3(ffl-yloxy)-tran s-cyclohexyll-arnino)- thiophene-2-carboxyiic acid

Compound 334

Compound 334 was prepared in a manner similar to Example 333 using A- (tetrahydro-furan-3(f?)-yloxy)-cyciohexanone in place of 4-(tetrahydro-furan-3(S)- yloxy)-cyclohexanone: MS (m/z): 516.0 [M+H]+; HPLC retention time 4.64 min (2- 98% acetonitrile: water with 0.05% triftuoroacetic acid): MS (m/z): 514.1 [IVH-H]+; HPLC retention time 4.53 min (2-98% acetonitrile: water with 0.05% trifiuoroacetic acid).

Example 335- Compound 335: S-O^-Dimethyi-but-i-ynyD-S-KI RH-methyi- cvclohex-3-enecarboπvi)-f4-ftetrahvdro-furan-3ff?Vyioxy)-tr ans-cvciohexyl1-aminoV- thiophene-2-carboxylic acid

Compound 335

Compound 335 was prepared in a manner similar to Example 333 using 4- (tetrahydro-furan-3(R)-yloxy)-cyclohexanone in place of 4-(tetrahydro-furan-3(S)- y!oxy)-cyclohexanone and (1f?)-4-methyi-cyciohex-3-ene-1-carboxyϊic acid in place of (1S)-4-methyl-cycSohex-3-ene-1-carboxylic acid: MS (m/z): 514.1 [M+H]+; HPLC retention time 4.52 min (2-98% acetonitrile: water with 0.05% trifiuoroacetic acid).

Example 336- Compound 336: 5-(3,3-Dimethyl-but-1-vnylV3-{f1 f?V4-methyl- cyclohex-3-enecarbonyl)-| ' 4-ftetrahvdro-furan-3(S)-vioχv)-trans-cvcioheχyn-ami no)- thiophene-2-carboxylic acid

Compound 336

Compound 336 was prepared in a manner similar to Example 333 using (1R)- 4-methyl-cyciohex-3-ene-1-carboxylic acid in place of (1 S)-4-methyl-cyclohex-3-ene- 1-carboxylic acid: MS (m/z): 514.0 [M+H]+; HPLC retention time 4.53 min (2-98% acetonitrϋe: water with 0.05% trifluoroacetic acid).

Example 337 - Compound 337: 3-[[4-(4-Dimethylaminomethyl-pyridin-2-ySoxy)- trans- cyciohexyπ-(4-methyl-cyciohex-3-enecarbonyl)-amino1-5-f3,3- dimethyi-but-1-vnyl)- thJophene-2-carboxylic acid

Compound 337

Scheme 19

(I SH-Methyl-cyclohex-S-ene-i-carboxylic acid (250 mg, 1.78 mmol), azeotropicaily dried by evaporation from toluene was dissolved in dichioromethane (4 ml_) and treated with dimethylformamide (1 drop). The reaction mixture was cooled to O 0 C and treated dropwise with oxalyl chloride (0.42 mL, 4.5 mmol). The reaction mixture was allowed to warm to ambient temperature while stirring for 4 h. The solution was concentrated, treated with hexanes and concentrated again to afford (1 S)-4-methyl-cyc!ohex-3-ene-1-carboxyiic acid chloride as a light yellow oil which was used immediately in the next step.

(1 S)-4-Methyl-cyclohex-3-ene-1-carboxylic acid chloride {1.8 mmol), 5-(3,3- dimethyl-but-1 -ynyi)-3-( 1 ^-dioxa-spiro^.Sldec-δ-ylaminoHhiophene^-carboxylic acid methyl ester (336 mg, 0.89 mmo!) and DMAP {217 mg, 1.8 mmo!) were dissolved in dichloroethane (2.2 mL), sealed with a cap and heated to 80 °C. After 2 h, the temperature was increased to 90 °C, and the solution was stirred 16 h. The reaction mixture was further heated to 100 °C, stirred 24 h and partitioned between water and ethyl acetate:hexanes (1:1 ). The layers were separated and the aqueous layer was extracted again with ethyl acetate:hexanes (1:1 ). The combined organic layers were dried over sodium sulfate, filtered and concentrated. Flash chromatography (5% EtOAc: hexanes 5 min then 5-40% EtOAc:hexanes, 20 min, 24 g coiumn) afforded 250 mg (56% yield of the desired 5-(3,3-dimethyl-but-1-ynyl)-3- [(1 ! 4-dioxa-spiro[4.5]dec-8-yi)-(4-methy[-cyclohex-3-eneca rbonyi)-amino]-thiophene- 2-carboxyiic acid methyl ester as a white foam.

5-{3,3-Dimethyl-but-1-ynyl}-3-[(1 ! 4-dioxa-spiro[4.5]dec-8-ylH4-methyl- cyclohex-3-enecarbonyl)-amino]-thiophene-2-carboxylic acid methyl ester (240 mg, 0.48 mmo!) was dissolved in THF (3.2 ml_) and treated with 4M HCI (1.6 ml_, 0.8 mmol). The reaction mixture was heated to 45 °C and stirred at ambient temperature. After 2 h, methanol was added (15-20 drops) and the solution was stirred for 3 h. An additional portion of 4M HCI (1.6 mL, 0.8 mmol) and methanol (1 ml_) were added and the solution was stirred 16 h at ambient temperature followed by 40 0 C for 4 h. The solution was partitioned between water and ethyl acetate. The organic layer was washed with sodium bicarbonate (sat. aq.) and brine, dried over sodium sulfate and concentrated to 233 mg (quantitative yield) of the desired 5-(3,3-dmethyl-but-1-yny!)- 3-[(4-methyl-cyciohex-3-enecarbonyl)-(4-oxo-cyclohexyl)-amin o]4hiophene-2- carboxyiic acid methyl ester as a white foam.

5-(3 ! 3-DimethyI-but-1-ynyl)-3-[(4-methyl-cyclohex-3-enecarb onyi)-(4-oxo- cyclohexyl)-amino]-thiophene-2-carboxyiic acid methyl ester (233 mg, 0.51 mmol) in THF (3 mL) and water (0.3 mL) was treated with sodium borohydride (19 mg, 0.48 mmol). After stirring for 20 min, water was added and the reaction mixture was extract twice with ethyl acetate. The combined organic layers were dried over sodium sulfate, filtered and concentrated. Flash chromatography (10%

EtOAc: hexanes, 4 min, 10-70% EtOAc: hexanes, 12 min, 12 g column) afforded 130 mg (59% yield) of the desired 5-(3,3-Dimethy!-but-1-ynyl)-3-[(4-hydroxy-trans- cyclohexylH4-methyl-cyclohex-3-enecarbonyl)-amino3-thiophene -2-carboxylic acid methyl ester.

5-(3,3-Dimethyl-but-1-ynyl)-3-[(4-hydroxy-trans-cyclohexyl)- (4-methyi- cyclohex-3-enecarbonyl)-amino]-thiophene-2-carboxylic acid methyl ester (40 mg, 0.088 mmol) and (N,N-dimethyl)-(2-fluoro-pyridin-4-ylmethyl)amine (84 mg, 0.55 mmol) in DMF (1 mL) were treated with sodium hydride (22 mg, 60% oil dispersion, 0.55 mmol). After the bubbling slowed, the reaction mixture was sealed and heated by microwave (90 0 C) for 70 min. The reaction mixture was treated with 10% citric acid (2-3 mL) followed by water (2-3 mL). The organics were extracted twice with ethyl acetate, dried over sodium sulfate, filtered and concentrated. The residue was purified by reverse phase HPLC to give the Compound 337, 18 mg (35% yield): MS (rn/z): 578.0 (M+HJ+; HPLC retention time 3.53 min (2-98% acetonitrile: water with 0.05% trifiuoroacetic acid).

Example 338 - Compound 338: 3-((1 r.4R)-N-ff 1 r.4R)-4-(4-

((dimethylamino)methyl)pyridin-2-yloxy)cvc[ohexy!)-4- methγlcyclohexanecarboxarnido)-5-((1 -

(trifluoromethvDcyclobutvDethyn y , l Jthj o ph en e - 2 : ca rboxylic acid

Scheme 20

0.1 % TFA

HATU (14.9 g, 39.3 mmoi, 1.1 equiv) and N,O-dimethylhydroxylamine hydrochloride (3.9 g, 39.3 mmol, 1.1 equiv) were charged into a round-bottomed fiask containing 200 mL dichloromethane. Triethylamine (14.9 mL, 107 mmol, 3.0 equiv) was added, and then a solution of (i-trifluoromethyl)cyclobutanecarboxylic acid (6.0 g, 35.7 mmol, 1.0 equiv) in DCM (25 mL) was added. The reaction was stirred at room temperature untii complete consumption of the carboxylic acid. The solution was concentrated in vacuo to remove voiatiles and the residue was partitioned between DCM (200 mL) and saturated NH 4 CI (100 mL). The aqueous was extract with DCM (200 mL) and the combined organics were dried over Na 2 SO 4 and then concentrated. The crude was purified by silica gel chromatography to afford the product (5.64 g, 27 rnmoi, 75% yield) as colorless oil.

A solution of methyl 3-((1r,4R)-N-((1 r,4R)-4-(tert- butyldimethylsilyloxy)cyclohexy!)-4-methylcyclohexanecarboxa mido)-5- iodothiophene-2-carboxylate (620 mg, 1 mmol, 1.0 equiv) in DCM (5.0 mL) in a round-bottomed flask was cooled to -78 0 C. lsopropyimagnesium chloride (2.0 M in THF 1 1.1 mmol, 1.1 equiv) was added dropwise and then stirred for 30 min. A solution of amide (232 mg, 1.1 mmol, 1.1 equiv) in DCM (1.0 mL) was added slowly and the resulting solution was gradually warmed to room temperature and stirred overnight. The reaction was poured into saturated NH 4 Cl (100 mL) and extracted with DCM (2 x 100 mL). The combined organics were dried over Na 2 SO 4 and then concentrated. The crude was purified by silica gel chromatography to afford the product (186 mg, 0.3 mmol, 29% yieid) as a white solid.

Methyl 3-((1 r, 4R)-N-((1 r, 4R}-4-(tert-butyidimethylsilyloxy)cyclohexyl)-4- methylcyclohexanecarboxamido)-5-( 1 -

(trifiuoromethy!)cyclobutanecarbonyl)thiopbene-2-carboxyl ate (550 mg, 0.85 mmol) was dissolved in acetonitrile (5 mL), H 2 O (1 mL) and trifluoroacetic acid (0.1 mL). The reaction was stirred at room temperature until complete consumption of the starting material. After concentrated to remove volatϋes, the resulting residue was purified by silica gel chromatography to afford the product (430 mg, 0.81 mmol, 95% yield) as a white solid.

To a solution of methyl 3-((1 r,4R)-N-((1 r,4R)-4-hydroxycyciohexyl)-4- methylcyclohexanecarboxamido)-5-(1- (trifluoromethyl)cyclobutanecarbonyl)thiophene-2-carboxylate (400 mg, 0.75 mmol, 1.0 equiv) and dimethyl 1-diazo-2-oxoproρylphosphonate (232 mg, 1.2 mmole, 1.6 equiv) in MeOH (3.0 mL) was added K 2 CO 3 (259 mg, 1.9 mmol, 2.5 equiv). The resulting solution was stirred over night at room temperature. The reaction was then partitioned between EtOAc (100 mL) and 1 N HCI (100 mL). The organic was dried over Na 2 SO 4 and then concentrated. The crude was purified by reverse phase preparative HPLC to afford the product (200 mg, 0.38 mmol, 51 % yield) as a white solid.

To a solution of methyl 3-((1r,4R)-N-((1 r,4R)-4-hydroxycyciohexyl)-4- methylcyclohexanecarboxamido)-5-((1-(trif!uoromethyl)cyclobu tyl)ethynyi)thiophene- 2-carboxylate (50 mg, 0.1 mmol) in DMF (1 ml_) was added NaH (20 mg, 0.5 mmol). After stirred at room temperature for 10 min, (N,N-dimethyl)~(2~fiuoro-pyridin-4- y!methyl)amine (46 mg, 0.3 mmol) was added and reaction was stirred over night at 80 0 C. After cooling to room temperature, the mixture was purified by reverse phase preparative HPLC to afford Compound 338 (27 mg, 0.042 mmol, 42%) as a white solid: MS (m/z) 646.3 [M+Hf HPLC retention time: 1.76 min (50-95% acetonitrile with 0.05% TFA: water with 0.05% TFA).

Exam ple 339 - Compound 339: 3-(( 1 r, 4R)-N-f ( 1 r,4R)-4-(6- hvd roxypyridazin-3-yloxγ)cyciohexyl )-4- methylcyciohexanecarboxamido)-5-((1 - (trifluoromethyl )cvclobutyl )ethvnyl)thiophene-2-carboxyiic acid

Scheme 21

0C

Compound 339

To a solution of methyl 3-((1r,4R)-N-{(1 r,4R)-4-hydroxycyclohexyl)-4- methylcyc!ohexanecarboxamido)-5-((1-(trifluoromethyl)cyclobu tyl)ethynyl)thiophene- 2-carboxySate (50 mg, 0.1 mmol) in DIVlSO (2 mL) was added KOtBu (168 mg, 1.5 mmol). After stirred at room temperature for 10 min, 1-(2-fluoropyridin-4-yl)-N,N- dimethylmethanamine (149 mg, 1.0 mmol) was added and reaction was heated at 80 0 C in a microwave reactor for 1 h. After cooiing to room temperature, the mixture was purified by reverse phase preparative HPLC.

The product from previous step was dissolved in AcOH (2.0 rnL) and NaOAc (82 mg, 1 mmof) was added. The mixture was stirred overnight at 1 10 0 C. After removing the volatiles, the resulting residue was purified by reverse phase preparative HPLC to afford Compound 338 (18 mg, 0.03 mmoi, 30%) as a white soiid: MS (m/z) 606.1 [M+Hf HPLC retention time: 3.1 min (5-95% acetonitrile with 0.05% TFA: water with 0.05% TFA). Example 340 - Com pound 340 : 3-(( 1 r,4R)-N-f ( 1 r,4R)-4-(4- ((dimethylam i no) ri! ,, g , l.b.yiJ£ , .yi.[d , i.Dz.^.:.yJ , . o .M].9.Y£.! , o , h l §x.y ι !Jz4- methylcyclohexanecarboxamido)-5-(( 1 - (tπf)uoromethyl)cvclopropyl)ethynyl)thiophene-2-carboxyriC acid

Scheme 22

Compound 340

Compound 340 was prepared by the same route as example 338, excepting the substitution of 1-(trif!uoromethyl)cyciopropanecarboxylic acid in the formation of the Weinreb amide: MS (m/z) 632.2 [M+H] + HPLC retention time: 3.96 min (50-95% acetonitrile with 0.05% TFA: water with 0.05% TFA). Example 341 - Compound 341: 3-((1 r,4R)-N-((1 r,4R)-4-(4- ((dimethviamino)methyl)pyridin-2-yloxy)cyciohexyi)-4- methylcvcgohexanecarboxamido)-5-(4,4,4-trifluoro-3,3-dimethv ibut-1' ynyl)thiophene-2-carboxyiic acid

Scheme 23

Compound 341

Compound 341 was prepared by the same route as example 338, excepting the substitution of 3,3,3-trif!uoro-2,2-dimethylpropanoic acid in the formation of the Wetnreb amide: MS (m/z) 634.3 [M+H] + HPLC retention time: 4.10 min (5-95% acetonitrile with 0.05% TFA: water with 0.05% TFA).

Example 342 - Compound 342: 3-ff 1 r,4R)-N-f(1 r,4R)-4-(6- hγdroxγpyridazin-3-yloxy)cγclohexyl)-4- m ethyl cyclohexaneca rboxa mido)-5-((1-

(trif[uoromethv0cvcloproρv0ethynvQthioρhene-2-carboxyii c acid

Scheme 24 I KOtBu 1 DMSO 1 SO 0 C

2. NaOAc, AcOH, 100 0 C

Compound 342

Compound 342 was prepared by the same route as example 339, excepting the substitution of the different secondary alcohol intermediate shown above in the ether formation: MS (m/z) 614.1 [M+Naf HPLC retention time: 4.34 min (5-95% acetonitrile with 0.05% TFA: water with 0.05% TFA).

Example 343 - Compound 343: 3-f (1 r, 4R)-N-(M r, 4R)-4-(6- hvdroxypyridazin-3-yloxy)cvciohexyO-4- methylcvclohexanecarboxamido)-5-f4.4,4-trifiuoro-3,3-dimethy lbut-1 - vnyl)thiophene-2-carboxγlic acid

Scheme 25

1. KOtBu, DMSO, 80 X

2. NaOAc 1 AcOH, 100 0 C

Compound 343

Compound 343 was prepared by the same route as example 339, excepting the substitution of the different secondary aicohol intermediate shown above in the ether formation: MS (m/z) 616.2 [M+Na] + HPLC retention time: 4.48 min (5-95% acetonitrile with 0.05% TFA: water with 0.05% TFA).

ExampJe 344 - Compound 344: 3-f f 1 r.4R)-4-methyl-N-(f 1 r,4R)-4- (pyrimidin-4-yloxy)cyclohexy0cvclohexanecarboxamido)-5-{2-f 1 - (trifluoromethv[)cvclopropyl)ethynyl)thiophene-2-carboxylic acid

Scheme 26

Compound 344

Compound 344 was prepared by the same route as example 340, excepting the substitution of 4-chloropyrιmιdtne hydrochloride as the electrophile in the final ether formation step MS (m/z) 574 3 [M-H] HPLC retention time. 4 38 mm (5-95% acetonitriie with 0 05% TFA: water with 0.05% TFA)

Exam ple 345 - Compou nd 345 3-(( 1 r,4R)-4-methyl-N-{( 1 r.4R)-4- (tetrahvd rofuran-3-yloxy)cyclohexyl)cyclohexanecarboxam ιdo)-5-(2-( 1 - (tπfluoromethyl )cvciopropyl )ethynyl)thiθphene-2-carboxyiιc a cid

Scheme 27

To a suspension of methyl 3-((1r,4R)-4~methyl~N-({1r,4R)-4-(oxocyclohexyi) cyclohexanecarboxamιdo)-5-(2-(1-(tπfluoromethyl)cyc(opropy ()ethynyl)thιoρhene-2- carboxylate (82mg, 0.161mmol) and FeCI 3 (3mg, 0.016 mmol) in nitromethane (1 mL), cooled to 0 0 C, was added neat trimethyl-(tetrahydro-furan-3-yloxy)-silane (200 mg, 1.25mmoi) followed by the addition of triethylsilane (0.051 mL, 0.322 mmoS). The reaction was allowed to slowly warm to room temperature and stirred overnight.

Phosphate buffer of pH 4.5 was added and the reaction mixture was extracted with dichloromethane. The organic layer was dried over Na 2 SO 4 and concentrated.

Purification by HPLC with CH 3 CN(0.1 % TFA)/H 2 O(0.1 % TFA) provided both methyl 3-({1r,4R)-4-methyl-N-((1 r,4R)-4-(tetrahydrofuran-3- yIoxy)cyclohexyl)cyclohexanecarboxamido)-5-(2-(1- (trifluoromethyl)cycloρropyl)ethynyl)thϊophene-2-carboxyla te and methyl 3-((1 r,4R}-4- methyl-N-((1s,4S)-4-(tetrahydrofuran-3-yioxy)cyclohexyl)cyci ohexanecarboxamido)- 5-(2-( 1 -(trifluoromethyl)cyc!opropyt)ethynyi)thiophene-2 -carboxylate. Each of the diastereomers was then subjected to ester hydrolysis separately.

Methyl 3-((1 r,4R)-4-methyl-N-((1r,4R)-4-(tetrahydrofuran-3- yioxy)cyclohexyl)cyciohexanecarboxamido)-5-(2-( 1 -

{trifluoromethyl)cycloproρyl)ethynyl)thioρhene-2-carbox ylate (75mg, 0.129mmo!) was dissolved in tetrahydrofuran (1 mL). To this was added 2N NaOH (0.65mL, 1.3mmol) and 2 drops of methanol, and the solution was stirred 1 hour at room temperature. The reaction solution was then partitioned between ethyl acetate (30 mL) and 2N HCI (3OmL). The aqueous ϊayer was washed once with 3OmL ethyl acetate, and the combined organic layer was concentrated. Purification by HPLC with CH 3 CN(0.1 % TFA)/H 2 O(0.1 % TFA) provided 3-((1 r,4R)-4-methyl-N-((1 r,4R)-4-(tetrahydrofuran-3- yloxy)cyclohexyl)cyclohexanecarboxamido)-5-(2-(1- (trifluoromethyl)cyclopropyl)ethynyl)thiophene-2-carboxyiic acid (31 mg, 0.55mmol, 42%) as a white solid: MS (m/z) 568.2 [M+H] + HPLC retention time: 2.61 min (50- 95% acetonitrile with 0.05% TFA: water with 0.05% TFA).

Example 346 - Compound 346 : 3-(f 1 r.4R>-4-methvi-N-({ 1 s,4S)-4- (tetrahvdrofu ra n-3-yloxy)cγclohexyl)cycio hexanecarboxa rn ido)-5-(2-( 1 - (trifiuoromethy[)cvciopropyDethvnv0thiophene-2-carboxyiic acid Scheme 28

Compound 346

Methyl 3-((1 r,4R)-4-methyl-N-((1s,4S)-4-(tetrahydrofuran-3- yloxy)cyclohexy!)cyclohexanecarboxamido)-5-{2-(1-

(triflϋoromethySJcyclopropyOethyπyOthiophene^-carboxyia te {12mg, 0.021 mmol) was dissolved in tetrahydrofuran (1 mL). To this was added 2N NaOH (0.2mL, 0,4 mmoi) and 2 drops of methanol, and the solution was stirred 1 hour at room temperature. The reaction solution was then partitioned between ethyl acetate (15 mL) and 2N HCi (15mL). The aqueous layer was washed once with 15mL ethyi acetate, and the combined organic layer was concentrated. Purification by HPLC with CH 3 CN(0.1% TFA)/H 2 O(0.1% TFA) provided 3-((1 r,4R)-4-methyl-N-((1 s,4S)-4-(tetrahydrofuran-3- yloxy)cyciohexyl)cyclohexanecarboxamido)-5-(2-(1- (trifluoromethy[)cyclopropyl)ethynyl)thiophene-2-carboxylic acid (4mg, 0.007mmoi, 34%) as a white solid: MS (m/z) 568.2 [M+H] + HPLC retention time: 2.73 min (50- 95% acetonitriie with 0.05% TFA: water with 0.05% TFA).

Example 347 - Compound 347 : 5-( 3.3-dimethvibut-1 -vnyl)-3-f ( 1 r,4R)- 4-methvi-N-({ 1 s,4S)-4-(oxetan-3- yloxy)cyclohexyi)cyclohexanecarboxam ido)-5-f2-( 1 - {trifiuoromethyl )cyclopropyl )ethynvOthiophene-2-carboxylic acid

Scheme 29

To a suspension of Methyl 5-(3,3-dimethyfbut-1 -ynyi)-3-((1 r,4r)-4-methyl-N-4- (oxocyciohexyl) cyclohexanecarboxamιdo)thιophene-2-carboxyiate (1 Og,

2 185mmol) and FeCI 3 (35mg, 0 22 mmoi) in nitromethane (15 ml) cooled to 0 0 C was added neat (1 ,3-dιoxan-3-yloxy)tπmethylsιlane (1 05, 6mmot) followed by the addition of tπethylsifane (0 59 ml, 4 37 mmoi) The reaction was allowed to slowly warm to room temperature and stirred overnight Phosphate buffer of pH 4 5 was added and the reaction mixture was extracted with dichloromethane The organic layer was dried over Na 2 SO 4 and concentrated The crude was purified by silica gel chromatography (45% ethyl acetate in hexanes) to afford the ether product (704 mg, 1 29 mmoi, 59% yield) as a yeliow oil MS (m/z) 546 5 [M+H] + HPLC retention time 4 13 mm (50-95% acetonstriie with 0 05% TFA water with 0 05% TFA). This oil was dissolved in 1 ,4-dιoxane (3 mL) and 2N HCI (7mL) and was then heated and stirred at reflux overnight The reaction was then cooled and diluted with water (50 mL) The mixture was then extracted twice with ethyl acetate (100 mL) The combined ethyl acetate extracts were washed with brine, dried over Na 2 SO 4 and concentrated. The crude product was taken on directly to the next step.

Dimethytaminopyridine (175mg, 1.434mmol) and the crude diol starting material (255mg, 0.478mmol) were dissolved in dichloromethane (4 ml_). 4- Toiuenesulfonyi chloride {109mg, 0.574 mmo!) was then added and the solution was stirred 30 minutes at room temperature. The excess tosyl chloride was then quenched by aqueous NH 4 Cl (30 ml_), and the mixture was extracted twice with dichloromethane (30 mL). The organic layer was dried over Na 2 SO 4 and

concentrated. The residue was dissolved in tetrahydrofuran (4 mL) and sodium hydride (50 mg, 1 .195 mmol) was added. The reaction was stirred at 5O 0 C overnight. 2N HCI (30 mL) was then added, and the solution was extracted twice with ethyl acetate (50 mL). The organic layer was dried over Na 2 SO 4 and concentrated.

Purification by HPLC with CH 3 CN(0.1 % TFA)/H 2 O{0.1% TFA) provided methyl 5-(3,3- dimethylbut-1 -ynyl)-3-((1r,4RH-methyi-N-((1s,4S)-4-(oxetan-3- yloxy)cyclohexyl)cyclohexanecarboxamido)-5-(2-(1-

(trifluoromethyOcyclopropyOethynyOthiophene^-carboxylate (32 mg, 0.62mmol, 13%) as an off-white solid: MS (m/z) 516.2 [M+H] + HPLC retention time: 3.02 min (50-95% acetonitrile with 0.05% TFA: water with 0.05% TFA).

Methyl 5~(3,3-dimethylbut-1-ynyl)-3-((1r,4R)-4-methyl-N-((1s,4SH-(o xetan-3- yloxy)cyclohexyl)cyclohexanecarboxamido)-5-(2-(1 -

(trifiuoromethyl)cyclopropyl)ethynyl)thioρhene-2-carboxy iate (92mg, Q.184mmo!) was dissolved in tetrahydrofuran (5 mL). To this was added 2N NaOH (0.92ιmL, 1.84 mmol) and 2 drops of methanol, and the solution was stirred 30 minutes at room temperature. The reaction solution was then acidified with 2N HCI (1.5mL). The mixture was extracted twice with ethyl acetate (30 mL) and concentrated. Purification by HPLC with CH 3 CN(0.1% TFA)/H 2 O(0.1% TFA) provided 5-(3,3-dimethylbut-1- ynyi)-3-((1 r,4R)-4-methyl-N-((1s,4S)-4-(oxetan-3- yioxy)cyclohexyl)cyclohexanecarboxamido)-5-(2-(1 - (trifluoromethyl)cyclopropyl)ethynyl)thiophene-2~carboxyfic acid (10mg, 0.02mmol, 11%) as a white solid: MS (m/z) 502.2 [M+H] + HPLC retention time: 5.03 min (5-95% acetonitrile with 0.05% TFA: water with 0.05% TFA). Example 348 - Synthesis 5-(3,3-Dimethyl-but-1-vnylV3-rf 1 S,6S)-(4,6-dimethyl- cvclohex-3-enecarbonylH4-hydroxy-cyciohexyl)-amino]-thioρhe ne-2-carpoxyJic acid

Scheme 1 (1 S,6S)-4,6-dimethy(-cyclohex-3-eπe-carboxylic acid (3.04g, 19.7mmol) was dissolved in CH 2 Ci 2 (3OmL) and DMF (20μL) was added. The solution was cooled to 0 0 C and then (COCI) 2 (3.7mL, 39mmol) was added siowly. The reaction was stirred in an ice bath for 2 hours and then concentrated. The residue was taken up in hexanes and concentrated; this hexanes coevaporation was repeated once more. To the residue was added [5-(3,3-Dimethyl-but-1-ynyl)4hiophen-3-yl]-(1 ,4-dioxa- spiro[4.5]dec-8-yl)-amine (4.16g, 13mmol), diisopropylethylamine (4.5ml_, 26mmol), and 1 ,2-dichloroethane (4OmL) at 0 0 C. The solution was warmed to room

temperature and stirred overnight. The reaction was diluted with CH 2 CI 2 , twice washed with saturated NH 4 Ci (aq) , dried over MgSO 4 , filtered, concentrated, and purified by silica gei column chromatography, eluting with a mixture of 0-75%

EtOAc/hexanes, to give (1S,6S)-4,6-dimethyi-cyclohex-3-ene-carboxylic acid [5-(3,3- dimethyi-but-1-ynyl)-thiophen-3-yl]-(1 ,4-dioxa-spiro[4.5]dec-8-yl)-amide (5.6g, 12mmoϊ) as a single isomer.

(1 S,6S)-4,6-Dimethyl-cyclohex-3-ene-carboxyiic acid [5-(3,3-dimethyl-but-1 - ynyl)-thiophen-3-yl]-(1 ,4-dioxa-spiro[4.5]dec-8-yi)-amide (5.6g, 12mmol) was dissolved in THF (7OmL) and treated with 4M HCI (35mL). The reaction mixture was heated to 45 0 C and stirred for 2.5 h. THF was removed in vacuo, and the aqueous layer was thrice extracted into ethyl acetate. The combined organic layers were washed with saturated NaHCO 3(aq) , water, and brine, dried over MgSO 4 , filtered, and concentrated to give (1S,6S)-4,6-dimethyi-cyclohex-3-ene-carboxylic acid [5-(3,3- dimethyl-but-1-ynyl)-thiophen-3-yl]-(4~oxo~cyc!ohexyl)-amide (5.05g, 12mmol).

( 1 S,6S)-4,6-Dimethyl-cyclohex-3-ene-carboxylic acid [5-(3,3-dimethyl-but-1 - ynyl)-thiophen-3-yl]-(4-oxo-cyc!ohexyl)-amide (2.Og, 4.9mmol) in MeOH (10OmL) was treated with sodium borohydride (230mg, δ.Ommol) at 0 0 C. After stirring for 30 min, 4M HCI (6mL) was added and the reaction mixture was twice extracted with ethyl acetate. The combined organic layers washed with saturated NaHCO 3 ^ q)1 brine, dried over MgSO 4 , filtered, and concentrated. Silica gel chromatography (20-60% ethyl acetate/hexanes) gave the desired (1S,6S)-4,6-dimethyl-cyclohex-3-ene- carboxylic acid [5-(3,3-dimethyl-but-1 -ynyl)-thiophen-3-yl]-(frans-4-hydroxy- cycloheχyi)-amide (1.74g, 4.2mmo!).

(1 S,6S)-4,6-Dimethyl-cyclohex-3-ene-carboxyIic acid [5-(3,3-dimethyI-but-1- ynyl)-thiophen-3-yl]-(frans-4-hydroxy-cyciohexyl)-amide (1.74g, 4.2mmol) in THF

(5OmL) was cooled to -78 0 C and treated with lithium diisopropylamine (8.4mL, 2.0M in heptane/THF/PhEt, 16.8mmol) and allowed to warm to 0 0 C over the course of 2 hours. CO 2 was vigorously bubbled through the reaction solution for 10 minutes. The reaction was then quenched with the addition of iPrOH, diluted with ethyl acetate, washed with saturated NH 4 CI (aq) , dried over MgSO 4 , filtered, and concentrated. Silica gel chromatography (0-100% ethyi acetate/dichloromethane) afforded 530mg (1.2mmol) of the title compound: MS (m/z): 458.1 [M+H]+; HPLC retention time 4.35 min (2-98% acetonitriie: water with 0.05% trifiuoroacetic acid).

Example 349 - Compound 349: 5-(3,3-Dimethyl-but-1-vnviV3-(f1S.6S)-(4.6-dimethyi- cvc!ohex-3-enecarbonyl)-rfrans-4-(pyrimidin-2-yloxy)-cvclohe xyπ-aminoHhiophene-2- carboxylic acid

Scheme 2

Compound 349

5-(3,3-Dimethyl-but-1-ynyl)-3S-[(4,6S-dimethyl-cyclohex-3-en ecarbonyl)-(4- hydroxy-cyciohexyl)-amino]-thiophene-2~carboxy!ic acid (51 mg, 0.11mmol) and 2- chloropyrimidine {100mg, 0.87mmol) were dissolved in DMF (0.5 mL) and treated with sodium hydride (49 mg, 60% oil dispersion, 1.2 mmol). The mixture was stirred under nitrogen until the bubbling slowed, then was sealed and heated by microwave (90 °C, 30 min). After cooϋng, the mixture was diluted with ethyl acetate (about 5 mL) and quenched with 10% citric acid (about 2 mL). Water was added and the mixture was extracted twice with ethyS acetate. After concentration the resulting residue was purified by HPLC with CH 3 CN (0.1% TFA)/H 2 O(0.1% TFA) to afford 39 mg of Compound 349: MS (m/z): 533.9 [M-H]-; HPLC retention time 4.93 min (2-98% acetonitriie: water with 0.05% trifiuoroacetic acid). Example 350 - Compound 350: 5-(3.3-Dimethyl-but-1-vnvi)-3-(f1S.6SH4.6-dimethyl- cvciohex-3-enecarbonvπ-ffraA7s-4-(pyrimidin-4-yloxy)-cycloh exyl]-amino|-thiophene-2- carboxylic acid

Compound 350

Compound 350 was synthesized in a manner similar to Example 349 using 4- chioropyrimidine hydrochloride in place of 2-chloropyrimidine: MS (m/z): 534.0 [M- H]-; HPLC retention time 4.27 min (2-98% acetonitrile: water with 0.05%

trifluoroacetic acid).

Example 351 - Compound 351 : 5-(3,3-Dimethyl-but-1-vnvi)-3S-((4,6S-dimethyl- cvclohex-3-enecarbonv[)-ffrans-4-fpyridin-2-yioxy)-cvciohexy n-amino)-thiophene-2- carboxylic acid

Compound 351

Compound 351 was synthesized in a manner similar to Example 349 using 2- chioropyridine in place of 2-chloroρyrimidiπe: MS (m/z): 532.9 [M-H]-; HPLC retention time 4.75 min (2-98% acetonitriie: water with 0.05% trifluoroacetic acid). Example 352 - Compound 352: 5-f3,3-Dtmethy[-but-1-vnvi)-3-(f1 S,6SH4.6-dimethyl- cvclohex-3-enecarbonviHfrans-4-(1-oxy-pyridin-3-yloxy)-cvcio hexyπ-amino}- thiophene-2-carboxylic acid

Compound 352

Compound 352 was synthesized in a manner similar to Example 349 using 3- fSuoropyridine-N-oxide in place of 2-chloropyrimidine: MS (m/z): 551.1 [M+H]+; HPLC retention time 4.21 min (2-98% acetonitrile: water with 0.05% trifluoroacetic acid).

Example 353-Compound 353

Compound 353

Compound 353 may be prepared in the same manner as in Example 314.

Exampie 354-Compound 354

Compound 354

Compound 354 may be prepared in the same manner as in Example 314.

Exampie 355-Compound 355

Compound 355

Compound 355 may be prepared in the same manner as in Example 314. Example 356-Compound 356

Compound 356

Compound 356 may be prepared in the same manner as Example 349 using 3,6-dichSoropyridazine in place of 2-chloropyrimidine.

Example 357-Compound 357

Compound 357

Compund 357 may be prepared in the same manner as Example 349 using 2-chloro-4-{2-(trimethylsiiyl)ethoxy)pyrimidine in place of 2-chloropyrimidine.

Additional Prophetic Examples

The following prophetic examples may be synthesized using analogous techniques.

Aromatic heterocycle variants , where R ah is

Saturated heterocycle variants

where R sh is

Cyclic and Acyclic Rlyariants

Substituted R 3 and other R 3 variant

Biological Examples

Antiviral Activity

Another aspect of the invention relates to methods of inhibiting viral infections, comprising the step of treating a sample or subject suspected of needing such inhibition with a composition of the invention.

Within the context of the invention samples suspected of containing a virus include natural or man-made materials such as living organisms; tissue or cell cultures; bioiogical samples such as biological material samples (blood, serum, urine, cerebrospinal fluid, tears, sputum, saliva, tissue samples, and the like); laboratory samples; food, water, or air samples; bioproduct samples such as extracts of cells, particularly recombinant cells synthesizing a desired glycoprotein; and the like.

Typically the sample will be suspected of containing an organism which induces a viral infection, frequently a pathogenic organism such as a tumor virus. Samples can be contained in any medium including water and organic solvent\water mixtures. Samples include living organisms such as humans, and man made materials such as cell cultures.

If desired, the anti-virus activity of a compound of the invention after application of the composition can be observed by any method including direct and indirect methods of detecting such activity. Quantitative, qualitative, and

semiquantitative methods of determining such activity are all contemplated. Typically one of the screening methods described above are applied, however, any other method such as observation of the physiological properties of a living organism are also applicable.

The antiviral activity of a compound of the invention can be measured using standard screening protocols that are known. For example, the antiviral activity of a compound can be measured using the following general protocols. Cell-based Fiavivirus Immunodetection assay

BHK21 or A549 cells are trypsinized, counted and diluted to 2x10 s cells/mL in Hams F-12 media (A549 cells) or RPMI-1640 media (BHK21 cells) supplemented with 2% fetal bovine serum (FBS) and 1% penicillin/streptomycin. 2x10 4 cells are dispensed in a dear 96-well tissue culture plates per well and palced at 37° C, 5% CO 2 overnight. On the next day, the cells are infected with viruses at multiplicity of infection (MOS) of 0.3 in the presence of varied concentrations of test compounds for 1 hour at 37° C and 5% CO 2 for another 48 hours. The ceils are washed once with PBS and fixed with cold methanol for 10 min. After washing twice with PBS, the fixed ceils are blocked with PBS containing 1 % FBS and 0.05% Tween-20 for 1 hour at room temperature. The primary antibody solution (4G2) is then added at a concentration of 1 :20 to 1 :100 in PBS containing 1 % FBS and 0.05% Tween-20 for 3 hours. The cells are then washed three times with PBS followed by one hour incubation with horseradish peroxidase(HRP)-conjugated anti-mouse IgG (Sigma, 1 :2000 dilution). After washing three times with PBS, 50 microliters of 3, 3', 5,5'- tetramethylbenzidine (TMB) substrate solution (Sigma) is added to each well for two minutes. The reaction is stopped by addition of 0.5 M sulfuric acid. The plates are read at 450 nm abosorbance for viral load quantification. After measurement, the cells are washed three times with PBS followed by incubation with propidium iodide for 5 min. The plate is read in a Tecan Safire™ reader (excitation 537 nm, emission 617 nm) for cell number quantification. Dose response curves are plotted from the mean absorbance versus the log of the concentration of test compounds. The EC 50 is calculated by non-linear regression analysis. A positive control such as N-nonyi- deoxynojirimycin may be used.

Cell-based Flavivirus cytopathic effect assay For testing against West Nile virus or Japanese encephalitis virus, BHK21 cells are trypsinized and diluted to a concentration of 4 x 10 5 cells/mL in RPMI-1640 media supplemented with 2% FBS and 1% penicillin/streptomycin. For testing against dengue virus, Huh7 ceils are trypsinized and diluted to a concentration of 4 x 10 s cells/mL in DMEM media supplemented with 5% FBS and 1%

penicillin/streptomycin. A 50 microliter of eel! suspension (2 x 10 4 ceils) is dispensed per weil in a 96-well optical bottom PIT polymer-based plates (Nunc). Cells are grown overnight in culture medium at 37° C, 5% CO 2 , and then infected with West Nile virus (e.g. B956 strain) or Japanese encephalitis virus (e.g. Nakayama strain) at MOI = 0.3, or with dengue virus (e.g. DEN-2 NGC strain) at MOI = 1 , in the presence of different concentrations of test compounds. The plates containing the virus and the compounds are further incubated at 37 0 C, 5% CO 2 for 72 hours. At the end of incubation, 100 microliters of CellTiter-Glo™ reagent is added into each well.

Contents are mixed for 2 minutes on an orbital shaker to induce cell lysis. The plates are incubated at room temperature for 10 minutes to stabilize luminescent signal. Lumnescence reading is recorded using a plate reader. A positive control such as N- nonyl-deoxynojirimycin may be used.

Antiviral Activity in a Mouse Model of Dengue Infection. Compounds are tested in vivo in a mouse model of dengue virus infection (Schul et al. J. Infectious Dis. 2007; 195:665-74). Six to ten week old AG129 mice (B&K Universa! Ltd, Hit, UK) are housed in individually ventilated cages. Mice are injected intra peritoneal Iy with 0,4 mL TSV01 dengue virus 2 suspension. Biood samples are taken by retro orbital puncture under isoflurane anaesthesia. Blood samples are collected in tubes containing sodium citrate to a final concentration of 0.4%, and immediately centrifuged for 3 minutes at 600Og to obtain plasma. Plasma (20 microliters) is diluted in 780 microiiters RPMI-1640 medium and snap frozen in liquid nitrogen for plaque assay analysis. The remaining plasma is reserved for cytokine and NS1 protein level determination. Mice develop dengue viremia rising over several days, peaking on day 3 post-infection.

For testing of antiviral activity, a compound of the invention is dissolved in vehicle fluid, e.g. 10% ethanol, 30% PEG 300 and 60% D5W (5% dextrose in water; or 6N HCI (1.5 eq):1 N NaOH (pH adjusted to 3.5): 100 mM citrate buffer pH 3.5 (0.9% v/v:2.5% v/v: 96.6% v/v). Thirty six 6-10 week old AG129 mice are divided into six groups of six mice each. Ail mice are infected with dengue virus as described above (day 0). Group 1 is dosed by oral gavage of 200 mL/mouse with 0.2 mg/kg of a compound of the invention twice a day (once early in the morning and once late in the afternoon) for three consecutive days starting on day 0 (first dose just before dengue infection). Groups 2, 3 and 4 are dosed the same way with 1 mg/kg, 5 mg/kg and 25 mg/kg of the compound, respectively. A positive control may be used, such as (2R,3R,4R > 5R)-2-(2-amino-6-hydroxy-purin-9-yl)-5-hydroxymethyl-3 -methyl- tetrahydro-furan-3,4-diol, dosed by oral gavage of 200 microliters/mouse the same way as the previous groups. A further group is treated with only vehicle fluid. On day 3 post-infection approximately 100 microliter blood samples (anti- coaguiated with sodium citrate) are taken from the mice by retro-orbital puncture under isoflurane anaesthesia. Plasma is obtained from each blood sample by centrifugation and snap frozen in liquid nitrogen for plague assay analysis. The collected plasma samples are analyzed by plague assay as described in Schui ef a/. Cytokines are also analysed as as described by Schul. NS1 protein levels are analysed using a Platelia™ kit (BioRad Laboratories). An anti-viral effect is indicated by a reduction in cytokine levels and/or NS1 protein levels. Typically, reductions in viremia of about 5-100 fold, more typically 10-60 fold, most typically 20-30 fold, are obtained with 5-50 mg/kg bid dosages of the compounds of the invention. HCV Assay Protocol

The anti-HCV activity of the compounds of this invention was tested in a human hepatoma Huh-7 cell iine harboring a HCV replicon. The assay comprised the following steps:

Step 1 : compound preparation and serial dilution,

Serial dilution was performed in 100% DMSO in a 384-well plate. A solution containing a compound at 225-fold concentration of the starting final serial dilution concentration was prepared in 100% DMSO and 15 uL added to the pre-specified wells in column 3 or 13 of a polypropylene 384-well plate. The rest of the 384-welt plate was filled with 10 uL 100% DMSO except for columns 23 and 24, where 10 uL of 500 uM a HCV protease inhibitor (ITMN-191 ) in 100% DMSO was added. The

HCV protease inhibitor was used a control of 100% inhibition of HCV replication. The plate was then placed on a Biomek FX Workstation to start the serial dilution. The serial dilution was performed for ten cycles of 3-fold dilution from column 3 to 12 or from column 13 to 22.

Step 2: cell culture plate preparation and compound addition

To each well of a black polypropylene 384-well plate, 90 μL of cell media containing 1600 suspended Huh-7 HCV replicon cells was added with a Biotek uFlow Workstation. A volume of 0.4 μL of the compound solution was transferred from the serial dilution plate to the cell culture plate on a Biomek FX Workstation. The DMSO concentration in the final assay condition was 0.44%. The plates were incubated for 3 days at 37 0 C with 5% CO2 and 85% humidity.

Step 3: detection of cytotoxicity and inhibition of viral replication

a) Assessment of cytotoxicity: The media in the 384-weil cell culture plate was aspirated with a Biotek EL405 plate-washer. A volume of 50 μL of a solution containing 400 nM Calcein AM in 100% PBS was added to each well of the plate with a Biotek uFlow Workstation. The plate was incubated for 30 minutes at room temperature before the fluorescence signal (emission 490 nm, exitation 520 nm) was measured with a Perkin Elmer Envision Plate Reader. b) Assessment of inhibition of viral replication: The calcein-PBS solution in the 384-weS! cell culture plate was aspirated with a Biotek EL405 plate-washer, A volume of 20 μL of Dual-Glo luciferase buffer (Promega, Dual-G!o Luciferase Assay Reagent, cat. #E298B) was added to each well of the plate with a Biotek uFlow Workstation. The plate was incubated for 10 minutes at room temperature. A volume of 20 μL of a solution containing 1 :100 mixture of Dual-Glo Stop & Gio substrate(Promega, Dual- Glo Luciferase Assay Reagent, cat. #E313B) and Dual-Glo Stop & GIo buffer (Promega, Dual-Glo Luciferase Assay Reagent, cat. #E314B) was then added to each well of the plate with a Biotek uFiow Workstation. The plate was incubated at room temperature for 10 minutes before the luminescence signal was measured with a Perkin Elmer Envision Plate Reader.

Step 4: calculation

The percent cytotoxicity was determined by calcein AM conversion to fluorescent product. The average fluorescent signal from the DMSO control wells were defined as 100% nontoxic. The individual fluorescent signal from testing compound treated well was divided by the average signal from DMSO control wells and then multiplied by 100% to get the percent viability. The percent anti-HCV replication activity was determined by the luminescence signal from the testing well compared to DMSO controls wells. The background signal was determined by the average luminescence signal from the HCV protease inhibitor treated welis and was subtracted from the signal from the testing wells as well as the DMSO control wells. Following 3-fold serial dilutions, the EC 50 and CC 50 values were calculated by fitting % inhibition at each concentration to the following equation:

% inhibition = 100%/[(EC 50 /[l]) b + 1]

Where b is Hill's coefficient. See, for reference, Hill, A. V., The Possible

Effects of the Aggregation of the Molecules of Haemoglobin on its Dissociation Curves, J. Physiol. 40: ϊv-vii. (1910).

% inhibition values at a specific concentration, for example 2μM, can also be derived from the formula above.

When tested, certain compounds of this invention were found to inhibit viral replication as listed in Table 1 :

Table 1

Non-limiting preferred compounds of Table 1 include Compounds 3, 14, 24, 27, 29, 85, 96, 101 , 104, 110, 143, 147, 155, 156, 158, 162, 169, 178, 179, 180, 183, 185, 187, 190, 191 , 192, 193, 196, 197, 198, 199, 200, 209, 210, 220, 221 , and 224.

The specific pharmacological and biochemical responses observed may vary according to and depending on the particular active compound selected or whether there are present pharmaceutical carriers, as weil as the type of formulation and mode of administration employed, and such expected variations or differences in the results are contempiated in accordance with practice of the present invention.

Although specific embodiments of the present invention are herein illustrated and described in detail, the invention is not limited thereto. The above detailed descriptions are provided as exemplary of the present invention and should not be construed as constituting any limitation of the invention. Modifications will be obvious to those skilled in the art, and aii modifications that do not depart from the spirit of the invention are intended to be included with the scope of the appended claims,