Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PIPERAZINE-PIPERIDINE COMPOUNDS AS HEPATITIS C VIRUS INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2013/163262
Kind Code:
A1
Abstract:
The invention provides compounds of formula (I) wherein the variables are defined in the specification, or a pharmaceutically-acceptable salt thereof, that are inhibitors of replication of the hepatitis C virus. The invention also provides pharmaceutical compositions comprising such compounds, methods of using such compounds to treat hepatitis C viral infections, and processes and intermediates useful for preparing such compounds.

Inventors:
LONG DANIEL D (US)
MCKINNELL ROBERT MURRAY (US)
VAN ORDEN LORI JEAN (US)
OGAWA GAVIN (US)
WILTON DONNA (US)
Application Number:
PCT/US2013/037920
Publication Date:
October 31, 2013
Filing Date:
April 24, 2013
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
THERAVANCE INC (US)
LONG DANIEL D (US)
MCKINNELL ROBERT MURRAY (US)
VAN ORDEN LORI JEAN (US)
OGAWA GAVIN (US)
WILTON DONNA (US)
International Classes:
C07D403/14; C07D403/04; C07D407/14
Foreign References:
US20110064695A12011-03-17
Other References:
MACDONALD ET AL., JOURNAL OF GENERAL VIROLOGY, vol. 85, 2004, pages 2485 - 2502
T. W. GREENE; G. M. WUTS: "Protecting Groups in Organic Synthesis", WILEY
MIYAURA; SUZUKI, CHEM. REV., vol. 95, 1995, pages 2457 - 2483
"Remington: The Science and Practice of Pharmacy", 2000, LIPPINCOTT WILLIAMS & WHITE
H.C. ANSEL ET AL.: "Pharmaceutical Dosage Forms and Drug Delivery Systems", 1999, LIPPINCOTT WILLIAMS & WHITE
Attorney, Agent or Firm:
HAGENAH, Jeffrey A. et al. (Inc.901 Gateway Boulevar, South San Francisco California, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A compound of formula (I):

(I)

wherein

R1 is selected from Ci-6alkyl, Ci-6alkoxy, phenyl, C3_6cycloalkyl, heterocycle, and heteroaryl, wherein Ci-6alkyl is optionally substituted with -0Ra amino, -SRe, heterocycle, or heteroaryl, Ci-6alkoxy is optionally substituted with -0Ra, and heterocycle is optionally substituted with -ORa amino, or -C(0)OCi_6alkyl, or with one or two Ci-3alkyl;

R2 is selected from hydrogen and Ci-6alkyl;

R3 is selected from hydrogen, Chalky!, -C(0)OCi_6alkyl,

-C(0)NRmRn, -C(0)C3-6cycloalkyl, and -S(0)2C1-3alkyl;

R4, R5, and R6 are each hydrogen;

or R4 is selected from C1-6alkyl, -NRbRc, -ORd, -CN, -C(0)NRaRb, and halo; and R5 and R6 are hydrogen;

or R4 and R5 are independently Ci_6alkyl or halo and R6 is hydrogen;

or R4 and R5 taken together form -0-(CH2)2-0- and R6 is hydrogen;

or R4 is hydrogen and R5 and R6 taken together form -(CH2)n- wherein n is 1, 2,

3, or 4;

or R4 and R5 are hydrogen, and R6 is Ci-6alkyl,

R7 is selected from halo, Ci_3alkyl, and and

Ci-3alkoxy are optionally substituted with one, two, three, four, or five halo;

a is 0, 1, or 2;

R 8 is Ci h

-3alkyl optionally substituted with -OR ;

b is 0, 1 or 2;

or when b is 2, two R8 can be joined to form -(CH2)2-;

R9 is Ci_3alkyl; c is 0, 1 or 2;

R10 is hydrogen, halo, or Chalky! substituted with one, two, or three halo, or with

-ORh;

R11 is selected from Ci-6alkyl, C3_iocycloalkyl, -NRfRs, heteroaryl, heterocycle, and -CH2-heteroaryl;

wherein:

Ci-6alkyl is optionally substituted with one or two substituents independently selected from -ORh, -NRjRk, and phenyl;

Ci_6alkoxy is optionally substituted with -ORh or with phenyl;

any C3_iocycloalkyl is optionally substituted with one or two substituents independently selected from -CD3, halo, and -ORh;

any heterocycle is optionally substituted with one, two, or three substituents independently selected from halo, -C(0)OCi_3alkyl, and -C(0)Ci-6alkyl, wherein any -C(0)Ci_6alkyl is optionally substituted with -NHC(0)OCi_3alkyl;

any heteroaryl is optionally substituted with one or two Ci_3alkyl;

R12 is hydrogen or R10 and R12 taken together form -CH=CH- or -(CH2)2-;

Ra, Rb, Rc, Rd, Re, Rf, Rh, Rj, Rm, and Rn are each independently hydrogen or Ci-3alkyl;

Rs is selected from hydrogen, Ci-6alkyl, and C3_6cycloalkyl;

Rk is selected from hydrogen, -C(0)C1-3alkyl, -C(0)OC1-3alkyl, -C(0)ONRbRc; and -C(0)NRbRc; and

X is N and Y is CH or X is CH and Y is N;

provided that when X is CH, b is 0 and when Y is CH, c is 0;

or a pharmaceutically-acceptable salt or stereoisomer thereof.

2. The compound of Claim 1 wherein the compound of formula (I) is a compound of formula (II):

(II)

3. The compound of Claim 1 wherein the compound of formula (I) is a compound of formula (III):

(HI)

4. The compound of Claim 1 wherein R1 is selected from Ci-6alkyl optionally substituted with hydroxy or methoxy, and tetrahydropyran.

5. The compound of Claim 1 wherein R4 is methyl, methoxy, fluoro, or -C(0)NH2 and R5 and R 6 are hydrogen.

6. The compound of Claim 1 wherein R10 and R12 taken together form -CH=CH-. 7. The compound of Claim 1 wherein a is 1 or 2.

8. The compound of Claim 1 wherein the compound is a compound of formula (IV):

(TV)

wherein:

R1 is selected from Ci-6alkyl optionally substituted with hydroxy or methoxy, tetrahydropyran and phenyl;

R2 is hydrogen;

R3 is -C(0)OCi_6alkyl;

R7 is selected from fluoro, chloro, -CF3; and -OCF3;

R11 is selected from Ci-6alkyl, C3_6cycloalkyl, and wherein Ci-6alkyl is optionally substituted with -ORh and C3_6cycloalkyl is optionally substituted with one or two Ci-3alkyl;

R10 is hydrogen, chloro, or fluoro; and

a is 1 or 2;

or a pharmaceutically-acceptable salt thereof.

9. The compound of Claim 8 wherein R11 is selected from tert-butyl, cyclopropyl, 2,2-dimethylcyclopropyl, cyclobutyl, 2,2-dimethylcyclobutyl, 3-hydroxy- 2,2-dimethylpropyl, and tert-butoxy.

10. The compound of Claim 1 wherein the compound is a compound of formula (V):

wherein:

R1 is selected from Ci-6alkyl optionally substituted with hydroxy or methoxy, tetrahydropyran and phenyl;

R2 is hydrogen;

R3 is -C(0)OCi_6alkyl;

R7 is selected from fluoro, chloro, -CF3; and -OCF3;

R11 is selected from Ci-6alkyl, C3_6cycloalkyl, and wherein Ci-6alkyl is optionally substituted with -ORh and C3_6cycloalkyl is optionally substituted with one or two Ci-3alkyl;and

a is 1 or 2;

or a pharmaceutically-acceptable salt thereof.

11. The compound of Claim 10 wherein R11 is selected from tert-butyl, cyclopropyl, 2,2-dimethylcyclopropyl, cyclobutyl, 2,2-dimethylcyclobutyl, 3-hydroxy- 2,2-dimethylpropyl, and tert-butoxy.

The compound of Claim 1 wherein the compound is selected from

and pharmaceutically-acceptable salts thereof.

13. A pharmaceutical composition comprising a compound of any one of Claims 1 to 12 and a pharmaceutically-acceptable carrier.

14. The pharmaceutical composition of Claim 13 further comprising one or more other therapeutic agents useful for treating hepatitis C viral infections.

15. The pharmaceutical composition of Claim 14 wherein the one or more other therapeutic agents is selected from HCV NS3 protease inhibitors, and HCV NS5B nucleoside and non-nucleoside polymerase inhibitors.

16. A compound of any one of Claims 1 to 12 for use in therapy. 17. A compound of any one of Claims 1 to 12 for use in the treatment of a hepatitis C viral infection in a mammal.

18. A compound of Claim 17 for use in combination with one or more other therapeutic agents.

19. The compound of Claim 18 wherein the one or more other therapeutic agents is selected from HCV NS3 protease inhibitors, HCV NS5B nucleoside and non- nucleoside polymerase inhibitors, interferons and pegylated interferons, cyclophilin inhibitors, HCV NS5A inhibitors, and ribavirin and related nucleoside analogs.

20. A compound of any one of Claims 1 to 12 for use in inhibiting replication of the hepatitis C virus in a mammal.

21. The compound of Claim 20 for use in combination with one or more other therapeutic agents selected from HCV NS3 protease inhibitors, HCV NS5B nucleoside and non-nucleoside polymerase inhibitors, interferons and pegylated interferons, cyclophilin inhibitors, HCV NS5A inhibitors, and ribavirin and related nucleoside analogs.

22. A method of treating hepatitis C viral infection in a mammal, the method comprising administering to the mammal a pharmaceutical composition comprising a compound of any one of Claims 1 to 12 and a pharmaceutically-acceptable carrier.

23. The method of Claim 22 wherein the method further comprises administering one or more other therapeutic agents useful for treating hepatitis C viral infections.

24. The method of Claim 23 wherein the one or more other therapeutic agents is selected from HCV NS3 protease inhibitors, HCV NS5B nucleoside and non- nucleoside polymerase inhibitors, interferons and pegylated interferons, cyclophilin inhibitors, HCV NS5A inhibitors, and ribavirin and related nucleoside analogs.

25. A method of inhibiting replication of the hepatitis C virus in a mammal, the method comprising administering to the mammal a pharmaceutical composition comprising a compound of any one of Claims 1 to 12 and a pharmaceutically-acceptable carrier.

26. The method of Claim 25 wherein the method further comprises administering to the mammal one or more other therapeutic agents useful for inhibiting replication of the hepatitis C virus in a mammal.

27. The method of Claim 26 wherein the one or more other therapeutic agents is selected from HCV NS3 protease inhibitors, HCV NS5B nucleoside and non- nucleoside polymerase inhibitors, interferons and pegylated interferons, cyclophilin inhibitors, HCV NS5A inhibitors, and ribavirin and related nucleoside analogs.

Description:
PIPERAZINE-PIPERIDINE COMPOUNDS AS

HEPATITIS C VIRUS INHIBITORS

BACKGROUND OF THE INVENTION

Field of the Invention

The invention is directed to compounds useful as inhibitors of replication of the hepatitis C virus (HCV). The invention is also directed to pharmaceutical compositions comprising such compounds, methods of using such compounds to treat HCV infection, and processes and intermediates useful for preparing such compounds.

State of the Art

Recent estimates place the number of people infected with the hepatitis C virus (HCV) worldwide at more than 170 million, including 3 million people in the United States. The infection rate is thought to be roughly 4 to 5 times that of the human immunodeficiency virus (HIV). While in some individuals, the natural immune response is able to overcome the virus, in the majority of cases, a chronic infection is established, leading to increased risk of developing cirrhosis of the liver and hepatocellular carcinomas. Infection with hepatitis C, therefore, presents a serious public health problem.

Prior to mid-2011 , the accepted standard of care for HCV involved the use of a pegylated interferon which is believed to act by boosting the body's immune response, together with ribavirin. Unfortunately, the course of treatment is lengthy, typically 48 weeks, often accompanied by serious adverse side effects, including depression, flu-like symptoms, fatigue, and hemolytic anemia, and ineffective in up to 50 % of patients. In mid-201 1, two HCV protease inhibitors were approved in the United States to be used in combination with interferon and ribavirin. Although better cure rates have been reported, the course of therapy is still lengthy and accompanied by undesirable side effects.

Accordingly, there remains a serious unmet need in HCV treatment.

The virus responsible for HCV infection has been identified as a positive-strand RNA virus belonging to the family Flaviviridae. The HCV genome encodes a polyprotein that during the viral lifecycle is cleaved into ten individual proteins, including both structural and non-structural proteins. The six non-structural proteins, denoted as NS2, NS3, NS4A, NS4B, NS5A, and NS5B have been shown to be required for RNA replication. In particular, the NS5A protein appears to play a significant role in viral replication, as well as in modulation of the physiology of the host cell. Effects of NS5A on interferon signaling, regulation of cell growth and apoptosis have also been identified. (Macdonald et al, Journal of General Virology (2004), 85, 2485-2502.) Compounds which inhibit the function of the NS5A protein are expected to provide a new approach to HCV therapy.

SUMMARY OF THE INVENTION

In one aspect, the invention provides novel compounds which inhibit replication of the HCV virus.

Accordingly, the invention provides a compound of formula (I):

(I)

wherein

R 1 is selected from Ci- 6 alkyl, Ci- 6 alkoxy, phenyl, C3_ 6 cycloalkyl, heterocycle, and heteroaryl, wherein Ci- 6 alkyl is optionally substituted with -OR a amino, -SR e , heterocycle, or heteroaryl, Ci- 6 alkoxy is optionally substituted with -OR a , and heterocycle is optionally substituted with -OR a amino, or -C(0)OCi- 6 alkyl, or with one or two Ci -3 alkyl;

R 2 is selected from hydrogen and Ci- 6 alkyl;

R 3 is selected from hydrogen, Ci_ 6 alkyl, -C(0)OC 1-6 alkyl, -C(0)NR m R n ,

-C(0)C 3 - 6 cycloalkyl, and -S(0) 2 C 1-3 alkyl;

R 4 , R 5 , and R 6 are each hydrogen; or R 4 is selected from C 1-6 alkyl, -NR b R c , -OR d , -CN, -C(0)NR a R b , and halo; and R 5 and R 6 are hydrogen;

or R 4 and R 5 are independently Ci_ 6 alkyl or halo and R 6 is hydrogen;

or R 4 and R 5 taken together form -0-(CH 2 ) 2 -0- and R 6 is hydrogen;

or R 4 is hydrogen and R 5 and R 6 taken together form -(CH 2 ) n - wherein n is 1, 2,

3, or 4;

or R 4 and R 5 are hydrogen, and R 6 is Ci- 6 alkyl,

R 7 is selected from halo, Ci_ 3 alkyl, and wherein Ci_ 3 alkyl and Ci- 3 alkoxy are optionally substituted with one, two, three, four, or five halo;

a is 0, 1, or 2;

R 8 is Ci- 3 alkyl optionally substituted with -OR h ;

b is 0, 1 or 2;

or when b is 2, two R 8 can be joined to form -(CH 2 ) 2 -;

R 9 is Ci_ 3 alkyl;

c is 0, 1 or 2;

R 10 is hydrogen, halo, or substituted with one, two, or three halo, or with

-OR h ;

R 11 is selected from Ci- 6 alkyl, C 3 _iocycloalkyl, Ci- 6 alkoxy, -NR f R s , heteroaryl, heterocycle, and -CH 2 -heteroaryl;

wherein:

Ci-6alkyl is optionally substituted with one or two substituents independently selected from -OR h , -NR j R k , and phenyl;

Ci_ 6 alkoxy is optionally substituted with -OR h or with phenyl;

any C 3 _iocycloalkyl is optionally substituted with one or two substituents independently selected from -CD 3 , halo, and -OR h ;

any heterocycle is optionally substituted with one, two, or three substituents independently selected from halo, -C(0)OCi_ 3 alkyl, and -C(0)Ci- 6 alkyl, wherein any -C(0)Ci_ 6 alkyl is optionally substituted with -NHC(0)OCi_ 3 alkyl;

any heteroaryl is optionally substituted with one or two

R 12 is hydrogen or R 10 and R 12 taken together form -CH=CH- or -(CH 2 ) 2 -;

R a , R b , R c , R d , R e , R f , R h , R j , R m , and R n are each independently hydrogen or d -3 alkyl;

R s is selected from hydrogen, Ci- 6 alkyl, and C 3 - 6 cycloalkyl; R k is selected from hydrogen, -C(0)d_ 3 alkyl, -C(0)OC 1-3 alkyl, -C(0)ONR b R c ; and -C(0)NR b R c ; and

X is N and Y is CH or X is CH and Y is N;

provided that when X is CH, b is 0 and when Y is CH, c is 0;

or a pharmaceutically-acceptable salt or stereoisomer thereof.

As used hereinafter, the phrase "compound of formula (I)" means a compound of formula (I) or a pharmaceutically acceptable salt thereof; i.e., this phrase means a compound of formula (I) in free base form or in a pharmaceutically acceptable salt form unless otherwise indicated.

The invention also provides a pharmaceutical composition comprising a compound of the invention and a pharmaceutically-acceptable carrier. In addition, the invention provides a pharmaceutical composition comprising a compound of the invention, a pharmaceutically-acceptable carrier and one or more other therapeutic agents useful for treating hepatitis C viral infections.

The invention also provides a method of treating a hepatitis C viral infection in a mammal, the method comprising administering to the mammal a therapeutically effective amount of a compound or of a pharmaceutical composition of the invention. In addition, the invention provides a method of treating a hepatitis C viral infection in a mammal, the method comprising administering to the mammal a compound or a pharmaceutical composition of the invention and one or more other therapeutic agents useful for treating hepatitis C viral infections. Further, the invention provides a method of inhibiting replication of the hepatitis C virus in a mammal, the method comprising administering a compound or a pharmaceutical composition of the invention.

In separate and distinct aspects, the invention also provides synthetic processes and intermediates described herein, which are useful for preparing compounds of the invention.

The invention also provides a compound of the invention as described herein for use in medical therapy, as well as the use of a compound of the invention in the manufacture of a formulation or medicament for treating a hepatitis C viral infection in a mammal.

DETAILED DESCRIPTION OF THE INVENTION

Among other aspects, the invention provides inhibitors of HCV replication of formula (I), pharmaceutically-acceptable salts thereof, and intermediates for the preparation thereof. The following substituents and values are intended to provide representative examples of various aspects of this invention. These representative values are intended to further define such aspects and are not intended to exclude other values or limit the scope of the invention.

In a specific aspect, R 1 is selected from Ci- 6 alkyl, Ci_ 6 alkoxy, phenyl,

C3_ 6 cycloalkyl, heterocycle, and heteroaryl, wherein Ci- 6 alkyl is optionally substituted with -OR a amino, -SR e , heterocycle, or heteroaryl, Ci- 6 alkoxy is optionally substituted with -OR a , and heterocycle is optionally substituted with -OR a amino,

or -C(0)OCi_ 6 alkyl, or with one or two Ci_ 3 alkyl.

In another specific aspect, R 1 is selected from Ci_ 6 alkyl, phenyl, and heterocycle, wherein Ci_ 6 alkyl is optionally substituted with -OR a , and heterocycle has six ring atoms and is optionally substituted with -OR a or amino or with one or two methyl.

In another specific aspect, R 1 is selected from Ci_ 6 alkyl, phenyl, and

tetrahydropyranyl, wherein Ci_ 6 alkyl is optionally substituted with -OR a ; wherein R a is hydrogen or Ci_ 3 alkyl.

In another specific aspect, R 1 is Ci- 6 alkyl, optionally substituted with hydroxy or methoxy, or tetrahydropyran. In another specific aspect, R 1 is selected from Ci_ 6 alkyl and phenyl.

In a specific aspect, R 1 is C 1-3 alkyl.

In another specific aspect, R 1 is isopropyl.

In yet another specific aspect, R 1 is phenyl.

In still another specific aspect, R 1 is tetrahydropyranyl.

In still another specific aspect, R 1 is tetrahydropyran-4-yl.

In a specific aspect, R 2 is hydrogen or Ci- 6 alkyl.

In other specific aspects, R 2 is hydrogen or Ci_ 3 alkyl; or R 2 is hydrogen.

In a specific aspect, R 3 is selected from hydrogen, Ci- 6 alkyl,

-C(0)OCi ealkyl, -C(0)NR m R n , -C(0)C 3 _ 6 cycloalkyl, and -S(0) 2 Ci_ 3 alkyl;

In another specific aspect, R 3 is selected from hydrogen, Ci_ 6 alkyl,

and -C(0)OCi_ 6 alkyl.

In yet another specific aspect, R 3 is -C(0)OCi_ 3 alkyl.

In a specific aspect, R 1 is Ci- 6 alkyl, R 2 is hydrogen, and R 3 is -C(0)OCi_ 6 alkyl. In another specific aspect, R 1 is isopropyl, R 2 is hydrogen, and R 3 is -C(0)OCH 3 . In yet other specific aspects, R 1 is phenyl, R 2 is hydrogen, and R 3

is -C(0)OCi_ 3 alkyl; or R 1 is tetrahydropyranyl, R 2 is hydrogen, and R 3

is -C(0)OCi_ 3 alkyl.

In a specific aspect, R 4 , R 5 , and R 6 are each hydrogen; or R 4 is selected from Ci_ 6 alkyl, -NR b R c , -OR d , -CN, -C(0)NR a R b and halo; and R 5 and R 6 are hydrogen; or R 4 and R 5 are independently Ci- 6 alkyl or halo and R 6 is hydrogen; or R 4 and R 5 taken together form -0-(CH 2 ) 2 -0- and R 6 is hydrogen; or R 4 is hydrogen and R 5 and R 6 taken together form -(CH 2 ) n -, wherein n is 1, 2, 3, or 4; or R 4 and R 5 are hydrogen, and R 6 is Ci- 6 alkyl.

In a specific aspect, R 4 , R 5 , and R 6 are each hydrogen.

In another specific aspect, R 4 is selected from Ci_ 6 alkyl, -OR d , halo,

and -C(0)NR a R b and R 5 and R 6 are hydrogen.

In another specific aspect, R 4 is selected from methyl, methoxy, fluoro, and -C(0)NH 2 , and R 5 and R 6 are hydrogen.

In atill another aspect, R 4 is methyl or methoxy, and R 5 and R 6 are hydrogen.

In a further aspect, R is methyl and R and R are hydi

IInn another specific aspect, R 4 and R 5 are independently Ci- 6 alkyl and R 6 is hydrogen

In yet another specific aspect, R 4 and R 5 taken together form -0-(CH 2 ) 2 -0- and R 6 is hydrogen.

In additional aspects, R 4 is hydrogen and R 5 and R 6 taken together form -(CH2) n - wherein n is 1 or 4; or R 4 is hydrogen and R 5 and R 6 taken together form -(CH 2 ) 4 -

In still further aspects, R 4 and R 5 are each hydrogen and R 6 is Ci- 6 alkyl, or R 4 and R 5 are each hydrogen and R 6 is methyl.

In a specific aspect, R 7 is selected from halo, wherein

Ci- 6 alkyl and C^alkoxy are optionally substituted with one, two, three, four, or five halo.

In another specific aspect R 7 is selected from halo, Ci_ 3 alkyl, and

wherein Ci- 6 alkyl and Ci- 6 alkoxy are substituted with one, two, or three halo.

In yet another specific aspect, R 7 is selected from fluoro, chloro, -CF 3; and -OCF 3 . In still another aspect, R 7 is -OCF 3 .

In a specific aspect, R 8 is Ci_ 3 alkyl.

In another specific aspect, R 8 is methyl.

In a specific aspect, R 9 is Ci_ 3 alkyl.

In another specific aspect, R 9 is methyl. In a specific aspect, R is hydrogen, halo, or Ci_ 3 alkyl substituted with one, two, or three halo, or with -OR h .

In other specific aspects, R 10 is hydrogen or halo; or R 10 is hydrogen, chloro, or fluoro; or R 10 is hydrogen or -CH 2 OH.

In another specific aspect, R 10 is chloro.

In yet another specific aspect, R 10 is hydrogen.

In a still further aspect, R 10 is -CH 2 OH.

In a specific aspect, R 11 is defined as in formula (I).

In another specific aspect, R 11 is selected from Ci- 6 alkyl, C3_ 6 cycloalkyl,

Ci_ 6 alkoxy, -NR f R s , heteroaryl, heterocycle, and -CH2-heteroaryl; wherein any heteroaryl or heterocycle has 5 or 6 ring atoms; Ci- 6 alkyl is optionally substituted with one or two substituents independently selected from -OR h and -NR j R k ; any C3-6cycloalkyl is optionally substituted with one or two Ci_ 3 alkyl or halo; any heterocycle is optionally substituted with one, or two substituents independently selected from

and -C(0)Ci- 6 alkyl, wherein any -C(0)Ci_ 6 alkyl is optionally substituted

with -NHC(0)OCi_ 3 alkyl,

In yet another aspect, R 11 is selected from Ci- 6 alkyl, C3_ 6 cycloalkyl, and

Ci_ 6 alkoxy wherein Ci_ 6 alkyl is optionally substituted with -OR h and C3_ 6 cycloalkyl is optionally substituted with one or two

In still another aspect, R 11 is selected from tert-butyl, cyclopropyl,

2,2-dimethylcyclopropyl, cyclobutyl, 2,2-dimethylcyclobutyl, 3 -hydroxy -2,2- dimethylpropyl, and tert-butoxy.

In a specific aspect, R 12 is hydrogen.

In another specific aspect, R 10 and R 12 taken together form -CH=CH- or -(CH2)2-.

In another specific aspect, R 10 and R 12 taken together form -CH=CH-.

In a specific aspect, a is 0, 1, or 2.

In another specific aspect, a is 1 or 2.

In another specific aspect, a is 1.

In a specific aspect, b is 0, 1, or 2.

In other specific aspects, b is 0 or b is 1.

In a specific aspect, c is 0, 1, or 2.

In other specific aspects, c is 0 or c is 1.

In a specific aspect, X is N, Y is CH, and c is 0.

In another specific aspect, X is CH, Y is N, and b is 0. In another aspect, the invention provides compounds of formula (II):

(II)

wherein the variables of formula (II) are as defined herein.

In another aspect, the invention provides compounds of formula (III)

(HI)

wherein the variables of formula (III) are as defined herein.

In yet another aspect, the invention provides compounds of formula (IV):

(IV)

wherein:

R 1 is selected from Ci- 6 alkyl optionally substituted with hydroxy or methoxy, tetrahydropyran and phenyl; R 2 is hydrogen; and R 3 is -C(0)OCi_ 6 alkyl;

R 2 is hydrogen; and R 3 is -C(0)OCi_ 6 alkyl;

R 7 is selected from fluoro, chloro, -CF 3; and -OCF 3 ;

R 11 is selected from Ci- 6 alkyl, C3_ 6 cycloalkyl, and wherein Ci- 6 alkyl is optionally substituted with -OR h and C3- 6 cycloalkyl is optionally substituted with one or two Ci_ 3 alkyl; R 10 is hydrogen, chloro, or fluoro; and

a is 1 or 2;

or a pharmaceutically-acceptable salt thereof.

Included within this aspect are compounds wherein R 11 is selected from tert-butyl, cyclopropyl, 2,2-dimethylcyclopropyl, cyclobutyl, 2,2-dimethylcyclobutyl, 3-hydroxy- 2,2-dimethylpropyl, and tert-butoxy.

In yet another aspect, the invention provides compounds of formula (V):

(V)

wherein:

R 1 is selected from Ci- 6 alkyl optionally substituted with hydroxy or methoxy, tetrahydropyran and phenyl; R 2 is hydrogen; and R 3 is -C(0)OCi_ 6 alkyl;

R 7 is selected from fluoro, chloro, -CF3, and -OCF3;

R 11 is selected from Ci- 6 alkyl, C3_ 6 cycloalkyl, and Ci- 6 alkoxy wherein Ci- 6 alkyl is optionally substituted with -OR h and C3- 6 cycloalkyl is optionally substituted with one or two Ci_ 3 alkyl; and

a is 1 or 2;

or a pharmaceutically-acceptable salt thereof.

Included within this aspect are compounds wherein R 11 is selected from tert-butyl, cyclopropyl, 2,2-dimethylcyclopropyl, cyclobutyl, 2,2-dimethylcyclobutyl, 3-hydroxy- 2,2-dimethylpropyl, and tert-butoxy.

In one aspect, the invention provides the compounds of Examples 1-17 and Tables 1-9 below

In another aspect, the invention provides a compound selected from the following compounds

and pharmaceutically-acceptable salts thereof.

The chemical naming convention used herein is illustrated for the compound of Example 7:

which is [(5)-l-((25,45)-2-{5-Chloro-4-[4'-({4-[l-(2,2-dimethyl-propi onyl)-piperidin- yl]-piperazine-l-carbonyl}-amino)-2'-trifluoromethoxy-biphen yl-4-yl]-lH-imidazol-2 yl}-4-methyl-pyrrolidine-l-carbonyl)-2-methyl-propyl]-carbam ic acid methyl ester according to the IUPAC conventions as implemented in AutoNom software, (MDL Information Systems, GmbH, Frankfurt, Germany).

Furthermore, the imidazole moiety in the structure of formula (I) exists in tautomeric forms, illustrated below for a fragment of the compound of Example 2

A B

According to the IUPAC convention, these representations give rise to different numbering of the atoms of the imidazole moiety: {(5)-2-methyl-l-[(25',45)-4-methyl-2-(4 I phenyl- lH-imidazol-2-yl -pyrrolidine- 1 -carbonyll -propyl} -carbamic acid methyl ester (structure A) vs. {(5)-2-methyl-l-[(25,45)-4-methyl-2-(5 pJienyJ-lH-imidazol-2-yl)- pyrrolidine-l-carbonyl]-propyl}-carbamic acid methyl ester (structure B).

Similarly, the napthimidazole structure where the variables R 10 and R 12 taken together form -CH=CH-, illustrated for a fragment of the compound of Example 15, exists in tautomeric forms:

D

which also give rise to different numbering: {(5)-2-methyl-l-[(25',45 , )-4-methyl-2-(3H- naphtho[l,2-d]imidazol-2-yl)-pyrrolidine-l-carbonyl]-propyl} -carbamic acid methyl ester (structure C) vs. {(5')-2-methyl-l-[(25 , ,45 , )-4-methyl-2-(lH-naphtho[l,2-d]imidazol-2-yl)- pyrrolidine-l-carbonyl]-propyl}-carbamic acid methyl ester (structure D). It will be understood that although structures are shown, or named, in a particular form, the invention also includes the tautomer thereof.

The compounds of the invention contain one or more chiral centers and therefore, such compounds (and intermediates thereof) can exist as racemic mixtures; pure stereoisomers (i.e., enantiomers or diastereomers); stereoisomer-enriched mixtures and the like. Chiral compounds shown or named herein without a defined stereochemistry at a chiral center are intended to include any or all possible stereoisomer variations at the undefined stereocenter unless otherwise indicated. The depiction or naming of a particular stereoisomer means the indicated stereocenter has the designated

stereochemistry with the understanding that minor amounts of other stereoisomers may also be present unless otherwise indicated, provided that the utility of the depicted or named compound is not eliminated by the presence of another stereoisomer.

Compounds of formula (I) also contain several basic groups (e.g., amino groups) and therefore, such compounds can exist as the free base or in various salt forms, such a mono-protonated salt form, a di-protonated salt form, a tri-protonated salt form, or mixtures thereof. All such forms are included within the scope of this invention, unless otherwise indicated.

This invention also includes isotopically-labeled compounds of formula (I), i.e., compounds of formula (I) where an atom has been replaced or enriched with an atom having the same atomic number but an atomic mass different from the atomic mass that predominates in nature. Examples of isotopes that may be incorporated into a compound of formula (I) include, but are not limited to, ¾, 3 H, U C, 13 C, 14 C, 13 N, 15 N, 15 0, 17 0, 18 0, 35 S, 36 C1, and 18 F. Of particular interest are compounds of formula (I) enriched in tritium or carbon-14, which compounds can be used, for example, in tissue distribution studies. Also of particular interest are compounds of formula (I) enriched in deuterium especially at a site of metabolism, which compounds are expected to have greater metabolic stability. Additionally of particular interest are compounds of formula (I) enriched in a positron emitting isotope, such as n C, 18 F, 15 0 and 13 N, which compounds can be used, for example, in Positron Emission Tomography (PET) studies.

Definitions

When describing this invention including its various aspects and embodiments, the following terms have the following meanings, unless otherwise indicated.

The term "alkyl" means a monovalent saturated hydrocarbon group which may be linear or branched or combinations thereof. Unless otherwise defined, such alkyl groups typically contain from 1 to 10 carbon atoms. Representative alkyl groups include, by way of example, methyl (Me), ethyl (Et), w-propyl (n-Pr) or (nPr), isopropyl (i-Pr) or (iPr), M-butyl (n-Bu) or (nBu), sec -butyl, isobutyl, tert-butyl (t-Bu) or (tBu), w-pentyl, w-hexyl, 2,2-dimethylpropyl, 2-methylbutyl, 3-methylbutyl, 2-ethylbutyl, 2,2-dimethylpentyl, 2-propylpentyl, and the like

When a specific number of carbon atoms are intended for a particular term, the number of carbon atoms is shown preceding the term. For example, the term "C 1-3 alkyl" means an alkyl group having from 1 to 3 carbon atoms wherein the carbon atoms are in any chemically-acceptable configuration, including linear or branched configurations..

The term "alkoxy" means the monovalent group -O-alkyl, where alkyl is defined as above. Representative alkoxy groups include, by way of example, methoxy, ethoxy, propoxy, butoxy, and the like.

The term "cycloalkyl" means a monovalent saturated carbocyclic group which may be monocyclic or multicyclic. Unless otherwise defined, such cycloalkyl groups typically contain from 3 to 10 carbon atoms. Representative cycloalkyl groups include, by way of example, cyclopropyl (cPr), cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, adamantyl, and the like.

The term "heterocycle", "heterocyclic", or "heterocyclic ring" means a monovalent saturated or partially unsaturated cyclic non-aromatic group, having from 3 to

10 total ring atoms, wherein the ring contains from 2 to 9 carbon ring atoms and from 1 to

4 ring heteroatoms selected from nitrogen, oxygen, and sulfur. Heterocyclic groups may be monocyclic or multicyclic (i.e., fused or bridged). Representative heterocyclic groups include, by way of example, pyrrolidinyl, piperidinyl, piperazinyl, imidazolidinyl, morpholinyl, thiomorpholyl, indolin-3-yl, 2-imidazolinyl, tetrahydropyranyl, 1,2,3,4- tetrahydroisoquinolin-2-yl, quinuclidinyl, 7-azanorbornanyl, nortropanyl, and the like, where the point of attachment is at any available carbon or nitrogen ring atom. Where the context makes the point of attachment of the heterocyclic group evident, such groups may alternatively be referred to as a non-valent species, i.e. pyrrolidine, piperidine, piperazine, imidazole, etc.

The term "heteroaryl" or "heteroaryl ring" means a monovalent aromatic group having from 5 to 10 total ring atoms, wherein the ring contains from 1 to 9 carbon ring atoms and from 1 to 4 ring heteroatoms selected from nitrogen, oxygen, and sulfur.

Heteroaryl groups may be monocyclic or multicyclic. Representative heteroaryl groups include, by way of example, pyrroyl, isoxazolyl, isothiazolyl, pyrazolyl, oxazolyl, oxadiazolyl, thiadiazolyl, thiazolyl, imidazolyl, triazolyl, tetrazolyl, furanyl, triazinyl, thienyl, pyridyl (or, equivalently, pyridinyl), pyrimidyl, pyridazinyl, pyrazinyl, indolyl, benzofuranyl, benzothienyl, benzimidazolyl, benzthiazolyl, and the like, where the point of attachment is at any available carbon or nitrogen ring atom. Where the context makes the point of attachment of the heteroaryl group evident, such groups may alternatively be referred to as a non-valent species, i.e.pyrrole, isoxazole, isothiazole, pyrazole, imidazole, etc.

The term "halo" means fluoro, chloro, bromo or iodo.

The term "therapeutically effective amount" means an amount sufficient to effect treatment when administered to a patient in need of treatment.

The term "treatment" as used herein means the treatment of a disease, disorder, or medical condition in a patient (such as hepatitis C viral infection), such as a mammal (particularly a human) which includes one or more of the following:

(a) preventing the disease, disorder, or medical condition from occurring, i.e., preventing the reoccurrence of the disease or medical condition or prophylactic treatment of a patient that is pre-disposed to the disease or medical condition;

(b) ameliorating the disease, disorder, or medical condition, i.e., eliminating or causing regression of the disease, disorder, or medical condition in a patient, including counteracting the effects of other therapeutic agents;

(c) suppressing the disease, disorder, or medical condition, i.e., slowing or arresting the development of the disease, disorder, or medical condition in a patient; or

(d) alleviating the symptoms of the disease, disorder, or medical condition in a patient. The term "pharmaceutically acceptable salt" means a salt that is acceptable for administration to a patient or a mammal, such as a human (e.g., salts having acceptable mammalian safety for a given dosage regime). Representative pharmaceutically acceptable salts include salts of acetic, ascorbic, benzenesulfonic, benzoic,

camphorsulfonic, citric, ethanesulfonic, edisylic, fumaric, gentisic, gluconic, glucoronic, glutamic, hippuric, hydrobromic, hydrochloric, isethionic, lactic, lactobionic, maleic, malic, mandelic, methanesulfonic, mucic, naphthalenesulfonic, naphthalene- 1,5- disulfonic, naphthalene-2,6-disulfonic, nicotinic, nitric, orotic, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic and xinafoic acid, and the like.

The term "salt thereof means a compound formed when the hydrogen of an acid is replaced by a cation, such as a metal cation or an organic cation and the like. For example, the cation can be a protonated form of a compound of formula (I), i.e. a form where one or more amino groups have been protonated by an acid. Typically, the salt is a pharmaceutically acceptable salt, although this is not required for salts of intermediate compounds that are not intended for administration to a patient.

The term "amino-protecting group" means a protecting group suitable for preventing undesired reactions at an amino nitrogen. Representative amino-protecting groups include, but are not limited to, formyl; acyl groups, for example alkanoyl groups, such as acetyl and tri-fluoroacetyl; alkoxycarbonyl groups, such as tert butoxycarbonyl (Boc); arylmethoxycarbonyl groups, such as benzyloxycarbonyl (Cbz) and

9-fluorenylmethoxycarbonyl (Fmoc); arylmethyl groups, such as benzyl (Bn), trityl (Tr), and l,l-di-(4'-methoxyphenyl)methyl; silyl groups, such as trimethylsilyl (TMS), tert- butyldimethylsilyl (TBDMS), [2-(trimethylsilyl)ethoxy]methyl (SEM); and the like. Numerous protecting groups, and their introduction and removal, are described in T. W. Greene and G. M. Wuts, Protecting Groups in Organic Synthesis, Third Edition, Wiley, New York.

General Synthetic Procedures

Compounds of this invention, and intermediates thereof, can be prepared according to the following general methods and procedures using commercially-available or routinely-prepared starting materials and reagents. The substituents and variables (e.g.,

R 1 , R 2 , R 3 , R 4 , etc.) used in the following schemes have the same meanings as those defined elsewhere herein unless otherwise indicated. Additionally, compounds having an acidic or basic atom or functional group may be used or may be produced as a salt unless otherwise indicated (in some cases, the use of a salt in a particular reaction will require conversion of the salt to a non-salt form, e.g., a free base, using routine procedures before conducting the reaction).

Although a particular embodiment of the present invention may be shown or described in the following procedures, those skilled in the art will recognize that other embodiments or aspects of the present invention can also be prepared using such procedures or by using other methods, reagents, and starting materials know to those skilled in the art. In particular, it will be appreciated that compounds of the invention may be prepared by a variety of process routes in which reactants are combined in different orders to provide different intermediates en route to producing final products.

In one exemplary method of synthesis, compounds of formula (1-6) in which X is

N and Y is CH are prepared as shown in Scheme 1 :

Scheme 1

where G represents the group

L and L' represent leaving groups, and Pg represents an amino-protecting group. For example, 1 , l'-carbonyldiimidazole (CDI) or 4-nitrophenyl chloro formate or triphospgene may be used as the acylating agent, L-C(0)-L'. Aniline intermediate 1-1 is reacted with the acylating agent to form the adduct 1-2 which is reacted with a protected piperazine- piperidine intermediate 1-3 to form a protected intermediate 1-4. Intermediate 1-4 is then deprotected, for example, by treatment with an acid to provide compound 1-5, which is reacted with an acid chloride in the presence of base or with a carboxylic acid under amide bond formation conditions to prepare a compound of the invention of formula 1-6. The amide bond formation reaction may utilize coupling agents such as Ν,Ν,Ν',Ν'- tetramethyl-0-(7-azabenzotriazol-l-yl)uronium hexafluorophosphate (HATU), or as 1,3- dicyclohexylcarbodiimide (DCC), l-(3-dimethylaminopropyl)-3-ethylcarbodiimide (EDC), or other coupling agents known in the art.

Protected intermediate 1-4 may also be conveniently prepared by alternative processes, for example, by the Suzuki coupling reaction in the presence of a palladium catalyst (Miyaura and Suzuki, Chem. Rev. 1995, 95, 2457-2483). As shown in Scheme 2 below, either coupling partner may bear the boronate moiety. Alternatively, a boronic acid reagent may be used in place of a boronate reagent, such as the pinacol boronate depicted below.

Scheme 2

If protected intermediate 2-2 were replaced by an intermediate 2-2'

then the Suzuki coupling of the boronate 2-1 in the first line of Scheme 2 would directly provide a final compound of the invention. Similarly, if the protected intermediate 2-4 were replaced by an intermediate 2-4',

then the Suzuki coupling of the bromo intermediate 2-3 in the second line of Scheme 2 would directly provide a final compound of the invention.

As described in the examples below, napthimidazole compounds of the invention, of formula (V), may be prepared by a Suzuki coupling reaction analogous to that of the first line of Scheme 2, in which a suitable boronate intermediate corresponding to 2-1 is reacted with intermediate 2-2.

Compounds of the invention in which R 10 is chloro or fluoro are conveniently prepared by the Suzuki reaction of an intermediate 2-3'

which may be prepared by reacting intermediate 2-3 with a chlorinating agent, such as N-chlorosuccinimide, or with a fluorinating agent, such as l-fluoro-4-hydroxy-l,4- diazoniabicyclo [2.2.2] octane bis(tetrafluoroborate) .

The bromo intermediate 2-2 may be prepared, for example, by reactions analogous to the acylation and leaving group replacement steps of Scheme 1 as shown in Scheme 3 below. Scheme 3

3-1 3-2 1-3

The protected piperazine-piperidine intermediate 1-3 may be prepared by the reductive amination of a protected piperazine with a protected carboxy piperidine as shown in Scheme 4 below where Pg' and Pg represent amino-protecting groups removable under different conditions. For example, to provide an intermediate 1-3 where Pg is Boc, it is useful to use Cbz as protecting group Pg'.

Scheme 4

Pg,-

4-1 4-2 4-3 1-3

Compounds of the invention in which X is CH and Y is N may be prepared by reactions analogous to the reactions of Schemes 1 to 4 in which the intermediate 1-3 is replaced by a protected piperidine-piperazine intermediate 5-4 which may be prepared as shown in Scheme 5.

Scheme 5

5-1 5-2 5-3 5-4

The biaryl aniline intermediate 1-1 may be prepared by the Suzuki coupling reaction of Scheme 6, where, as shown, either coupling partner may bear the boronate moiety. Scheme 6

Intermediates 6-1 and 6-3 used in the Suzuki reaction of Scheme 6 may be prepared, for example, as shown in Schemes 7 and 8.

Scheme 7

Reagent 7-1, where X represents bromo or chloro is reacted with a protected proline carboxylic acid 7-2 to provide intermediate 7-3 which is converted to intermediate 7-4 in the presence of an excess of ammonium acetate. The ring closure reaction typically is performed at a temperature between about 100 °C and about 120 °C for a period of about 4 to about 24 hours. To provide compound 6-1, intermediate 7-4 is typically deprotected and coupled with a reagent HO-G to provide compound 6-1.

Finally, to provide boronate intermediate 6-3, intermediate 6-1 is reacted with 8-1 in the presence of a palladium catalyst as shown in Scheme 8. Scheme 8

Details regarding specific reaction conditions and other procedures for preparing representative compounds of the invention or intermediates thereto are described in the examples below.

Thus, in one of its method aspects, the invention provides the processes of Schemes 1-5 and variations thereto described above as well as the processes exemplified below.

It will further be understood, this disclosure encompasses compounds of formula (I) when prepared by synthetic processes such as those described above and below or by metabolic processes including those occuring in vivo in human or animal body or in vitro.

Pharmaceutical Compositions

The compounds of the invention and pharmaceutically-acceptable salts thereof are typically used in the form of a pharmaceutical composition or formulation. Such pharmaceutical compositions may be administered to a patient by any acceptable route of administration including, but not limited to, oral, rectal, vaginal, nasal, inhaled, topical (including transdermal) and parenteral modes of administration.

Accordingly, in one of its compositions aspects, the invention is directed to a pharmaceutical composition comprising a pharmaceutically-acceptable carrier or excipient and a compound of formula (I), where, as defined above, "compound of formula (I)" means a compound of formula (I) or a pharmaceutically-acceptable salt thereof. Optionally, such pharmaceutical compositions may contain other therapeutic and/or formulating agents if desired. When discussing compositions and uses thereof, the "compound of the invention" may also be referred to herein as the "active agent". As used herein, the term "compound of the invention" is intended to include all compounds encompassed by formula (I) as well as the species embodied in formulas (II), (III), (IV), and (V) and pharmaceutically-acceptable salts thereof

The pharmaceutical compositions of the invention typically contain a

therapeutically effective amount of a compound of the present invention. Those skilled in the art will recognize, however, that a pharmaceutical composition may contain more than a therapeutically effective amount, i.e., bulk compositions, or less than a therapeutically effective amount, i.e., individual unit doses designed for multiple administration to achieve a therapeutically effective amount.

Typically, such pharmaceutical compositions will contain from about 0.1 to about

95% by weight of the active agent; preferably, from about 5 to about 70% by weight; and more preferably from about 10 to about 60% by weight of the active agent.

Any conventional carrier or excipient may be used in the pharmaceutical compositions of the invention. The choice of a particular carrier or excipient, or combinations of carriers or excipients, will depend on the mode of administration being used to treat a particular patient or type of medical condition or disease state. In this regard, the preparation of a suitable pharmaceutical composition for a particular mode of administration is well within the scope of those skilled in the pharmaceutical arts.

Additionally, the carriers or excipients used in the pharmaceutical compositions of this invention are commercially-available. By way of further illustration, conventional formulation techniques are described in Remington: The Science and Practice of Pharmacy, 20th Edition, Lippincott Williams & White, Baltimore, Maryland (2000); and H.C. Ansel et al, Pharmaceutical Dosage Forms and Drug Delivery Systems, 7th Edition, Lippincott Williams & White, Baltimore, Maryland (1999).

Representative examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, the following: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, such as microcrystalline cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen- free water; isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical compositions.

Pharmaceutical compositions are typically prepared by thoroughly and intimately mixing or blending the active agent with a pharmaceutically-acceptable carrier and one or more optional ingredients. The resulting uniformly blended mixture can then be shaped or loaded into tablets, capsules, pills and the like using conventional procedures and equipment.

The pharmaceutical compositions of the invention are preferably packaged in a unit dosage form. The term "unit dosage form" refers to a physically discrete unit suitable for dosing a patient, i.e., each unit containing a predetermined quantity of active agent calculated to produce the desired therapeutic effect either alone or in combination with one or more additional units. For example, such unit dosage forms may be capsules, tablets, pills, and the like, or unit packages suitable for parenteral administration.

In one embodiment, the pharmaceutical compositions of the invention are suitable for oral administration. Suitable pharmaceutical compositions for oral administration may be in the form of capsules, tablets, pills, lozenges, cachets, dragees, powders, granules; or as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water or water-in-oil liquid emulsion; or as an elixir or syrup; and the like; each containing a predetermined amount of a compound of the present invention as an active ingredient.

When intended for oral administration in a solid dosage form (i.e., as capsules, tablets, pills and the like), the pharmaceutical compositions of the invention will typically comprise the active agent and one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate. Optionally or alternatively, such solid dosage forms may also comprise: fillers or extenders, such as starches, microcrystalline cellulose, lactose, sucrose, glucose, mannitol, and/or silicic acid; binders, such as

carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; humectants, such as glycerol; disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and/or sodium carbonate; solution retarding agents, such as paraffin; absorption accelerators, such as quaternary ammonium compounds; wetting agents, such as cetyl alcohol and/or glycerol monostearate;

absorbents, such as kaolin and/or bentonite clay; lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and/or mixtures thereof; coloring agents; and buffering agents.

Release agents, wetting agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the

pharmaceutical compositions of the invention. Examples of pharmaceutically-acceptable antioxidants include: water-soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfate, sodium sulfite and the like; oil- soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, lecithin, propyl gallate, alpha-tocopherol, and the like; and metal- chelating agents, such as citric acid, ethylenediamine tetraacetic acid, sorbitol, tartaric acid, phosphoric acid, and the like. Coating agents for tablets, capsules, pills and like, include those used for enteric coatings, such as cellulose acetate phthalate, polyvinyl acetate phthalate, hydroxypropyl methylcellulose phthalate, methacrylic acidmethacrylic acid ester copolymers, cellulose acetate trimellitate, carboxymethyl ethyl cellulose, hydroxypropyl methyl cellulose acetate succinate, and the like.

Pharmaceutical compositions of the invention may also be formulated to provide slow or controlled release of the active agent using, by way of example, hydroxypropyl methyl cellulose in varying proportions; or other polymer matrices, liposomes and/or microspheres. In addition, the pharmaceutical compositions of the invention may optionally contain opacifying agents and may be formulated so that they release the active ingredient only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active agent can also be in microencapsulated form, if appropriate, with one or more of the above-described excipients.

Suitable liquid dosage forms for oral administration include, by way of illustration, pharmaceutically-acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. Liquid dosage forms typically comprise the active agent and an inert diluent, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (esp., cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Suspensions, in addition to the active ingredient, may contain suspending agents such as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters,

microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.

The compounds of this invention can also be administered parenterally (e.g. by intravenous, subcutaneous, intramuscular or intraperitoneal injection). For parenteral administration, the active agent is typically admixed with a suitable vehicle for parenteral administration including, by way of example, sterile aqueous solutions, saline, low molecular weight alcohols such as propylene glycol, polyethylene glycol, vegetable oils, gelatin, fatty acid esters such as ethyl oleate, and the like. Parenteral formulations may also contain one or more anti-oxidants, solubilizers, stabilizers, preservatives, wetting agents, emulsifiers, buffering agents, or dispersing agents. These formulations may be rendered sterile by use of a sterile injectable medium, a sterilizing agent, filtration, irradiation, or heat.

Alternatively, the pharmaceutical compositions of the invention are formulated for administration by inhalation. Suitable pharmaceutical compositions for administration by inhalation will typically be in the form of an aerosol or a powder. Such compositions are generally administered using well-known delivery devices, such as a metered-dose inhaler, a dry powder inhaler, a nebulizer or a similar delivery device.

When administered by inhalation using a pressurized container, the

pharmaceutical compositions of the invention will typically comprise the active ingredient and a suitable propellant, such as dichlorodifluoromethane,

trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. Additionally, the pharmaceutical composition may be in the form of a capsule or cartridge (made, for example, from gelatin) comprising a compound of the invention and a powder suitable for use in a powder inhaler. Suitable powder bases include, by way of example, lactose or starch.

The compounds of the invention can also be administered transdermally using known transdermal delivery systems and excipients. For example, the active agent can be admixed with permeation enhancers, such as propylene glycol, polyethylene glycol monolaurate, azacycloalkan-2-ones and the like, and incorporated into a patch or similar delivery system. Additional excipients including gelling agents, emulsifiers and buffers, may be used in such transdermal compositions if desired.

The following non-limiting examples illustrate representative pharmaceutical compositions of the present invention.

Oral solid dosage form

A compound of the invention is dissolved in polyethylene glycol acidified to pH < 2 with optional heating to form a solution comprising 10 % w/w or 40 % w/w active agent. The solution is spray dried to form a powder. The resulting powder is loaded into capsules, for example gelatin or hydroxypropyl methylcellulose capsules, to provide a unit dosage of 14 mg or 56 mg, respectively, active agent per capsule. Oral liquid formulation

A compound of the invention (100 mg) is added to a mixture of ethanol (5 mL), propylene glycol (10 mL), and polyethylene glycol (25 mL). Once dissolution is achieved, acidified distilled water (q.s. to 100 mL) is added to provide a liquid formulation at a concentration of 1 mg/mL active agent.

Lipid emulsion formulation

A lipid emulsion formulation comprising a compound of the invention (10 %), oleic acid (78 %) polyethylene glycol (10 %), and polysorbate 20 (2 %) w/w is formed by adding a compound of the invention to a mixture of the remaining ingredients.

Lipid emulsion formulation

A lipid emulsion formulation comprising a compound of the invention (10 %) and oleic acid (90 %) w/w is formed by adding a compound of the invention to oleic acid.

Micro-emulsion formulation

A compound of the invention (1 g) is dissolved in a mixture of ethanol (2 mL), propylene glycol (2 mL), polyethylene glycol 400 (4 mL), and polyethylene glycol- 15- hydroxystearate (4 mL). Acidified distilled water (q.s. to 100 mL) is added to form a self-emulsifying micro-emulsion formulation.

Utility

The compounds of the invention have been shown to inhibit viral replication in HCV replicon assays and therefore are expected to be useful for the treatment of hepatitis C viral infections.

In one aspect, therefore, the invention provides a method of inhibiting replication of the hepatitis C virus in a mammal (e.g., a human), the method comprising

administering to the mammal a therapeutically-effective amount of a compound of the invention or of a pharmaceutical composition comprising a pharmaceutically-acceptable carrier and a compound of the invention.

The invention further provides a method of treating hepatitis C viral infections in a mammal (e.g., a human), the method comprising administering to the mammal a therapeutically-effective amount of compound of the invention or of a pharmaceutical composition comprising a pharmaceutically-acceptable carrier and a compound of the invention.

The compounds of the invention may inhibit viral replication by inhibiting the function of the NS5A protein encoded by the HCV genome. In one aspect, therefore, the invention provides a method of inhibiting the NS5A protein of HCV in a mammal, the method comprising administering to the mammal, a compound or a composition of the invention.

When used to treat HCV infections, the compounds of the invention will typically be administered orally in a single daily dose or in multiple doses per day, although other forms of administration may be used. The amount of active agent administered per dose or the total amount administered per day will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered and its relative activity, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.

Suitable doses for treating HCV infections will range from about 1 to about 2000 mg/day of active agent, including from about 5 to about 300 mg/day and from about 10 to about 200 mg per day of active agent for an average 70 kg human.

Combination therapy

Compounds of the invention may also be used in combination with one or more agents which act by the same mechanism or by different mechanisms to effect treatment of HCV. Useful classes of agents for combination therapy include, but are not limited to, HCV NS3 protease inhibitors, HCV NS5B nucleoside and non-nucleoside polymerase inhibitors, helicase inhibitors, NS4B protein inhibitors, HCV viral entry inhibitors, cyclophyllin inhibitors, toll-like receptor agonists, inhibitors of heat shock proteins, interfering RNA, antisense RNA, HCV internal ribosome entry site (IRES) inhibitors, thiazolides, nucleoside analogs such as ribavirin and related compounds, interferons and other immunomodulatory agents, inosine 5 '-monophosphate dehydrogenase (IMPDH) inhibitors, and other NS5A protein inhibitors. Agents which act to inhibit HCV replication by any other mechanism may also be used in combination with the present compounds.

HCV NS3 protease inhibitors which may be used in combination therapy include, but are not limited to, Incivek® (telaprevir,VX-950), boceprevir (SCH-503034), simeprevir (TMC-435), narlaprevir (SCH-900518), vaniprevir (MK-7009), danoprevir (ITMN-191, R-7227), BI-201335, ABT-450/r, asunaprevir (BMS-650032), GS-9256, GS-9451, sovaprevir (ACH-1625), ACH-2684, BMS-605339, VX-985, PHX-1766, BMS-791325, IDX-320, and MK-5172.

Examples of HCV NS5B nucleoside polymerase inhibitors include, but are not limited to, mericitabine (RG7128), IDX-184, sofosbuvir (GS-7977, PSI-7977), PSI-7851, PSI-938, BMS-986094 (ΓΝΧ-189, ΓΝΧ-08189), RG7348, MK-0608, TMC-649128, HCV-796, and ALS-2200 (VX-135),while, non-nucleoside HCV NS5B polymerase inhibitors, include but are not limited to, filibuvir (PF-8685540), tegobuvir (GS-9190), VX-222, VX-759, setrobuvir (ANA-598), ABT-072, ABT-333, BI-207127, BMS- 791325, MK-3281, IDX-37, BMS-824393, TMC-647055.

A wide variety of interferons and pegylated interferons, including alpha, beta, omega, and gamma interferons, having antiviral, antiproliferative or immunomodulatory effects, can be combined with the present compounds. Representative examples include, but are not limited to, Intron® A (interferon-alpha2b), Actimmune® (interferon-gamma- lb), Alferon N, Advaferon®, Roferon-A (interferon alpha-2a), Peglntron®

(peginterferon-alpha 2b), Alfaferone, Pegasys® (peginterferon alpha-2a), Alfanative (interferon alpha), Zalbin™ (albinterferon alpha-2b), Infergon® (interferon alfacon-1), Omega DUROS® (omega interferon), Locteron™ (interferon alpha), PEG-rIL-29 (pegylated interferon lambda), and Rebif® (interferon beta- la).

Nucleoside analog antiviral agents include, but are not limited to, ribavirin

(Copegus®, Rebetol®,Virazole®) and Viramidine (taribavirin). Interferons and ribavirin are also provided in in the form of kits which include, for example, but are not limited to, Rebetron® (interferon alpha-2b/ribavirin) and Pegetron® (Peginterferon alpha- 2b/ribavirin).

Useful compounds acting by other mechanisms include, but are not limited to: cyclophilin inhibitors, such as DEB-025, SCY-635, NIM-81 1, and cyclosporine and derivatives; toll-like receptor agonists, such as resiquimod, IMO-2125, and ANA-773, HCV viral entry inhibitors, such as civacir, thiazolides, such as nitazoxanide, and broad- spectrum viral inhibitors, such as, inosine-5' -monophosphate dehydrogenase (IMPDH) inhibitors.

In addition, compounds of the invention may be combined with an NS5A inhibitor, for example, daclatasvir (BMS-790052), AZD-7295, PPI-461, PPI-1301, GS- 5885, GSK2336805, ABT-267, ACH-2928, ACH-3102, EDP-239, IDX-719, MK-8742, or PPI-668.

In another aspect, therefore, the invention provides a therapeutic combination for use in the treatment of hepatitis C viral infections, the combination comprising a compound of the invention and one or more other therapeutic agents useful for treating HCV. For example, the invention provides a combination comprising a compound of the invention and one or more agents selected from HCV NS3 protease inhibitors, HCV NS5B nucleoside and non-nucleoside polymerase inhibitors, interferons and pegylated interferons, cyclophilin inhibitors, HCV NS5A inhibitors, and ribavirin and related nucleoside analogs. Also provided, therefore, is a pharmaceutical composition comprising a compound of the invention and one or more other therapeutic agents useful for treating HCV.

Further, in a method aspect, the invention provides a method of treating a hepatitis C viral infection in a mammal, the method comprising administering to the mammal a compound of the invention and one or more other therapeutic agents useful for treating HCV.

In another method aspect, the invention provides a method of inhibiting replication of the hepatitis C virus in a mammal, the method comprising administering to the mammal a compound of the invention and one or more other therapeutic agents useful for inhibiting replication of the hepatitis C virus.

For example, in one method aspect, the invention provides a method of treating a hepatitis C viral infection in a mammal, the method comprising administering to the mammal a compound of the invention, an interferon or pegylated interferon, and ribavirin.

In another exemplary method aspect, the invention provides a method of treating a hepatitis C viral infection in a mammal, the method comprising administering to the mammal a compound of the invention, an interferon or pegylated interferon, ribavirin, and an HCV NS3 protease inhibitor.

In still another method aspect, the invention provides a method of treating a hepatitis C viral infection in a mammal, the method comprising administering to the mammal a compound of the invention, an HCV NS3 protease inhibitor, and ribavirin.

Still other all-oral combination therapies useful in other method aspects, include, for example, a compound of the invention and an HCV NS3 protease inhibitor; a compound of the invention and an HCV NS5B nucleoside polymerase inhibitor; a compound of the invention, an HCV NS5B nucleoside polymerase inhibitor, and ribavirin; a compound of the invention, an HCV NS3 protease inhibitor, and an HCV NS5B nucleoside polymerase inhibitor; a compound of the invention, an HCV NS3 protease inhibitor, an HCV NS5B nucleoside polymerase inhibitor and ribavirin; a compound of the invention, an HCV NS3 protease inhibitor, and an HCV NS5B non- nucleoside polymerase inhibitor; and a compound of the invention, an HCV NS3 protease inhibitor, an HCV NS5B non-nucleoside polymerase inhibitor and ribavirin. In another method aspect, the invention provides a method of inhibiting replication of the hepatitis C virus in a mammal, using a compound of the invention in combination with other agents, as described above.

When used in combination therapy, the agents may be formulated in a single pharmaceutical composition, as disclosed above, or the agents may be provided in separate compositions that are administered simultaneously or at separate times, by the same or by different routes of administration. When administered separately, the agents are administered sufficiently close in time so as to provide a desired therapeutic effect. Such compositions can be packaged separately or may be packaged together as a kit. The two or more therapeutic agents in the kit may be administered by the same route of administration or by different routes of administration.

Finally, the compounds of the invention may also find utility as research tools, for example, for discovering new HCV NS5A protein inhibitors or explicating mechanisms of HCV replication.

Compounds of the invention have been demonstrated to be potent inhibitors of

HCV replication in HCV replicon assays, as described in the following examples.

EXAMPLES

The following synthetic and biological examples are offered to illustrate the invention, and are not to be construed in any way as limiting the scope of the invention. In the examples below, the following abbreviations have the following meanings unless otherwise indicated. Abbreviations not defined below have their generally accepted meanings.

ACN acetonitrile

AcOH acetic acid

CDI 1 , 1 '-carbonyldiimidazole

DCM dichloromethane

DIPEA N,N-diisopropylethylamine

DMA N,N-dimethylacetamide

DMF N,N-dimethylformamide

DMSO dimethyl sulfoxide

EDC N-(3-dimethylaminopropyl)-N-ethylcarbodiimide

hydrochloride

EtOAc ethyl acetate

Et 3 N triethylamine

h hour(s)

HATU N,N,N',N'-tetramethyl-0-(7-azabenzotriazol- 1 -yl)uronium

hexafluorophosphate

HCTU 2-(6-chloro- lH-benzotriazole- 1 - yl)- 1, 1,3,3- tetramethylaminium hexafluorophosphate HOAt = l-hydroxy-7-azabenzotriazole

IPA = isopropylalcohol

MeOH = methanol

min = minute(s)

Pd 2 (dba) 3 = tris(dibenzylideneacetone)dipalladium(0)

Pd(dppf)Cl 2 = dichloro( 1,1 ' -bis(diphenylphosphino)- ferrocene)dipalladium(II)

MTBE = methyl tert-butyl ether

RT = room temperature

TFA = trifluoroacetic acid

THF = tetrahydrofuran

bis(pinacolato)diboron = 4,4,5, 5,4',4',5',5'-octamethyl-

[2,2']bi[[l,3,2]dioxaborolanyl]

Reagents and solvents were purchased from commercial suppliers (Aldrich, Fluka, Sigma, etc.), and used without further purification. Reactions were run under nitrogen atmosphere, unless noted otherwise. Progress of reaction mixtures was monitored by thin layer chromatography (TLC), analytical high performance liquid chromatography (anal. HPLC), and mass spectrometry. Reaction mixtures were worked up as described specifically in each reaction; commonly they were purified by extraction and other purification methods such as temperature-, and solvent-dependent crystallization, and precipitation. In addition, reaction mixtures were routinely purified by preparative HPLC, typically using CI 8 or BDS column packings and conventional eluents. Typical preparative HPLC conditions are described below.

Characterization of reaction products was routinely carried out by mass and X H-NMR spectrometry. For NMR analysis, samples were dissolved in deuterated solvent ( such as CD 3 OD, CDCI 3 , or i¾-DMSO), and 1 H-NMR spectra were acquired with a Varian Gemini 2000 instrument (400 MHz) under standard observation conditions. Mass spectrometric identification of compounds was performed by an electrospray ionization method (ESMS) with an Applied Biosystems (Foster City, CA) model API 150 EX instrument or an Agilent (Palo Alto, CA) model 1200 LC/MSD instrument.

General Preparative HPLC Conditions

Column: C18, 5 μιη. 21.2 x 150 mm or C18, 5 μιη 21 x 250 or C14 21x150

Column temperature: Room Temperature

Flow rate: 20.0 mL/min

Mobile Phases: A = Water + 0.05 % TFA

B = ACN + 0.05 % TFA,

Injection volume: (100-1500 μί)

Detector wavelength: 214 nm Crude compounds were dissolved in 1 : 1 water: acetic acid at about 50 mg/mL. A 4 minute analytical scale test run was carried out using a 2.1 x 50 mm C18 column followed by a 15 or 20 minute preparative scale run using 100 μϊ ^ injection with the gradient based on the % B retention of the analytical scale test run. Exact gradients were sample dependent. Samples with close running impurities were checked with a 21 x 250 mm C18 column and/or a 21 x 150 mm C14 column for best separation. Fractions containing desired product were identified by mass spectrometric analysis.

Preparation 1: 4-(4-bro -phenyl)-2-(S)-pyrrolidin-2-yl-lH-imidazole

(a) 2-Bromo-l-(4-bromo-phenyl)-ethanone

Bromine (80 g, 500 mmol) was added dropwise to a solution of l-(4-bromo- phenyl)-ethanone (100 g, 500 mmol) in dichloromethane (1500 mL) at ambient temperature. The reaction mixture was stirred for 3 h and then concentrated. The residue was washed with dichloromethane (100 mL) to give the crude title compound (120 g, 86 % yield) as a white solid. X H NMR (CDC1 3 , 400 MHz) δ (ppm): 7.78 (d, J=8.4 Hz, 2H), 7.57 (d, J=8.4 Hz, 2H), 4.32 (s, 2H).

(b) (^-Pyrrolidine- 1 ,2-dicarboxylic acid 2-r2-(4-bromo-phenyl)-2-oxo-ethyl1 ester \-tert- butyl ester

Diisopropylethylamine (67 g, 518 mmol) was added dropwise to a solution of the product of the previous step (120 g, 432 mmol) and (5)-pyrrolidine-l,2-dicarboxylic acid 1-tert-butyl ester ( -Boc proline) (102 g, 475 mmol) in acetonitrile (2 L) at room temperature. The reaction mixture was stirred overnight and concentrated to dryness. The residue was dissolved in ethyl acetate (2 L) and washed with water (2 L). The organic layer was dried over sodium sulfate and concentrated to give crude title compound (178 g, 100 % yield).

(c) (5 f )-2-r4-(4-Bromo-phenyl)-lH-imidazol-2-yl1-pyrrolidine- l-carboxylic acid fe/t-butyl ester

A solution of the product of the previous step (178 g, 432 mmol) and ammonium acetate (500 g, 6.5 mol) in toluene (2 L) was heated at reflux overnight. The solvent was removed and the residue was dissolved in ethyl acetate (2 L) and washed with water (2 L). The organic layer was dried over sodium sulfate and concentrated. The residue was purified by silica gel column chromatography in 1:3 petroleum ether: ethyl acetate to give the title compound (120 g, 71 % yield) as a yellow solid. ¾ NMR (CDC1 3 , 400 MHz) δ (ppm): 7.56 (s, 1H), 7.39 (d, J=8.0 Hz, 2H), 7.24 (m, 1H), 7.14 (s, 1H), 4.88 (m, 1H), 3.33 (m, 2H), 2.94 (s, 1H), 2.07 (m, 2H), 1.88 (m, 1H), 1.42 (s, 9H).

(d) 4-(4-Bromo-phenyl)-2-(y)-pyrrolidin-2-yl-lH-imidazole

To a solution of (5)-2-[4-(4-bromo-phenyl)-lH-imidazol-2-yl]-pyrrolidine-l- carboxylic acid tert-butyl ester (3 g, 7.6 mmol) in methanol (3 mL) was added 4N HQ in methanol (60 mL) at 0 °C. The reaction mixture was stirred for 2 h and then concentrated to give crude hydrochloride salt of the title compound (2.51 g 100 % yield) as a yellow solid.

Preparation 2: (5)-2-Methoxycarbonylamino-3-methyl-butyric acid

Methylchloroformate (14.5 mL, 0.188 mol) was added over 15 min to a cooled (0- 6 °C) mixture of (5)-2-amino-3 -methyl-butyric acid (20.0 g, 0.171 mol), NaOH (6.80 g, 0.171 mol) and sodium carbonate (18.1 g, 0.171 mol) in water (200 mL). The cooling bath was removed and the mixture was stirred at ambient temperature overnight. Cone, aqueous HC1 (30 mL) was added to the reaction mixture to adjust pH to ~1. A solid formed and the mixture was stirred for 90 min. The mixture was filtered and the solid was dried overnight under reduced pressure at 40 °C t to provide the title intermediate (27.8 g, 93 % yield). X H NMR (CD 3 OD, 400 MHz) δ (ppm) 4.87 (br. s, 2H), 4.05 (d, J = 5.49, 1 H), 3.65 (s, 3 H), 2.25-2.05 (m, 1 H), 0.98 (d, J = 6.87, 3 H), 0.94 (d, J = 6.87, 3 H).

Preparation 3: ((S)-l-{(S)-2-[4-(4-Bromo-phenyl)-lH-imidazol-2-yl]- pyrrolidine-l-carbonyl}-2-met methyl ester

Triethylamine (2.3 g, 11.4 mmol) was added to a solution of 4-(4-bromo-phenyl)- 2-(5)-pyrrolidin-2-yl-lH- imidazole hydrochloride (2 g, 11.4 mol), (5)-2- methoxycarbonylamino-3-methyl-butyric acid (2.5 g, 7.6 mmol), and HATU (4.3 g, 11.4 mmol) in dimethylformamide (50 niL) at 0 °C under nitrogen. The reaction mixture was stirred at room temperature overnight and treated with ethyl acetate (100 mL) and water (1000 mL). The organic layer was washed with water (2 x 100 mL) and brine (100 mL), dried over anhydrous sodium sulfate and concentrated. The residue was purified by silica gel column chromatography in 1 : 1 petroleum ether: ethyl acetate to give the title compound (2.5 g 74 % yield) as a yellow solid. X H NMR (i¾-DMSO, 400 MHz) δ (ppm) 7.63 (d, J=8.8 Hz, 2H), 7.54 (m, IH), 7.47 (m, 2H), 7.26 (d, J=8.4 Hz, IH), 5.03 (m, IH), 4.02 (t, J =8.4 Hz, IH), 3.76 (m, 2H), 3.51 (s, 3H), 2.10 (m, 2H), 1.93 (m, 3H), 0.85 (d, J=6.8 Hz, 3H), 0.81 (d, J=6.8 Hz, 3H).

Preparation 4: [(S)-2-Methyl-l-((S)-2-{4-[4-(4,4,5,5-tetramethyl-

[l,3,2]dioxaborolan-2-yl)-phenyl]-lH-imidazol-2-yl}-pyrro lidine-l-carbonyl)- propylj-carbamic acid met

To a solution of ((5)-l-{(5)-2-[4-(4-bromo-phenyl)-lH-imidazol-2-yl]- pyrrolidine- l-carbonyl}-2-methyl-propyl)-carbamic acid methyl ester (50 g, 0.11 mol), 4,4,5,5,4',4',5',5'-octamethyl-[2,2']bi[[l,3,2]dioxaborolany l] (57 g, 0.22 mol) and potassium acetate (108 g, 1.1 mol) in dioxane (1000 mL) was added Pd(dppf)Ci 2 -CH 2 Ci 2 (4.5 g, 5.5 mmol) under nitrogen. The reaction mixture was stirred at 85 °C overnight and then ethyl acetate (100 mL) and water (1000 mL) were added. The organic layer was washed with water (2 x 1000 mL) and brine (1000 mL), dried over anhydrous Na 2 S0 4 and concentrated. The residue was purified by silica gel column chromatography (1 : 1 petroleum ethenethyl acetate) to give the title compound (22.5 g) as a yellow solid. X H NMR (CDC1 3 , 400 MHz) δ (ppm) 7.71 (m, 3H), 7.32 (m, IH), 7.19 (m, IH), 5.56 (m, IH), 5.18 (m, IH), 4.23 (m, IH), 3.73 (m, IH), 3.61 (s, 3H), 3.55 (m, IH), 2.95 (m, IH), 2.38 (s, IH), 2.13 (m, IH), 2.02 (m, IH), 1.89 (m, 2H), 1.22 (s, 12H), 0.79 (d, 6H). Preparation 5: ((S)-l-{(S)-2-[4-(4'-Amino-2'-trifluoromethoxy-biphenyl-4-yl )- lH-imidazol-2-yl]-pyrrolidine-l-carbonyl}-2-methyl-propyl)-c arbamic acid methyl ester

To a solution of [(5)-2-methyl-l-((5)-2-{4-[4-(4,4,5,5-tetramethyl-

[1,3 ,2] dioxaborolan-2-yl)-phenyl] - lH-imidazol-2-yl} -pyrrolidine- 1 -carbonyl)-propyl] - carbamic acid methyl ester (1.80 g, 3.63 mmol) and 4-bromo-3-trifluoromethoxy- phenylamine (1.00 g, 3.90 mmol) dissolved in toluene (5.4 mL) and water (2.7 mL) was added potassium carbonate (2.50 g, 18.1 mmol). The reaction mixture was sparged with nitrogen. Tetrakis(triphenylphosphine)-palladium(0) (0.21 g, 0.18 mmol) was added and the reaction mixture was sparged with nitrogen and heated at 100 °C in a sealed tube overnight, diluted in ethyl acetate (100 mL) and washed with water, brine. The organic layer was dried over sodium sulfate, filtered, and concentrated to produce a red paste (6.5 g), which was purified by silica gel chromatography (120 g silica, 0-3 %

MeOH/DCM) to provide the title intermediate (1.05 g) and a second fraction which was purified by reverse phase HPLC to provide the di-TFA salt of the title intermediate (800 mg). (Total yield 80 %).

Preparation 6: Cyclopropyl-[4-((S) -2-methyl-piperazin-l-yl)-piperidin-l-yl]- methanone

To a mixture of (5)-3-methyl-piperazine-l-carboxylic acid tert-butyl ester (220 mg, 1.1 mmol) and l-(cyclopropylcarbonyl)piperidin-4-one (150 mg, 0.9 mmol) in methanol (5 mL, 120 mmol) was added slowly trifluoroacetic acid (6.9 μί, 0.09mmol) followed by sodium cyanoborohydride (56 mg, 0.9 mmol). The reaction mixture was stirred overnight at RT, concentrated by rotary evaporation, washed with 1 N NaOH (20 mL) and extracted with EtOAc (3 x 15 mL). Combined organic fractions were washed with brine, dried with sodium sulfate, and concentrated by rotary evaporation to afford a white solid (320 mg). A mixture of the white solid in 4 M HC1 in 1,4-dioxane (5 mL) was stirred at RT for 30 min and concentrated to provide the di-HCl salt of the title intermediate (180 mg, 62 % yield) as a white suspension.

Preparation 7: (2S,4S)-2-[4-(4-Bromo-phenyl)-lH-imidazol-2-yl]-4-methyl- pyrrolidine-l-carboxylic acid

(a) (^-Pyrrolidine- 1 ,2-dicarboxylic acid 2-r2-(4-bromo-phenyl)-2-oxo-ethyl1 ester l-tert- butyl ester

To a mixture ofp-bromophenacyl bromide (242 mg, 0.87 mmol) in DCM

(1.5 mL) and DMA (1.5 mL), under nitrogen, was added (25,4S)-4-methyl-pyrrolidine- 1,2-dicarboxylic acid 1 -tert-butyl ester (200 mg, 0.87 mmol) and N,N- diisopropylethylamine (531.8 μί, 3.05 mmol) and the resulting mixture was stirred at 35 °C for 3 h, concentrated under vacuum, dissolved in DCM (30 mL), and washed with water (2 x 5 ml). The organic layer was dried over magnesium sulfate, filtered, and concentrated under vacuum to provide the title intermediate.

(b) (25 , .45 f )-2-r4-(4-Bromo-phenyl)-lH-imidazol-2-yl1-4-methyl-pyr rolidine-l-carboxylic acid tert-butyl ester

The product of the previous step was dissolved in toluene (20.0 mL), ammonium acetate (1.345 g, 17.45 mmol) was added, and resulting mixture was stirred at 95 °C overnight, concentrated and purified by silica gel chromatography (24 g silica, 0-80 % EtOAc/hexanes)) to give the title product (265 mg) (m/z): [M+H] + calcd for

Ci9H 24 Br 3 02 406.1 1, 408.1 1 found 408.5.

Preparation 8: 5-(4-Bromo-phenyl)-2-((2S,4S)-4-methyl-pyrrolidin-2-yl)-lH- imidazole A mixture of (25',45)-2-[4-(4-bromo-phenyl)-lH-imidazol-2-yl]-4-methyl- pyrrolidine- 1-carboxylic acid tert-butyl ester (265 mg, Preparation 7) and 4 M HCl in 1,4- dioxane (2.0 mL) was left for 1 h and then concentrated by rotary evaporation to provide the di-HCl salt of the title intermediate (224 mg, 68 % yield), (m/z): [M+H] + calcd for Ci 4 H 16 BrN 3 306.06, 308.06 found 306.3.

Preparation 9: ((S)-l-{(2S,4S)-2-[5-(4-Bromo-phenyl)-lH-imidazol-2-yl]-4- methyl-pyrrolidine-l-carbon -2-methyl-propyl)-carbamic acid methyl ester

A mixture of (5)-2-methoxycarbonylamino-3 -methyl-butyric acid (103 mmol, Preparation 2) and HATU (270 mg, 0.71 mmol) were stirred in DMA (2 mL) for 10 min and then 5-(4-bromo-phenyl)-2-((25',45)-4-methyl-pyrrolidin-2-yl)-lH- imidazole 2 HCl (224 mg, 0.59 mmol, Preparation 8) and N,N-diisopropylethylamine (0.31 mL, 1.8 mmol) were added. The resulting mixture was stirred at RT overnight, diluted with ethyl acetate (50 mL), and washed with water (2 x 5 mL). The organic layer was dried over magnesium sulfate, filtered, concentrated and purified by silica gel chromatography (0- 100% EtOAc/hexanes)). Fractions with desired product were combined and concentrated to give the title compound (183 mg, 66 % yield) (m/z): [M+H] + calcd for C 2 iH 27 BrN 4 0 3 463.13, 465.12 found 465.3.

Preparation 10: [(S)-2-Methyl-l-((2S,4S)-4-methyl-2-{4-[4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)-phenyl]-lH-imidazol-2-y l}-pyrrolidine-l- carbonyl)-propyl]-carba

To a solution of ((5)-l-{(2S,4S)-2-[4-(4-bromo-phenyl)-lH-imidazol-2-yl]-4- methyl-pyrrolidine-l-carbonyl}-2-methyl-propyl)-carbamic acid methyl ester (183 mg, 0.39 mmol; Preparation 9) and 4,4,5,5,4',4',5',5'-octamethyl-

[2,2']bi[[l,3,2]dioxaborolanyl] (120 mg, 0.47mmol) in 1,4-dioxane (5 mL) was added potassium acetate (56 mg, 0.59 mmol). The resulting mixture was sparged with nitrogen, Pd(dppf)Cl 2 -CH 2 Cl 2 (27 mg, 0.033 mmol) was added, and the reaction mixture was capped and heated at 100 °C overnight. The reaction was cooled to RT and partitioned between EtOAc (50 mL) and water (10 mL). The organic layer was washed with water (5 mL), brine (2 mL), dried over magnesium sulfate, filtered ,and concentrated to give a dark-brown oil, which was purified by silica gel chromatography (24g silica gel, 0-100% EtOAc/hexanes). Fractions with desired product were combined and dried to give the title compound (94 mg, 47 % yield) as a white foam, (m/z): [M+H] + calcd for C27H 39 BN4O5 51 1.30 found 511.7.

Preparation 11: 4-Bromo- -chloro-5-trifluoromethoxy-phenylamine

To a mixture of 4-bromo-3-trifluoromethoxy-phenylamine (2.0 g, 7.8 mmol) in ACN (60 mL) was slowly added a solution of N-chlorosuccinimide (1.0 g, 7.8 mmol) in ACN (40 mL). The reaction mixture was heated to at 60 °C overnight and extracted with ethyl actetate/water. The organic layer was dried over sodium sulfate and purified by silica gel chromatography (40 g silica, 100% hexanes to 10% EtOAc: hexanes) to produce the desired product as an orange colored oil (1.4 g, 64 % yield).

Preparation 12: 4-Pip rboxylic acid tert-butyl ester

(a) 4-(l-ter?-Butoxycarbonyl-piperidin-4-yl)-piperazine-l-carbox ylic acid benzyl ester

A mixture of piperazine- 1 -carboxylic acid benzyl ester (6.6 g, 29.96 mmol) and 4- oxo-piperidine- 1 -carboxylic acid tert-butyl ester in dichloroethane (150 mL) containing two drops of acetic acid was stirred at RT for 1 h and then sodium triacetoxyborohydride (19.05 g, 89.89 mmol) was added in portions. The reaction mixture was stirred at RT overnight, water (150 mL) was added, and the reaction mixture was stirred at RT for 2 h. The reaction mixture was extracted with DCM (4 x 30 mL). Combined organic layers were dried over sodium sulfate, filtered, concentrated under reduced pressure, and purified by silica gel chromatography (20-30 % EtO Ac/petroleum ether) to provide the title intermediate (9 g, 74 % yield) as a colorless oil. (m/z): [M+H] + calcd for C22H 33 3 O4 404.25, found 404.3.

(b) 4-piperazin-l-yl-piperidine-l-carboxylic acid fe/ -butyl ester

The product of the previous step (9 g, 22.30 mmol) and palladium on carbon (1 g) in IPA (150 mL) were stirred at RT under hydrogen (50 psi) overnight, filtered, and the solvent was removed under vacuum to provide the title intermediate (5.57 g, 93 % yield) as a colorless solid, (m/z): [M+H] + calcd for Ci 4 H 27 N 3 0 2 270.21, found 270.2. X H NMR (MeOD, 400 MHz): δ (ppm) 4.1 1 (d, J= 13.5 Hz, 2H), 2.98-2.64 (m, 1 1H), 1.85 (d, J = 12.4 Hz, 2H), 1.44 (s, 9H), 1.44-1.34 (m, 2H).

Preparation 13: 4-[4-(4-Bromo-2-chloro-5-trifluoromethoxy- phenylcarbamoyl)-p tert-butyl ester

To a solution of 4-bromo-2-chloro-5-trifluoromethoxy-phenylamine (0.55 g, 1.90 mmol) in DCM (4.9 mL) was added CDI (0.34 g, 2.09 mmol). The reaction solution was stirred at RT overnight and then 4-piperazin- 1 -yl-piperidine- 1 -carboxylic acid tert-butyl ester (0.56 g, 2.09 mmol; Preparation 12) was added and the reaction mixture was stirred for 30 min at RT, concentrated under vacuum and purified by silica gel chromatography (0-5% MeOH/DCM). Pure fractions were combined and concentrated under vacuum to provide the title intermediate (0.67 g, 60 % yield) as a white foam, (m/z): [M+H] + calcd for C 2 2H 29 BrClF 3 N 4 0 4 585.1 found 584.6.

Preparation 14: 4-[4-(5-Chloro-4'-{2-[(2S,4S)-l-((S)-2- methoxycarbonylamino-3-methyl-butyryl)-4-methyl-pyrrolidin-2 -yl]-lH-imidazol-4- yl}-2-trifluoromethoxy-biphenyl-4-ylcarbamoyl)-piperazin-l-y l]- piperidine-1- carboxylic

A solution of [(5')-2-methyl-l-((25,45 , )-4-methyl-2- {4-[4-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)-phenyl]-lH-imidazol-2-yl} -pyrrolidine- l-carbonyl)-propyl]- carbamic acid methyl ester (0.12 g, 0.24 mmol), 4-[4-(4-bromo-2-chloro-5- trifluoromethoxy-phenylcarbamoyl)-piperazin-l-yl]-piperidine -l-carboxylic acid tert- butyl ester (0.138 g, 0.235 mmol; Preparation 13) in 1,4-dioxane (1.84 mL) and water (0.28 mL) was degassed with nitrogen for 20 min and then Pd2(dba) 3 (32.3 mg,

0.0353 mmol) and 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (0.1 12 g, 0.235 mmol) were added and the reaction was degassed with nitrogen for a further 15 min. The resulting solution was heated at 90 °C for 60 h, cooled to RT and diluted with EtOAc (3 mL). The layers were separated and the aqueous layer was washed with EtOAc (3 x 3 mL). The combined organic extracts were washed with water and brine, dried over MgS0 4 , filtered, concentrated, and purified by reverse phase HPLC. Fractions were combined and lyophilised overnight to provide the di-TFA salt of the title intermediate (65 mg, 25 % yield).

Preparation 15: ((S)-l-{(2S,4S)-2-[4-(4-Bromo-phenyl)-5-chloro-lH-imidazol-

2-yl]-4-methyl-pyrrolidine-l-carbonyl}-2-methyl-propyl)-c arbamic acid methyl ester

A mixture of ((5)-l- {(25',45 , )-2-[4-(4-bromo-phenyl)-lH-imidazol-2-yl]-4-methyl- pyrrolidine-l-carbonyl}-2-methyl-propyl)-carbamic acid methyl ester (175 mg,

0.38 mmol) and N-chlorosuccinimide (202 mg, 1.51 mmol) in acetonitrile (4 mL) was stirred at RT overnight. The reaction mixture was concentrated by rotary evaporation and purified by silica gel chromatography (24 g silica, 0-100% hexanes/EtOAc) to provide the title intermediate (208 mg) as a brown solid, (m/z): [M+Hf calcd for C 2 iH 26 BrClN 4 0 3 497.09, found 497.4.

Preparation 16: 4-[4-(4-Bromo-3-trifluoromethoxy-phenylcarbamoyl)- piperazin-l-yl]-piperi

To a solution of 4-bromo-3-trifluoromethoxy-phenylamine (500 mg, 2 mmol) in DCM (5 mL) was added N,N-carbonyldiimidazole (348 mg, 2.15 mmol). The reaction mixture was stirred at RT overnight and then 4-piperazin-l-yl-piperidine-l-carboxylic acid tert-butyl ester (579 mg, 2.15 mmol, Preparation 12) was added and the reaction mixture was stirred at RT for 15 min, and concentrated by rotary evaporation and purified by silica gel chromatography (80 g silica, 0-10% MeOH/DCM) to provide the title intermediate (776 mg, 70 % yield) as a yellow powder, (m/z): [M+H] + calcd for C22H 3 oBrF 3 4 0 4 551.14, found 551.3.

Preparation 17: 4-{4-[4-(4,4,5,5-Tetramethyl-l,3,2-dioxaborolan-2-yl)-3- trifluoromethoxy-phenylcarbamoyl]-piperazin-l-yl}-piperidine -l-carboxylic acid tert-butyl ester

To a solution of 4-[4-(4-bromo-3-trifluoromethoxy-phenylcarbamoyl)-piperazin- l-yl]-piperidine-l-carboxylic acid tert-butyl ester (200 mg, 0.4 mmol, Preparation 16) and 4,4,5,5,4',4',5',5'-octamethyl-[2,2']bi[[l,3,2]dioxaborolany l] (1 15 mg, 0.45 mmol), potassium acetate (53.4 mg, 0.54 mmol) was added 1,4-dioxane (4 mL, 50 mmol) . The resulting mixture was sparged with nitrogen, Pd(dppf)Ci 2 -CH 2 Ci 2 (88.9 mg, 0.11 mmol) was added, and the reaction mixture was heated at 100 °C overnight, and then filtered through a Celite® pad, which was washed with methanol. The eluent was concentrated, diluted in a minimal amount of DCM and purified via silica gel chromatography (40 g silica, 0-100% EtOAc/hexanes, flushed with 100 % EtOAc) to provide the title intermediate (70 mg, 30 % yield) as a brown solid, (m/z): [M+H] + calcd for

C 2 8H4 2 BF 3 4 0 6 599.31, found 599.5.

Preparation 18: {(S)-l-[(2S,4S)-2-(5-Chloro-4-{4'-[(4-piperidin-4-yl- piperazine-l-carbonyl)-amino]-2'-trifluoromethoxy-biphenyl-4 -yl}-lH-imidazol-2- yl)-4-methyl-py methyl ester

(a) 4-r4-(4'- {5-Chloro-2-r(25'.4y)-l-((y)-2-methoxycarbonylamino-3-methyl -butyryl)-4- methyl-pyrrolidin-2 -yll - 1 H-imidazol-4-yl } -2 -trifluoromethoxy-biphenyl-4- ylcarbamovD-piperazin-l-yll-piperidine-l-carboxylic acid ferf-butyl ester

A mixture of 4- {4-[4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3- trifluoromethoxy-phenylcarbamoyl] -piperazin- 1 -yl} -piperidine- 1 -carboxylic acid tert- butyl ester (70 mg, 0.12 mmol; Preparation 17), ((S)-l- {(2S,4S)-2-[4-(4-bromo-phenyl)-5- chloro- lH-imidazol-2-yl]-4-methyl-pyrrolidine- 1 -carbonyl} -2-methyl-propyl)-carbamic acid methyl ester (58 mg, 0.12 mmol; Preparation 15), and sodium bicarbonate (49 mg, 0.58 mmol) in 1,4-dioxane (2 mL) and water (0.5 mL) was purged with nitrogen and then 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (Xphos) (56 mg, 0.12 mmol) and Pd 2 (dba) 3 (21 mg, 0.023 mmol) were added. The reaction mixture was heated at 100 °C overnight, concentrated by rotary evaporation, dissolved in 1 : 1 acetic acid:water (4 mL), purified by reverse phase HPLC, and lyophilized to provide the TFA salt of the title intermediate (20 mg) as a yellow solid, (m/z): [M+H] + calcd for C 4 3H 56 C1F 3 8 0 7 889.39, found 889.4.

2'-trifluoromethoxy-biphenyl-4-yl} - lH-imidazol-2-yl)-4-methyl-pyrrolidine- 1 - carbonyll-2-methyl-propyll-carbamic acid methyl ester

The product of the previous step was diluted in 4 M HQ in 1,4-dioxane (3 mL), stirred at RT, and concentrated to provide the tri-HCl salt of the title intermediate (20 mg, yield 20 %) as a yellow solid, (m/z): [M+H calcd for CssHuClFsNgOs 789.34, found 789.3.

Preparation 19: ((2S,4S)-2-[4-(4-Bromo-phenyl)-5-fluoro-lH-imidazol-2-yl]- 4-methyl-pyrrolidine-l-carboxylic acid tert-butyl ester

A solution of (25 , ,45')-2-[4-(4-bromo-phenyl)-lH-imidazol-2-yl]-4-methyl - pyrrolidine-l-carboxylic acid tert-butyl ester (500 mg, 1.23 mmol) and l-fluoro-4- hydroxy-l,4-diazoniabicyclo[2.2.2]octane bis(tetrafluoroborate) (436 m g, 1.36 mmol) in DMF (20 mL) was stirred at RT for 1 h, concentrated by rotary evaporation, and purified by silica gel chromatography (0-100 % EtOAc/hexanes with 1 % Et 3 N). Desired fractions were collected and concentrated to provide the title intermediate (180 mg, 35 % yield), (m/z): [M+H] + calcd for Ci 9 H 24 BrFN 3 02 424.10, 426.09 found 424.1.

Preparation 20: ((S)-l-{(2S,4S)-2-[4-(4-Bromo-phenyl)-5- fluoro -1H- imidazol-2-yl]-4-methyl-pyrrolidine-l-carbonyl}-2-methyl-pro pyl)-carbamic acid methyl ester

A solution of (25 , ,45)-2-[4-(4-bromo-phenyl)-5-fluoro-lH-imidazol-2-yl]- 4- methyl-pyrrolidine-l-carboxylic acid tert-butyl ester (180 mg, 0.42 mmol; Preparation 19) in 4 M HC1 in l,4-dioxane(0.53 mL) and 1,4-dioxane (0.9 mL) was stirred at RT for 30 min and concentrated by rotary evaporation.

The crude material was dissolved in DMA (5.54 g) and then («S)-2- methoxycarbonylamino-3 -methyl-butyric acid (89.2 mg, 0.51 mmol; Preparation 2), HCTU (0.21 g, 0.51 mmol), and DIPEA (0.27 g, 2.12 mmol) were added. The reaction mixture was stirred at RT overnight, concentrated by rotary evaporation, and purified by silica gel chromatography (0- 100 % EtOAc/hexanes). Desired fractions were collected and concentrated by rotary evaporation to provide the title intermediate (200 mg, 98 % yield), (m/z): [M+H] + calcd for C 2 iH 26 BrFN 4 0 3 481.12, 483.12 found 480.7.

Preparation 21 : 4-[4-(4'-{5-Fluoro-2-[(2S,4S)-l-((S) 2- methoxycarbonylamino-3-methyl-butyryl)-4-methyl-pyrrolidin-2 -yl]-lH-imidazol-4- yl}-2-trifluoromethoxy-biphenyl-4-ylcarbamoyl) -piperazin-l-yl]- piperidine-1- carboxylic

A solution of 4- {4-[4-(4,4,5,5-tetramethyl- l ,3,2-dioxaborolan-2-yl)-3- trifluoromethoxy-phenylcarbamoyl]-piperazin-l -yl} -piperidine- l-carboxylic acid tert- butyl ester (31.1 mg, 0.05 mmol), ((5')- l - {(25,45 , )-2-[4-(4-bromo-phenyl)-5-fluoro-lH- imidazol-2-yl]-4-methyl-pyrrolidine- l-carbonyl} -2-methyl-propyl)-carbamic acid methyl ester (25 mg, 0.05 mmol; Preparation 20) and sodium bicarbonate (22.7 mg, 0.27 mmol) in 1,4-dioxane (2.1 1 mL) and water (0.10 mL,) was degassed with nitrogen for 15 min and then Pd2(dba) 3 (9.51 mg, 0.01 mmol) and 2-dicyclohexylphosphino-2',4',6'- triisopropylbiphenyl (24.8 mg, 0.05mmol) were added and the purple solution was degassed with nitrogen for a further 15 min. The solution was capped and heated at 90 °C for 12 h, concentrated under vacuum, dissolved in 1 : 1 acetic acid:water (5 mL), filtered through a pad of Celite®, and purified by reverse phase HPLC. Pure fractions were combined and lyophilised overnight to provide the di-TFA salt of the title intermediate (40 mg, 69 % yield), (m/z): [M+H] + calcd for C 43 H 56 F 4 N 8 0 7 873.42 found 873.6.

Preparation 22: (5)-3-Methyl-4-piperidin-4-yl-piperazine-l-carboxylic acid teri-butyl ester (a) ffl-4-(l-Benzyloxycarbonyl-piperidin-4-yl -3-methyl-piperazine-l-carboxylic acid fe/t-butyl ester

To a solution of (5)-3-methyl-piperazine-l-carboxylic acid tert-butyl ester (1.0 g, 5 mmol) and N-benzyloxycarbonyl-4-piperidone (1.2 g, 5 mmol) in methanol (8 mL) was added acetic acid (0.28 mL) and the solution was stirred at RT for 1 h. Sodium cyanoborohydride (310 mg, 5 mmol) was added and the reaction mixture was stirred at 45 °C overnight and then 1 Ν NaOH (5 mL) was added. The aqueous phase was extracted with EtOAc (3 x 15 mL). The combined organic extracts were washed with brine, dried over MgS04, filtered, concentrated under vacuum, and purified by silica gel

chromatography (0-5% MeOH/ DCM). Pure fractions were combined and concentrated under vacuum to provide the title intermediate (0.71 g, 30 % yield) as a clear oil.

(m/z): [M+H] + calcd for C23H35N3O4 418.26 found 418.1.

(b) y)-3-Methyl-4-piperidin-4-yl-piperazine-l-carboxylic acid fe/ -butyl ester

A solution of the product of the previous step and 10% Pd/C, wet 50%

(0.05:0.45:0.5, Palladium:carbon blackwater, (362 mg, 0.17 mmol) in THF (7.5 mL) was purged with nitrogen. A balloon of hydrogen was bubbled through the solution for 15 min and the reaction mixture was stirred at RT under an atmosphere of hydrogen for 16 h, cooled, and filtered through a pad of Celite® and washed with THF. The crude product was purified by silica gel chromatography (10 % MeOH in DCM). Desired fractions were combined and concentrated under vacuum to provide the title intermediate.

Preparation 23: (5)-4-[l-(4-Bromo-2-chloro-5-trifluoromethoxy- phenylcarbamoyl)-piperidin-4-yl]-3-methyl-piperazine-l-carbo xylic acid tert-but l ester

To a solution of 4-bromo-2-chloro-5-trifluoromethoxy-phenylamine (92.5 mg,

0.32 mmol) in DCM (0.82 mL) was added CDI (56.8 mg, 0.35 mmol). The reaction mixture was stirred at RT for 6 h and a solution of (5)-3-methyl-4-piperidin-4-yl- piperazine-l-carboxylic acid tert-butyl ester (99.3 mg, 0.35 mmol; Preparation 22) in DCM (1 mL) was added. The solution was concentrated under vacuum and purified by silica gel chromatography (0-5% MeOH/DCM). Product fractions were combined and concentrated under vacuum to provide the title intermediate (32 mg, 17 % yield) as a clear oil. (m/z): [M+H] + calcd for C2 3 H 31 BrClF 3 N 4 0 4 599.12, 601.1 1 found 601.4.

Preparation 24: (S)-4-[l-(5-Chloro-4'-{2-[(2S,4S)-l-((S)-2- methoxycarbonylamino-3-methyl-butyryl)-4-methyl-pyrrolidin-2 -yl]-lH-imidazol-4- yl}-2-trifluoromethoxy-biphenyl-4-ylcarbamoyl)-piperidin-4-y l]-3-methyl- piperazine- -carboxylic acid teri-butyl ester

A solution of [(5)-2-methyl-l-((25,45)-4-methyl-2- {4-[4-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)-phenyl]-lH-imidazol-2-yl} -pyrrolidine- l-carbonyl)-propyl]- carbamic acid methyl ester (27.8 mg, 0.05 mmol, (5)-4-[l-(4-bromo-2-chloro-5- trifluoromethoxy-phenylcarbamoyl)-piperidin-4-yl]-3-methyl-p iperazine-l-carboxylic acid tert-butyl ester (32.7 mg, 0.05 mmol; Preparation 23) and sodium bicarbonate (23.8 mg, 0.28 mmol) in 1,4-dioxane (2.2 mL) and water (0.1 mL) was degassed with nitrogen for 20 min and Pd2(dba) 3 (10.0 mg, 0.01 mmol) and 2-dicyclohexylphosphino-2',4',6'- triisopropylbiphenyl (26.0 mg, 0.05 mmol) were added. The reaction mixture was purged with nitrogen for 15 min, heated at 90 °C for 60 h, concentrated, diluted with methanol, filtered through a pad of Celite®, concentrated under vacuum, and purified by reverse phase HPLC. Desired fractions were combined and lyophilised overnight to provide the di-TFA salt of the title intermediate (13 mg, 20 % yield) as a white powder.

(m/z): [M+H] + calcd for C 4 4H 58 C1F 3 8 0 7 903.41 found 903.6.

Preparation 25: ((S)-l-{(2S,4S)-2-[5-(4-Bromo-phenyl)-lH-imidazol-2-yl]-4- methoxy-pyrrolidine-l-carb amic acid methyl ester

(a) (2£,4<S 2-r5-(4-Bromo-phenyl - lH-imidazol-2-yl1-4-methoxy-pyrrolidine- 1 - carboxylic acid /ert-butyl ester

To a solution of (2S,45)-4-methoxy -pyrrolidine- 1 ,2-dicarboxylic acid 1 -tert-butyl ester (900 mg, 3.67 mmol), and -bromophenacyl bromide (1.02 g, 3.67 mmol) in DCM (20 mL) under nitrogen, was added N,N-diisopropylethylamine (1.92 mL, 11.01 mmol). The resulting mixture was stirred at 35 °C for 3 h and concentrated under vacuum. The crude intermediate was dissolved in toluene (150 mL), ammonium acetate (5.66 g, 73.4 mmol) was added, and the resulting mixture was stirred at 95 °C overnight, cooled to RT, and washed with water (2 x 10 mL). The organic layer was dried over magnesium sulfate, filtered, concentrated and purified by silica gel chromatography (24 g, ethyl acetate/hexanes 0 to 60%) to give the title intermediate (1.49 g, 96 % yield).

(b) 5-(4-Bromo-phenyl)-2-((25',4y)-4-methoxy-pyrrolidin-2-yl)-lH -imidazole

The product of the previous step was treated with 4 M HQ in 1,4-dioxane (2 mL) for 1 h and concentrated by rotary evaporation to give the di-HCl salt of the title intermediate (1.40 g, 97 % yield), (m/z): [M+H] + calcd for C 14 H 16 BrN 3 0 322.05, 324.05 found 324.

(c) ((y)-l-{(25',4y)-2-r5-(4-Bromo-phenyl)-lH-imidazol-2-yl1-4-m ethoxy-pyrrolidine-l- carbonyl}-2-methyl-propyl)-carbamic acid methyl ester

A mixture of (5)-2-methoxycarbonylamino-3 -methyl-butyric acid (643 mg, 3.67 mmol) and HATU (1.67 g, 4.40 mmol) in DMA (5 mL) was stirred for 10 min, and then the product of the previous step (1.40 g, 3.54 mmol) and N,N-diisopropylethylamine (1.92 mL, 11.01 mmol) were added, and the resulting mixture was stirrred at room temperature overnight, concentrated by rotary evaporation, dissolved in ethyl acetate (100 mL), and washed with water (2 x 10 mL). The organic layer was dried over magnesium sulfate, filtered, concentrated, and purified by silica gel chromatography (ethyl acetate/hexanes 30 to 80%) to give the title product (1.38 g, 79 % yield),

(m/z): [M+H] + calcd for C 21 H 27 BrN 4 0 4 479.12, 481.12 found 481. Preparation 26: [(S)-l-((2S,4S)-4-Methoxy-2-{4-[4-(4,4,5,5-tetramethyl-l,3,2 - dioxaborolan-2-yl)-phenyl]-lH-imidazol-2-yl}-pyrrolidine-l-c arbonyl)-2-methyl- propyl]- carbamic acid

A solution of ((S 1 - {(25,45)-2-[5-(4-bromo-phenyl)- lH-imidazol-2-yl]-4- methoxy-pyrrolidine-l-carbonyl}-2-methyl-propyl)-carbamic acid methyl ester (1.05 g, 2.18 mmol; Preparation 25), 4,4,5,5,4',4',5',5'-octamethyl-[2,2']bi[[l,3,2]dioxaborolany l] (0.83 g, 3.27 mmol), and potassium acetate (0.39 g, 3.93 mmol) in 1,4-dioxane (5 mL) was purged with nitrogen for 5 min, then Pd(dppf)Ci2 (95.80 mg, 0.13 mmol) was added, and the resulting mixture was stirred at 100 °C for 3 h, diluted with ethyl acetate (50 mL), and filtered through a pad of Celite® and silica gel. The pad was washed with ethyl acetate (150 ml); the filtrate was concentrated and purified by silica gel chromatography (ethyl acetate/hexanes 30 to 100 %) to give the title intermediate (669 mg 58 % yield), (m/z): [M+H] + calcd for C 27 H 39 BN 4 0 6 527.30 found 527.2.

Preparation 27: 7-Bromo-2-((2S,4S)-4-methyl-pyrrolidin-2-yl)-3H- naphtho [l,2-< ]imidazole

(a) (2£.4»S)-2-(2-Amino-6-bromo-naphthalen- 1 -ylcarbamoyl)-4-methyl-pyrrolidine- 1 - carboxylic acid fert-butyl ester

To a solution of 6-bromo-naphthalene-l,2-diamine (2.0 g, 8.5 mmol) in DMF (150 mL) was added (2S,45)-4-methyl-pyrrolidine-l,2-dicarboxylic acid 1 -tert-butyl ester (2.14 g, 10.2 mmol), DIPEA (3.29 g, 25.5 mmol), and HATU (4.84 g, 12.7 mmol). The reaction mixture was stirred at RT overnight and extracted with EtOAc/ELO (150 mL). The organic layer was dried over Na 2 S0 4 , filtered, concentrated, and purified by silica gel chromatography (2: 1 EtOAc:petroleum ether) to provide the title intermediate (1.6 g ) (b) f2S,4SV2-f 7-Bromo-3H-naphthor 1 ,2-^1imidazol-2-yl)-4-methyl-pyrrolidine- 1 - carboxylic acid tert-butyl ester

A solution of the product of the previous step (1.6 g, 3.57 mmol) in acetic acid (15 mL) was heated to 60 °C under nitrogen for lh, adjusted to pH 8-10 with 1 N NaOH, and extracted with DCM. The organic layer was dried over a 2 S04, filtered, and concentrated to give the title intermediate (1.5 g). (m/z): [M+H] + calcd for C 2 iH 24 Br 3 0 2 430.1 1, 432.11 found 432.1.

(c) 7-Bromo-2-((25 , ,4y)-4-methyl-pyrrolidin-2-yl -3H-naphthori,2-(i1imidazole

To a solution of the product of the previous step (1.5 g, 3.5mmol) in DCM (20 mL) was added TFA (2 mL). The mixture was stirred at RT for 5 h, adjusted to pH -10 with 1 N NaOH (5 mL) and extracted with DCM (3 x 200 mL). The reaction mixture was dried over a2S0 4 , filtered, and concentrated to give the title intermediate (1.1 g). (m/z): [M+H] + calcd for C 16 H 16 BrN 3 330.05, 332.05 found 330.2, 332.1.

Preparation 28: ((S)-2-Methyl-l-{(2S,4S)-4-methyl-2-[7-(4,4,5,5-tetramethyl- [l,3,2]dioxaborolan-2-yl)-3H-naphtho[l,2-< |imidazol-2-yl]-pyrrolidine-l-carbonyl}- propyl)-carbamic acid m

(a) {(^-l-r(25 , .4y)-2-(7-Bromo-3H-naphthorL2-t/1imidazol-2-yl)-4-meth yl-pyrrolidine- l-carbonyll-2-methyl-propyl}-carbamic acid methyl ester

To a solution of 7-bromo-2-((25',45)-4-methyl-pyrrolidin-2-yl)-3H-naphtho[l,2 -

( jimidazole (1.05 g, 3.2 mmol; Preparation 27) in DCM (20 mL) was added (5)-2- methoxycarbonylamino-3 -methyl-butyric acid (672 mg, 3.84 mmol), DIPEA (825 mg, 6.4 mmol), and HATU (1.82 g, 4.8 mmol). The reaction mixture was stirred at RT for 4 h and extracted with DCM (3 x 250 mL). The organic layer was dried over a 2 S0 4 , filtered, concentrated, and purified by silica gel chromatography (1 : 1 EtOAc:petroleum ether) to provide the title intermediate (1.2 g) (m/z): [M+H] + calcd for C 23 H 27 Br 4 0 3 487.13, 489.12 found 489.2. (b) (ffl-2-Methyl-l - { (2S.45V4-methyl-2-r7-(4 A5 tetramethyl-r 1.3.21dioxaborolan-2- yl -3H-naphthorL2-^imidazol-2-yl1-pyrrolidine-l-carbonyl}-propy l -carbamic acid methyl ester

To a solution of the product of the previous step (1.1 g, 2.2 mmol) in dioxane (20 mL) was added 4,4,5,5,4',4',5',5'-octamethyl-[2,2']bi[[l,3,2]dioxaborolany l] (838 mg, 3.3 mmol), Pd(dppf)Ci2 (161 mg, 0.22 mmol) and potassium acetate (646 mg, 6.6 mmol) at RT under nitrogen. The reaction mixture was stirred at 90 °C overnight and extracted with EtOAc/H 2 0 (3 x 150 mL). The organic layer was dried over Na 2 S0 4 , filtered, concentrated, and purified by silica gel chromatography (2: 1 petroleum ether: EtOAc) to provide the title intermediate (450 mg. (m/z): [M+H] + calcd for C 2 ,H 39 BN 4 0 5 535.30 found 535.3. H NMR: (DMSO-i¾, 400 MHz) 8(ppm) 0.64~0.86(m, 6H), 1.09(s, 3H), 1.31(s, 12H), 1.85~1.90(m, 2H), 2.20~2.38(m, 1H), 2.51(s, 1H), 3.31~3.33(m, 1H), 3.51(s, 3H), 4.01~4.07(m, 1H), 4.11~4.16(m, 1H), 5.09-5.10(m, 1H), 7.15(d, J=8.0Hz„ 2H), 7.60~7.79(m, 3H), 8.23~8.37(m, 2H), 12.5~13.55(d, 1H).

Preparation 29: 4-[4-(4-{2-[(2S,4S)-l-((S)-2-Methoxycarbonylamino-3- methyl-butyryl)-4-methyl-pyrrolidin-2-yl]-3H-naphtho[l,2-< ; |imidazol-7-yl}-3- trifluoromethoxy-phenylcarbamoyl)-piperazin-l-yl]-piperidine -l-carboxylic acid tert-but l ester

A solution of ((5)-2-methyl-l-{(25,45)-4-methyl-2-[7-(4,4,5,5-tetramethyl- l,3,2- dioxaborolan-2-yl)-3H-naphth[ 1 ,2-<i]imidazol-2-yl]-pyrrolidine- 1 -carbonyl} -propyl)- carbamic acid methyl ester (72.7 mg, 0.13 mmol; Preparation 28), 4-[4-(4-bromo-3- trifluoromethoxy-phenylcarbamoyl)-piperazin- 1 -yl]-piperidine- 1 -carboxylic acid tert- butyl ester (75 mg, 0.14 mmol) and sodium bicarbonate (59.4 mg, 0.71 mmol) in 1,4- dioxane (5.7 mL) and water (260 μί) was degassed with nitrogen for 20 min. Pd 2 (dba) 3 (24.91 mg, 0.027 mmol) and 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (64.8 mg, 0.14 mmol) were added and the reaction mixture was degassed with nitrogen for 15 min, heated at 90 °C for 12 h, concentrated, diluted with methanol and filtered through a pad of Celite®. The solution was then concentrated under vacuum and purified by reverse phase HPLC. Fractions containing product were combined and lyophilised overnight to afford a white powder, which was dissolved in 1 : 1 acetic acid:water and purified by reverse phase HPLC to provide the di-TFA salt of the title intermediate (47 mg, 31 % yield), (m/z): [M+H] + calcd for C 45 H 54 F 3 8 0 7 879.43 found 879.5.

Preparation 30: 4-[l-(2,2-Dimethyl-propionyl)-piperidin-4-yl]-piperazine-l- carboxylic acid (4-bro -3-trifluoromethoxy-phenyl)-amide

(a) 4-Piperidin-4-yl-piperazine- 1 -carboxylic acid (4-bromo-3-trifluoromethoxy-phenyl)- amide

A solution of 4- [4-(4-bromo-3 -trifluoromethoxy-phenylcarbamoyl)-piperazin- 1 - yl]-piperidine-l -carboxylic acid tert-butyl ester (61.8 mg, 0.1 1 mmol) in 4 M of HC1 in 1,4-dioxane (2.8 mL) was stirred at RT for 1 h to provide the title intermediate,

(b) 4-ri-(2,2-Dimethyl-propionyl)-piperidin-4-yl1-piperazine-l -carboxylic acid (4- bromo-3-trifluoromethoxy-phenyl)-amide

To a solution of the product of the previous step in DMA (5.6mL) was added 2,2- dimethylpropanoyl chloride (13.8 μί, 0.11 mmol) and DIPEA (97.6 μΐ ^ , 0.56 mmol). The reaction mixture was stirred at RT for 1 h, concentrated under vacuum, and purified by silica gel chromatography (0-5% MeOH in DCM). The pure fractions were combined and concentrated under vacuum to provide the title intermediate (45 mg, 75 % yield) as a white foam, (m/z): [M+H] + calcd for CABrFs ^ 535.15, 537.14 found 535.5.

Preparation 31 : 4-[l-(2,2-Dimethyl-propionyl)-piperidin-4-yl]-piperazine-l- carboxylic acid (4-br

(a) 4-Piperidin-4-yl-piperazine- 1 -carboxylic acid (4-bromo-3-trifluoromethoxy-phenyl)- amide

A solution of 4- [4-(4-bromo-3 -trifluoromethoxy-phenylcarbamoyl)-piperazin- 1 - yl]-piperidine-l -carboxylic acid tert-butyl ester (61.8 mg, 0.1 1 mmol) in 4 M of HC1 in 1,4-dioxane (2.8 mL) was stirred at RT for 1 h to provide the title intermediate. (b) 4- Γ 1 -(3 -Hydroxy-2 ,2-dimethyl-propionyl)-piperidin-4-yl1 -piperazine- 1 -carboxylic acid (4-bromo-3-trifluoromethoxy-phenyl)-amide

A solution of 2,2-dimethyl-3-hydroxypropionic acid (15.9 mg, 0.13mmol) and HATU (51.1 mg, 0.13 mmol) in DMA (3 mL) was stirred at 50 °C for 15 min and then a solution of the product of the previous step in DMA (2.63 mL) was added, followed by DIPEA (97.6 μί, 0.56 mmol). The solution was stirred at 50 °C for 4 hours, concentrated under vacuum, and purified by silica gel chromatography (0-5% MeOH in DCM). The pure fractions were combined and concentrated under vacuum to provide the title intermediate (42 mg, 68 % yield) as a white foam, (m/z): [M+H] + calcd for

C 22 H 3 oBrF 3 4 0 4 551.14, 553.14 found 553.4.

Preparation 32: (2S,4S)-4-Methyl-2-{4-[4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)- phenyl]-lH-imidazol-2-yl}-pyrrolidine-l-carboxylic acid tert- butyl ester

A solution of (25',45)-2-[4-(4-bromo-phenyl)-lH-imidazol-2-yl]-4-methyl- pyrrolidine- 1 -carboxylic acid tert-butyl ester (2.0 g, 4.9 mmol), bis(pinacolato)diboron (1.9 g, 7.4 mmol) and potassium acetate (0.72 g, 7.4 mmol) in 1,4-dioxane (16 mL) was degassed with nitrogen for 20 min and then Pd(dppf)Ci 2 -CH 2 Ci 2 (160 mg, 0.20 mmol) was added. The solution was degassed with nitrogen for 20 min, sealed, stirred at 100 °C overnight, cooled to RT, filtered through a pad of Celite®, washed with EtOAc (180 mL) and concentrated to give a black oil. The crude material was purified by silica gel chromatography (20-100% EtOAc/hexanes). Desired fractions were combined and concentrated to give the title intermediate (1.55 g, 69 % yield) as a yellow solid, (m/z): [M+H] + calcd for 454.28 found 454.4. Preparation 33: (2S,4S)-2-{4-[4'-({4-[4-(2,2-Dimethyl-propionyl)-piperazin-l - yl]-piperidine-l-carbonyl}-amino)-2 , -trifluoromethoxy-biphenyl-4-yl]-lH-imidazol- 2-yl}-4-meth -pyrrolidine-l-carboxylic acid tert-but l ester

A solution of (25 , ,45')-4-methyl-2- {4-[4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)-phenyl]-lH-imidazol-2-yl}-pyrrolidine-l-carboxylic acid tert-butyl ester (104 mg, 0.23 mmol; Preparation 32), 4-[l-(2,2-dimethyl-propionyl)-piperidin-4-yl]-piperazine-l- carboxylic acid (4-bromo-3-trifluoromethoxy-phenyl)-amide (122 mg, 0.23 mmol;

Preparation 30) and sodium bicarbonate (100 mg, 1.19 mmol) in 1,4-dioxane (9.6 mL) and water (0.44 mL) was degassed with nitrogen for 20 min and then Pd2(dba) 3 (41.8 mg, 0.046 mmol) and 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (109 mg, 0.23 mmol) were introduced and the reaction was degassed with nitrogen for 15 min. The reaction solution was heated at 90 °C for 60 h, concentrated, diluted with methanol, filtered through a pad of Celite®, concentrated under vacuum, and purified by reverse phase HPLC. Desired fractions were combined and lyophilised overnight to afford a white powder. Impure fractions were combined and concentrated under vacuum, dissolved in a 1 : 1 acetic acid:water solution and purified by reverse phase HPLC to provide the di-TFA salt of the title intermediate (total 14.6 mg, 6 % yield),

(m/z): [M+H] + calcd for C 4 iH 54 F 3 7 0 6 782.41 found 782.9.

Preparation 34: (S)-Methoxycarbonylamino-(tetrahydro-pyran-4-yl)-acetic acid

A solution of (5)-amino-(tetrahydro-pyran-4-yl)-acetic acid (1 g, 6.28 mmol) in saturated aqueous sodium bicarbonate solution (12.32 mL, 125.6 mmol) was stirred until all solids were dissolved. Methyl chloroformate (0.97 mL, 12.56 mmol) was added dropwise, the reaction mixture was stirred for 1 h, and IN HC1 was added to adjust pH to 1. The reaction mixture was extracted with ethyl acetate (3 x 15 mL) and the combined organic extracts were dried over sodium sulfate, filtered, concentrated and dried overnight under vacuum to give the title intermediate (1.36 g, 99 % yield) as a white, sticky solid, (m/z): [M+H] + calcd for C 9 H 15 N0 5 218.10 found 218.3.

Example 1: ((S)-l-{(S)-2-[4-(4'-{[(S)-4-(l-Cyclopropanecarbonyl-piperid in-4- yl)-3-methyl-piperazine-l-carbonyl]-amino}-2'-trifluorometho xy-biphenyl-4-yl)-lH- imidazol-2-yl]- pyrrolidine-l-carbonyl}-2-methyl-propyl) -carbamic acid methyl ester

A mixture of ((5)-l- {(5)-2-[4-(4'-amino-2'-trifluoromethoxy-biphenyl-4-yl)-lH- imidazol-2-yl]-pyrrolidine-l-carbonyl}-2-methyl-propyl)-carb amic acid methyl ester (20 mg, 0.04 mmol) and / nitrophenyl chloroformate (8.9 mg, 0.04 mmol) in DMA (5 mL) was stirred at RT for 30 min and then cyclopropyl-[4-((5)-2-methyl-piperazin-l-yl)- piperidin-l-yl]-methanone 2 HCl (18 mg, 0.06 mmol; Preparation 6) was added followed by DIPEA (32 μϊ ^ , 0.18 mmol). The reaction mixture was stirred at RT for 1 h, concentrated, dissolved in 1 : 1 acetic acid:water (1.5 mL), and purified by reverse phase HPLC to provide the di-TFA salt of the title product (6.8 mg, 20 % yield) (m/z): [M+H] + calcd for C 4 2H 5 3F 3 8 0 6 823.40 found 823.6.

Example 2: [(S)-l-((S)-2-{4-[5'-Chloro-4'-({4-[l-(2,2-dichloro- cyclopropanecarbonyl)-piperidin-4-yl]-piperazine-l-carbonyl} -amino)-2'- trifluoromethoxy-biphenyl-4-yl]-lH-imidazol-2-yl}-4-methyl-p yrrolidine-l- carbonyl)- -methyl-propyl]-carbamic acid methyl ester

A solution of 4-[4-(5-chloro-4'- (2-[(25',45)- ^((^^-methoxycarbonylamino-S- methyl-butyryl)-4-methyl-pyrrolidin-2-yl]-lH-imidazol-4-yl}- 2-trifluoromethoxy- biphenyl-4-ylcarbamoyl)-piperazin-l-yl]-piperidine-l-carboxy lic acid tert-butyl ester 2TFA (12.94 mg, 0.012 mmol; Preparation 14) in 4 M HCl in 1,4-dioxane (0.21 mL) was stirred at RT for 30 min to provide the intermediate product {(S)-l-[(2S,4S)-2-(4- {5'- Chloro-4'- [(4-piperidin-4-yl-piperazine-l-carbonyl)-amino]-2'-trifluor omethoxy- biphenyl-4-yl}-lH-imidazol-2-yl)-4-methyl-pyrrolidine-l-carb onyl]-2-methyl-propyl}- carbamic acid methyl ester.

A solution of 0.5 M 2,2-dichlorocyclopropane- 1 -carboxylic acid in DMA (27.8 μί, 0.014 mmol), and HATU (5.28 mg, 0.014 mmol) in DMA (0.4 mL) was stirred at 50 °C for 15 min and then a solution of the intermediate product of the previous step (9.1 mg, 0.012 mmol) in DMA (0.58 mL, 6.25 mmol) was added followed by DIPEA (10.1 μί, 0.058 mmol). The reaction solution was stirred at 50 °C for 4 h, concentrated under vacuum and purified by reverse phase HPLC. Desired fractions were combined and lyophilized to provide the di-TFA salt of the title compound (8.2 mg, 61 % yield). (m/z): [M+H] + calcd for C 4 2H 5 oCl 3 F 3 8 0 6 925.29 found 925.8.

Examples 3-6:

Following the procedure of Example 2, the intermediate product of the first step (9.1 mg, 0.012 mmol) in DMA (0.58 mL, 6.25 mmol) was reacted with the appropriate reagents to provide the following compounds:

Example 7: [(S)-l-((2S,4S)-2-{5-Chloro-4-[4'-({4-[l-(2,2-dimethyl-propi onyl)- piperidin-4-yl]-piperazine-l-carbonyl}-amino)-2'-trifluorome thoxy-biphenyl-4-yl]- lH-imidazol-2-yl}-4-methyl-pyrrolidine-l-carbonyl)-2-methyl- propyl]-carbamic acid methyl ester

A mixture of {(5)-l-[(25',45 , )-2-(5-chloro-4-{4'-[(4-piperidin-4-yl-piperazine-l- carbonyl)-amino]-2'-trifluoromethoxy-biphenyl-4-yl}-lH-imida zol-2-yl)-4-methyl- pyrrolidine-l-carbonyl]-2-methyl-propyl}-carbamic acid methyl ester tri-HCl (10 mg, 0.01 mmol; Preparation 18) in DMA (1.5 mL) was stirred at RT to dissolution and then 2,2-dimethylpropanoyl chloride (1.4 μΙ_,, 0.01 1 mmol) was added followed by DIPEA (9.7 μΐ ^ , 0.056 mmol). The reaction mixture was stirred at RT for 5 min, concentrated by rotary evaporation, dissolved in 1 : 1 acetic acid:water (4 mL), purified by reverse phase HPLC, and lyophilized to provide the TFA salt of the title intermediate (4 mg) as a yellow powder, (m/z): [M+H] + calcd for C4 3 H 56 C1F 3 8 0 6 873.40, found 873.4.

Example 8: [(S)-l-((2S,4S)-2-{4-[4'-({4-[l-(2, 2-Dimethyl-propionyl)- piperidin-4-yl]-piperazine-l-carbonyl}-amino)-2'-trifluorome thoxy-biphenyl-4-yl]-5- fluoro-lH-imidazol-2-yl}-4-methyl-pyrrolidine-l-carbonyl)-2- methyl-propyl]- carbamic a

trifluoromethoxy-biphenyl-4-yl} - lH-imidazol-2-yl)-4-methyl-pyrrolidine- 1 - carbonyll-2-methyl-propyll-carbamic acid methyl ester

A solution of 4-[4-(4'- {5-fluoro-2-[(25',45)-l-((5 , )-2-methoxycarbonylamino-3- methyl-butyryl)-4-methyl-pyrrolidin-2-yl]- lH-imidazol-4-yl} -2-trifluoromethoxy- biphenyl-4-ylcarbamoyl)-piperazin-l-yl]-piperidine-l-carboxy lic acid tert-butyl ester di- TFA (11.5 mg, 0.01 mmol; Preparation 21) in 4 M of HQ in l,4-dioxane(0.26 mL) was stirred at RT for 30 min, and concentrated under vacuum to afford a cream-colored solid.

(b) \(S)-l-((2SAS)-2-l4-\4'-(l4-\l-(2, 2-Dimethyl-propionyl)-piperidin-4-yll-piperazine- 1 -carbonyl} -amino)-2'-trifluoromethoxy-biphenyl-4-yl1-5-fluoro- lH-imidazol-2-yl} -

4-methyl-pyrrolidine-l-carbonyl -2-methyl-propyl1-carbamic acid methyl ester

To the product of the previous step (8.1 mg, 0.01 mmoL) in DMA (0.52 mL) was added 2,2-dimethylpropanoyl chloride (1.29 μΐ,, 0.01 mmol) and DIPEA (9.01 μΐ,, 0.05 mmol). The reaction mixture was stirred at RT for 1 h and purified by reverse phase HPLC. Pure fractions were combined and lyophilised to provide the di-TFA salt of the title compound (4 mg, 33 % yield), (m/z): [M+H] + calcd for C 43 H 56 F 4 N 8 0 6 857.43, 858.43, found 858.7.

Examples 9-10:

Following the procedure of Example 8, the product of step (a) (8.1 mg,

0.01 mmol) in DMA (0.52 mL) was reacted with the appropriate reagents to provide the following compounds:

Ex. No. R Reagent Product cyclopropanoyl chloride 2 TFA salt (2.6 mg)

9 (0.959 μί, 0.010 mmol) (m/z): [M+H] + calcd for

DIPEA (9.01 μΐ, 0.05mmol) C 42 H 52 F 4 N 8 0 6 841.29 found 841.8

0.5 M (5)-(+)-2,2- 2 TFA salt (3 mg) (m/z): [M+H] + dimethylcyclopropane carboxylic calcd for C 44 H 56 F 4 8 0 6 869.43

10 >^ acid in DMA (25.1 μί, 0.013 found 870.0

mmol)

DIPEA (9.01 μί, 0.05mmol)

HATU (4.77mg, 0.013 mmol)

Example 11 : ((S)-l-{(2S,4S)-2-[4-(5'-Chloro-4'-{[4-((S)-4- dimethylcyclopropane-carbonyl-2-methyl-piperazin-l-yl)-piper idine-l-carbonyl]- amino}-2'-trifluoromethoxy-biphenyl-4-yl)-lH-imidazol-2-yl]- 4-methyl-pyrrolidine- l-carbonyl}- -methyl-propyl)-carbamic acid methyl ester

(a) ((S)- 1 - {(2 .45)-2-r4-(5'-Chloro-4'- { r4-((5)-2-methyl-piperazin- 1 -yl)-piperidine- 1 - carbonyll-amino} -2'-trifluoromethoxy-biphenyl-4-yl)-lH-imidazol-2-yll-4-meth yl- pyrrolidine- l-carbonyl}-2-methyl-propyl)-carbamic acid methyl ester

A solution of (5)-4-[l -(5-Chloro-4'- {2-[(25,45)-l -((5)-2-methoxycarbonylamino-

3-methyl-butyryl)-4-methyl-pyrrolidin-2-yl]- lH-imidazol-4-yl} -2-trifluoromethoxy- biphenyl-4-ylcarbamoyl)-piperidin-4-yl] -3 -methyl-piperazine- 1 -carboxylic acid tert-butyl ester 2 TFA (6.35 mg, 0.006 mmol; Preparation 24) in 4.0 M HCl in 1 ,4-dioxane (0.1 mL) was stirred at RT for 30 min and concentrated under vacuum to provide the title

intermediate, (m/z): [M+H] + calcd for CssHsoClFs sOj 803.35 found 803.8. (b) ((5y (2£45y2-r4-(5'-Chloro-4'- { r4^

methyl-piperazin-l-yl)-piperidine-l-carbonyl1-a

4-yl -lH-imidazol-2-yl1- 4-methyl-pyrrolidine-l-carbonyl}-2-methyl-propyl - carbamic acid methyl ester

A solution of 0.5 M (5)-(+)-2,2-dimethylcyclopropane carboxylic acid in DMA

(13.5 μί, 0.007 mmol) and HATU (2.56 mg, 0.007 mmol) in DMA (0.3 mL) was stirred at 50 °C for 15 min. and then the product of the previous step (4.5 mg, 0.006 mmol) and DIPEA (4.9 μί, 0.028 mmol) in DMA (0.3 mL) was added. The solution was stirred at 50 °C for 2 h, dissolved in 1 : 1 acetic acid:water (1.5 mL), purified by reverse phase HPLC to provide the di-TFA salt of the title product (2 mg, 40 % yield), (m/z): [M+H] + calcd for C 4 5H 58 C1F 3 N 8 0 6 899.41 found 901.8.

Example 12: [(S)-l-((2S,4S)-2-{4-[5'-Chloro-4'-({4-[(S)-4-(2,2-dimethyl- propionyl)-2-methyl-piperazin-l-yl]-piperidine-l-carbonyl}-a mino)-2'- trifluoromethoxy-biphenyl-4-yl]-lH-imidazol-2-yl}-4-methyl-p yrrolidine-l- carbonyl)-2-

Following a procedure similar to that of Example 11, the product of Example 1 1, step (a) (4.5 mg, 0.006 mmol) in DMA (0.3 mL) was combined with 2,2-dimethyl- propanoyl chloride (0.7 μί, 0.006 mmol) and DIPEA (4.89 μΓ) to provide the di-TFA salt of the title compound (2 mg, 40 % yield), (m/z): [M+H] + calcd for C 4 4H 58 C1F 3 N 8 0 6 887.41 found 887.8. Example 13: 4-[4-(4'-{2-[(2S,4S)-4-Methoxy-l-((S)-2- methoxycarbonylamino-3-methyl-butyryl)- pyrrolidin-2-yl]-lH-imidazol-4-yl}-2- trifluoromethoxy-biphenyl-4-ylcarbamoyl)- piperazin-l-yl]-piperidine-l-carboxylic acid teri-b

A solution of [(5)-l-((25,45)-4-methoxy-2-{4-[4-(4,4,5,5-tetramethyl-l,3,2 - dioxaborolan-2-yl)-phenyl]-lH-imidazol-2-yl}-pyrrolidine-l-c arbonyl)-2-methyl-propyl]- carbamic acid methyl ester (35.5 mg, 0.067 mmol; Preparation 26), 4-[4-(4-bromo-3- trifluoromethoxy-phenylcarbamoyl)-piperazin- 1 -yl]-piperidine- 1 -carboxylic acid tert- butyl ester (37.1 mg, 0.067 mmol) and sodium bicarbonate (29.4 mg, 0.35 mmol) in 1,4- dioxane (2.8 mL) and water (129 μΚ) was degassed with nitrogen for 20 min. Pd2(dba)3 (12.33 mg, 0.013 mmol) and 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (Xphos) (32.09 mg, 0.067 mmol) were added and the reaction was degassed with nitrogen for 15 min. The resulting solution was heated at 90 °C for 60 h, concentrated, diluted with methanol, filtered through a pad of Celite®, concentrated under vacuum, and purified by reverse phase HPLC. Fractions containing product were combined and lyophilised overnight to afford a white powder. Impure fractions were combined and concentrated under vacuum, dissolved in 1 : 1 acetic acid:ACN solution to provide the di-TFA salt of the title compound (40 mg, 54 % yield) (m/z): [M+H] + calcd 871.43 found 871.8.

Example 14: [(S)-l-((2S,4S)-2-{4-[4'-({4-[4-(2,2-Dimethyl- cyclobutanecarbonyl)-piperazin-l-yl]-piperidine-l-carbonyl}- amino)-2'- trifluoromethoxy-biphenyl-4-yl]-lH-imidazol-2-yl}-4-methoxy- pyrrolidine-l- carbonyl)- -methyl-propyl]-carbamic acid methyl ester

(a) {ffl-l-r(2 ^^-4-Methoxy-2-( ' 4- {4'-r( ' 4-piperidin-4-yl-piperazine-l-carbonyl - amino1-2'-trifluoromethoxy-biphenyl-4-yl}-lH-imidazol-2-yl -pyrrolidine-l- carbonyll-2-methyl-propyll-carbamic acid methyl ester

A solution of 4-[4-(4'- {2-[(25',45)-4-Methoxy-l-((5 , )-2-methoxycarbonylamino-3- methyl-butyryl)-pyrrolidin-2-yl]-lH-imidazol-4-yl}-2-trifluo romethoxy-biphenyl-4- ylcarbamoyl)-piperazin-l-yl]-piperidine-l-carboxylic acid tert-butyl ester 2 TFA (9.7 mg, 0.009 mmol; Example 13) in 4.0 M HC1 in 1,4-dioxane (0.22 mL) was stirred at RT for 30 min, and concentrated under vacuum to afford the title intermediate as a cream-colored powder.

(b) \(S)- 1 -((2SAS)-2- { 4-Γ4'-( { 4-r4-(2,2-Dimethyl-cvclobutanecarbonyl)-piperazin- 1 -yll- piperidine-l-carbonyl}-amino)-2'-trifluoromethoxy-biphenyl-4 -yl1-lH-imidazol-2- yl}-4-methoxy-pyrrolidine-l-carbonyl)-2-methyl-propyl1-carba mic acid methyl ester A solution of 2,2-dimethyl-cyclobutane carboxylic acid (1.36 mg, 0.01 1 mmol) and HATU (4.03 mg, 0.01 1 mmol) in DMA (0.44 mL) was stirred at 50 °C for 15 min. A solution of the intermediate product of the previous step in DMA (0.2 mL) was added, followed by DIPEA (7.7 μί, 0.044 mmol). The reaction mixture was stirred at 50 °C for 2 h, concentrated under vacuum, dissolved in 1 : 1 acetic acid:ACN (1.5 mL) and purified by reverse phase HPLC to provide the di-TFA salt of the title compound (0.9 mg). (m/z): [M+H] + calcd for C44H59F3N8O7881.45 found 881.8. Example 15: [(S)-l-((2S,4S)-2-{7-[4-({4-[l-(2,2-Dimethyl-propionyl)- piperidin-4-yl]-piperazine-l-carbonyl}-amino)-2-trifluoromet hoxy-phenyl]-3H- naphtho[l,2-< ]imidazol-2-yl}-4-methyl-pyrrolidine-l-carbonyl)-2-methyl-pr opyl]- carbamic ac

(a) {ffl-2-Methyl (2 ^^-4-methyl-2-( ' 7- {4-r( ' 4-piperidin-4-yl-piperazine-l-carbonyl)- amino1-2-trifluoromethoxy-phenyl} -3H-naphthor 1 ,2-^1imidazol-2-yl)-pyrrolidine- 1 - carbonyll-propyll-carbamic acid methyl ester

A solution of 4-[4-(4- {2-[(2S,4S)- 1 -((S)-2-methoxycarbonylamino-3-methyl- butyryl)-4-methyl^yrrolidin-2-yl]-3H-naphth[l,2-(i]imidazol- 7-yl}-3-trifluoromethoxy- phenylcarbamoyl)-piperazin-l-yl]-piperidine-l-carboxylic acid tert-butyl ester 2 TFA (8.8 mg, 0.008 mmol; Preparation 29) in 4.0 M HCl in l,4-dioxane(0.20 mL) was stirred at RT for 30 min and concentrated under vacuum to afford the title intermediate as a white powder, (m/z): [M+H] + calcd for C 40 H 49 F 3 8 O 5 779.38 found 779.8.

(b) \(S)-l -((2SAS)-2- Ι7-Γ4-Π4-Γ1 -(2 ,2-Dimethyl-propionyl)-piperidin-4-yl1-piperazine- 1 - carbonyl}-amino)-2-trifluoromethoxy-phenyl1-3H-naphthori,2-( i1imidazol-2-yl}-4- methyl-pyrrolidine-l-carbonyl)-2-methyl-propyl1-carbamic acid methyl ester

To a solution of the product of the previous step in DMA (0.40 mL) was added 2,2-dimethylpropanoyl chloride (1.0 μί, 0.008 mmol) and DIPEA (6.9 μί, 0.04 mmol). The reaction mixture was stirred at RT for 30 min, concentrated undervacuum, dissolved in 1 : 1 acetic acid:water (1.5 mL) and purified by reverse phase HPLC to provide the di- TFA salt of the title compound (5 mg, 54 % yield), (m/z): [M+H] + calcd for

C 45 H 57 F 3 8 0 6 863.44 found 863.8. Example 16: ((S)-1-{(2S,4S 2-[7-(4-{[4-(l-Cyclopropanecarbonyl-piperidin- 4-yl)-piperazine-l-carbonyl]- amino}-2-trifluoromethoxy-phenyl)-3H-naphth[l, 2- d]imidazol-2-yl]-4-methyl-pyrrolidine-l-carbonyl}-2-methyl-p ropyl)-carbami acid methyl ester

Following the procedure of Example 15, substituting cylopropanecarbonyl chloride (0.76 μΐ,, 0.008 mmol) for the 2,2-dimethylpropanoyl chloride (1.0 μΐ,, 0.008 mmol), the di-TFA salt of the title compound was obtained (5 mg, 63 % yield), (m/z): [M+H] + calcd for C 4 4H5 3 F 3 8 0 6 847.40 found 847.8.

Example 17: [(S)-2-((2S,4S)-2-{4-[4'-({4-[4-(2,2-Dimethyl-propionyl)- piperazin-l-yl]-piperidine-l-carbonyl}-amino)-2'-trifluorome thoxy-biphenyl-4-yl]- lH-imidazol-2-yl}-4-methyl-pyrrolidin-l-yl)-2-oxo-l-(tetrahy dro-pyran-4-yl)-ethyl]- carbamic acid methyl ester

(a) 4-ri-(2.2-Dimethyl-propionyl)-piperidin-4-yl1-piperazine-l-c arboxylic acid {4'-Γ2-

((2 ^<S f )-4-methyl-pyrrolidin-2-yl -lH-imidazol-4-yl1-2-trifluoromethoxy-biphenyl-4- yl| -amide

A solution of (25 , ,45')-2- {4-[4'-({4-[4-(2,2-dimethyl-propionyl)-piperazin-l-yl]- piperidine-l-carbonyl}-amino)-2'-trifluoromethoxy-biphenyl-4 -yl]-lH-imidazol-2-yl}-4- methyl-pyrrolidine- 1 -carboxylic acid tert-butyl ester 2 TFA (12.3 mg, 0.012 mmol; Preparation 33) in 4 M HC1 in 1,4-dioxane (0.23 mL) was stirred at RT for 30 min and concentrated under vacuum to provide the title intermediate as a cream-colored solid, (b) rffl-2-((2 ^5)-2-{4 4'-({4 4-(2.2-Dimethyl-propionyl)-piperazin-l-yl1-piperidine- l-carbonyl}-amino)-2'-trifluoromethoxy-biphenyl-4-yl1-lH-imi dazol-2-yl}-4-methyl- pyrrolidin-l-yl)-2-oxo-l-( ' tetrahvdro-pyran-4-yl)-ethyl1-carbamic acid methyl ester

A solution of (5)-methoxycarbonylamino-(tetrahydro-pyran-4-yl)-acetic acid (3.44 mg, 0.016 mmol; Preparation 34) and HATU (6.02 mg, 0.016 mmol) in DMA (0.3 mL) was stirred at 50 °C for 30 min. A solution of the product of the previous step in DMA (0.61 mL) was added, followed by DIPEA (10.6 μί, 0.061 mmol). The solution was stirred at 50 °C for 1 h, concentrated under vacuum, and purified by reverse phase HPLC. Pure fractions were combined and lyophilised overnight to provide the di-TFA salt of the title compound (2 mg, 16 % yield), (m/z): [M+H] + calcd for C45H5 9 F 3 8 O7 881.45 found 881.7.

Using similar synthetic procedures, the compounds of Tables 1 to 9 were prepared, where a blank in any column denotes hydrogen.

Table 1

(#) For all compounds, when the substituent R is present, the orientation of the chiral carbon atom bearing the substituent R is (R). Table 2

Table 3

Table 4

Table 5

Table 6

Table 8

Table 9

Biological Assays

The hepatitis C virus has been classified into six major different genotypes on the basis of nucleotide sequence, and further divided into subtypes within genotypes.

Compounds of the invention demonstrated inhibition of HCV replication in one or more of the following HCV replicon assays.

Assay 1: HCV Genotype lb Replicon Assay

The HCV genotype lb replicon cell line was obtained from Apath LLC

(Brooklyn, NY) (APC144; Huh7 cell background). This subgenomic replicon contains the N-terminus of the HCV core protein fused to the neomycin-resistance selectable marker. The EMCV IRES lies downstream and drives expression of humanized Renilla luciferase fused to the non-structural proteins NS3-NS5B. This cell line was used to determine compound potency using the luciferase activity readout as a measurement of compound inhibition of replicon levels.

Cells were grown at 37°C in a 5% CO 2 humidified incubator in DMEM

(Invitrogen) with 10% FBS (HyClone), lx NEAA (Invitrogen), lx Pen-Strep

(Invitrogen), and 500μg/mL G418 (Invitrogen). On day 1 of the assay, cells were plated at 10,000 cells/well in white 96-well tissue culture plates (Costar) in 200μί media lacking G418. Four hours later, once the cells have adhered, the media was removed and replaced with media (no G418) containing dose-responses of test compounds.

Compounds were initially diluted in DMSO and then diluted another 200 x in media to bring the final DMSO concentration down to 0.5%. The cells were incubated with test compounds for 48 hours. At the end of the incubation period, media and compound were removed from the plates and the luciferase activity was determined using Promega Renilla-Glo reagents.

To analyze the data, the luciferase activity was plotted vs. the compound concentration, and EC5 0 values were determined from a 4-parameter robust fit model with the GraphPad Prism software package (GraphPad Software, Inc., San Diego, CA).

Results are expressed as the negative decadic logarithm of the EC5 0 value, pECso.

Test compounds having a higher pECso value in this assay show greater inhibition of HCV genotype lb replication. Compounds of the invention tested in this assay typically exhibited pECso values between about 7 and about 12. Assay 2: HCV Genotype la Replicon Assay

The HCV genotype la replicon cell line was obtained from Apath LLC (APC89; Huh7.5 cell background). This subgenomic replicon contains the N-terminus of the HCV core protein fused to the neomycin-resistance selectable marker. The EMCV IRES lies downstream and drives expression of the non-structural proteins NS3-NS5B. Compound potencies were determined using the NS3 -specific protease activity in lysates as a measurement of compound inhibition of replicon levels.

Cells were grown at 37°C in a 5% CO 2 humidified incubator in DMEM

(Invitrogen) with 10% FBS (HyClone), lx NEAA (Invitrogen), lx Pen-Strep

(Invitrogen), and 850μg/mL G418 (Invitrogen). On day 1 of the assay, cells were plated at 15,000 cells/well in black 96-well tissue culture plates (Costar) in 200μί media lacking G418. Four hours later, once the cells had adhered, the media was removed and replaced with media (no G418) containing dose-responses of test compounds. Compounds were initially diluted in DMSO and then diluted another 200x in media to bring the final DMSO concentration down to 0.5%. The cells were incubated with test compounds for 48 or 72 hours. At the end of the incubation period, media and compound were removed from the plates.

To determine the NS3 -specific protease activity in lysates, the cells were lysed at room temperature in 50μΕΛνε11 of 50mM Hepes pH 7.5, 150mM NaCl, 15% Glycerol, 0.15% Triton X-100, lOmM DTT for 20 minutes with shaking. 50μί of an S3/4a protease-specific FRET substrate (Anaspec RET SI Cat#22991) was then added to the wells at a final concentration of 15μΜ. The plates were incubated at 37°C for 20 minutes, which corresponds to a timepoint at which the protease activity is still in the linear phase. Protease activity was determined by measuring fluorescence (Excitation: 340 nm;

Emission: 509nm).

To analyze the data, the fluorescence was plotted vs. the compound concentration, and EC50 values were determined from a 4-parameter robust fit model using GraphPad Prism software. Compounds of the invention tested in this assay typically exhibited PEC5 0 values between about 6 and about 1 1.5.

Assay 3: Replicon Assays Against Resistant Mutants

To create replicon cells with resistant mutations of interest, the mutation was first introduced into the parental plasmid by site-directed mutagenesis. Mutations in genotype lb included L3 IV, Y93H, and the L31 V/Y93H double mutant. Mutations in genotype la included Q30R and L3 IV. The replicon plasmid was then linearized and in vitro transcribed to RNA. The RNA was used to stably transfect Huh7 cells by electroporation, and new cell lines were selected with 50C^g/mL G418. Potencies of test compounds against these mutant cell lines were determined as previously described above for the HCV Genotype lb and la replicon assays.

Potencies of test compounds against additional mutations of interest were determined using transient transfection assays. These mutants included and genotype la Y93C, Y93H, M28T, Q30E, Q30K. The mutation was first introduced into the parental plasmid by site-directed mutagenesis. The replicon plasmid was then linearized and in vitro transcribed to RNA. The RNA was used to transiently transfect Huh-LUNET cells (obtained from ReBLikon GmbH, Schriesheim, Germany) by electroporation, and the potencies of test compounds against the mutants were determined as previously described.

Assay 4: Additional Replicon Assays

Potencies of test compounds against NS5A sequences of other genotypes were determined by creating intergenotypic chimeras. The entire NS5A gene from genotypes 2-6, or the nucleotide sequence encoding amino acids 1 1-1 18 of NS5A, was subcloned into a genotype lb replicon. These chimeric replicon plasmids were then linearized and in vitro transcribed to RNA. The RNA was used to transiently or stably transfect Huh- LUNET cells by electroporation, and the potencies of test compounds against the chimeras were determined as previously described.

Assay Results

All of the compounds of Examples 1 to 17 and Tables 1 to 9 were tested in one or more of the assays described above. For example, the following results were obtained in the HCV genotype la and lb replicon assays where A represents a pECso value between 6 and 8 (EC5 0 between 1 μΜ and 10 nM), B represents pECso between 8 and 9 (EC5 0 between 1 and 10 nM), C represents pECso between and 9 and about 10, (EC5 0 between 1 nM and 0.1 nM), and D represents pEC 50 >10 (EC50 < 0.1 nM). Example Genotype Genotype Example : Genotype Genotype

No. la lb No. la lb

1 C 1-22 C

2 B 1-23 c

3 C 1-24 c

4 c 1-25 c

5 c 1-26 c

6 c 1-27 c

7 D D 1-28 c

8 D 1-29 A

9 D 1-30 A

10 D 1-31 A

11 D D 1-32 A

12 C 1-33 C

13 C 1-34 c

14 C 1-35 c

15 D 1-36 c

16 D D 1-37 B

17 C D 1-38 B

1-39 B

Table 1 1-40 B

1-1 C D 1-41 B

1-2 B 1-42 B

1-3 C 1-43 A

1-4 B 1-44 A

1-5 B 1-45 B

1-6 A

1-7 B Table 2

1-8 C D 2-1 D

1-9 B 2-2 D

1-10 B 2-3 D

1-11 C 2-4 D

1-12 C D 2-5 D D

1-13 C D 2-6 D D

1-14 C D 2-7 c

1-15 B 2-8 C

1-16 C D 2-9 D D

1-17 C D 2-10 C

1-18 C 2-11 D

1-19 C 2-12 C

1-20 C 2-13 D

1-21 B 2-14 C While the present invention has been described with reference to the specific embodiments thereof, it should be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the true spirit and scope of the invention. In addition, many modifications may be made to adapt a particular situation, material, composition of matter, process, process step or steps, to the objective, spirit and scope of the present invention. All such modifications are intended to be within the scope of the claims appended hereto. Additionally, all publications, patents, and patent documents cited hereinabove are incorporated by reference herein in full, as though individually incorporated by reference.