Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
HEPATITIS C VIRUS INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2014/070964
Kind Code:
A1
Abstract:
Hepatitis C virus inhibitors having the general formula (I) are disclosed. Compositions comprising the compounds and methods for using the compounds to inhibit HCV are also disclosed.

Inventors:
SUN LI-QIANG (US)
GILLIS ERIC P (US)
MULL ERIC (US)
BOWSHER MICHAEL S (US)
ZHAO QIAN (US)
SCOLA PAUL MICHAEL (US)
Application Number:
PCT/US2013/067649
Publication Date:
May 08, 2014
Filing Date:
October 31, 2013
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BRISTOL MYERS SQUIBB CO (US)
International Classes:
C07K5/083; A61K38/06; A61P31/14
Foreign References:
US6608027B12003-08-19
Other References:
"Remington's Pharmaceutical Sciences", 1999, MACK PUBLISHING COMPANY
BERGER A.; SCHECHTER I., TRANSACTIONS OF THE ROYAL SOCIETY LONDON SERIES, vol. B257, 1970, pages 249 - 264
PHARMACEUTICAL RESEARCH, vol. 3, no. 6, 1986, pages 318
P SIMMONDS; KA ROSE; S GRAHAM; SW CHAN; F MCOMISH; BC DOW; EA FOLLETT; PL YAP; H MARSDEN, J. CLIN. MICROBIOL., vol. 31, no. 6, 1993, pages 1493 - 1503
YANAGI,M.; PURCELL,R.H.; EMERSON,S.U.; BUKH, J., PROC. NATL. ACAD. SCI. U.S.A., vol. 94, no. 16, 1997, pages 8738 - 8743
YANAGI,M.; ST CLAIRE,M.; SHAPIRO,M.; EMERSON,S.U.; PURCELL,R.H.; BUKH, J., VIROLOGY, vol. 244, no. 1, 1998, pages 161 - 172
GALLINARI P; PAOLINI C; BRENNAN D; NARDI C; STEINKUHLER C; DE FRANCESCO R., BIOCHEMISTRY., vol. 38, no. 17, 1999, pages 5620 - 32
GALLINARI P; BRENNAN D; NARDI C; BRUNETTI M; TOMEI L; STEINKUHLER C; DE FRANCESCO R, J VIROL., vol. 72, no. 8, 1998, pages 6758 - 69
TALIANI ET AL., ANAL. BIOCHEM., vol. 240, no. 2, 1996, pages 60 - 67
LOHMANN V; KOMER F; KOCH J; HERIAN U; THEILMANN L; BARTENSCHLAGER R., SCIENCE, vol. 285, no. 5424, 1999, pages 110 - 3
KRIEGER N; LOHMANN V; BARTENSCHLAGER R, J. VIROL., vol. 75, no. 10, 2001, pages 4614 - 4624
BLIGHT KJ; KOLYKHALOV, AA; RICE, CM, SCIENCE, vol. 290, no. 5498, pages 1972 - 1974
YANAGI M; PURCELL RH; EMERSON SU ET AL.: "Transcripts from a single full-length cDNA clone of hepatitis C virus are infectious when directly transfected into the liver of a chimpanzee.", PROC NATL ACAD SCI USA, vol. 94, no. 16, 1997, pages 8738 - 8743, XP002918288, DOI: doi:10.1073/pnas.94.16.8738
Attorney, Agent or Firm:
MINGO, Pamela A. et al. (P.O. Box 4000Princeton, New Jersey, US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A compound of formula (I)

(I),

pharmaceutically acceptable salt thereof, wherein

p is 1 or 2;

is a single or double bond;

m is 0, 1, or 2;

R1 is selected from

wherein R1 is attached to the parent molecular moiety through any substitutable carbon atom in the group;

n is 0, 1, 2, 3, 4, 5, or 6;

o is 0, 1, 2, 3, 4, or 5;

q is 0, 1, 2, 3, or 4; X is selected from CH and N;

XI is selected from CH and N;

X2 and X3 are independently selected from CH, C(Ra) and N; provided that at least one of X1, X2, and X3 is other than N;

X4 is selected from CH and CRa;

one of X5, X6, X7, and X8 is N and the others are selected from CH and CRa; X9 is selected from CRa, CH, and N;

each Ra is independently selected from alkenyloxy, alkoxy, alkoxyalkoxy, alkoxycarbonyl, alkyl, benzodioxanyl, carboxamido, carboxy, carboxyalkoxy, cyano, cycloalkyl, cycloalkylalkoxy, cycloalkyloxy, deuteroalkoxy, dialkylamino, halo, haloalkyl, haloalkoxy, haloalkoxycarbonyl, hydroxy, imidazolyl, morpholinyl, oxazolyl, phenyl, piperazinyl, pyrazolyl, pyridinyl, pyrrolidinyl, thiazolyl, and - NRqRq , wherein the imidazolyl, the morpholinyl, the oxazolyl, the phenyl, the piperazinyl, the pyridinyl, the pyrrolidinyl, and the thiazolyl are optionally substituted with one or two groups independently selected from alkoxy, alkyl, alkylsulfonyl, halo, haloalkoxy, haloalkyl, and morpholinyl; and wherein two adjacent Ra groups, together with the carbon atoms to which they are attached, can optionally form a ring selected from dioxanyl, dioxolanyl, furanyl, morpholinyl, pyranyl, and phenyl, wherein the ring is optionally substituted with one or two groups independently selected from alkyl and halo;

Rb is alkyl;

Rx is selected from methyl and ethyl;

Rz and Rz are independently selected from hydrogen and hydroxy; provided that when is a double bond, Rz and Rz are each hydrogen;

R2 is selected from hydrogen, alkyl, deuteroalkyl, halo, haloalkoxy, haloalkyl, and hydroxyalkyl;

R3 is selected from hydrogen, alkoxyalkoxycarbonyl, alkoxycarbonyl, alkylaminocarbonyl, alkylcarbonyl, cycloalkylalkoxycarbonyl, cycloalkylcarbonyl, cycloalkyloxycarbonyl, deuteroalkoxycarbonyl, deuterohaloalkoxycarbonyl, dialkylaminocarbonyl, dialkylaminocarbonylcarbonyl, haloalkoxycarbonyl, haloalkylaminocarbonyl, haloalkylcarbonyl, heterocyclylcarbonyl,

heterocyclyloxycarbonyl, phenylcarbonyl, and phenyloxycarbonyl, wherein the cycloalkyl part of the cycloalkylalkoxycarbonyl, the cycloalkylcarbonyl, and the cycloalkyloxycarbonyl, the heterocyclyl part of the heterocyclylcarbonyl and the heterocyclyloxycarbonyl, and the phenyl part of the phenylcarbonyl and the phenyloxycarbonyl, is optionally substituted with one , two, or three groups independently selected from alkyl, alkylamino, alkylcarbonyl, cycloalkyl, dialkylamino, halo, haloalkoxy, and haloalkyl;

one of Rq and Rq is selected from hydrogen and alkyl and the other is selected from alkylcarbonyl and phenylcarbonyl; and

one of Y and Y' is oxygen and the other is CH2.

2. A compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein p is 1.

3. A compound of claim 2, or a pharmaceutically acceptable salt thereof, wherein is a double bond.

4. A compound of claim 3, or a pharmaceutically acceptable salt thereof, wherein m is 1.

5. A compound of claim 4 wherein R1 is

; wherein

n is 1, 2, 3, or 4; and

each Ra is independently selected from alkoxy, halo, and pyridinyl, wherein the pyridinyl is optionally substituted with one alkoxy group or wherein two adjacent R' groups, together with the carbon atoms to which they are attached, can optionally form a ring selected from dioxanyl and pyranyl.

6. A compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R2 is selected from alkyl and haloalkyl.

7. A compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from alkoxycarbonyl and haloalkoxycarbonyl.

8. A compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein

p is 1;

is a double bond;

m is 1.

R1 is

; wherein

n is 1, 2, 3, or 4; and

each Ra is independently selected from alkoxy, halo, and pyridinyl, wherein the pyridinyl is optionally substituted with one alkoxy group or wherein two adjacent Ra groups, together with the carbon atoms to which they are attached, can optionally form a ring selected from dioxanyl and pyranyl;

R2 is selected from alkyl and haloalkyl; and

R3 is selected from alkoxycarbonyl and haloalkoxycarbonyl.

9. A compound selected from

1,1,1 -trifhioro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7- ethyl-2-((7-fluoro-6-methoxyisoquinolin- 1 -yl)oxy)-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifhioro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-

((3 ,6-dimethoxyisoquinolin- 1 -yl)oxy)-7-ethyl-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate; 1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7- ethyl-2-((4-methoxyisoquinolin- 1 -yl)oxy)-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)- 14a- ((cyclopropylsulfonyl)carbamoyl)-7-ethyl-2-((4-methoxyisoquinolin-l-yl)oxy)-9- methyl-5,16-dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S, 13aS, 14aR, 16aS,Z)-7-ethyl-2 ((6-methoxyisoquinolin- 1 -yl)oxy)- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)- 5,16-dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7- ethyl-2-((6-methoxyisoquinolin- 1 -yl)oxy)-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate]oxadiazacyclopentadecin-6- yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)- 14a- ((cyclopropylsulfonyl)carbamoyl)-7-ethyl-2-((6-methoxyisoquinolin-l-yl)oxy)-9- methyl-5,16-dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)- 14a- ((cyclopropylsulfonyl)carbamoyl)-2-((3 ,6-dimethoxyisoquinolin- 1 -yl)oxy)-7-ethyl-9- methyl-5,16-dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S, 13aS, 14aR, 16aS,Z)-7-ethyl-2 ((7-fluoro-6-methoxyisoquinolin- 1 -yl)oxy)- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate; 1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S, 13aS, 14aR, 16aS,Z)-2-((3 ,6- dimethoxyisoquinolin- 1 -yl)oxy)-7-ethyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S, 13aS, 14aR, 16aS,Z)-7-ethyl-2 ((4-methoxyisoquinolin- 1 -yl)oxy)- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)- 5,16-dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7- ethyl-2-((7-fluoro-6-methoxyisoquinolin- 1 -yl)oxy)- 14a-((( 1 - (fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-9-methyl-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2- ((3 ,6-dimethoxyisoquinolin- 1 -yl)oxy)-7-ethyl- 14a-((( 1 - (fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-9-methyl-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7- ethyl- 14a-((( 1 -(fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-2-((4- methoxyisoquinolin- 1 -yl)oxy)-9-methyl-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7- ethyl-2-((7-methoxy-3 -(trifluoromethyl)quinoxalin-2-yl)oxy)-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

3,3-difluoro-2-methylbutan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2 ((7-methoxy-3 -(trifluoromethyl)quinoxalin-2-y l)oxy)-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7- ethyl-2-((8-fluoro-9-methoxy-2,3-dihydro-lH-pyrano[2,3-c]isoquinolin-6-yl)oxy)-9- methyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7- ethyl-2-((6-fluoro-2,7-dimethoxyquinolin-4-yl)oxy)-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2- ((2,7-dimethoxyquinolin-4-yl)oxy)-7-ethyl-9-methyl-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

3,3-difluoro-2-methylbutan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((2,7- dimethoxyquinolin-4-yl)oxy)-7-ethyl-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

tert-butyl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((6-methoxyisoquinolin-l - yl)oxy)-7,9-dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((6 methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((4 methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((4 ethoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((7 fluoro-6-methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

3,3-difluoro-2-methylbutan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((7- fluoro-6-methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((7 fluoro-4-methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

3,3-difluoro-2-methylbutan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((7- fluoro-4-methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2- ((2,3-dihydro-[l,4]dioxino[2,3-i^isoquinolin-7-yl)oxy)-7,9-dimethyl-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate; 3,3-difluoro-2-methylbutan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((2,3- dihydro-[ 1 ,4]dioxino[2,3-f]isoquinolin-7-yl)oxy)-7,9-dimethyl- 14a-(((l - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((3 (5-isopropoxypyridin-2-yl)-6-methoxyisoquinolin-l-yl)oxy)-7,9-dimethyl-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

3,3-difluoro-2-methylbutan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((3-(5- isopropoxypyridin-2-yl)-6-methoxyisoquinolin-l-yl)oxy)-7,9-dimethyl-14a-(((l- methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)- 14a- ((( 1 -(fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-2-((6-methoxyisoquinolin- 1 - yl)oxy)-7,9-dimethyl-5,16-dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15, 16,16a- tetradecahydro- 1 H-cyclopropa[i]pyrrolo[ 1 ,2-e] [ 1 ,5 ,8]oxadiazacyclopentadecin-6- yl)carbamate;

3,3-difluoro-2-methylbutan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-14a-(((l- (fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-2-((6-methoxyisoquinolin-l-yl)oxy) 7,9-dimethyl-5,16-dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)- 14a- ((( 1 -(fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-2-((4-methoxyisoquinolin- 1 - yl)oxy)-7,9-dimethyl-5,16-dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15, 16,16a- tetradecahydro- 1 H-cyclopropa[i]pyrrolo[ 1 ,2-e] [ 1 ,5 ,8]oxadiazacyclopentadecin-6- yl)carbamate;

l,l-difluoro-2-methylpropan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-14a- ((( 1 -(fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-2-((4-methoxyisoquinolin- 1 - yl)oxy)-7,9-dimethyl-5,16-dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15, 16,16a- tetradecahydro- 1 H-cyclopropa[i]pyrrolo[ 1 ,2-e] [ 1 ,5 ,8]oxadiazacyclopentadecin-6- yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((7 fluoro-5 -methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

3,3-difluoro-2-methylbutan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((7- fluoro-5 -methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1 , 1 -difluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((7- fluoro-5 -methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((7 fluoro-5 -methoxyisoquinolin- 1 -yl)oxy)- 14a-((( 1 -

(fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-7,9-dimethyl-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

3,3-difluoro-2-methylbutan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((7- fluoro-5 -methoxyisoquinolin- 1 -yl)oxy)- 14a-((( 1 -

(fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-7,9-dimethyl-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1 , 1 -difluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((7- fluoro-5 -methoxyisoquinolin- 1 -yl)oxy)- 14a-((( 1 -

(fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-7,9-dimethyl-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate; 3,3-difluoro-2-methylbutan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((2,3- dihydro- 1 H-pyrano [2,3 -c]isoquinolin-6-yl)oxy)-7-ethyl-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2- ((2,3-dihydro-lH-pyrano[2,3-c]isoquinolin-6-yl)oxy)-7-ethyl-9-methyl-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

3,3-difluoro-2-methylbutan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((2,3- dihydro- 1 H-pyrano[2,3-c]isoquinolin-6-yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

l,l-difluoro-2-methylpropan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((2,3 dihydro- 1 H-pyrano[2,3-c]isoquinolin-6-yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2- ((2,3-dihydro-lH-pyrano[2,3-c]isoquinolin-6-yl)oxy)-7,9-dimethyl-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((9- methoxy-2,3-dihydro-lH-pyrano[2,3-c]isoquinolin-6-yl)oxy)-7,9-dimethyl-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7- ethyl-2-((9-methoxy-2,3-dihydro-lH-pyrano[2,3-c]isoquinolin-6-yl)oxy)-9-methyl- 14a-((( 1 -methylcy clopropyl)sulfonyl)carbamoy l)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

3,3-difiuoro-2-methylbutan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2- ((9-methoxy-2,3-dihydro-lH-pyrano[2,3-c]isoquinolin-6-yl)oxy)-9-methyl-14a-(((l- methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

3,3-difiuoro-2-methylbutan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((9- methoxy-2,3-dihydro-lH-pyrano[2,3-c]isoquinolin-6-yl)oxy)-7,9-dimethyl-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1 , 1 -difiuoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((9- methoxy-2,3-dihydro-lH-pyrano[2,3-c]isoquinolin-6-yl)oxy)-7,9-dimethyl-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1 , 1 -difluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7-ethyl- 2-((9-methoxy-2,3-dihydro-lH-pyrano[2,3-c]isoquinolin-6-yl)oxy)-9-methyl-14a- ((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

tert-butyl ((2R,6S,7R,9S, 13aS, 14aR, 16aS,Z)-2-((4-methoxyisoquinolin-l - yl)oxy)-7,9-dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,8,9,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[e]pyrrolo[2,l- i] [ 1 ,7, 10]oxadiazacyclopentadecin-6-yl)carbamate;

tert-butyl ((2R,6S,7R,9S, 13aS, 14aR, 16aS,Z)-2-((6-methoxyisoquinolin-l - yl)oxy)-7,9-dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,8,9,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[e]pyrrolo[2,l- i] [ 1 ,7, 10]oxadiazacyclopentadecin-6-yl)carbamate;

tert-butyl ((2R,6S,7R,9S,13aS,14aR,16aS,Z)-14a- ((cyclopropylsulfonyl)carbamoyl)-2-((6-methoxyisoquinolin-l-yl)oxy)-7,9-dimethyl- 5,16-dioxo-2,3,5,6,7,8,9,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[e]pyrrolo[2, 1 -i] [ 1 ,7, 10]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7R,9S, 13aS, 14aR, 16aS,Z)- 14a- ((cyclopropylsulfonyl)carbamoyl)-2-((6-methoxyisoquinolin-l-yl)oxy)-7,9-dimethyl- 5,16-dioxo-2,3,5,6,7,8,9,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[e]pyrrolo[2, 1 -i] [ 1 ,7, 10]oxadiazacyclopentadecin-6-yl)carbamate;

3,3-difluoro-2-methylbutan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2- ((7-fluoro-4-methoxyisoquinolin- 1 -yl)oxy)-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7- ethyl-2-((7-fluoro-4-methoxyisoquinolin- 1 -yl)oxy)-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

3,3-difluoro-2-methylbutan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((6- ethoxy-7-fluoroisoquinolin- 1 -yl)oxy)-7-ethyl-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((6- ethoxy-7-fluoroisoquinolin- 1 -yl)oxy)-7-ethyl-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

3,3-difluoro-2-methylbutan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((2,3- dihydro-[l,4]dioxino[2,3-i^isoquinolin-7-yl)oxy)-7-ethyl-9-methyl-14a-(((l- methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2- ((2,3-dihydro-[l,4]dioxino[2,3-i^isoquinolin-7-yl)oxy)-7-ethyl-9-methyl-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

tert-butyl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((2,3-dihydro- [l,4]dioxino[2,3-f]isoquinolin-7-yl)oxy)-7-ethyl-9-methyl-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1,1,1 -trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7- ethyl-2-((3 -(5 -isopropoxypyridin-2-y l)-6-methoxyisoquinolin- 1 -yl)oxy)-9-methyl- 14a-((( 1 -methylcy clopropyl)sulfonyl)carbamoy l)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

3,3-difluoro-2-methylbutan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2- ((3-(5-isopropoxypyridin-2-yl)-6-methoxyisoquinolin-l-yl)oxy)-9-methyl-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1 , 1 -difluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7-ethyl- 2-((3-(5-isopropoxypyridin-2-yl)-6-methoxyisoquinolin-l-yl)oxy)-9-methyl-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1 , 1 -difluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((6- ethoxy-7-fluoroisoquinolin- 1 -yl)oxy)-7-ethyl-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

1 , 1 -difluoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7-ethyl- 2-((7-fluoro-6-methoxyisoquinolin- 1 -yl)oxy)-9-methyl- 14a-(((l - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate; 1 , 1 -difhioro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7-ethyl- 2-((7-fluoro-4-methoxyisoquinolin- 1 -yl)oxy)-9-methyl- 14a-(((l - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate; and

I, l-difluoro-2-methylpropan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-((4- methoxyisoquinolin- 1 -yl)oxy)-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate;

or a pharmaceutically acceptable salt thereof.

10. A composition comprising a compound of claim 1, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.

I I . The composition of claim 10 further comprising at least one additional compound having anti-HCV activity.

12. The composition of claim 11 wherein at least one of the additional compounds is an interferon or a ribavirin.

13. The composition of claim 12 wherein the interferon is selected from interferon alpha 2B, pegylated interferon alpha, consensus interferon, interferon alpha 2A, and lymphoblastoid interferon tau.

14. The composition of claim 11 wherein at least one of the additional compounds is selected from interleukin 2, interleukin 6, interleukin 12, Imiquimod, ribavirin, an inosine 5 '-monophosphate dehydrogenase inhibitor, amantadine, and rimantadine.

15. The composition of claim 11 wherein at least one of the additional compounds is effective to inhibit the function of a target selected from HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, and IMPDH for the treatment of an HCV infection.

16. A method of treating an HCV infection in a patient, comprising administering to the patient a therapeutically effective amount of a compound of claim 1 , or a pharmaceutically acceptable salt thereof.

17. The method of claim 16 further comprising administering at least one additional compound having anti-HCV activity prior to, after, or simultaneously with the compound of formula (I), or a pharmaceutically acceptable salt thereof.

18. The method of claim 17 wherein at least one of the additional compounds is an interferon or a ribavirin. 19. The method of claim 18 wherein the interferon is selected from interferon alpha 2B, pegylated interferon alpha, consensus interferon, interferon alpha 2A, and lymphoblastoid interferon tau.

20. The method of claim 17 wherein at least one of the additional compounds is selected from interleukin 2, interleukin 6, interleukin 12, Imiquimod, ribavirin, an inosine 5 '-monophosphate dehydrogenase inhibitor, amantadine, and rimantadine.

21. The method of claim 17 wherein at least one of the additional compounds is effective to inhibit the function of a target selected from HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, and IMPDH for the treatment of an HCV infection.

Description:
HEPATITIS C VIRUS INHIBITORS

The present disclosure is generally directed to antiviral compounds, and more specifically directed to compounds which inhibit the function of the NS3 protease (also referred to herein as "serine protease") encoded by Hepatitis C virus (HCV), compositions comprising such compounds, and methods for inhibiting the function of the NS3 protease.

HCV is a major human pathogen, infecting an estimated 170 million persons worldwide - roughly five times the number infected by human immunodeficiency virus type 1. A substantial fraction of these HCV infected individuals develop serious progressive liver disease, including cirrhosis and hepatocellular carcinoma.

Presently, the most effective HCV therapy employs a combination of alpha- interferon and ribavirin, leading to sustained efficacy in 40% of patients. Recent clinical results demonstrate that pegylated alpha-interferon is superior to unmodified alpha-interferon as monotherapy. However, even with experimental therapeutic regimens involving combinations of pegylated alpha-interferon and ribavirin, a substantial fraction of patients do not have a sustained reduction in viral load. Thus, there is a clear and unmet need to develop effective therapeutics for treatment of HCV infection.

HCV is a positive-stranded RNA virus. Based on a comparison of the deduced amino acid sequence and the extensive similarity in the 5 ' untranslated region, HCV has been classified as a separate genus in the Flaviviridae family. All members of the Flaviviridae family have enveloped virions that contain a positive stranded RNA genome encoding all known virus-specific proteins via translation of a single, uninterrupted, open reading frame.

Considerable heterogeneity is found within the nucleotide and encoded amino acid sequence throughout the HCV genome. Six major genotypes have been characterized, and more than 50 subtypes have been described. The major genotypes of HCV differ in their distribution worldwide, and the clinical significance of the genetic heterogeneity of HCV remains elusive despite numerous studies of the possible effect of genotypes on pathogenesis and therapy.

The single strand HCV RNA genome is approximately 9500 nucleotides in length and has a single open reading frame (ORF) encoding a single large polyprotein of about 3000 amino acids. In infected cells, this polyprotein is cleaved at multiple sites by cellular and viral proteases to produce the structural and non- structural (NS) proteins. In the case of HCV, the generation of mature non-structural proteins (NS2, NS3, NS4A, NS4B, NS5A, and NS5B) is effected by two viral proteases. The first one cleaves at the NS2-NS3 junction; the second one is a serine protease contained within the N-terminal region of NS3 and mediates all the subsequent cleavages downstream of NS3, both in cis, at the NS3-NS4A cleavage site, and in trans, for the remaining NS4A- NS4B, NS4B-NS5A, NS5A-NS5B sites. The NS4A protein appears to serve multiple functions, acting as a co-factor for the NS3 protease and possibly assisting in the membrane localization of NS3 and other viral replicase components. The complex formation of the NS3 protein with NS4A is essential for efficient polyprotein processing, enhancing the proteolytic cleavage at all of the sites. The NS3 protein also exhibits nucleoside triphosphatase and RNA helicase activities. NS5B is a RNA-dependent RNA polymerase that is involved in the replication of HCV.

The present disclosure provides peptide compounds that can inhibit the functioning of the NS3 protease, e.g., in combination with the NS4A protease.

Further, the present disclosure describes the administration of combination therapy to a patient whereby a compound in accordance with the present disclosure, which is effective to inhibit the HCV NS3 protease, can be administered with additional compounds having anti-HCV activity.

In its first as ect the present disclosure provides a compound of formula (I)

is a single or double bond; m is 0, 1, or 2;

1 is selected from

wherein R 1 is attached to the parent molecular moiety through any substitutable carbon atom in the group;

n is 0, 1, 2, 3, 4, 5, or 6;

o is 0, 1, 2, 3, 4, or 5;

q is 0, 1, 2, 3, or 4;

X° is selected from CH and N;

X 1 is selected from CH and N;

X 2 and X 3 are independently selected from CH, C(R a ) and N; provided that at least one of X 1 , X 2 , and X 3 is other than N;

X 4 is selected from CH and CR a ;

one of X 5 , X 6 , X 7 , and X 8 is N and the others are selected from CH and CR a ; X 9 is selected from CR a , CH, and N;

each R a is independently selected from alkenyloxy, alkoxy, alkoxyalkoxy, alkoxycarbonyl, alkyl, benzodioxanyl, carboxamido, carboxy, carboxyalkoxy, cyano, cycloalkyl, cycloalkylalkoxy, cycloalkyloxy, deuteroalkoxy, dialkylamino, halo, haloalkyl, haloalkoxy, haloalkoxycarbonyl, hydroxy, imidazolyl, morpholinyl, oxazolyl, phenyl, piperazinyl, pyrazolyl, pyridinyl, pyrrolidinyl, thiazolyl, and - NR q R q , wherein the imidazolyl, the morpholinyl, the oxazolyl, the phenyl, the piperazinyl, the pyridinyl, the pyrrolidinyl, and the thiazolyl are optionally substituted with one or two groups independently selected from alkoxy, alkyl, alkylsulfonyl, halo, haloalkoxy, haloalkyl, and morpholinyl; and wherein two adjacent R a groups, together with the carbon atoms to which they are attached, can optionally form a ring selected from dioxanyl, dioxolanyl, furanyl, morpholinyl, pyranyl, and phenyl, wherein the ring is optionally substituted with one or two groups independently selected from alkyl and halo;

R b is alkyl;

R x is selected from methyl and ethyl;

R z and R z are independently selected from hydrogen and hydroxy; provided that when is a double bond, R z and R z are each hydrogen;

R 2 is selected from hydrogen, alkyl, deuteroalkyl, halo, haloalkoxy, haloalkyl, and hydroxyalkyl;

R 3 is selected from hydrogen, alkoxyalkoxycarbonyl, alkoxycarbonyl, alkylaminocarbonyl, alkylcarbonyl, cycloalkylalkoxycarbonyl, cycloalkylcarbonyl, cycloalkyloxycarbonyl, deuteroalkoxycarbonyl, deuterohaloalkoxycarbonyl, dialkylaminocarbonyl, dialkylaminocarbonylcarbonyl, haloalkoxycarbonyl, haloalkylaminocarbonyl, haloalkylcarbonyl, heterocyclylcarbonyl,

heterocyclyloxycarbonyl, phenylcarbonyl, and phenyloxycarbonyl, wherein the cycloalkyl part of the cycloalkylalkoxycarbonyl, the cycloalkylcarbonyl, and the cycloalkyloxycarbonyl, the heterocyclyl part of the heterocyclylcarbonyl and the heterocyclyloxycarbonyl, and the phenyl part of the phenylcarbonyl and the phenyloxycarbonyl, is optionally substituted with one , two, or three groups independently selected from alkyl, alkylamino, alkylcarbonyl, cycloalkyl, dialkylamino, halo, haloalkoxy, and haloalkyl;

one of R q and R q is selected from hydrogen and alkyl and the other is selected from alkylcarbonyl and phenylcarbonyl; and

one of Y and Y' is oxygen and the other is C¾.

In a first embodiment of the first aspect the present disclosure provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein p is

1. In a second embodiment of the first aspect is a double bond. In a third embodiment of the first aspect m is 1. In a fourth embodiment of the first aspect R 1 is

; wherein n is 1, 2, 3, or 4; and

each R a is independently selected from alkoxy, halo, and pyridinyl, wherein the pyridinyl is optionally substituted with one alkoxy group or wherein two adjacent R a groups, together with the carbon atoms to which they are attached, can optionally form a ring selected from dioxanyl and pyranyl.

In a fifth embodiment of the first aspect the present disclosure provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein R 2 is selected from alkyl and haloalkyl.

In a sixth embodiment of the first aspect the present disclosure provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein R 3 is selected from alkoxycarbonyl and haloalkoxycarbonyl.

In a seventh embodiment of the first aspect the present disclosure provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein

p is 1;

is a double bond;

m is 1.

R 1 is

; wherein

n is 1, 2, 3, or 4; and

each R a is independently selected from alkoxy, halo, and pyridinyl, wherein the pyridinyl is optionally substituted with one alkoxy group or wherein two adjacent R a groups, together with the carbon atoms to which they are attached, can optionally form a ring selected from dioxanyl and pyranyl;

R 2 is selected from alkyl and haloalkyl; and

R 3 is selected from alkoxycarbonyl and haloalkoxycarbonyl.

In a second aspect the present disclosure provides a composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. In a first embodiment of the second aspect the present disclosure provides a composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, at least one additional compound having anti-HCV activity, and a pharmaceutical carrier. In a second embodiment at least one of the additional compounds is an interferon or a ribavirin. In a third embodiment the interferon is selected from interferon alpha 2B, pegylated interferon alpha, consensus interferon, interferon alpha 2A, and lymphoblastiod interferon tau. In a fourth embodiment of the second aspect the present disclosure provides a composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, at least one additional compound having anti-HCV activity, and a pharmaceutical carrier, wherein at least one of the additional compounds is selected from interleukin 2, interleukin 6, interleukin 12, Imiquimod, ribavirin, an inosine 5'-monophospate dehydrogenase inhibitor, amantadine, and rimantadine. In a fifth embodiment of the second aspect the present disclosure provides a composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, at least one additional compound having anti-HCV activity, and a pharmaceutical carrier, wherein at least one of the additional compounds is effective to inhibit the function of a target selected from HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, and IMPDH for the treatment of an HCV infection.

In a third aspect the present disclosure provides a method of treating an HCV infection in a patient, comprising administering to the patient a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof. In a first embodiment of the third aspect the method further comprises administering at least one additional compound having anti-HCV activity prior to, after, or simultaneously with the compound of formula (I), or a pharmaceutically acceptable salt thereof. In a second embodiment of the third aspect at least one of the additional compounds is an interferon or a ribavirin. In a third embodiment the interferon is selected from interferon alpha 2B, pegylated interferon alpha, consensus interferon, interferon alpha 2A, and lymphoblastiod interferon tau. In a fourth embodiment of the third aspect the present disclosure provides a method of treating an HCV infection in a patient, comprising administering to the patient a

therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and at least one additional compound having anti-HCV activity prior to, after, or simultaneously with the compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein at least one of the additional compounds is selected from interleukin 2, interleukin 6, interleukin 12, Imiquimod, ribavirin, an inosine 5'-monophospate dehydrogenase inhibitor, amantadine, and rimantadine. In a fifth embodiment of the third aspect the present disclosure provides a method of treating an HCV infection in a patient, comprising administering to the patient a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and at least one additional compound having anti-HCV activity prior to, after, or simultaneously with the compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein at least one of the additional compounds is effective to inhibit the function of a target selected from HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, and IMPDH for the treatment of an HCV infection.

Other aspects of the present disclosure may include suitable combinations of embodiments disclosed herein.

Yet other aspects and embodiments may be found in the description provided herein.

The description of the present disclosure herein should be construed in congruity with the laws and principals of chemical bonding. In some instances it may be necessary to remove a hydrogen atom in order to accommodate a substitutent at any given location.

It should be understood that the compounds encompassed by the present disclosure are those that are suitably stable for use as pharmaceutical agent.

It is intended that the definition of any substituent or variable at a particular location in a molecule be independent of its definitions elsewhere in that molecule.

For example, when n is 2, each of the two R 1 groups may be the same or different.

All patents, patent applications, and literature references cited in the specification are herein incorporated by reference in their entirety. In the case of inconsistencies, the present disclosure, including definitions, will prevail.

As used herein, the singular forms "a", "an", and "the" include plural reference unless the context clearly dictates otherwise.

The term "alkenyl," as used herein, refers to a group derived from a straight or branched chain saturated hydrocarbon containing from two to ten carbon atoms and at least one double bond. In one embodiment the alkenyl groups contain from two to six carbon atoms. In another embodiment the alkenyl groups contain from two to four carbon atoms.

The term "alkenyloxy," as used herein, refers to an alkenyl group attached to the parent molecular moiety through an oxygen atom.

The term "alkoxy," as used herein, refers to an alkyl group attached to the parent molecular moiety through an oxygen atom.

The term "alkoxyalkoxy," as used herein, refers to an alkoxyalkyl group attached to the parent molecular moiety through an oxygen atom.

The term "alkoxyalkoxycarbonyl," as used herein, refers to an alkoxyalkoxy group attached to the parent molecular moiety through a carbonyl group.

The term "alkoxyalkyl," as used herein, refers to an alkyl group substituted with one, two, or three alkoxy groups.

The term "alkoxycarbonyl," as used herein, refers to an alkoxy group attached to the parent molecular moiety through a carbonyl group.

The term "alkyl," as used herein, refers to a group derived from a straight or branched chain saturated hydrocarbon containing from one to ten carbon atoms. In one embodiment the alkyl groups contain from one to six carbon atoms. In another embodiment the alkyl groups contain from one to four carbon atoms.

The term "alkylamino," as used herein, refers to -NHR, wherein R is an alkyl group.

The term "alkylaminocarbonyl," as used herein, refers to an alkylamino group attached to the parent molecular moiety through a carbonyl group.

The term "alkylcarbonyl," as used herein, refers to an alkyl group attached to the parent molecular moiety through a carbonyl group.

The term "alkylsulfonyl," as used herein, refers to an alkyl group attached to the parent molecular moiety through a sulfonyl group.

The term "carbonyl," as used herein, refers to -C(O)-.

The term "carboxamido," as used herein, refers to -C(0)NR x R y , wherein R x and R y are independently selected from hydrogen and alkyl.

The term "carboxy," as used herein, refers to -C0 2 H.

The term "carboxyalkoxy," as used herein, refers to a carboxyalkyl group attached to the parent molecular moiety through an oxygen atom. The term "carboxyalkyl," as used herein, refers to an alkyl group substituted with one, two, or three carboxy groups.

The term "cyano," as used herein, refers to -CN.

The term "cycloalkyl," as used herein, refers to a saturated monocyclic or bicyclic hydrocarbon ring system having three to seven carbon atoms and zero heteroatoms. Representative examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, and cyclopentyl.

The term "cycloalkylalkoxy," as used herein, refers to a (cycloalkyl)alkyl group attached to the parent molecular moiety through an oxygen atom.

The term "cycloalkylalkoxycarbonyl," as used herein, refers to a

cycloalkylalkoxy group attached to the parent molecular moiety through a carbonyl group.

The term "(cycloalkyl)alkyl," as used herein, refers to an alkyl group substituted with one, two, or three cycloalkyl groups.

The term "cycloalkylcarbonyl," as used herein, refers to a cycloalkyl group attached to the parent molecular moiety through a carbonyl group.

The term "cycloalkyloxy," as used herein, refers to a cycloalkyl group attached to the parent molecular moiety through an oxygen atom.

The term "cycloalkyloxycarbonyl," as used herein, refers to a cycloalkyloxy group attached to the parent molecular moiety through a carbonyl group.

The term "deuteroalkoxy," as used herein, refers to an alkoxy group wherein at least one hydrogen atom is replaced by a deuterium atom.

The term "deuteroalkoxycarbonyl," as used herein, refers to a deuteroalkoxy group attached to the parent molecular moiety through a carbonyl group.

The term "deuteroalkyl," as used herein, refers to an alkyl group wherein at least one hydrogen atom is replaced by a deuterium atom.

The term "deuterohaloalkoxy," as used herein, refers to a haloalkoxy group wherein at least one hydrogen atom is replaced by a deuterium atom.

The term "deuterohaloalkoxycarbonyl," as used herein, refers to a

deuterohaloalkoxy group attached to the parent molecular moiety through a carbonyl group.

The term "dialkylamino," as used herein, refers to -NR p R q , wherein R p and R q are alkyl groups. The alkyl groups may be the same or different. The term "dialkylaminocarbonyl," as used herein, refers to a dialkylamino group attached to the parent molecular moiety through a carbonyl group.

The term "dialkylaminocarbonylcarbonyl," as used herein, refers to a dialkylaminocarbonyl group attached to the parent molecular moiety through a carbonyl group.

The terms "halo" and "halogen," as used herein, refer to F, CI, Br, and I.

The term "haloalkoxy," as used herein, refers to a haloalkyl group attached to the parent molecular moiety through an oxygen atom.

The term "haloalkoxycarbonyl," as used herein, refers to a haloalkoxy group attached to the parent molecular moiety through a carbonyl group.

The term "haloalkyl," as used herein, refers to an alkyl group substituted with one, two, three, or four halogen atoms.

The term "haloalkylamino," as used herein, refers to an alkyl amino group wherein the alkyl is substituted with one, two, three, or four halogen atoms.

The term "haloalkylaminocarbonyl," as used herein, refers to a

haloalkylamino group attached to the parent molecular moiety through a carbonyl group.

The term "haloalkylcarbonyl," as used herein, refers to a haloalkyl group attached to the parent molecular moiety through a carbonyl group.

The term "heterocyclyl," as used herein, refers to a four-, five-, six-, or seven- membered ring containing one, two, three, or four heteroatoms independently selected from nitrogen, oxygen, and sulfur. The four-membered ring has zero double bonds, the five-membered ring has zero to two double bonds, and the six- and seven- membered rings have zero to three double bonds. The term "heterocyclyl" also includes bicyclic groups in which the heterocyclyl ring is fused to another monocyclic heterocyclyl group, or a four- to six-membered aromatic or non-aromatic carbocyclic ring; as well as bridged bicyclic groups such as 7-azabicyclo[2.2.1]hept- 7-yl, 2-azabicyclo[2.2.2]oc-2-tyl, and 2-azabicyclo[2.2.2]oc-3-tyl. The heterocyclyl groups of the present disclosure can be attached to the parent molecular moiety through any carbon atom or nitrogen atom in the group. Examples of heterocyclyl groups include, but are not limited to, benzothienyl, furyl, imidazolyl, indolinyl, indolyl, isothiazolyl, isoxazolyl, morpholinyl, oxazolyl, piperazinyl, piperidinyl, pyrazolyl, pyridinyl, pyrrolidinyl, pyrrolopyridinyl, pyrrolyl, thiazolyl, thienyl, thiomorpholinyl, 7-azabicyclo[2.2.1]hept-7-yl, 2-azabicyclo[2.2.2]oc-2-tyl, and 2- azabicyclo[2.2.2]oc-3-tyl.

The term "heterocyclylcarbonyl," as used herein, refers to a heterocyclyl group attached to the parent molecular moiety through a carbonyl group.

The term "heterocyclyloxy," as used herein, refers to a heterocyclyl group attached to the parent molecular moiety through an oxygen atom.

The term "heterocyclyloxycarbonyl," as used herein, refers to a

heterocyclyloxy group attached to the parent molecular moiety through a carbonyl group.

The term "hydroxy," as used herein, refers to -OH.

The term "hydroxyalkyl," as used herein, refers to an alkyl group substituted with one, two, or three hydroxy groups.

The term "phenylcarbonyl," as used herein, refers to a phenyl group attached to the parent molecular moiety through a carbonyl group.

The term "phenyloxy," as used herein, refers to a phenyl group attached to the parent molecular moiety through an oxygen atom.

The term "phenyloxycarbonyl," as used herein, refers to a phenyloxy group attached to the parent molecular moiety through a carbonyl group.

The compounds of the present disclosure can exist as pharmaceutically acceptable salts. The term "pharmaceutically acceptable salt," as used herein, represents salts or zwitterionic forms of the compounds of the present disclosure which are water or oil-soluble or dispersible, which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of patients without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio, and are effective for their intended use. The salts can be prepared during the final isolation and purification of the compounds or separately by reacting a suitable basic functionality with a suitable acid. Representative acid addition salts include acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate,

camphorsulfonate; digluconate, glycerophosphate, hemisulfate, heptanoate, hexanoate, formate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2- hydroxyethanesulfonate, lactate, maleate, mesitylenesulfonate, methanesulfonate, naphthylenesulfonate, nicotinate, 2-naphthalenesulfonate, oxalate, palmoate, pectinate, persulfate, 3-phenylproprionate, picrate, pivalate, propionate, succinate, tartrate, trichloroacetate, trifluoroacetate, phosphate, glutamate, bicarbonate, para- toluenesulfonate, and undecanoate. Examples of acids which can be employed to form pharmaceutically acceptable addition salts include inorganic acids such as hydrochloric, hydrobromic, sulfuric, and phosphoric, and organic acids such as oxalic, maleic, succinic, and citric.

Basic addition salts can be prepared during the final isolation and purification of the compounds by reacting an acidic group with a suitable base such as the hydroxide, carbonate, or bicarbonate of a metal cation or with ammonia or an organic primary, secondary, or tertiary amine. The cations of pharmaceutically acceptable salts include lithium, sodium, potassium, calcium, magnesium, and aluminum, as well as nontoxic quaternary amine cations such as ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, diethylamine, ethylamine, tributylamine, pyridine, N,N-dimethylaniline, N- methylpiperidine, N-methylmorpholine, dicyclohexylamine, procaine, dibenzylamine, N,N-dibenzylphenethylamine, and N,N'-dibenzylethylenediamine. Other

representative organic amines useful for the formation of base addition salts include ethylenediamine, ethanolamine, diethanolamine, piperidine, and piperazine.

As used herein, the term "anti-HCV activity" means the compound is effective to treat the HCV virus.

The term "compounds of the disclosure", and equivalent expressions, are meant to embrace compounds of formula (I), and pharmaceutically acceptable enantiomers, diastereomers, and salts thereof. Similarly, references to intermediates, are meant to embrace their salts where the context so permits.

The term "patient" includes both human and other mammals.

The term "pharmaceutical composition" means a composition comprising a compound of the disclosure in combination with at least one additional

pharmaceutical carrier, i.e., adjuvant, excipient or vehicle, such as diluents, preserving agents, fillers, flow regulating agents, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavoring agents, perfuming agents, antibacterial agents, antifungal agents, lubricating agents and dispensing agents, depending on the nature of the mode of administration and dosage forms. Ingredients listed in Remington's Pharmaceutical Sciences, 18 th ed., Mack Publishing Company, Easton, PA (1999) for example, may be used.

The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of patients without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable risk/benefit ratio.

The term "sulfonyl," as used herein, refers to -S0 2 -.

The term "therapeutically effective amount" means the total amount of each active component that is sufficient to show a meaningful patient benefit, e.g., a sustained reduction in viral load. When applied to an individual active ingredient, administered alone, the term refers to that ingredient alone. When applied to a combination, the term refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially or

simultaneously.

The terms "treat" and "treating" refers to: (i) preventing a disease, disorder or condition from occurring in a patient which may be predisposed to the disease, disorder and/or condition but has not yet been diagnosed as having it; (ii) inhibiting the disease, disorder or condition, i.e., arresting its development; and/or (iii) relieving the disease, disorder or condition, i.e., causing regression of the disease, disorder and/or condition.

Where used in naming compounds of the present disclosure, the designations PI ', PI, P2, P2*, P3, and P4, as used herein, map the relative positions of the amino acid residues of a protease inhibitor binding relative to the binding of the natural peptide cleavage substrate. Cleavage occurs in the natural substrate between PI and PI ' where the nonprime positions designate amino acids starting from the C-terminus end of the peptide natural cleavage site extending towards the N-terminus; whereas, the prime positions emanate from the N-terminus end of the cleavage site designation and extend toward the C-terminus. For example, PI ' refers to the first position away from the right hand end of the C-terminus of the cleavage site (i.e. N-terminus first position); whereas PI starts the numbering from the left hand side of the C-terminus cleavage site, P2: second position from the C-terminus, etc.). (see Berger A. & Schechter I., Transactions of the Royal Society London series (1970), B257,

249-264]. Asymmetric centers exist in the compounds of the present disclosure. For example, the compounds may include PI cyclopropyl element of formula

P1

wherein Ci and C 2 each represent an asymmetric carbon atom at positions 1 and 2 of the cyclopropyl ring.

(IR, 2S) (IS, 2R)

R 2 is syn to carbonyl R 2 is syn to carbonyl

(1R, 2R) (1S, 2S)

R 2 is syn to amide R 2 is syn to amide

It should be understood that the disclosure encompasses all stereochemical forms, or mixtures thereof, which possess the ability to inhibit HCV protease.

Certain compounds of the present disclosure may also exist in different stable conformational forms which may be separable. Torsional asymmetry due to restricted rotation about an asymmetric single bond, for example because of steric hindrance or ring strain, may permit separation of different conformers. The present disclosure includes each conformational isomer of these compounds and mixtures thereof.

Certain compounds of the present disclosure may exist in zwitterionic form and the present disclosure includes each zwitterionic form of these compounds and mixtures thereof.

When it is possible that, for use in therapy, therapeutically effective amounts of a compound of formula (I), as well as pharmaceutically acceptable salts thereof, may be administered as the raw chemical, it is possible to present the active ingredient as a pharmaceutical composition. Accordingly, the disclosure further provides pharmaceutical compositions, which include therapeutically effective amounts of compounds of formula (I) or pharmaceutically acceptable salts thereof, and one or more pharmaceutically acceptable carriers, diluents, or excipients. The compounds of formula (I) and pharmaceutically acceptable salts thereof, are as described above. The carrier(s), diluent(s), or excipient(s) must be acceptable in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. In accordance with another aspect of the disclosure there is also provided a process for the preparation of a pharmaceutical formulation including admixing a compound of formula (I), or a pharmaceutically acceptable salt thereof, with one or more pharmaceutically acceptable carriers, diluents, or excipients.

Pharmaceutical formulations may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose. Dosage levels of between about 0.01 and about 150 milligram per kilogram ("mg/kg") body weight per day, preferably between about 0.05 and about 100 mg/kg body weight per day of the compounds of the disclosure are typical in a monotherapy for the prevention and treatment of HCV mediated disease. Typically, the pharmaceutical compositions of this disclosure will be administered from about 1 to about 5 times per day or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy. The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending on the condition being treated, the severity of the condition, the time of administration, the route of administration, the rate of excretion of the compound employed, the duration of treatment, and the age, gender, weight, and condition of the patient. Preferred unit dosage formulations are those containing a daily dose or sub-dose, as herein above recited, or an appropriate fraction thereof, of an active ingredient. Generally, treatment is initiated with small dosages substantially less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached. In general, the compound is most desirably administered at a concentration level that will generally afford antivirally effective results without causing any harmful or deleterious side effects.

When the compositions of this disclosure comprise a combination of a compound of the disclosure and one or more additional therapeutic and/or

prophylactic agent, both the compound and the additional agent can be present in a dose that is less than or equal to the dosage normally administered in a monotherapy regimen. The compositions of this disclosure may be co-formulated with one or more additional therapeutic or prophylactic agents, for example, in the form of a monolithic and/or bi/multi-layer tablet or may be administered separately from the therapeutic or prophylactic agent(s).

Pharmaceutical formulations may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual, or transdermal), vaginal, or parenteral (including subcutaneous, intracutaneous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, intralesional, intravenous, or intradermal injections or infusions) route. Such formulations may be prepared by any method known in the art of pharmacy, for example by bringing into association the active ingredient with the carrier(s) or excipient(s).

Pharmaceutical formulations adapted for oral administration may be presented as discrete units such as capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in-oil emulsions.

For instance, for oral administration in the form of a tablet or capsule, the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, and the like. Powders are prepared by comminuting the compound to a suitable fine size and mixing with a similarly comminuted pharmaceutical carrier such as an edible carbohydrate, as, for example, starch or mannitol. Flavoring, preservative, dispersing, and coloring agent can also be present.

Capsules are made by preparing a powder mixture, as described above, and filling formed gelatin sheaths. Glidants and lubricants such as colloidal silica, talc, magnesium stearate, calcium stearate, or solid polyethylene glycol can be added to the powder mixture before the filling operation. A disintegrating or solubilizing agent such as agar-agar, calcium carbonate, or sodium carbonate can also be added to improve the availability of the medicament when the capsule is ingested.

Moreover, when desired or necessary, suitable binders, lubricants,

disintegrating agents, and coloring agents can also be incorporated into the mixture. Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, and the like. Lubricants used in these dosage forms include sodium oleate, sodium chloride, and the like.

Disintegrators include, without limitation, starch, methyl cellulose, agar, betonite, xanthan gum, and the like. Tablets are formulated, for example, by preparing a powder mixture, granulating or slugging, adding a lubricant and disintegrant, and pressing into tablets. A powder mixture is prepared by mixing the compound, suitable comminuted, with a diluent or base as described above, and optionally, with a binder such as carboxymethylcellulose, an aliginate, gelating, or polyvinyl pyrrolidone, a solution retardant such as paraffin, a resorption accelerator such as a quaternary salt and/or and absorption agent such as betonite, kaolin, or dicalcium phosphate. The powder mixture can be granulated by wetting with a binder such as syrup, starch paste, acadia mucilage, or solutions of cellulosic or polymeric materials and forcing through a screen. As an alternative to granulating, the powder mixture can be run through the tablet machine and the result is imperfectly formed slugs broken into granules. The granules can be lubricated to prevent sticking to the tablet forming dies by means of the addition of stearic acid, a stearate salt, talc, or mineral oil. The lubricated mixture is then compressed into tablets. The compounds of the present disclosure can also be combined with a free flowing inert carrier and compressed into tablets directly without going through the granulating or slugging steps. A clear or opaque protective coating consisting of a sealing coat of shellac, a coating of sugar or polymeric material, and a polish coating of wax can be provided. Dyestuffs can be added to these coatings to distinguish different unit dosages.

Oral fluids such as solution, syrups, and elixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of the compound. Syrups can be prepared by dissolving the compound in a suitably flavored aqueous solution, while elixirs are prepared through the use of a non-toxic vehicle.

Solubilizers and emulsifiers such as ethoxylated isostearyl alcohols and

polyoxyethylene sorbitol ethers, preservatives, fiavor additive such as peppermint oil or natural sweeteners, or saccharin or other artificial sweeteners, and the like can also be added.

Where appropriate, dosage unit formulations for oral administration can be microencapsulated. The formulation can also be prepared to prolong or sustain the release as for example by coating or embedding particulate material in polymers, wax, or the like.

The compounds of formula (I), and pharmaceutically acceptable salts thereof, can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles.

Liposomes can be formed from a variety of phopholipids, such as cholesterol, stearylamine, or phophatidylcho lines.

The compounds of formula (I) and pharmaceutically acceptable salts thereof may also be delivered by the use of monoclonal antibodies as individual carriers to which the compound molecules are coupled. The compounds may also be coupled with soluble polymers as targetable drug carriers. Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamidephenol, polyhydroxyethylaspartamidephenol, or polyethyleneoxidepolylysme substituted with palitoyl residues. Furthermore, the compounds may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates, and cross-linked or amphipathic block copolymers of hydrogels.

Pharmaceutical formulations adapted for transdermal administration may be presented as discrete patches intended to remain in intimate contact with the epidermis of the recipient for a prolonged period of time. For example, the active ingredient may be delivered from the patch by iontophoresis as generally described in Pharmaceutical Research, 3(6), 318 (1986).

Pharmaceutical formulations adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols, or oils.

For treatments of the eye or other external tissues, for example mouth and skin, the formulations are preferably applied as a topical ointment or cream. When formulated in an ointment, the active ingredient may be employed with either a paraffinic or a water-miscible ointment base. Alternatively, the active ingredient may be formulated in a cream with an oil-in-water cream base or a water-in oil base.

Pharmaceutical formulations adapted for topical administrations to the eye include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent.

Pharmaceutical formulations adapted for topical administration in the mouth include lozenges, pastilles, and mouth washes.

Pharmaceutical formulations adapted for rectal administration may be presented as suppositories or as enemas.

Pharmaceutical formulations adapted for nasal administration wherein the carrier is a solid include a course powder which is administered in the manner in which snuff is taken, i.e., by rapid inhalation through the nasal passage from a container of the powder held close up to the nose. Suitable formulations wherein the carrier is a liquid, for administration as a nasal spray or nasal drops, include aqueous or oil solutions of the active ingredient.

Pharmaceutical formulations adapted for administration by inhalation include fine particle dusts or mists, which may be generated by means of various types of metered, dose pressurized aerosols, nebulizers, or insufflators.

Pharmaceutical formulations adapted for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulations.

Pharmaceutical formulations adapted for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats, and soutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules, and tablets.

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

Table 1 below lists some illustrative examples of compounds that can be administered with the compounds of this disclosure. The compounds of the disclosure can be administered with other anti-HCV activity compounds in combination therapy, either jointly or separately, or by combining the compounds into a composition.

Table 1

Type of Inhibitor or

Brand Name Physiological Class Source Company

Target

Pyrazolopyrimidine

compounds and salts

Arrow Therapeutics From WO- Antiviral HCV Inhibitors

Ltd.

2005047288

26 May 2005

Ribapharm Inc.,

Levovirin Antiviral IMPDH inhibitor

Costa Mesa, CA

Vertex

Merimepodib Pharmaceuticals

Antiviral IMPDH inhibitor

(VX-497) Inc., Cambridge,

MA

XTL

XTL-6865 (XTL-

Antiviral monoclonal antibody Biopharmaceuticals 002)

Ltd., Rehovot, Isreal

Vertex

Pharmaceuticals

Telaprevir

NS3 serine protease Inc., Cambridge, (VX-950, LY- Antiviral

inhibitor MA/ Eli Lilly and 570310)

Co. Inc.,

Indianapolis, IN

NS5B Replicase

HCV-796 Antiviral Wyeth / Viropharma

Inhibitor

NS5B Replicase

NM-283 Antiviral Idenix / Novartis

Inhibitor

NS5B Replicase Gene Labs /

GL-59728 Antiviral

Inhibitor Novartis

NS5B Replicase Gene Labs /

GL-60667 Antiviral

Inhibitor Novartis

NS5B Replicase

2'C MeA Antiviral Gilead

Inhibitor

NS5B Replicase

PSI 6130 Antiviral Roche

Inhibitor Type of Inhibitor or

Brand Name Physiological Class Source Company

Target

NS5B Replicase

R1626 Antiviral Roche

Inhibitor

2'C Methyl NS5B Replicase

Antiviral Merck

adenosine Inhibitor

Japan Tobacco Inc.,

JTK-003 Antiviral RdRp inhibitor

Tokyo, Japan

ICN

Levovirin Antiviral ribavirin Pharmaceuticals,

Costa Mesa, CA

Schering-Plough

Ribavirin Antiviral ribavirin Corporation,

Kenilworth, NJ

Ribapharm Inc.,

Viramidine Antiviral Ribavirin Prodrug

Costa Mesa, CA

Ribozyme

Heptazyme Antiviral ribozyme Pharmaceuticals

Inc., Boulder, CO

Boehringer serine protease Ingelheim Pharma

BILN-2061 Antiviral

inhibitor KG, Ingelheim,

Germany serine protease

SCH 503034 Antiviral Schering Plough inhibitor

SciClone

Zadazim Immune modulator Immune modulator Pharmaceuticals

Inc., San Mateo, CA

Maxim

Ceplene Immunomodulator immune modulator Pharmaceuticals

Inc., San Diego, CA

F. Hoffmann-La

HCV IgG immuno¬

CellCept Immunosuppressant Roche LTD, Basel, suppressant

Switzerland Type of Inhibitor or

Brand Name Physiological Class Source Company

Target

Nabi

HCV IgG immuno¬

Civacir Immunosuppressant Biopharmaceuticals suppressant

Inc., Boca Raton, FL

Human Genome

Albuferon - a Interferon albumin IFN-a2b Sciences Inc.,

Rockville, MD

InterMune

IFN

Infergen A Interferon Pharmaceuticals alfacon- 1

Inc., Brisbane, CA

Omega IFN Interferon IFN-ω Intarcia Therapeutics

Transition

IFN- and EMZ701 Interferon IFN- and EMZ701 Therapeutics Inc.,

Ontario, Canada

Serono, Geneva,

Rebif Interferon TFN^la

Switzerland

F. Hoffmann-La

Roferon A Interferon IFN-a2a Roche LTD, Basel,

Switzerland

Schering-Plough

Intron A Interferon IFN-a2b Corporation,

Kenilworth, NJ

RegeneRx

Biopharma. Inc.,

Intron A and IFN-a2b/al- Bethesda, MD/

Interferon

Zadaxin thymosin SciClone

Pharmaceuticals Inc, San Mateo, CA

Schering-Plough

Rebetron Interferon IFN-a2b/ribavirin Corporation,

Kenilworth, NJ

InterMune Inc.,

Actimmune Interferon INF-γ

Brisbane, CA

Interferon- Interferon Interferon- -la Serono Type of Inhibitor or

Brand Name Physiological Class Source Company

Target

Viragen/

Multiferon Interferon Long lasting IFN

Valentis

Lympho-blastoid Glaxo SmithKline

Wellferon Interferon

IFN-anl pic, Uxbridge, UK

Viragen Inc.,

Omniferon Interferon natural IFN-a

Plantation, FL

F. Hoffmann-La

Pegasys Interferon PEGylated IFN-a2a Roche LTD, Basel,

Switzerland

Maxim

PEGylated IFN-a2a/

Pegasys and Ceplene Interferon Pharmaceuticals immune modulator

Inc., San Diego, CA

F. Hoffmann-La

Pegasys and PEGylated IFN-

Interferon Roche LTD, Basel, Ribavirin a2a/ribavirin

Switzerland

Schering-Plough

PEG-Intron Interferon PEGylated IFN-a2b Corporation,

Kenilworth, NJ

Schering-Plough

PEG-Intron / PEGylated IFN-

Interferon Corporation, Ribavirin a2b/ribavirin

Kenilworth, NJ

Indevus

IP-501 Liver protection antifibrotic Pharmaceuticals

Inc., Lexington, MA

Idun

IDN-6556 Liver protection caspase inhibitor Pharmaceuticals

Inc., San Diego, CA

InterMune serine protease

ITMN-191 (R-7227) Antiviral Pharmaceuticals inhibitor

Inc., Brisbane, CA

NS5B Replicase

GL-59728 Antiviral Genelabs

Inhibitor

ANA-971 Antiviral TLR-7 agonist Anadys Type of Inhibitor or

Brand Name Physiological Class Source Company

Target

serine protease

Boceprevir Antiviral Schering Plough inhibitor

serine protease Tibotec BVBA,

TMS-435 Antiviral

inhibitor Mechelen, Belgium

Boehringer serine protease Ingelheim Pharma

BI-201335 Antiviral

inhibitor KG, Ingelheim,

Germany serine protease

MK-7009 Antiviral Merck

inhibitor

PF-00868554 Antiviral replicase inhibitor Pfizer

Anadys

Non-Nucleoside

Pharmaceuticals,

ANA598 Antiviral NS5B Polymerase

Inc., San Diego, CA, Inhibitor

USA

Idenix

Non-Nucleoside Pharmaceuticals,

IDX375 Antiviral

Replicase Inhibitor Cambridge, MA,

USA

Boehringer

NS5B Polymerase Ingelheim Canada

BILB 1941 Antiviral

Inhibitor Ltd R&D, Laval,

QC, Canada

Nucleoside Pharmasset,

PSI-7851 Antiviral

Polymerase Inhibitor Princeton, NJ, USA

Nucleotide NS5B Pharmasset,

PSI-7977 Antiviral

Polymerase Inhibitor Princeton, NJ, USA

Antiviral NS5B Polymerase

VCH-759 ViroChem Pharma

Inhibitor

Antiviral Nucleotide NS5B

I X- 189 Inhibitex

Polymerase Inhibitor

Antiviral NS5B Polymerase

VCH-916 ViroChem Pharma

Inhibitor Type of Inhibitor or

Brand Name Physiological Class Source Company

Target

Antiviral NS5B Polymerase

GS-9190 Gilead

Inhibitor

Peg-interferon Antiviral ZymoGenetics/Brist

Interferon

lamda ol-Myers Squibb

Antiviral Bristol-Myers daclatasvir NS5A inhibitor

Squibb

Antiviral NS5B Polymerase Bristol-Myers

BMS-791325

Inhibitor Squibb

ACH-3102 Antiviral Bristol-Myers

NS5A inhibitor

Squibb

Antiviral Bristol-Myers

BMS-984478 NS5A inhibitor

Squibb

Antiviral serine protease Bristol-Myers asunaprevir

inhibitor Squibb

The compounds of the disclosure may also be used as laboratory reagents. Compounds may be instrumental in providing research tools for designing of viral replication assays, validation of animal assay systems and structural biology studies to further enhance knowledge of the HCV disease mechanisms. Further, the compounds of the present disclosure are useful in establishing or determining the binding site of other antiviral compounds, for example, by competitive inhibition.

The compounds of this disclosure may also be used to treat or prevent viral contamination of materials and therefore reduce the risk of viral infection of laboratory or medical personnel or patients who come in contact with such materials, e.g., blood, tissue, surgical instruments and garments, laboratory instruments and garments, and blood collection or transfusion apparatuses and materials.

This disclosure is intended to encompass compounds having formula (I) when prepared by synthetic processes or by metabolic processes including those occurring in the human or animal body (in vivo) or processes occurring in vitro.

The present disclosure will now be described in connection with certain embodiments which are not intended to limit its scope. On the contrary, the present disclosure covers all alternatives, modifications, and equivalents as can be included within the scope of the claims. Thus, the following examples, which include specific embodiments, will illustrate one practice of the present disclosure, it being understood that the examples are for the purposes of illustration of certain

embodiments and are presented to provide what is believed to be the most useful and readily understood description of its procedures and conceptual aspects.

The abbreviations used in the present application, including particularly in the illustrative schemes and examples which follow, are well-known to those skilled in the art. Some of the abbreviations used are as follows: THF for tetrahydrofuran; min for minutes; h or hr or hrs for hours; r.t. or RT or Rt for room temprature or retention time (context will dictate); EtOAC or EtOAc for ethyl acetate; TMS for

trimethylsilane; DMSO for Ν,Ν-dimethylsulfoxide; DCM for dichloromethane; DMF for Ν,Ν-dimethylformamide; Me for methyl; DPPA for diphenylphosphoryl azide; Et 2 0 for diethyl ether; Ph for phenyl; DCE for 1 ,2-dichloroethane; TFA for trifluoroacetic acid; Et 3 N or TEA for triethylamine; (BOC) 2 0 for di-tert-butyl dicarbonate; and HATU for <3-(7-azabenzotriazol- 1 -yl)-N,N,N',N'- tetramethyluronium phosphate.

The starting materials useful to synthesize the compounds of the present disclosure are known to those skilled in the art and can be readily manufactured or are commercially available.

The following methods set forth below are provided for illustrative purposes and are not intended to limit the scope of the claims. It will be recognized that it may be necessary to prepare such a compound in which a functional group is protected using a conventional protecting group then to remove the protecting group to provide a compound of the present disclosure. The details concerning the use of protecting groups in accordance with the present disclosure are known to those skilled in the art Compounds were named using ChemDraw.

Preparation of 3 ,3-dtfluoro-2-methylbutan-2-yl pyridin-2-yl carbonate Schem

Step 1 :

Methylmagnesium bromide(24.91 mL, 74.7 mmol) was added dropwise via syringe to a solution of ethyl 2,2-difluoropropanoate (3.44 g, 24.91 mmol) in Diethyl ether (50 mL) at -20 °C and stirred at this temp for 1 hr before warming up to RT. The reaction was quenched with sat. ammonium chloride and extracted with ether. The organic layer was washed with brine, collected, dried over MgS04, filtered and partially evaporated to give the crude product 3,3-difluoro-2-methylbutan-2-ol(1.84g, 59.5% yield) as an oil. 1H NMR (500MHz, CDC1 3 ) δ 1.68 - 1.58 (m, 3H), 1.31 (t, J=1.2 Hz, 6H).

Step 2:

To a suspension of sodium hydride, 60%> in mineral oil (0.652 g, 16.31 mmol) in THF (25 mL) was added 3,3-difluoro-2-methylbutan-2-ol (1.84 g, 14.82 mmol) at 0°C. After stirring 30 min, the solution was transferred to a solution of di(pyridin-2- yl) carbonate (3.20 g, 14.82 mmol) in THF (25 mL) through a cannula. The formed slurry was stirred at 0 °C for 30 min. The slurry was warmed to rt and stirred for 2 h. The reaction was diluted with EtOAc, washed with brine, dried over MgS04, filtered, concentrated to give a residue that was purified by Biotage eluting with 10- 50% EtOAc in hexanes to afford the desired product 3,3-difluoro-2-methylbutan-2-yl pyridin-2-yl carbonate(500mg, 13.76%) as an oil that later crystallized to a white solid upon standing. 1H NMR (500MHz, CDC1 3 ) δ 8.43 (ddd, J=4.9, 2.0, 0.7 Hz, 1H), 7.95 - 7.75 (m, 1H), 7.31 - 7.24 (m, 1H), 7.15 (dt, J=8.2, 0.8 Hz, 1H), 1.72 (s, 6H), 1.77 - 1.66 (m, 3H).

Preparation of l-fluoro-4-methoxyisoquinoline

Step 1: Preparation of l-chloro-4-methoxyisoquinoline

To a solution of l-chloroisoquinolin-4-ol (5.0 g, 27.8 mmol) in acetonitrile

(50 mL) was added TMS-diazomethane (12.73 g, 111.2 mmol) at 0 °C. The reaction mixture was allowed to come to room temperature and stirred for 2 h. Solvent was evaporated under reduced pressure to get crude compound. The crude compound was purified by silica gel chromatography to get l-chloro-4-methoxyisoquinoline (2.5 g, 46.4%) as off-white solid. 1H NMR (400 MHz, CD 3 OD): δ ppm 8.29-8.17 (m, 2H), 7.97 (s, 1H), 7.91-7.82 (m, 2H), 4.05 (s, 3H); MS: MS m/z 194.7 (M + +l).

Step 2: Preparation of l-Fluoro-4-methoxyisoquinoline

To a solution of l-chloro-4-methoxyisoquinolin (2.5 g, 12.91 mmol) in DMSO was added cesium fluoride (4.01 g, 25.82 mmol) at room temperature. The reaction vessel (Pressure tube) was sealed and heated at 145 °C for 18 h. The reaction mass was diluted with water and extracted with ethyl acetate. The organic layer was dried over anhydrous Na 2 S0 4 and evaporated under reduced pressure to get crude compound. The crude compound was purified by silica gel chromatography to get desired compound (700 mg, 62%) as a white solid. 1H NMR (400 MHz, CD 3 OD): δ ppm

8.10 (m, 1H), 8.08 (m, 1H), 7.78-7.75 (m, 1H), 7.69-7.65 (m, 1H), 7.49 (m, 1H), 4.04 (s, 3H); 19 F NMR: δ ppm -78.66 (1 F); MS: MS m/z 178.1 (M + +l).

Preparation of -fluoro-6-methoxyisoquinoline

To a 2-5mL microwave vial was added l-chloro-6-methoxyisoquinoline (300 mg 1.55 mmol) and cesium fluoride (941 mg, 6.20 mmol), then DMSO (4 mL) and a small stir bar. The vial was sealed, then irradiated at 200 °C for 40 minutes in a microwave reactor. The reaction mixture was transfered to a 125 mL separatory funnel and was diluted with EtOAc (50 mL). The solution was washed with water:brine (25 mL:25 mL); dried over MgSC^; filtered; then concentrated in vacuo. The residue was transfered subjected to silica gel chromatography (hexanes:EtOAc, 100:0 to 80:20) to afford l-fluoro-6-methoxyisoquinoline as a white solid (21 mg, 8%). 1H-NMR (400MHz, CDC1 3 ) δ 8.07 (d, J=9.0 Hz, 1H), 7.99 (dd, J=5.8, 1.0 Hz, 1H), 7.42 (dd, J=5.8, 1.5 Hz, 1H), 7.27 (dd, J=9.0, 2.5 Hz, 1H), 7.13 (t, J=2.0 Hz, 1H), 3.97 (s, 3H).

Preparation of 1 -fluoro-3 ' ,6-dimethoxyisoquinoline

Scheme:

Step 1 :

2-(2-cyano-5-methoxyphenyl)acetic acid (3.8 g, 19.88 mmol), and SOCl 2 (20 mL, 274 mmol) were stirred in dichloromethane (25 mL) at RT. The suspension became a solution over 8 h. The reaction was stirred overnight. The volitile organics were removed under vacuum and the residue was taken up in DCM and filtered. The filtrate was concentrated and then dissolved in 4 N HC1 dioxane (30 mL) and transfered to a sealed vessel and heated to 60 °C for 3 h. The reaction was cooled and the solid was collected, washed with dioxane, and dried under vacuum to give the product l-chloro-6-methoxyisoquinolin-3-ol (3.3g, 70% yield). 1H NMR (400MHz, CDC1 3 ) δ 8.10 (d, J=9.6 Hz, 1H), 7.12 (dd, J=9.3, 2.5 Hz, 1H), 6.95 (s, 2H), 3.98 (s, 3H). Step 2:

To a mixture of l-chloro-6-methoxyisoquinolin-3-ol (3.3 g, 15.74 mmol) in DMF (30 mL) was added potassium carbonate (2.61 g, 18.89 mmol) and iodomethane (1.969 mL, 31.5 mmol). It was then stirred at rt overnight. LC/MS showed 2 peaks with the desired mass and also starting material. An additional 1 equ. of Mel, and 1 equ of K 2 CO 3 was added and the reaction warmed to 40 °C for 2h. LC/MS showed all starting material had been consumed. The reaction was diluted with EtOAc and water. The organic layer was washed with water, brine, dried over sodium sulfate, and concentrated under vacuum. The crude material was pruifed by silica gel column using 20% EtOAc/Hexanes. The product fractions were collected and the solvent removed under vacuum to give the desired product l-chloro-3,6- dimethoxyisoquinoline (2.47g, 70% yield) as a white solid. MS: MS m/z

223.93(M + +1). 1H NMR (400MHz, CDC1 3 ) δ 8.10 (d, J=9.3 Hz, 1H), 7.08 (dd, J=9.3, 2.5 Hz, 1H), 6.93 (d, J=2.5 Hz, 1H), 6.85 (s, 1H), 4.07 - 3.99 (m, 3H), 3.95 (s, 3H).

Step 3:

To a solution of l-chloro-3,6-dimethoxyisoquinoline (300 mg, 1.341 mmol) in DMSO (5 mL) was added CsF (408 mg, 2.68 mmol) and heated to 140 °C for 2 hrs. LC/MS showed the desired product. The reaction was diluted with Ethylacteate and washed with water, and brine. The organic phase was collected, dried over MgS04, and concentrated under vacuum to give the crude product l-fluoro-3,6- dimethoxyisoquinoline(250mg, 90%> yield) as a reddish brown solid. 1H NMR (400MHz, CDCI3) δ 7.93 (d, J=9.0 Hz, 1H), 7.03 (dd, J=9.3, 2.3 Hz, 1H), 6.95 (t, J=1.9 Hz, 1H), 6.75 (s, 1H), 4.01 - 3.96 (m, 3H), 3.96 - 3.90 (m, 3H). MS: MS m/z 208.07(M + +1).

Preparation of 1, -difluoro-6-methoxyisoquinoline Scheme:

Step 1: Preparation of (E)-3-(4-fluoro-3-methoxyphenyl) acrylic acid

To a solution of 4-fluoro-3-methoxybenzaldehyde (30 g, 195 mmol) in pyridine (134 ml) and Piperidine (4.12 ml) was added malonic acid (30.4 g, 292 mmol) at room temperature. The reaction mass was refluxed for 18 h. The solvent was evaporated under reduced pressure and the residue was acidified with 1.5N HC1 solution. The precipitated solid was filtered washed with pet ether to get crude compound (37 g, 97%) as white solid. The crude compound was taken to the next step without further purification. 1H NMR (400 MHz, CDC1 3 ): δ ppm 12.37 (s, 1H), 7.59-7.55 (d, J= 16 Hz, 1H), 7.53 (s, 1H), 7.26-7.22 (m, 2H), 6.59-6.55 (d, J= 16 Hz, 1H), 3.89 (s, 3H); MS: MS m/z 195.0 (M + -l). Step 2: Preparation of (E)-3-(4-fluoro-3-methoxyphenyl) acryloyl azide

To a solution of (E)-3-(4-fluoro-3-methoxyphenyl) acrylic acid (5 g, 25.5 mmol) in benzene (30 ml) was added triethylamine (5.16 g, 51 mmol) followed by DPPA (7.01 g, 25.5 mmol) at room temperature. The reaction mass was stirred at the same temperature for 18 h. The solvent was evaporated under reduced pressure and the residue was diluted with water and extracted with dichloromethane. The organic layer was dried over anhydrous sodium sulfate, filtered and evaporated to get crude compound. The crude compound was purified by conventional column

chromatography (Silica gel, 60-120 mesh) using 10% ethyl acetate in pet ether as mobile phase to get the desire compound as white solid (4 g, 71%). 1H NMR (400 MHz, CDC1 3 ): δ ppm 7.70-7.66 (d, J= 16 Hz, 1H), 7.12-7.07 (m, 3H), 6.35-6.31 (d, J = 16 Hz, 1H), 3.92 (s, 3H). Step 3: Preparation of 7-fluoro-6-methoxyisoquinolin- 1 (2H)-one

To a hot (125 °C) diphenyl ether (20 ml) was added (E)-3-(4-fluoro-3- methoxyphenyl) acryloyl azide (4 g, 18.08 mmol) portion wise. The reaction was heated at 250°C for 4 h. The reaction mass was cooled to room temperature and diluted with pet ether. The precipitated solid was filtered washed with pet ether to get crude compound (3.1 g, 89%). The crude compound was taken to the next step without further purification. MS: MS m/z 194.1 (M + +l).

Step 4: Preparation of l-chloro-7-fluoro-6-methoxyisoquinoline

A solution of 7-fluoro-6-methoxyisoquinolin-l(2H)-one (3.1 g, 16.05 mmol) in POCI 3 (25 ml) was refluxed for 18 h. The solvent was evaporated under reduced pressure and the residue was diluted with cold water. The aqueous solution was basified by solid sodium carbonate and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, filtered and evaporated under reduced pressure to get crude compound. The crude compound was purified by silica gel chromatography (20% ethyl acetate in pet ether) to get desired compound (1.9 g, 55%) as off-white solid. 1H NMR (400 MHz, CDC1 3 ): δ ppm 8.22-8.20 (d, J= 8 Hz, 1H), 7.97-7.94 (m, 1H), 7.49-7.48 (m, 1H), 7.18-7.16 (d, J=8 Hz, 1 H), 4.04 (s, 3H); MS: MS m/z 211.7 (M + +l).

Step 5: Preparation of 1, 7-difluoro-6-methoxyisoquinoline

To a solution of l-chloro-7-fluoro-6-methoxyisoquinoline (1.5 g, 7.09 mmol) in DMSO was added cesium fluoride (2.15 g, 14.18 mmol) at room temperature. The reaction vessel (Pressure tube) was sealed and heated at 145°C for 18 h. The reaction mass was diluted with water and extracted with ethyl acetate. The combined organic layer was dried over anhydrous Na 2 S0 4 and evaporated under reduced pressure to get crude compound. The crude compound was purified by silica gel chromatography to get desired compound (950 mg, 68%) as white solid. 1H NMR (400 MHz, CDC1 3 ): δ ppm 8.0-7.98 (m, 1H), 7.77-7.74 (d, J= 12 Hz, 1H), 7.42-7.40 (m, 1H), 7.21-7.19 (m, 1H), 4.04 (s, 3H); 19 F NMR: δ ppm -129.05 (1 F), -71.98 (IF); MS: MS m/z 196.1

(M + +l). -chloro-7-fluoro-6-methoxyisoquinolin-3-ol

Step 1 :

To a Schlenk flask equipped with a stir bar was added Pd(OAc) 2 (0.307 g, 1.37 mmol) and 2-(dicyclohexylphosphino)-2',4',6'-triisopropylbiphenyl ("XPhos", 1.31 g, 2.74 mmol). The flask was sealed with a rubber septum and then the flask was thrice evacuated and placed under nitrogen atmosphere to establish a N 2 atmosphere. To the flask was added dry CH 2 C1 2 (lmL) and Et 3 N (0.1 mL). The stirred solution was warmed with a heat gun until the solvent condensed on the walls of the flask. The solvent was removed under vacuum with the exclusion of oxygen. To the flask, while under positive nitrogen pressure, was added l-bromo-5-fluoro-4-methoxy-2- methylbenzene (6.00 g, 27.4 mmol), Na 2 C0 3 (0.363 g, 3.42 mmol), and potassium hexacyanoferrate(II) trihydrate (5.78 g, 13.7 mmol). The flask was thrice evacuated and placed under nitrogen atmosphere. To the flask was added water (20 mL) and acetonitrile (20 mL). The flask was placed in a 70 °C oil bath with stirring for 18 h. The mixture was cooled to room temperature, transferred to a separatory funnel, and then diluted with EtOAc and water. The mixture was shaken; the phases were separated. The organic phase was washed with water; then brine; dried over MgSC^; filtered; and then concentrated in vacuo. The resulting residue was subjected to silica gel chromatography (hexanes:EtOAc, 95:5 to 50:50) to afford 5-fluoro-4-methoxy-2- methylbenzonitrile as a white solid (3.43 g, 76 %). 1H-NMR (500MHz, DMSO-d 6 ) δ 7.73 (d, J=11.3 Hz, 1H), 7.28 (d, J=8.4 Hz, 1H), 3.91 (s, 3H), 2.45 (s, 3H). Step 2:

To a round-bottom flask equipped with a stir bar was added THF (50 mL) and N,N- diisopropylamine (2.98 mL, 20.9 mmol). The solution was cooled -78°C and to the solution was added dropwise n-butyllithium in hexanes (8.36 mL, 20.9 mmol). The solution was stirred for 10 minutes. To the solution was added 5-fluoro-4-methoxy-2- methylbenzonitrile (3.45 g, 20.9 mmol) in THF (15 mL). The solution was stirred for 30 min. To the solution was added dropwise chlorotrimethylsilane (2.64 mL, 20.9 mmol). The solution was stirred for an 60 minutes at -78 °C and then was allowed to warm to room temperature with stirring for 30 minutes. To the solution was carefully added water (80 mL). The mixture was transferred to a separatory funnel and the phases were separated. The aqueous phase was extracted with EtOAc. The combined organics were washed with brine; dried over Na 2 S0 4 ; filtered; then concentrated in vacuo to afford 3-fluoro-4-methoxy-6-methyl-2-(trimethylsilyl)benzonitrile as a yellow solid (4.01 g, 81%). 1H-NMR (400MHz, CDC1 3 ) δ 6.84 (d, J=8.3 Hz, 1H), 3.91 (s, 3H), 2.52 (s, 3H), 0.50 - 0.45 (m, 9H).

Step 3:

To a round-bottom flask equipped with a stir bar was added THF (85 mL) and N,N- diisopropylamine (2.65 mL, 18.6 mmol). The solution was cooled to -78 °C, then to the solution was added dropwise n-butyllithium in hexanes (7.43 mL, 18.58 mmol). The solution was stirred for 10 min. To the solution was added 3-fluoro-4-methoxy- 6-methyl-2-(trimethylsilyl)benzonitrile (4.01 g, 16.9 mmol) in THF (5 mL). The solution was stirred for 30 min. Carbon dioxide gas was then bubbled through the solution until the solution became clear. To the solution was carefully added water (80 mL) and the resulting mixture was allowed to warmed to room temperature. The mixture was transferred to a separatory funnel and was diluted with aq. NaOH (10 N, 40 mL). The mixture was twice washed with Et 2 0. The aqueous phase was isolated, cooled in a 0 °C bath, and then acidified with concentrated aq. HC1. The resulting mixture was warmed to room temperature and then extracted with EtOAc. The organic phase was collected; dried over Na 2 S0 4 ; filtered; then concentrated in vacuo to afford the 2-(2-cyano-4-fluoro-5-methoxyphenyl)acetic acid as a white solid (2.16 g, 61%). 1H-NMR (400MHz, CDC1 3 ) δ 7.37 (d, J=10.3 Hz, 1H), 6.98 (d, J=7.8 Hz, 1H), 3.96 (s, 3H), 3.89 (s, 2H). Step 4:

To a round-bottom flask equipped with a stir bar was added 2-(2-cyano-4-fluoro-5- methoxyphenyl)acetic acid (3.34 g, 16.0 mmol) and CH 2 CI 2 . To the mixture was added S0C1 2 (5.00 mL, 68.5 mmol). The mixture was stirred at room temperatuer for 16 h to afford a homogeneous solution. The volatile organics were removed under vacuum and the residue was taken up in CH 2 C1 2 and filtered. The filtrate was concentrated in vacuo. The residue was dissolved in a solution of HC1 in dioxane (4. ON, 30 mL) and then transfered to a bomb flask. The flask was sealed and then heated at 60 °C with stirring for 3 h. The mixture was cooled to room temperature and the solids were collected via filtration. The solids were washed with dioxane and then dried under vacuum to afford l-chloro-7-fluoro-6-methoxyisoquinolin-3-ol as a colorless solid (1.83 g, 50%). 1H-NMR (500MHz, DMSO-d 6 ) δ 7.76 (d, J=12.0 Hz, 1H), 7.44 (d, J=8.4 Hz, 1H), 6.92 (s, 1H), 3.98 (s, 3H).

Pre aration of 1, 7-dtfluoro-3, 6-dimethoxyisoquinoline

Step 1 :

To a round-bottom flask equipped with a stir bar and charged with l-chloro-7-fluoro- 6-methoxyisoquinolin-3-ol (138 mg, 0.606 mmol) in DMF (5 mL) was added K 2 CO 3 (168 mg, 1.21 mmol) and iodomethane (0.760 mL, 1.20 mmol). The mixture was stirred at room temperature for 16 h. To the solution was added K 2 CO 3 (84 mg, 0.6 mmol) and iodomethane (0.38 mL, 0.6 mmol). The mixture was stirred at 40 °C for 2 h. The mixture was transferred to a separatory funnel and was diluted with water; then was extracted with EtOAc. The organic phase was washed with water; then brine; then dried over MgSC^; filtered; then concentrated in vacuo. The resulting residue was subjected to silica gel chromatography (hexanes:EtOAc, 95:5 to 80:20) to afford l-chloro-7-fluoro-3, 6-dimethoxyisoquinoline as a white solid (100 mg, 68%). 1H-NMR (500MHz, DMSO-d 6 ) δ 7.83 (dd, J=l 1.8, 0.5 Hz, 1H), 7.52 (d, J=8.4 Hz, 1H), 7.18 (s, 1H), 3.99 (s, 3H), 3.91 (s, 3H).

Step 2:

To a vial equipped with a stir bar was added l-chloro-7-fluoro-3,6- dimethoxyisoquinoline (100 mg, 0.414 mmol), CsF (94 mg, 0.62 mmol) and DMSO (2 mL). The mixture was heated at 140 °C with stirring for 2 h. The mixture was transferred to a separatory funnel and was diluted with EtOAc. The mixture was washed with water; then brine. The organic phase was collected; dried over MgSO; filtered; then concentrated under vacuum. The resulting residue was subjected to silica gel chromatography (hexanes:EtOAc, 100:0 to 60:40) to afford 1 ,7-difluoro- 3,6-dimethoxyisoquinoline as a yellow solid (80 mg, 86%). 1H-NMR (500MHz, CDCls) δ 7.64 (d, J=10.9 Hz, 1H), 7.04 (dd, J=7.8, 1.3 Hz, 1H), 6.79 (s, 1H), 4.04 (s, 3H), 4.00 - 3.98 (m, 3H).

Preparation of 6, 8-dtfluoro-9-methoxy-2, 3-dihydro- lH-pyrano [ 2, 3-cJisoquinoline

Step l :

To a round-bottom flask equipped with a stir bar and placed under nitrogen atmosphere was added DMF (10 mL) and NaH (60%> dispersion in oil, 55.0 mg, 1.38 mmol). The solution was cooled to 0 °C. To the solution was added l-chloro-7- fluoro-6-methoxyisoquinolin-3-ol (285 mg, 1.25 mmol). The solution was stirred for 10 minutes. To the solution was added allylbromide (0.119 mL, 1.38 mmol). The solution was allowed to warm to room temperature with stirring for 1 h. The solution was transferred to a separatory funnel and was diluted with water; then extracted with EtOAc. The organic layer was washed with brine; dried over MgS0 4 ; filtered; and then concentrated in vacuo to afford 3-(allyloxy)-l-chloro-7-fluoro-6- methoxyisoquinoline as a solid (340 mg).

Step 2:

To a 2 dram vial equipped with a stir bar was added 3-(allyloxy)-l-chloro-7-fluoro-6- methoxyisoquinoline (all material from step 1, 340 mg, 1.27 mmol) and diethylene glycol dimethyl ether (5 mL). The vial was sealed with a PTFE-lined cap and then placed in a 175 °C heating block with stirring for 16 h. The solution was cooled to room temperature and then was transferred to a separatory funnel and diluted with water. The mixture was thrice extracted and EtOAc. The combined organics were washed with brine; dried over MgS0 4 ; filtered; then concentrated in vacuo to afford 4-allyl-l-chloro-7-fluoro-6-methoxyisoquinolin-3-ol as an orange-brown solid (306 mg, 90% over two steps). 1H-NMR (500MHz, DMSO-d 6 ) δ 11.23 (s, 1H), 7.80 (d, J=11.8 Hz, 1H), 7.30 (d, J=8.4 Hz, 1H), 5.97 - 5.85 (m, 1H), 5.08 - 4.98 (m, 2H), 4.00 (s, 3H), 3.66 (d, J=6.1 Hz, 2H).

Step 3:

To a round-bottom flask equipped with a stir bar was added 4-allyl-l-chloro-7- fluoro-6-methoxyisoquinolin-3-ol (all material from step 2, 306 mg, 1.14 mmol) and THF (10 mL). To the solution was added 9-Borabicyclo[3.3. l]nonane ("9-BBN",

0.5M in THF, 6.86 mL, 3.43 mmol). The solution was stirred at room temperature until 1H-NMR analysis of a reaction aliquot indicated complete conversion of the alkene starting material. To the solution was added aq. NaOH (3.0 N, 3.43 mL, 10.3 mmol), then and aq. 30% H 2 0 2 (1.17 mL, 11.4 mmol). The mixture was stirred for 16 h. To the mixture was acidified with via addition of aq. HCl (1.0 N). The mixture was transferred to a separatory funnel and was extracted with EtOAc. The organic phase was washed with brine; dried over MgS0 4 ; filtered; then concentrated in vacuo. The resulting residue was subjected to silica gel chromatography (hexanes: EtOAc, 80:20 to 50:50) to afford l-chloro-7-fluoro-4-(3-hydroxypropyl)-6-methoxyisoquinolin-3 -ol as a white solid (210 mg, 64%). 1H-NMR (500MHz, acetone-d 6 ) δ 9.53 (br. s., 1H), 7.78 (d, J=12.0 Hz, 1H), 7.57 (d, J=8.2 Hz, 1H), 4.08 (s, 3H), 3.61 (br. s., 2H), 3.11 - 3.04 (m, 2H), 1.92 - 1.83 (m, 2H).

Step 4:

To a round-bottom flask equipped with a stir bar was added PPh 3 (347 mg, 1.32 mmol), l-chloro-7-fluoro-4-(3-hydroxypropyl)-6-methoxyisoquinolin-3 -ol (210 mg, 0.735 mmol), and THF (20 mL). The solution was cooled to 0 °C, then to the solution was added dropwise diisopropylazodicarboxylate ("DIAD", 0.286 mL, 1.47 mmol). The mixture was allowed to warm to room temperature with stirring for 4 h. The mixture was concentrated in vacuo and the resulting residue was subjected to silica gel purification to afford 6-chloro-8-fluoro-9-methoxy-2,3-dihydro-lH- pyrano[2,3-c]isoquinoline as a yellow solid (171 mg). 1H-NMR (500MHz, CDC1 3 ) δ 7.86 (d, J=11.7 Hz, 1H), 7.01 (d, J=7.9 Hz, 1H), 4.43 - 4.34 (m, 2H), 4.04 (s, 3H), 2.97 (t, J=6.5 Hz, 2H), 2.22 - 2.13 (m, 2H).

Step 5:

To a vial equipped with a stir bar was added 6-chloro-8-fluoro-9-methoxy-2,3- dihydro-lH-pyrano[2,3-c]isoquinoline (all material from step 4, 171 mg, 0.639 mmol), CsF (146 mg, 0.958 mmol), and DMSO (3 mL). The vial was placed in a 140 °C heating block with stirring for 2 h. The mixture was transferred to a separatory funnel and was diluted with water. The mixture was extracted with EtOAc. The organic phase was washed with water; then brine; then dried over MgS0 4 ; filtered; then concentrated in vacuo. The resulting residue was subjected to silica gel chromatography (hexanes:EtOAc, 100:0 to 60:40) to afford 6,8-difluoro-9-methoxy- 2,3-dihydro-lH-pyrano[2,3-c]isoquinoline as a yellow solid (114 mg, 71% over two steps). MS: MS m/z 252.2 (M + +l).

Preparation of 4 6-dtfluoro-2, 7-dimethoxyquinoline Scheme:

Step 1 :

To a large round-bottom flask equipped with a stir bar was added 4-fiuoro-3- methoxyaniline (4.00 g, 28.3 mmol) and malonic acid (2.95 g, 28.3 mmol). To the flask was added POCI 3 (4.76 mL, 101 mmol). The flask was fitted with a bump-trap and then heated to 105 °C. After 5 minutes vigorous effervescence and foaming was observed. When the foaming had subsided, to the flask was added POCI 3 (4.76 mL, 101 mmol) upon which the mixture slowly became a homogeneous solution. The solution was stirred at 105 °C for 16 h. The solution was cooled to room temperature and then concentrated in vacuo. The resulting oil residue was diluted with CH 2 C1 2 and to the mixture was added ice. Once the ice had melted, the flask was placed in a 0 °C bath and to the mixture was added aq. NaOH (10 N) to adjust the aq. phase to pH > 8. The mixture was transferred to a separatory funnel and the phases were separated. The organic phase was washed with brine; dried over MgS0 4 ; filtered; then concentrated in vacuo. The resulting residue was subjected to silica gel chromagraphy (hexanes:EtOAc, 95:5 to 80:20) to afford 2,4-dichloro-6-fluoro-7- methoxyquinoline as a white solid (3.7 g, 53%). 1H-NMR (500MHz, CDC1 3 ) δ 7.81 (d, J=l 1.3 Hz, 1H), 7.46 (d, J=7.9 Hz, 1H), 7.42 (s, 1H), 4.04 (s, 3H).

Step 2:

To a round-bottom flask equipped with a stir bar was added 2,4-dichloro-6- f uoro-7-methoxyquinoline (2.00 g, 8.13 mmol), CsF (3.09 g, 20.3 mmol) and DMSO (20 mL). The mixture was stirred at 140 °C for 2 h. The mixture was cooled to room temperature and then transferred to a separatory funnel; then diluted with water. The mixture was extracted with EtOAc. The organic phase was washed with water; then brine; dried over MgSC^; filtered; then concentrated in vacuo to afford as a light- orange solid (1.75 g, 100%). 1H-NMR (500MHz, CDC1 3 ) δ 7.66 (d, J=10.7 Hz, 1H), 7.39 (dd, J=7.6, 1.5 Hz, 1H), 6.72 (dd, J=9.1, 1.5 Hz, 1H), 4.05 - 4.02 (m, 3H). Step 3:

To a round-bottom flask equipped with a stir bar was added 2,4,6-trifluoro-7- methoxyquinoline (500 mg, 2.35 mmol) and THF (15 mL). To the solution was added NaOMe (25% in MeOH, 2.35 mmol). The solution was stirred at room temperature for 16 h. The solution was transferred to a separatory funnel and was diluted with EtOAc; then washed with brine. The organic solution was dried over MgSC^; filtered; then concentrated in vacuo. The resulting residue was subjected to silica gel chromatography (hexanes:EtOAc. 95:5 to 80:20) to afford 4,6-difluoro-2,7- dimethoxyquinoline as a white solid (268 mg, 51 >). NMR

Step l :

To a round-bottom flask equipped with a stir bar was added 2,4-dichloro-7- methoxyquinoline (905 mg, 3.97 mmol), CsF (1.51 g, 9.92 mmol) and DMSO (20 mL). The mixture was stirred at 140 °C for 2 h. The mixture was allowed to cool to room temperature; then was transferred to a separatory funnel and diluted with water. The mixture was extracted with EtOAc. The organic phase was dried over MgSC^; filtered; then concentrated in vacuo to afford 2,4-difluoro-7-methoxyquinoline as a light-orange solid (706 mg, 91%). MS: MS m/z 196.2 (M + +l). Step 2:

To a round-bottom flask equipped with a stir bar was added 2,4-difluoro-7- methoxyquinoline (all material from step 1, 706 mg, 3.62 mmol) and THF (15 mL). To the solution was added sodium methoxide (25% in methanol, 3.62 mmol). The solution was stirred at room temperature for 16 h. The solution was transferred to a separatory funnel and was diluted with EtOAc; then washed with brine. The organic phase was dried over MgSC^; filtered; then concentrated in vacuo. The resulting residue was subjected to silica gel chromatography (hexanes:EtOAc, 95:5 to 80:20) to afford 4-fluoro-2,7-dimethoxyquinoline as a white solid (381 mg, 1.84 mmol, 51%). 1H-NMR (500MHz, CDC1 3 ) δ 7.83 (d, J=9.0 Hz, IH), 7.22 (t, J=2.1 Hz, IH), 7.05 (dd, J=9.0, 2.5 Hz, IH), 6.45 (d, J=10.7 Hz, IH), 4.06 (s, 3H), 3.96 - 3.92 (m, 3H); 13 C-NMR (101MHz, CDC1 3 ) δ 167.8, 165.2, 164.0 (d, J=14.6 Hz), 161.9, 149.8 (d, J=6.9 Hz), 121.7 (d, J=4.6 Hz), 116.4, 106.5, 106.4, 94.4 (d, J=19.3 Hz), 55.5, 53.7.

After the desired product was eluted from silica gel a second major product was isolated: 2-fluoro-4,7-dimethoxyquinoline as a white solid (222 mg, 30%>). 1H-NMR (400MHz, CDC1 3 ) δ 8.00 (d, J=9.0 Hz, IH), 7.20 (d, J=2.5 Hz, IH), 7.09 (dd, J=9.0, 2.5 Hz, IH), 6.26 (s, IH), 4.06 - 4.02 (m, 3H), 3.92 (s, 3H); 13 C-NMR (101MHz, CDC1 3 ) 166.2 (d, J=13.1), 164.4, 162.0, 161.9, 147.9 (d, J=21.6 Hz), 123.2, 117.2 (d, J=2.3 Hz), 114.6, 106.8, 86.5 (d, J=47.0 Hz), 56.1, 55.5.

Preparation of 3-chlo -6-methoxy-2-(trifluoromethyl)quinoxaline

Scheme:

Step 1 Step 2 Step 3 Step 1: Preparation of 6-methoxy-3-(trifluoromethyl)quinoxalin-2 ( lH)-one and 7- methoxy-3- ( trifluoromethyl)quinoxalin-2 ( lH)-one

To a solution of 4-methoxybenzene-l,2-diamine (1 g, 7.24 mmol) in ethanol(10 ml) was added ethyl 3,3,3-trifluoro-2-oxopropanoate (1.23 g, 7.24 mmol)). The reaction mass was heated at reflux for overnight. The solvent was evaporated under reduced pressure and the residue was diluted with ethyl acetate and then evaporated to dryness to get the crude compound. The crude compound was washed with pet ether to get the product (1.55 g, 88 % yield) as a mixture of regioisomers (black solid). This crude compound was taken to the next step without separation of isomers. 1H NMR (400 MHz, DMSO-d 6 ): δ ppm 12.92(br, s, 2H), 7.84- 7.81(d, J=12 Hz, 1H), 7.44-7.33 (m, 4H), 7.82 (s, 1H), 3.87(s, 6H), MS: MS m/z 245.15 (M + +l).

Step 2: Preparation of2-chloro-6-methoxy-3-(trifluoromethyl)quinoxaline and 3- chloro-6-methoxy-2-(trifluoromethyl)quinoxaline

A solution of 6-methoxy-3-(trifluoromethyl)quinoxalin-2(lH)-one & 7- methoxy-3-(trifluoromethyl)quinoxalin-2(lH)-one (0.90 g, 3.69 mmol) in POCI 3 (10 ml) was refluxed for 3 h. The solvent was evaporated under reduced pressure and the residue was diluted with cold water. The aqueous solution was basified by solid sodium carbonate and extracted with ethyl acetate. The combine organic layer was dried over anhydrous sodium sulfate, filtered and evaporated under reduced pressure to get crude compound. The crude compound was purified by silica gel

chromatography (20% ethyl acetate in pet ether) to get regioisomers as mixture. Step 3:

The mixture of regioisomers were separated by SFC purification to afford 2- chloro-6-methoxy-3-methylquinoxaline (0.51 g, 38.7%) and 3-chloro-6-methoxy-2- methylquinoxaline (required isomer) (0.48 g, 36.5%>) as off white solids.

2- chloro-6-methoxy-3-(trifluoromethyl)quinoxaline: 1H NMR (400 MHz, DMSO- d 6 ): δ 8.10-8.07 (d, J= 12 Hz, 1H), 7.75-7.44 (m, 2H), 3.95 (s, 3H); 19 F -NMR: δ ppm -65.36(1F) MS: MS m/z 263.10 (M + +l).

3- chloro-6-methoxy-2-(trifluoromethyl)quinoxaline: 1H NMR (400 MHz, DMSO- d 6 ): δ 8.11-8.08 (d, J= 12 Hz, 1H), 7.78-7.75 (d, J= 12 Hz, 1H), 7.68 (s, 1H), 4.00 (s, 3H); 19 F- NMR: δ ppm -65.36(1F) MS: MS m/z 263.09 (M + +l).

Preparation of (2S,3R)-l-tert-butyl 2-methyl 3-ethylaziridine-l,2-dicarboxylate

Step 1 :

To a 500 mL round-bottom flask equipped with a stir bar was added (S)-2- (diphenyl((trimethylsilyl)oxy)methyl)pyrrolidine (2.60 g, 8.00 mmol) and CHCI 3

(200 mL). To the solution was added tert-butyl tosylcarbamate (10.9 g, 40.0 mmol). The flask was placed in a room temperature-water bath. To the stirred solution was added sodium acetate (9.84 g, 120 mmol), then (E)-pent-2-enal (4.04 g, 48.0 mmol). The mixture was stirred at room temperature for 40 minutes. The mixture was filtered, washing the filter cake with a small amount of CHCI 3 . The filtrate was concentrated in vacuo at 30 °C. The resulting orange liquid was dissolved in Et 2 0 (400 mL) and transfered to a 1L separatory funnel. The solution was washed with water:aq. sat. NaHC0 3 (200 mL:200 mL). The aqueous phase was extracted with Et 2 0 (100 mL). The combined organics were washed with brine (200 mL); dried over MgS0 4 ; filtered; then concentrated in vacuo to afford (2R,3S)-tert-butyl 2-ethyl- 3-formylaziridine-l-carboxylate as a pale orange liquid.

Step 2:

The following step was run duplicate. To a 250 mL round-bottom flask equipped with a stir bar was added (2R,3S)-tert-butyl 2-ethyl-3-formylaziridine-l-carboxylate (one-half of the material isolated in step 1 , assumed 20 mmol) as a solution in MeOH (100 ml). The solution was cooled to 0 °C. To the solution was added sodium cyanide (1.96 g, 40.0 mmol). The solution was stirred for 10 minutes. To the solution was added oxidation-grade Mn0 2 (34.8 g, 400 mmol). The mixture was stirred for 10 minutes, then the ice bath was removed and the solution was allowed to warm to room temperature with stirring for 18 hours. The reaction mixture was filtered through a pad of diatomaceous earth (Celite™) and the filter cake was extracted with EtOAc (100 mL). The filtrate was concentrated in vacuo and the resulting orange solid residue was dissolved in water (100 mL) and EtOAc (100 mL) and transfered to a 1L separatory funnel where the mixture was further diluted with saturated aq. NaCl ("brine", 100 mL) and EtOAc (100 mL). The mixture was shaken and the phases were separated. The aq. phase was extracted with EtOAc (100 mL). The combined organics were washed with brine (100 mL); dried over MgS0 4 ;

filtered; then concentrated in vacuo. The resulting residue was combined with the duplicate run and the combined material was subjected to silica gel chromatography (hexanes:EtOAc, 50:50 to 0: 100) to afford (2S,3R)-l-tert-butyl 2-methyl 3- ethylaziridine-l,2-dicarboxylate as a yellow liquid (3.055 g, 33% over two steps). 1H- NMR (500MHz, CDC1 3 ) δ 3.77 (s, 3H), 2.84 (d, J=2.7 Hz, 1H), 2.77 (td, J=6.0, 2.7 Hz, 1H), 1.63 - 1.52 (m, 2H), 1.47 - 1.45 (m, 9H), 1.05 (t, J=7.5 Hz, 3H).

Preparation of (2S,3S)-methyl 2-((tert-butoxycarbonyl)amino)-3-((R)-hex-5-en-2- yloxy)pentanoate

To a round-bottom flask equipped with a stir bar was added (2S,3R)-l-tert- butyl 2-methyl 3-ethylaziridine-l,2-dicarboxylate (691, 3.02 mmol) and CH 2 C1 2 (12 mL). To the solution was added (R)-hex-5-en-2-ol (363 mg, 3.62 mmol), then BF 3 - OEt 2 (1.0M in CH 2 C1 2 , 0.30 mL). The solution was stirred at room temperature for

16 h. The solution was transfered to a separatory funnel and was dilued with CH 2 C1 2 , then the solution was washed with aq. saturated NaHC0 3 ; then brine. The organic phase was dried over Na2S04; filtered; then concentrated in vacuo. The resulting residue was subjected to silica gel chromatography (hexanes:EtOAc 90: 10 to 70:30) to afford (2S,3S)-methyl 2-((tert-butoxycarbonyl)amino)-3-((R)-hex-5-en-2- yloxy)pentanoate as a colorless oil (426 mg, 43%). 1H-NMR (500MHz, CDC1 3 ) δ 5.81 (ddt, J=17.0, 10.4, 6.5 Hz, 1H), 5.19 (d, J=7.7 Hz, 1H), 5.08 - 5.00 (m, 1H), 4.96 (dd, J=10.2, 1.7 Hz, 1H), 4.50 (dd, J=8.3, 3.5 Hz, 1H), 3.75 (s, 3H), 3.65 - 3.58 (m, 1H), 3.57 - 3.50 (m, 1H), 2.19 - 2.06 (m, 2H), 1.66 - 1.56 (m, 2H), 1.55 - 1.38 (m, 11H), 1.13 (d, J=6.1 Hz, 3H), 0.97 (t, J=7.3 Hz, 3H).

Preparation of (2S,4R)-methyl l-((2S,3S)-2-((tert-butoxycarbonyl)amino)-3-(pent-4- en-l-yloxy)pentanoyl)-4-hydroxypyrrolidine-2-carboxylate

To a round-bottom flask equipped with a stir bar was added (2S,3R)-l-tert- butyl 2-methyl 3-ethylaziridine-l,2-dicarboxylate (1.50 g, 6.54 mmol) and CH 2 CI 2 (30 mL). To the solution was added pent-4-en-l-ol (0.676 g, 7.85 mmol), then BF 3 - OEt 2 (93 mg, 0.083 mL, 0.65 mmol). The solution was stirred at room temperature for 16 h. The solution was transferred to a separatory funnel and was diluted with CH 2 C1 2 . The solution was washed with aq. saturated NaHC0 3 ; then brine. The organic phase was dried over Na 2 S0 4 ; filtered; then concentrated in vacuo. The resulting residue was subjected to silica gel chromatography (hexane:EtOAc 90: 10 to 70:30) to afford (2S,4R)-methyl l-((2S,3S)-2-((tert-butoxycarbonyl)amino)-3-(pent- 4-en-l-yloxy)pentanoyl)-4-hydroxypyrrolidine-2-carboxylate as as an oil (1.06 g, 51%). 1H-NMR (500MHz, CDC1 3 ) δ 5.87 - 5.76 (m, 1H), 5.20 (d, J=7.9 Hz, 1H), 5.07 - 5.01 (m, 1H), 4.99 - 4.95 (m, 1H), 4.54 (dd, J=8.6, 3.9 Hz, 1H), 3.75 (s, 3H), 3.58 - 3.51 (m, 1H), 3.50 - 3.44 (m, 2H), 2.17 - 2.08 (m, 2H), 1.69 - 1.61 (m, 2H), 1.61 - 1.54 (m, 2H), 1.46 - 1.43 (m, 9H), 0.99 (t, J=7.4 Hz, 3H). Preparation of tert-butyl ((2R,6S, 7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-hydroxy-9- methyl-14a-( ( (l-methylcyclopropyl)sulfonyl)carbamoyl)-5, 16-dioxo- 2, 3,5,6, 7,9,10,11,13 a, 14,14a, 15,16,16a-tetradecahydro- 1H- cyclopropa[i] pyrrolo [1 ,2-e] [1 ,5,8] oxadiazacyclopentadecin-6-yl)carbamate

Step 1 :

To a round-bottom flask equipped with a stir bar was added (2S,3S)-methyl 2-((tert- butoxycarbonyl)amino)-3-((R)-hex-5-en-2-yloxy)pentanoate (458 mg, 1.39 mmol), THF (5 mL) and MeOH (5 mL). To the stirred solution was added aq. LiOH (2.0 M, 2 mL). The mixture was stirred for 16 h, then was concentrated in vacuo to afford an aqueous solution. The solution was acidified with aq. HCl (1 M) and then transferred to a separatory funnel. The mixture was twice extracted with EtOAc. The combined organics were washed with brine; dried over MgS0 4 ; filtered; then concentrated in vacuo to afford (2S,3S)-2-((tert-butoxycarbonyl)amino)-3-((R)-hex-5-en-2- yloxy)pentanoic acid as a white foam (438 mg, 100%). Step 2:

To a round-bottom flask equipped with a stir bar was added (2S,3S)-2-((tert- butoxycarbonyl)amino)-3-(pent-4-en-l-yloxy)pentanoic acid (all material from step 1, 438 mg, 1.39 mmol), (2S,4R)-methyl 4-hydroxypyrrolidine-2-carboxylate-HCl salt (277 mg, 1.53 mmol), l-[Bis(dimethylamino)methylene]-lH-l,2,3-triazolo[4,5- b]pyridinium 3-oxid hexafluorophosphate ("HATU", 528 mg, 1.39 mmol), and CH 2 CI 2 (10 mL). To the mixture was added N,N-diisopropylethylamine (0.728 mL, 4.17 mmol). The mixture was stirred at room temperature for 16 h. The reaction mixture was transferred to a separatory funnel and was thrice washed with aq. IN HC1; then brine. The organic phase was dried over MgSC^; filtered; then

concentrated in vacuo to afford (2S,4R)-methyl l-((2S,3S)-2-((tert- butoxycarbonyl)amino)-3-((R)-hex-5-en-2-yloxy)pentanoyl)-4-h ydroxypyrrolidine-2- carboxylate as a yellow oil. Step 3:

To a round-bottom flask equipped with a stir bar was added (2S,4R)-methyl 1- ((2S,3S)-2-((tert-butoxycarbonyl)amino)-3-((R)-hex-5-en-2-yl oxy)pentanoyl)-4- hydroxypyrrolidine-2-carboxylate (all material from step 2, 1.39 mmol), THF (5 mL) and MeOH (5 mL). To the solution was added aq. lithium hydroxide (2.0 M, 4.51 mmol). The mixture was stirred at room temperature for 16 h; then was concentrated to afford an aqueous solution. This solution was acidified and then transferred to a separatory funnel. The solution was twice extracted with EtOAc. The combined organics were washed with brine; dried over MgSC^; filtered; then concentrated in vacuo to afford (2S,4R)-l-((2S,3S)-2-((tert-butoxycarbonyl)amino)-3-((R)-hex -5-en- 2-yloxy)pentanoyl)-4-hydroxypyrrolidine-2-carboxylic acid as a white solid (489 mg).

Step 4:

To a round-bottom flask equipped with a stir bar was added (2S,4R)-l-((2S,3S)-2- ((tert-butoxycarbonyl)amino)-3-((R)-hex-5-en-2-yloxy)pentano yl)-4- hydroxypyrrolidine-2-carboxylic acid (all material from step 3, 489 mg, 1.14 mmol), ( 1 R,2S)- 1 -amino-N-(( 1 -methylcyclopropyl)sulfonyl)-2- vinylcyclopropanecarboxamide-trifluoroacetic acid salt (450 mg, 1.26 mmol), HATU (456 mg, 1.20 mmol) and DCM (10 mL). To the stirred solution was added N,N- diisopropylethylamine (0.80 mL, 4.56 mmol). The mixture was stirred for 16 h and then was transferred to a separatory funnel. The mixture was thrice washed with aq. IN HC1; then brine. The solution was dried over MgS0 4 ; filtered; then concentrated in vacuo to afford tert-butyl ((2S,3S)-3-((R)-hex-5-en-2-yloxy)-l-((2S,4R)-4- hydroxy-2-((( 1 R,2S)- 1 -((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-2- vinylcyclopropyl)carbamoyl)pyrrolidin-l-yl)-l-oxopentan-2-yl )carbamate as a light- orange solid foam (569 mg). Step 5:

To a round-bottom flask equipped with a stir bar was added tert-butyl ((2S,3S)-3- ((R)-hex-5-en-2-yloxy)-l-((2S,4R)-4-hydroxy-2-(((lR,2S)-l-(( (l- methylcyclopropyl)sulfonyl)carbamoyl)-2-vinylcyclopropyl)car bamoyl)pyrrolidin-l- yl)-l-oxopentan-2-yl)carbamate (all material from step 4, 569 mg, 0.869 mmol) and DCE (100 mL). The solution was sparged with nitrogen for 30 min. and then to the solution was added (l,3-Bis-(2,4,6-trimethylphenyl)-2- imidazolidinylidene)dichloro(o-isopropoxyphenylmethylene)rut henium ("Hoveyda- Grubbs Catalyst 2nd Generation", 27 mg, 0.043 mmol). The solution was stirred at 80°C for 2 h and then cooled to 45°C and stirred for 3 days. The solution was concentrated in vacuo and the resulting residue was subjected to silica gel purification (hexanes: acetone 80:20 to 40:60). The product containing fractions were pooled; concentrated; and re-subjected to silica gel chromatography (hexanes: acetone 80:20 to 40:60) to afford tert-butyl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2- hydroxy-9-methyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,1 l,13a,14,14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate as a light-brown solid foam (90 mg, 10% over 5 steps). 1H-NMR (500MHz, CD 3 OD) δ 8.93 (s, 1H), 5.61 (td, J=10.1, 6.2 Hz, 1H), 5.03 (t, J=10.3 Hz, 1H), 4.58 (br. s., 1H), 4.47 - 4.37 (m, 2H), 3.92 (br. s., 2H), 3.86 - 3.79 (m, 1H), 3.58 - 3.49 (m, 1H), 2.75 (q, J=9.5 Hz, 1H), 2.64 - 2.52 (m, 1H), 2.23 - 2.11 (m, 2H), 1.92 - 1.80 (m, 2H), 1.75 (dd, J=8.5, 5.5 Hz, 1H), 1.66 -

1.60 (m, 1H), 1.59 - 1.52 (m, 2H), 1.49 (s, 4H), 1.43 (s, 9H), 1.41 - 1.32 (m, 2H), 1.15 (d, J=6.3 Hz, 3H), 0.95 (t, J=7.4 Hz, 3H), 0.88 - 0.84 (m, 2H). Preparation of tert-butyl ((2R,6S, 7S,9R, 13aS, 14aR, 16aS,Z)-l 4a- ( ( cyclopropylsulfonyl)carbamoyl)- 7-ethyl-2-hydroxy-9-methyl-5, 16-dioxo- 2, 3,5,6, 7,9,10,11,13 a, 14,14a, 15,16,16a-tetradecahydro- 1H- cyclopropa[i] pyrrolo [1 ,2-e] [1 ,5,8] oxadiazacyclopentadecin-6-yl)carbamate

Step 1 :

To a round-bottom flask equipped with a stir bar was added (2S,4R)-l-((2S,3S)-2- ((tert-butoxycarbonyl)amino)-3-((R)-hex-5-en-2-yloxy)pentano yl)-4- hydroxypyrrolidine-2-carboxylic acid (700 mg, 1.63 mmol), (lR,2S)-l-amino-N- (cyclopropylsulfonyl)-2-vinylcyclopropanecarboxamide-p-tolue nesulfonic acid salt (723 mg, 1.80 mmol), HATU (652 mg, 1.72 mmol) and CH 2 C1 2 (10 mL). To the mixture was added N,N-diisopropylethylamine (1.14 mL, 6.53 mmol). The mixture was stirred for 16 h. The mixture was transferred to a separatory funnel and was thrice washed with aq. IN HC1; then brine. The organic solution was dried over MgSC^; filtered; then concentrated in vacuo. The resulting residue was subjected to silica gel chromatography (hexanes: acetone 95:5 to 60:40) to afford tert-butyl ((2S,3S)-l-((2S,4R)-2-(((lR,2S)-l-((cyclopropylsulfonyl)carb amoyl)-2- vinylcyclopropyl)carbamoyl)-4-hydroxypyrrolidin- 1 -yl)-3-((R)-hex-5-en-2-yloxy)- 1 - oxopentan-2-yl)carbamate as a white solid foam (755 mg, 1.18 mmol, 72%).

Step 2:

To a round-bottom flask equipped with a stir bar was added tert-butyl ((2S,3S)-1- ((2S,4R)-2-(((lR,2S)-l-((cyclopropylsulfonyl)carbamoyl)-2- vinylcyclopropyl)carbamoyl)-4-hydroxypyrrolidin- 1 -yl)-3-((R)-hex-5-en-2-yloxy)- 1 - oxopentan-2-yl)carbamate (755 mg, 1.178 mmol) and DCE (120 mL). The solution was sparged with nitrogen for 30 min. and then to the solution was added Hoveyda- Grubbs Catalyst 2nd Generation (37 mg, 0.059 mmol). The solution was stirred at 80°C for 4 h. The reaction solution was cooled to room temperature and then was concentrated in vacuo. The resulting residue was subjected to C 18 chromatography (water with 0.1% TFA:acetonitrile with 0.1% TFA 60:40 to 0: 100) to afford tert- butyl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)- 14a-((cyclopropylsulfonyl)carbamoyl)-7- ethyl-2-hydroxy-9-methyl-5,16-dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15, 16,16a- tetradecahydro- 1 H-cyclopropa[i]pyrrolo[ 1 ,2-e] [ 1 ,5 ,8]oxadiazacyclopentadecin-6- yl)carbamate as a brown solid foam (393 mg, 54%). 1H-NMR (500MHz, CD 3 OD) δ 8.87 (s, 1H), 5.60 (td, J=10.2, 6.1 Hz, 1H), 5.08 (t, J=10.2 Hz, 1H), 4.57 (br. s., 1H), 4.46 - 4.36 (m, 2H), 3.91 (d, J=2.8 Hz, 2H), 3.85 - 3.79 (m, 1H), 3.58 - 3.50 (m, 1H), 2.96 - 2.86 (m, 1H), 2.74 (q, J=9.3 Hz, 1H), 2.63 - 2.51 (m, 1H), 2.27 - 2.11 (m, 2H), 1.90 - 1.79 (m, 2H), 1.76 (dd, J=8.5, 5.5 Hz, 1H), 1.73 - 1.63 (m, 1H), 1.61 - 1.47 (m, 4H), 1.42 (s, 8H), 1.30 (ddt, J=9.9, 7.1, 4.8 Hz, 1H), 1.16 (d, J=6.3 Hz, 3H), 1.14 - 0.98 (m, 5H), 0.95 (t, J=7.4 Hz, 3H). Preparation of tert-butyl ((2R,6S, 7S,9R,13aS,14aR,16aS,Z)-7-ethyl-14a-(((l-

(fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-2-hydroxy-9 -methyl-5,16-dioxo- 2, 3,5,6, 7,9,10,11,13 a, 14,14a, 15,16,16a-tetradecahydro- 1H- cyclopropa[i] pyrrolo [1 ,2-e] [1 ,5,8] oxadiazacyclopentadecin-6-yl)carbamate

Scheme:

Step 1 :

To a round-bottom flask equipped with a stir bar was added was added (2S,4R)-1- ((2S,3S)-2-((tert-butoxycarbonyl)amino)-3-((R)-hex-5-en-2-yl oxy)pentanoyl)-4- hydroxypyrrolidine-2-carboxylic acid (700 mg, 1.63 mmol), (lR,2S)-l-amino-N-((l- (fluoromethyl)cyclopropyl)sulfonyl)-2-vinylcyclopropanecarbo xamide-trifluoroacetic acid salt (676 mg, 1.80 mmol), HATU (652 mg, 1.72 mmol) and CH 2 C1 2 (10 mL). To the mixture was added N,N-diisopropylethylamine (1.14, 6.53 mmol). The mixture was stirred at room temperature for 16 h. The reaction mixture was transfered to a separatory funnel and was thrice washed with aq. IN HCl; then brine. The organic phase was dried over MgSC^; filtered; then concentrated in vacuo. The resulting residue was subjected to silica gel chromatography (hexanes: acetone 95:5 to 60:40) to afford tert-butyl ((2S,3S)-l-((2S,4R)-2-(((lR,2S)-l-(((l- (fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-2-vinylcyclopr opyl)carbamoyl)-4- hydroxypyrrolidin- 1 -yl)-3-((R)-hex-5-en-2-yloxy)- 1 -oxopentan-2-yl)carbamate as a white solid foam (559 mg, 51%).

Step 2:

To a round-bottom flask equipped with a stir bar was added tert-butyl ((2S,3S)-1- ((2S,4R)-2-(((lR,2S)-l-(((l-(fluoromethyl)cyclopropyl)sulfon yl)carbamoyl)-2- vinylcyclopropyl)carbamoyl)-4-hydroxypyrrolidin- 1 -yl)-3-((R)-hex-5-en-2-yloxy)- 1 - oxopentan-2-yl)carbamate (559 mg, 0.831 mmol) and DCE (90 mL). The solution was sparged with nitrogen for 30 min. and then to the solution was added Ho vey da- Grubbs Catalyst 2nd Generation (26 mg, 0.042 mmol). The solution was stirred at 80 °C for 16 h. The solution was concentrated in vacuo and the resulting residue was subjected to C 18 chromatography (water with 0.1% TFA:acetonitrile with 0.1% TFA, 60:40 to 0: 100) to afford tert-butyl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-14a- ((( 1 -(fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-2-hydroxy-9-m ethyl-5 ,16- dioxo-2,3, 5,6, 7,9, 10, l l,13a,14, 14a, 15,16, 16a-tetradecahydro-lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecin-6- yl)carbamate as a brown solid foam (120 mg, 22%). 1H NMR (500MHz, CD 3 OD) δ 8.91 (s, IH), 5.60 (td, J=10.0, 6.5 Hz, IH), 5.03 (t, J=10.3 Hz, IH), 4.61 - 4.55 (m, J=l 1.2 Hz, 2H), 4.49 (d, J=11.2 Hz, IH), 4.43 (d, J=9.9 Hz, IH), 4.38 (t, J=7.8 Hz, IH), 3.96 - 3.87 (m, 2H), 3.82 (d, J=12.1 Hz, IH), 3.53 (dq, J=9.4, 6.5 Hz, IH), 2.71 (q, J=9.4 Hz, IH), 2.61 - 2.50 (m, IH), 2.23 - 2.13 (m, 2H), 1.84 (dd, J=10.9, 6.8 Hz, 2H), 1.74 - 1.60 (m, 5H), 1.59 - 1.54 (m, 2H), 1.54 - 1.46 (m, 2H), 1.43 (s, 9H), 1.26 - 1.20 (m, 2H), 1.16 (d, J=6.3 Hz, 3H), 0.95 (t, J=7.4 Hz, 3H).

Preparation of tert-butyl ((2R,6S, 7S,13aS,14aR,16aS,Z)-7-ethyl-2-hydroxy-14a-(((l- methylcyclopropyl)sulfonyl)carbamoyl)-5, 16-dioxo- 2, 3,5,6, 7,9,10,11,13 a, 14,14a, 15,16,16a-tetradecahydro- 1H- cyclopropa[i] pyrrolo [1 ,2-e] [1 ,5,8] oxadiazacyclopentadecin-6-yl)carbamate

cheme:

Step 1 :

To a round-bottom flask equipped with a stir bar was added (2S,3S)-methyl 2- ((tert-butoxycarbonyl)amino)-3-(pent-4-en-l-yloxy)pentanoate (1.06 g, 3.36 mmol), THF (10 mL) and MeOH (10 mL). To the solution was added aq. LiOH (1.0 M, 5.04 mL). The mixture was stirred for 16 h at room temperature. The mixture was concentrated in vacuo, and the resulting aqueous solution was acidified with aq HCl (1.0 M) and then transferred to a separatory funnel. The solution was twice extracted with EtOAc and the combined organics were dried over MgS0 4 ; filtered; then concentrated in vacuo to afford (2S,3S)-2-((tert-butoxycarbonyl)amino)-3-(pent-4-en- l-yloxy)pentanoic acid as a colorless solid foam (1.00 g, 99 % yield).

Step 2:

To a round-bottom flask equipped with a stir bar was added (2S,3S)-2-((tert- butoxycarbonyl)amino)-3-(pent-4-en-l-yloxy)pentanoic acid (all material from step 1, 1.00 g, 3.32 mmol), (2S,4R)-methyl 4-hydroxypyrrolidine-2-carboxylate-HCl salt (0.663 g, 3.65 mmol), HATU (1.26 g, 3.32 mmol) and CH 2 C1 2 (30 mL). To the mixture was added N,N-diisopropylethylamine (1.75 mL, 9.95 mmol). The mixture was stirred at room temperature for 16 h. The mixture was transferred to a separatory funnel and was thrice washed with aq. HC1 (1M); then brine. The organic solution was dried over MgSC^; filtered; then concentrated in vacuo to afford (2S,4R)-methyl l-((2S,3S)-2-((tert-butoxycarbonyl)amino)-3-(pent-4-en-l-ylo xy)pentanoyl)-4- hydroxypyrrolidine-2-carboxylate as a yellow oil (1.53 g).

Step 3:

To a round-bottom flask equipped with a stir bar was added (2S,4R)-methyl l-((2S,3S)-2-((tert-butoxycarbonyl)amino)-3-(pent-4-en-l-ylo xy)pentanoyl)-4- hydroxypyrrolidine-2-carboxylate (all material from step 2, 1.53 g, 3.57 mmol), THF (10 mL) and MeOH (10 mL). To the solution was added aq. LiOH (1.0 M, 5.36 mL). The mixture was stirred at room temperatuer for 16 h; then was concentrated in vacuo to afford an aqueous solution. The solution was acidified with aq HC1 (1M) and then transferred to a separatory funnel. The solution was twice extracted with EtOAc. The combined organics were dried over MgSC^; filtered; then concentrated in vacuo to afford (2S,4R)- 1 -((2S,3S)-2-((tert-butoxycarbonyl)amino)-3-(pent-4-en- 1 - yloxy)pentanoyl)-4-hydroxypyrrolidine-2-carboxylic acid as a white solid (1.28 g, 86% over two steps ). Step 4:

To a round-bottom flask equipped with a stir bar was added (2S,4R)-l-((2S,3S)-2- ((tert-butoxycarbonyl)amino)-3 -(pent-4-en- 1 -yloxy)pentanoyl)-4- hydroxypyrrolidine-2-carboxylic acid (1/2 of material from step 3, 600 mg, 1.45 mmol), ( 1 R,2S)- 1 -amino-N-(( 1 -methylcyclopropyl)sulfonyl)-2- vinylcyclopropanecarboxamide-trifluoroacetic acid salt (571 mg, 1.59 mmol), HATU

(578 mg, 1.52 mmol) and CH 2 CI 2 (10 mL). To the mixture was added N,N- diisopropylethylamine (1.01 mL, 5.79 mmol). The mixture was stirred at room temperature for 16 h. The mixture was transferred to a separatory funnel and was thrice washed with aq. HC1 (1M); then brine. The organic solution was dried over MgSC^; filtered; then concentrated in vacuo. The resulting residue was subjected to silica gel chromatography (hexanes:acetone, 94:6 to 60:40) to afford tert-butyl ((2S,3 S)- 1 -((2S,4R)-4-hydroxy-2-((( 1 R,2S)- 1 -((( 1 - methylcyclopropyl)sulfonyl)carbamoyl)-2-vinylcyclopropyl)car bamoyl)pyrrolidin-l- yl)-l-oxo-3-(pent-4-en-l-yloxy)pentan-2-yl)carbamate as a colorless solid foam (480 mg, 52%). MS: MS m/z 641.5 (M + +l).

Step 5:

To a round-bottom flask equipped with a stir bar was added tert-butyl ((2S,3S)-1- ((2S,4R)-4-hydroxy-2-((( 1 R,2S)- 1 -((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-2- vinylcyclopropyl)carbamoyl)pyrrolidin- 1 -yl)- 1 -oxo-3-(pent-4-en- 1 -yloxy)pentan-2- yl)carbamate (all material from step 4, 480 mg, 0.749 mmol) and DCE (100 mL). The solution was sparged with nitrogen for 30 minutes. To the solution was added Hoveyda-Grubbs Catalyst 2nd Generation (23 mg, 0.037 mmol). The solution was stirred at 80 °C for 2 h, then was cooled to 45 °C and was stirred for 3 days. The solution was concentrated in vacuo and the resulting residue was subjected to C 18 chromatography (water with 0.1% TFA:acetonitrile with 0.1% TFA, 60:40 to 0:100) to afford tert-butyl ((2R,6S,7S,13aS,14aR,16aS,Z)-7-ethyl-2-hydroxy-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate as a brown solid foam (100 mg, 22%). MS: MS m/z 613.4 (M + +l).

Preparation of Compound 1001

cheme:

Step 1 :

To a vial equipped with a stir bar was added tert-butyl

((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7-ethyl-2-hydroxy-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate (25 mg, 0.040 mmol), l-chloro-7- fluoro-6-methoxyisoquinoline (9.3 mg, 0.044 mmol) and DMSO (1 mL). The mixture was sonicated for 5 min. To the solution was added potassium tert-butoxide (22 mg, 0.20 mmol). The solution was stirred at room temperature for 2h and then was diluted with aq. HC1 (2N) and transferred to a separatory funnel. The mixture was extracted with EtOAc; washed with brine; dried over MgSC^; then concentrated in vacuo to afford tert-butyl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-((7-fiuoro- 6-methoxyisoquinolin- 1 -yl)oxy)-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrol o[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. Step 2:

To a vial equipped with a stir bar and charged with tert-butyl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-((7-fluoro-6-me thoxyisoquinolin-l- yl)oxy)-9-methyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate (all material from step 1, 0.04 mmol) was added CH 2 CI 2 , then trifluoroacetic acid (1.0 ml, 13 mmol). The solution was stirred for 1 h. The solution was concentrated in vacuo to afford

(2R,6S,7S,9R,13aS,14aR,16aS,Z)-6-amino-7-ethyl-2-((7-fluo ro-6- methoxyisoquinolin- 1 -yl)oxy)-9-methyl-N-(( 1 -methylcyclopropyl)sulfonyl)-5 ,16- dioxo-2,3, 5,6, 7,9, 10, l l,13a,14, 14a, 15,16, 16a-tetradecahydro-lH- cyclopropa[i]pyrrolo [ 1 ,2-e] [ 1 ,5 , 8]oxadiazacyclopentadecine- 14a-carboxamide as the trifluoroacetic acid salt. Step 3:

To a vial equipped with a stir bar and charged with

(2R,6S,7S,9R,13aS,14aR,16aS,Z)-6-amino-7-ethyl-2-((7-fluo ro-6- methoxyisoquinolin- 1 -yl)oxy)-9-methyl-N-(( 1 -methylcyclopropyl)sulfonyl)-5 ,16- dioxo-2,3, 5,6, 7,9, 10, l l,13a,14, 14a, 15,16, 16a-tetradecahydro-lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecine-1 4a-carboxamide- trifluoroacetic acid salt (all material from step 2, 0.04 mmol) was added CH 2 CI 2 (1 mL) and pyridin-2-yl (l,l,l-trifluoro-2-methylpropan-2-yl) carbonate (12 mg, 0.049 mmol). To the solution was added N,N-diisopropylethylamine (0.036 mL, 0.21 mmol). The reaction was stirred for 1 h and then the solvent was removed in vacuo. The resulting residue was subjected to C 18 HPLC purification (water with 0.1%

TFA:MeCN with 0.1% TFA) to afford Compound 1001 as a white solid (14.7 mg, 41 ) yield over 3 steps).

Compound 1001 : 1,1,1 -trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-((7-fiuoro-6-me thoxyisoquinolin-l- yl)oxy)-9-methyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H-NMR (400MHz, CD 3 OD) δ 8.93 (s, IH), 7.94 (d, J=6.0 Hz, IH), 7.76 (d, J=11.5 Hz, IH), 7.38 (d, J=8.3 Hz, IH), 7.29 (d, J=5.5 Hz, IH), 5.90 (br. s., IH), 5.62 (td, J=10.0, 6.5 Hz, IH), 5.06 (t, J=10.2 Hz, IH), 4.64 - 4.54 (m, 2H), 4.35 (d, J=10.0 Hz, IH), 4.12 (dd, J=11.7, 3.6 Hz, IH), 4.02 (s, 3H), 3.93 - 3.86 (m, IH), 3.60 - 3.48 (m, IH), 2.82 (q, J=9.4 Hz, IH), 2.75 - 2.65 (m, IH), 2.65 - 2.55 (m, IH), 2.45 (ddd, J=13.8, 9.7, 4.4 Hz, IH), 1.94 - 1.81 (m, 2H), 1.77 (dd, J=8.7, 5.6 Hz, IH), 1.74 - 1.68 (m, IH), 1.68 - 1.62 (m, IH), 1.58 (dd, J=9.5, 5.5 Hz, 2H), 1.52 (s, 3H), 1.43 (s, 3H), 1.42 - 1.35 (m, 4H), 1.17 (d, J=6.3 Hz, 3H), 1.11 (s, 3H), 0.91 (t, J=7.2 Hz, 3H), 0.89 - 0.85 (m, IH); MS: MS m/z 857.5 (M + +l).

Pre aration of Compound 1002

Compound 1002 was prepared using l-fluoro-3,6-dimethoxyisoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1002: l,l,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((3 ,6-dimethoxyisoquinolin- 1 -yl)oxy)-7-ethyl- 9-methy 1- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H-NMR (400MHz, CD 3 OD) δ 8.95 (s, IH), 7.92 (d, J=9.0 Hz, IH), 7.02 (d, J=2.3 Hz, IH), 6.86 (dd, J=9.0, 2.3 Hz, IH), 6.55 (s, IH), 5.88 (br. s., IH), 5.62 (td, J=10.2, 6.3 Hz, IH), 5.06 (t, J=10.3 Hz, IH), 4.64 - 4.54 (m, 2H), 4.38 (d, J=10.0 Hz, IH), 4.17 (dd, J=l 1.5, 3.8 Hz, IH), 3.98 (s, 3H), 3.95 - 3.90 (m, IH), 3.89 (s, 3H), 3.55 (td, J=6.6, 2.9 Hz, IH), 2.80 (q, J=9.3 Hz, IH), 2.70 (dd, J=13.7, 7.4 Hz, IH), 2.66 - 2.51 (m, IH), 2.45 (ddd, J=13.9, 9.5, 4.5 Hz, IH), 1.93 - 1.80 (m, 2H), 1.77 (dd, J=8.5, 5.5 Hz, IH), 1.75 - 1.62 (m, 2H), 1.57 (dd, J=9.5, 5.8 Hz, IH), 1.54 - 1.48 (m, 4H), 1.46 (s, 3H), 1.45 - 1.35 (m, 4H), 1.17 (d, J=6.3 Hz, 3H), 1.09 (s, 3H), 0.91 (t, J=7.4 Hz, 3H), 0.89 - 0.85 (m, 2H).

Preparation of Compound 1003

Compound 1003 was prepared using l-fluoro-4-methoxyisoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1003 : l , l ,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7-ethyl-2-((4-methoxyisoquinolin- 1 -yl)oxy)-9- methyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10, 1 1 ,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l , 2- e][l ,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H-NMR (400MHz, CD 3 OD) δ 8.95 (s, IH), 8.19 - 8.09 (m, 2H), 7.73 (ddd, J=8.3, 7.0, 1.3 Hz, IH), 7.63 - 7.54 (m, 2H), 5.85 (br. s., IH), 5.62 (td, J=10.0, 6.4 Hz, IH), 5.06 (t, J=10.3 Hz, IH), 4.66 - 4.56 (m, 2H), 4.36 (d, J=10.0 Hz, IH), 4.13 (dd, J=l 1.8, 3.5 Hz, IH), 4.02 (s, 3H), 3.93 - 3.83 (m, IH), 3.55 (td, J=6.7, 3.0 Hz, IH), 2.81 (q, J=9.4 Hz, IH), 2.71 (dd, J=13.6, 7.3 Hz, IH), 2.64 - 2.54 (m, IH), 2.44 (ddd, J=13.9, 9.7, 4.3 Hz, IH), 1.94 - 1.80 (m, 2H), 1.77 (dd, J=8.5, 5.5 Hz, IH), 1.75 - 1.62 (m, 2H), 1.62 - 1.54 (m, 2H), 1.52 (s, 3H), 1.51 - 1.42 (m, 2H), 1.42 - 1.35 (m, 4H), 1.17 (d, J=6.3 Hz, 3H), 0.96 (s, 3H), 0.93 - 0.86 (m, 5H); MS: MS m/z 838.5 (M + +l). Preparation of Compound 1004

Compound 1004 was prepared using l-fluoro-4-methoxyisoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1004: l , l ,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)- 14a-((cyclopropylsulfonyl)carbamoyl)-7-ethyl-2- ((4-methoxyisoquinolin- 1 -yl)oxy)-9-methyl-5 , 16-dioxo-

2,3,5,6,7,9,10, 1 1 ,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l , 2- e][l ,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H-NMR (500MHz, DMSO-d 6 ) δ 8.65 (br. s., 1H), 8.05 (d, J=8.5 Hz, 2H), 7.86 (d, J=9.2 Hz, 1H), 7.79 (t, J=7.5 Hz, 1H), 7.65 - 7.58 (m, 2H), 5.76 (br. s., 1H), 5.50 (d, J=8.5 Hz, 1H), 5.31 (br. s., 1H), 4.40 (t, J=8.2 Hz, 1H), 4.28 (d, J=1 1.0 Hz, 1H), 4.15 (t, J=9.3 Hz, 1H), 3.98 (br. s., 1H), 3.95 (s, 3H), 3.81 (br. s., 1H), 2.89 (s, 1H), 2.82 (br. s., 1H), 2.73 (s, 1H), 2.55 (d, J=2.1 Hz, 1H), 2.31 (br. s., 2H), 1.81 (br. s., 1H), 1.77 (br. s., 1H), 1.65 (br. s., 2H), 1.59 - 1.48 (m, 3H), 1.38 (br. s., 3H), 1.28 (br. s., 1H), 1.23 (br. s., 1H), 1.06 (d, J=5.8 Hz, 3H), 0.99 (br. s., 3H), 0.91 (br. s., 3H), 0.80 (t, J=7.0 Hz, 3H); MS: MS m/z 824.7 (M + +l). Preparation of Compound 1005

Compound 1005 was prepared using l-fluoro-6-methoxyisoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1005: l,l,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S, 13aS, 14aR, 16aS,Z)-7-ethyl-2-((6-methoxyisoquinolin- 1 -yl)oxy)- 14a- ((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1 H-NMR (500MHz, DMSO-d 6 ) δ 8.58 - 8.18 (m, 1H), 8.00 - 7.89 (m, 4H), 7.32 - 7.28 (m, 2H), 7.17 - 7.13 (m, 1H), 5.81 (br. s., 1H), 5.56 - 5.50 (m, 1H), 5.29 (br. s., 1H), 4.45 (t, J=8.2 Hz, 1H), 4.27 (d, J=11.0 Hz, 1H), 4.21 - 4.14 (m, 1H), 4.05 (d, J=7.6 Hz, 1H), 3.89 (s, 3H), 3.72 (d, J=10.1 Hz, 1H), 2.40 (br. s., 1H), 2.24 (br. s., 1H), 2.15 - 2.03 (m, 3H), 1.69 (br. s., 1H), 1.59 (d, J=8.9 Hz, 3H), 1.52 (br. s., 2H), 1.37 (s, 3H), 1.40 (s, 3H), 1.28 (br. s., 2H), 1.18 (d, J=10.1 Hz, 2H), 1.03 (s, 3H), 0.81 (t, J=7.3 Hz, 3H), 0.67 (br. s., 2H); MS: MS m/z 824.6 (M + +l). Preparation of Compound 1006

Compounds 1006 was prepared using l-fluoro-6-methoxyisoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1006: l,l,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7-ethyl-2-((6-methoxyisoquinolin- 1 -yl)oxy)-9- methyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H-NMR (500MHz, DMSO-d 6 ) δ 8.88 (br. s., IH), 7.99 - 7.88 (m, 3H), 7.33 - 7.29 (m, 2H), 7.14 (dd, J=9.0, 2.3 Hz, IH), 5.84 (br. s., IH), 5.56 - 5.49 (m, IH), 5.16 (br. s., IH), 4.44 (t, J=8.4 Hz, IH), 4.30 (d, J=11.6 Hz, IH), 4.16 (t, J=9.6 Hz, IH), 3.99 (d, J=8.2 Hz, IH), 3.89 (s, 4H), 3.85 (d, J=9.2 Hz, IH), 3.47 (d, J=4.0 Hz, 2H), 2.65 - 2.54 (m, 2H), 2.38 (br. s., IH), 2.36 - 2.28 (m, IH), 1.82 (d, J=6.1 Hz, IH), 1.73 - 1.63 (m, IH), 1.57 (d, J=8.2 Hz, 2H), 1.45 (br. s., IH), 1.40 (d, J=4.0 Hz, 6H), 1.34 - 1.22 (m, 3H), 1.09 - 1.04 (m, 6H), 0.81 (t, J=7.2 Hz, 5H); MS: MS m/z 838.7 (M + +l). Preparation of Compound 1007

Compound 1007 Compound 1007 was prepared using l-fluoro-6-methoxyisoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1007: l,l,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)- 14a-((cyclopropylsulfonyl)carbamoyl)-7-ethyl-2- ((6-methoxyisoquinolin- 1 -yl)oxy)-9-methyl-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H-NMR (500MHz, DMSO-d 6 ) δ 11.59 - 10.46 (m, 1H), 8.84 (br. s., 1H), 7.98 (d, J=6.1 Hz, 1H), 7.91 (d, J=9.2 Hz, 1H), 7.69 (d, J=l 1.3 Hz, 1H), 7.55 (d, J=8.2 Hz, 1H), 7.35 (d, J=6.1 Hz, 1H), 5.82

(br. s., 1H), 5.55 - 5.49 (m, 1H), 5.17 (br. s., 1H), 4.44 - 4.38 (m, 1H), 4.30 (d, J=11.6 Hz, 1H), 4.16 (t, J=9.6 Hz, 1H), 3.98 (s, 3H), 3.95 (d, J=7.9 Hz, 2H), 3.85 (d, J=9.8 Hz, 1H), 2.89 (d, J=3.7 Hz, 1H), 2.67 (br. s., 1H), 2.61 - 2.54 (m, 1H), 2.42 (br. s., 1H), 2.33 - 2.27 (m, 1H), 1.82 (d, J=6.4 Hz, 1H), 1.71 (br. s., 1H), 1.62 - 1.50 (m, 4H), 1.43 (s, 3H), 1.42 - 1.35 (m, 1H), 1.35 - 1.27 (m, 2H), 1.14 (s, 3H), 1.07 (d,

J=6.1 Hz, 3H), 0.97 (br. s., 2H), 0.81 (t, J=7.5 Hz, 3H); MS: MS m/z 842.7 (M + +l). Preparation of Compound 1008

Compound 1008 was prepared using l-fluoro-3,6-dimethoxyisoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1008: l,l,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-14a-((cyclopropylsulfonyl )carbamoyl)-2-((3,6- dimethoxyisoquinolin- 1 -yl)oxy)-7-ethyl-9-methyl-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H-NMR (500MHz, DMSO-d 6 ) δ 11.23 (s, 1H), 8.83 (br. s., 1H), 7.91 (d, J=9.2 Hz, 1H), 7.85 (d, J=9.2 Hz, 1H), 7.13 (s, 1H), 6.89 (d, J=9.2 Hz, 1H), 6.61 (s, 1H), 5.77 (br. s., 1H), 5.55 - 5.49 (m, 1H), 5.19 (br. s., 1H), 4.39 (t, J=8.1 Hz, 1H), 4.29 (d, J=11.0 Hz, 1H), 4.17 (t, J=9.5 Hz, 1H), 4.00 (d, J=7.6 Hz, 1H), 3.85 (s, 5H), 3.89 (s, 4H), 2.88 (d, J=8.5 Hz, 1H), 2.62 - 2.55 (m, 1H), 2.44 - 2.28 (m, 2H), 1.83 (br. s., 1H), 1.70 (br. s., 1H), 1.60 - 1.51 (m, 3H), 1.44 (s, 3H), 1.43 - 1.34 (m, 2H), 1.30 (d, J=11.9 Hz, 1H), 1.15 (s, 3H), 1.07 (d, J=6.1 Hz, 3H), 0.96 (br. s., 3H), 0.81 (t, J=7.2 Hz, 3H); MS: MS m/z 854.8 (M + +l). Preparation of Compound 1009

Compound 1009 was prepared using l,7-difluoro-6-methoxyisoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1009: l,l,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S, 13aS, 14aR, 16aS,Z)-7-ethyl-2-((7-fiuoro-6-methoxyisoquinolin- 1 - yl)oxy)- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H-NMR (500MHz, DMSO-d 6 ) δ 7.96 (d, J=5.8 Hz, 1H), 7.92 (d, J=9.2 Hz, 1H), 7.70 (d, J=l 1.3 Hz, 1H), 7.54 (d, J=8.5 Hz, 1H), 7.34 (d, J=5.8 Hz, 1H), 5.79 (br. s., 1H), 5.55 - 5.48 (m, 1H), 5.27 (br. s., 1H), 4.51 - 4.43 (m, 1H), 4.27 (d, J=11.3 Hz, 1H), 4.17 (t, J=9.6 Hz, 1H), 4.03 (d, J=7.9 Hz, 1H), 3.97 (s, 3H), 3.72 (d, J=9.5 Hz, 1H), 3.53 (br. s., 1H), 3.24 (br. s., 1H), 2.37 (br. s., 1H), 2.24 (br. s., 1H), 2.08 (br. s., 1H), 1.69 (br. s., 1H), 1.57 (d, J=6.1 Hz, 3H), 1.50 (d, J=14.6 Hz, 1H), 1.40 (s, 3H), 1.36 (s, 4H), 1.31 (br. s., 1H), 1.24 (d, J=14.0 Hz, 2H), 1.18 (d, J=10.7 Hz, 1H), 1.08 (s, 3H), 0.80 (t, J=7.2 Hz, 3H), 0.67 (br. s., 2H); MS: MS m/z 842.9 (M + +l). Preparation of Compound 1010

Compound 1010 was prepared using l-fluoro-3,6-dimethoxyisoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1010: l,l,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S, 13aS, 14aR, 16aS,Z)-2-((3 ,6-dimethoxyisoquinolin- 1 -yl)oxy)-7-ethyl- 14a- ((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H-NMR (500MHz, DMSO-d 6 ) δ 8.62 (s, 1H), 7.93 (d, J=9.2 Hz, 1H), 7.85 (d, J=8.9 Hz, 1H), 7.13 (d, J=2.1 Hz, 1H), 6.89 (dd, J=9.2, 2.1 Hz, 1H), 6.61 (s, 1H), 5.77 (br. s., 1H), 5.55 (q, J=8.7 Hz, 1H), 5.21 (br. s., 1H), 4.44 (t, J=8.2 Hz, 1H), 4.29 (d, J=11.3 Hz, 1H), 4.18 (t, J=9.8 Hz, 1H), 4.05 (d, J=8.5 Hz, 1H), 3.91 - 3.84 (m, 8H), 3.73 (d, J=9.2 Hz, 1H), 3.24 (br. s., 1H), 2.38 (br. s., 2H), 2.17 (br. s., 1H), 2.07 (br. s., 1H), 1.70 (br. s., 1H), 1.63 - 1.51 (m, 4H), 1.43 (s, 3H), 1.38 (s, 3H), 1.33 (br. s., 2H), 1.23 (br. s., 2H), 1.11 (s, 3H), 0.81 (t, J=7.2 Hz, 3H), 0.74 (br. s., 2H); MS: MS m/z 854.8 (M + +l). Preparation of Compound 1011

Compound 1011 was prepared using l-fluoro-4-methoxyisoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1011 : 1,1,1 -trifluoro-2-methylpropan-2-yl

((2R,6S,7S, 13aS, 14aR, 16aS,Z)-7-ethyl-2-((4-methoxyisoquinolin- 1 -yl)oxy)- 14a- ((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H-NMR (500MHz, DMSO-d 6 ) δ 8.58 (br. s., 1H), 8.04 (d, J=8.2 Hz, 2H), 7.88 (d, J=9.2 Hz, 1H), 7.78 (t, J=7.5 Hz, 1H), 7.68 - 7.59 (m, 2H), 5.76 (br. s., 1H), 5.53 (d, J=9.2 Hz, 1H), 5.21 (br. s., 1H), 4.48 - 4.43 (m, 1H), 4.27 (d, J=l 1.3 Hz, 1H), 4.17 - 4.13 (m, 1H), 4.02 (d, J=8.9 Hz, 1H), 3.97 - 3.92 (m, 3H), 3.71 (d, J=9.5 Hz, 1H), 3.52 (br. s., 1H), 3.23 (br. s., 1H), 2.54 (br. s., 1H), 2.36 (br. s., 1H), 2.07 (br. s., 1H), 1.58 (br. s., 2H), 1.46 (br. s., 2H), 1.37 (br. s., 7H), 1.31 (br. s., 3H), 1.23 (d, J=10.7 Hz, 2H), 0.94 (s, 3H), 0.79 (t, J=7.2 Hz, 4H), 0.72 (br. s., 2H); MS: MS m/z 824.8 (M + +l). Preparation of Compound 1012

Compound 1012 was prepared using l,7-difluoro-6-methoxyisoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1012: l,l,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-((7-fiuoro-6-me thoxyisoquinolin-l- yl)oxy)- 14a-((( 1 -(fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-9-methyl-5 ,16- dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecin-6- yl)carbamate. 1H-NMR (500MHz, DMSO-de) δ 11.66 - 10.74 (m, 1H), 9.12 - 8.58 (m, 1H), 7.98 (d, J=5.8 Hz, 1H), 7.93 (d, J=9.2 Hz, 1H), 7.69 (d, J=l 1.3 Hz, 1H), 7.55 (d, J=8.2 Hz, 1H), 7.36 (d, J=5.8 Hz, 1H), 5.82 (br. s., 1H), 5.51 (d, J=7.3 Hz, 1H), 5.15 (br. s., 1H), 4.87 - 4.68 (m, 1H), 4.61 (d, J=10.4 Hz, 1H), 4.41 (t, J=8.1 Hz, 1H), 4.29 (d, J=11.0 Hz, 1H), 4.16 (t, J=9.6 Hz, 1H), 4.00 - 3.98 (m, 3H), 3.97 - 3.94 (m, 1H), 3.85 (d, J=9.2 Hz, 1H), 2.55 (br. s., 1H), 2.39 (br. s., 1H), 2.31 (t, J=9.5 Hz, 1H), 1.81 (br. s., 1H), 1.70 (br. s., 1H), 1.62 (br. s., 1H), 1.52 (d, J=7.9 Hz, 2H), 1.48 - 1.46 (m, 1H), 1.43 (s, 4H), 1.39 (d, J=8.5 Hz, 1H), 1.31 (d, J=12.2 Hz, 2H), 1.23 (br. s., 2H), 1.15 (s, 3H), 1.07 (d, J=6.1 Hz, 3H), 0.81 (t, J=7.3 Hz, 4H); MS: MS m/z 874.8 (M + +l). Preparation of Compound 1013

Compound 1013 was prepared using l-fluoro-3,6-dimethoxyisoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1013: l,l,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((3 ,6-dimethoxyisoquinolin- 1 -yl)oxy)-7-ethyl- 14a-((( 1 -(fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-9-methyl-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H-NMR (500MHz, DMSO-d 6 ) δ 11.32 (s, 1H), 9.11 - 8.59 (m, 1H), 7.92 (d, J=9.2 Hz, 1H), 7.85 (d, J=9.2 Hz, 1H), 7.14 (s, 1H), 6.89 (d, J=9.2 Hz, 1H), 6.61 (s, 1H), 5.77 (br. s., 1H), 5.51 (d, J=7.6 Hz, 1H), 4.86 - 4.67 (m, 1H), 4.65 - 4.55 (m, 1H), 4.40 (t, J=8.2 Hz, 1H), 4.29 (d, J=11.0 Hz, 1H), 4.20 - 4.14 (m, 1H), 4.01 (d, J=8.5 Hz, 1H), 3.86 (s, 4H), 3.89 (s, 4H), 2.55 (br. s., 1H), 2.34 (d, J=9.2 Hz, 2H), 1.83 (br. s., 1H), 1.69 (br. s., 1H), 1.52 (br. s., 3H), 1.45 (s, 4H), 1.43 - 1.34 (m, 2H), 1.29 (br. s., 2H), 1.23 (br. s., 1H), 1.15 (s, 3H), 1.06 (d, J=5.8 Hz, 4H), 0.82 (t, J=7.3 Hz, 4H); MS: MS m/z 886.9 (M + +l). Preparation of Compound 1014

Compound 1014 was prepared using l-fluoro-4-methoxyisoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1014: l,l,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7-ethyl- 14a-(((l - (fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-2-((4-methoxyi soquinolin- 1 -yl)oxy)- 9-methyl-5,16-dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecin-6- yl)carbamate. 1H-NMR (500MHz, DMSO-de) δ 11.37 (s, 1H), 8.94 (d, J=11.6 Hz, 1H), 8.05 (d, J=8.5 Hz, 2H), 7.89 (d, J=9.5 Hz, 1H), 7.79 (t, J=7.8 Hz, 1H), 7.66 - 7.60 (m, 2H), 5.78 (br. s., 1H), 5.51 (br. s., 1H), 4.87 - 4.67 (m, 1H), 4.52 (br. s., 1H), 4.42 (t, J=7.9 Hz, 1H),

4.30 (d, J=9.5 Hz, 1H), 4.15 (t, J=9.6 Hz, 1H), 3.99 (br. s., 1H), 3.96 (s, 4H), 3.85 (br. s., 1H), 2.55 (br. s., 1H), 2.31 (br. s., 1H), 1.83 (br. s., 1H), 1.68 (br. s., 1H), 1.52 (d, J=8.5 Hz, 3H), 1.49 - 1.44 (m, 2H), 1.39 (s, 4H), 1.37 - 1.34 (m, 1H), 1.30 (br. s., 1H), 1.25 (br. s., 2H), 1.07 (d, J=5.8 Hz, 4H), 1.02 (s, 3H), 0.81 (t, J=7.3 Hz, 4H); MS: MS m/z 856.8 (M + +l). Preparation of Compound 1015

Compound 1015 was prepared using 3-chloro-6-methoxy-2-

(trifluoromethyl)quinoxaline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1015 : l , l ,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R, 13aS,14aR,16aS,Z)-7-ethyl-2-((7-methoxy-3- (trifluoromethyl)quinoxalin-2-yl)oxy)-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10, 1 1 ,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l , 2- e][l ,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H-NMR (500MHz, DMSO-d 6 ) δ 8.88 (br. s., 1H), 7.76 (d, J=9.2 Hz, 1H), 7.57 (d, J=9.5 Hz, 1H), 7.16 (dd, J=9.2, 2.4 Hz, 1H), 7.09 (s, 1H), 5.69 (br. s., 1H), 5.32 - 5.24 (m, 1H), 4.81 (br. s., 1H), 4.15 (t, J=8.4 Hz, 1H), 4.04 (d, J=1 1.6 Hz, 1H), 3.82 - 3.76 (m, 1H), 3.74 - 3.69 (m, 3H), 3.58 (d, J=9.8 Hz, 1H), 3.23 - 3.16 (m, 1H), 2.39 (br. s., 1H), 2.33 (dd, J=13.4, 6.7 Hz, 1H), 2.21 - 2.05 (m, 2H), 1.58 (br. s., 1H), 1.40 (br. s., 1H), 1.31 (d, J=10.1 Hz, 3H), 1.22 (br. s., 1H), 1.14 (s, 3H), 1.12 - 1.05 (m, 2H), 1.02 (s, 5H), 0.98 (br. s., 1H), 0.83 - 0.77 (m, 6H), 0.61 (br. s., 1H), 0.56 (d, J=7.0 Hz, 1H), 0.49 (t, J=7.3 Hz, 3H); MS: MS m/z 907.7 (M + +l). Preparation of Compound 1016

Compound 1016 Compound 1016 was prepared using 3-chloro-6-methoxy-2-

(trifluoromethyl)quinoxaline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1016: 3,3-difluoro-2-methylbutan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-((7-methoxy-3- (trifluoromethyl)quinoxalin-2-yl)oxy)-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H-NMR (500MHz, DMSO-d 6 ) δ 11.16 (br. s., 1H), 9.16 (br. s., 1H), 8.01 (d, J=9.2 Hz, 1H), 7.63 (d, J=9.8 Hz, 1H),

7.41 (dd, J=9.3, 2.6 Hz, 1H), 7.34 (d, J=2.7 Hz, 1H), 5.94 (br. s., 1H), 5.56 - 5.50 (m, 1H), 5.03 (br. s., 1H), 4.44 - 4.38 (m, 1H), 4.31 (d, J=11.6 Hz, 1H), 4.10 - 4.01 (m, 2H), 4.01 - 3.94 (m, 4H), 3.81 (d, J=11.0 Hz, 1H), 2.73 (s, 1H), 2.58 (dd, J=13.7, 6.7 Hz, 1H), 2.40 - 2.27 (m, 1H), 1.84 (br. s., 1H), 1.67 (d, J=7.6 Hz, 1H), 1.62 - 1.56 (m, 2H), 1.51 (t, J=19.7 Hz, 3H), 1.40 (s, 3H), 1.38 - 1.21 (m, 5H), 1.16 (s, 3H), 1.05 (d, J=6.1 Hz, 3H), 0.90 (s, 6H), 0.75 (t, J=7.3 Hz, 3H); MS: MS m/z 903.9 (M + +l). Preparation of Compound 1017

Compound 1017 Compound 1017 was prepared using 6,8-difluoro-9-methoxy-2,3-dihydro-lH- pyrano[2,3-c]isoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1017: l,l,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-((8-fiuoro-9-me thoxy-2,3-dihydro-lH- pyrano[2,3-c]isoquinolin-6-yl)oxy)-9-methyl-14a-(((l- methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H-NMR (500MHz, DMSO-d 6 ) δ 11.08 (s, 1H), 8.89 (br. s., 1H), 7.93 (d, J=9.2 Hz, 1H), 7.59 (d, J=l 1.6 Hz, 1H), 7.17 (d, J=8.2 Hz, 1H), 5.69 (br. s., 1H), 5.55 - 5.49 (m, 1H), 5.13 (br. s., 1H), 4.44 - 4.37 (m, 1H), 4.31 - 4.22 (m, 3H), 4.15 (t, J=9.5 Hz, 1H), 3.99 (s, 3H), 3.95 - 3.90 (m, 1H), 3.85 (d, J=9.2 Hz, 1H), 2.95 - 2.87 (m, 1H), 2.85 (br. s., 2H), 2.61 - 2.53 (m, 1H), 2.41 (br. s., 1H), 2.26 (t, J=9.5 Hz, 1H), 2.07 - 1.98 (m, 2H), 1.82 (br. s., 1H), 1.69 (br. s., 1H), 1.65 - 1.51 (m, 3H), 1.44 (s, 3H), 1.39 (s, 5H), 1.31 (br. s., 1H), 1.26 (br. s., 2H), 1.19 - 1.12 (m, 4H), 1.06 (d, J=6.1 Hz, 3H), 0.81 (t, J=7.3 Hz, 4H); MS: MS m/z 912.8 (M + +l). Preparation of Compound 1018

Compound 1018 was prepared using 4,6-difluoro-2,7-dimethoxyquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1018: l,l,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-((6-fiuoro-2,7- dimethoxyquinolin-4- yl)oxy)-9-methyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H-NMR (500MHz, DMSO-d 6 ) δ 11.09 (br. s., 1H), 9.04 (br. s., 1H), 7.85 (d, J=9.8 Hz, 1H), 7.51 (d, J=11.9 Hz, 1H), 7.27 (d, J=7.6 Hz, 1H), 6.44 (br. s., 1H), 5.52 (br. s., 1H), 5.41 (br. s., 1H), 5.02 (br. s., 1H), 4.38 (br. s., 1H), 4.33 (d, J=10.7 Hz, 2H), 4.13 (br. s., 2H), 3.92 (br. s., 6H), 2.66 (br. s., 1H), 2.56 (br. s., 2H), 2.39 (br. s., 1H), 2.22 (br. s., 1H), 1.82 (br. s., 1H), 1.69 (br. s., 1H), 1.57 (br. s., 1H), 1.51 (br. s., 2H), 1.41 - 1.35 (m, 8H), 1.26 (br. s., 3H), 1.09 - 1.01 (m, 6H), 0.86 (br. s., 2H), 0.78 (br. s., 3H); MS: MS m/z 886.9 (M + +l). Preparation of Compound 1019

Compound 1019 Compound 1019 was prepared using 4-fluoro-2,7-dimethoxyquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1019: l,l,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((2,7-dimethoxyquinolin -4-yl)oxy)-7-ethyl-9- methyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H-NMR (500MHz, DMSO-d 6 ) δ 11.12 (br. s., 1H), 9.08 (br. s., 1H), 7.92 (d, J=9.5 Hz, 1H), 7.79 (d, J=8.9 Hz, 1H), 7.11 (br. s., 1H), 6.91 (d, J=8.2 Hz, 1H), 6.39 (s, 1H), 5.54 (d, J=7.9 Hz, 1H), 5.42

(br. s., 1H), 5.02 (br. s., 1H), 4.36 (d, J=10.7 Hz, 2H), 4.15 (t, J=9.0 Hz, 1H), 3.94 (s, 4H), 3.85 (br. s., 4H), 2.71 (d, J=8.9 Hz, 1H), 2.59 (br. s., 1H), 2.41 (br. s., 1H), 2.23 (br. s., 1H), 1.84 (br. s., 1H), 1.70 (br. s., 1H), 1.59 (br. s., 1H), 1.52 (br. s., 2H), 1.40 (s, 4H), 1.42 (s, 3H), 1.37 - 1.20 (m, 4H), 1.10 (br. s., 3H), 1.06 (d, J=5.5 Hz, 3H), 0.89 (br. s., 2H), 0.80 (t, J=6.9 Hz, 3H); MS: MS m/z 868.5 (M + +l). Preparation of Compound 1020

Compound 1020 was prepared using 4-fluoro-2,7-dimethoxyquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compound 1020: 3,3-difluoro-2-methylbutan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((2,7-dimethoxyquinolin-4- yl)oxy)-7-ethyl-9- methyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H-NMR (500MHz, DMSO-d 6 ) δ 11.12 (br. s., 1H), 9.05 (br. s., 1H), 7.79 (d, J=9.2 Hz, 1H), 7.72 (d, J=9.2 Hz, 1H), 7.12 (br. s., 1H), 6.91 (d, J=8.9 Hz, 1H), 6.39 (br. s., 1H), 5.53 (br. s., 1H), 5.42 (br. s., 1H), 5.03 (br. s., 1H), 4.38 (br. s., 2H), 4.17 (t, J=9.9 Hz, 1H), 3.94 (s, 4H), 3.86 (s, 5H), 2.73 (br. s., 1H), 2.58 (br. s., 1H), 2.24 (br. s., 1H), 1.86 (br. s., 1H), 1.71 (br. s., 1H), 1.59 (br. s., 1H), 1.56 - 1.47 (m, 4H), 1.46 (br. s., 2H), 1.40 (br. s., 4H), 1.33 (br. s., 3H), 1.23 (br. s., 3H), 1.07 (d, J=5.5 Hz, 3H), 1.03 (br. s., 3H), 0.89 (br. s., 2H), 0.82 (t, J=7.2 Hz, 3H); MS: MS m/z 864.7 (M + +l).

Preparation of intermediate 7-chloro-2,3-dihydro-[l,4]dioxino[2,3-fJisoquinoline

S

Step 1 :

(E)-3-(2,3-dihydrobenzo[b][l,4]dioxin-5-yl)acrylic acid (5 g, 24.25 mmol), diphenylphosphoryl azide (4.96 mL, 23.04 mmol), and Et 3 N (6.76 mL, 48.5 mmol) were dissolved in benzene and stirred for 16 h. The solution was concentrated under vacuum and the residue was purified by silica gel chromatography using 20%

EtOAc/hexanes to give 4.5 g of (E)-3-(2,3-dihydrobenzo[b][l,4]dioxin-5-yl)acryloyl azide as a yellow solid, which was taken into PhCH 2 Ph (50 mL). The resulting solution was slowly heated to 80°C for lh and then to reflux for 3h. After cooling to rt, the solid was collected washing with benzene to give 3.5 g of the desired product 2,3-dihydro-[l,4]dioxino[2,3-fJisoquinolin-7-ol as a solid. 1H NMR (400 MHz, CD 3 OD) δ ppm 4.37 (m, 4 H), 6.83 (d, J=7.09 Hz, 1 H), 7.02 (d, J=8.80 Hz, 1 H), 7.12 (d, J=7.34 Hz, 1 H), 7.79 (d, J=8.80 Hz, 1 H); MS: (M+H) + 204.

Step 2:

A solution of 2,3-dihydro-[l,4]dioxino[2,3-f]isoquinolin-7-ol (5 g, 24.61 mmol) in POCl 3 (50 mL) was refluxed for 14 h. After Concentration, the residue was taken into the mixture of DCM and 4N NaOH solution. The organic phase was collected and dried over sodium sulfate, filtered, then concentrated under vacuum. The crude material was purified by silica gel chromatography using 20% EtOAc/Hexanes as eluent to give 4 g of the desired product 7-chloro-2,3-dihydro-[l,4]dioxino[2,3- fjisoquinoline as a solid. 1H NMR (400 Hz, CDC1 3 ) δ ppm 4.42 (m, 4 H), 7.24 (d, J=9.05 Hz, 1 H), 7.77 (d, J=5.87 Hz, 1 H), 7.84 (d, J=9.05 Hz, 1 H), 8.18 (d, J=5.87 Hz, 1 H); MS: (M+H) + 222. Preparation of intermediate l-chloro-3-(5-isopropoxypyridin-2-yl)-6- methoxyisoquinoline

Step 1 :

To a solution of N,N-diethyl-4-methoxy-2-methylbenzamide (4.43 g, 20 mmol) in THF (100 ml) at -78°C, tert-butyllithium 1.7 M in pentane (17.65 ml, 30.0 mmol) solution was added dropwise. The reaction mixture was stirred for 0.5 h before addition of 5-isopropoxypicolinonitrile (3.41 g, 21.00 mmol) in THF (2 mL). The resulting solution was warmed to rt and stirred for 16 h. The reaction mixture was quenched with water, neutralized with 1 N HC1. The precipitated solid (5 g) was collected and washed with water to give 4 g of the product 3-(5-isopropoxypyridin- 2-yl)-6-methoxyisoquinolin-l-ol as a white solid. MS: MS m/z 311.11 (M + +l).

Step 2:

A solution of 3-(5-isopropoxypyridin-2-yl)-6-methoxyisoquinolin-l-ol (4 g, 12.89 mmol) in POCI 3 (20 mL) was refluxed for 2 hs. After concentration, the residue was taken into the mixture of DCM and 4N NaOH solution. The organic phase was collected and dried over sodium sulfate, filtered, then concentrated under vacuum. The crude material was purified by silica gel chromatography using CH 2 CI 2 as eluent to give 3.4 g of the desired product l-chloro-3-(5-isopropoxypyridin-2-yl)- 6-methoxyisoquinoline as a solid. 1H NMR (400MHz, CHLOROFORM-d) δ ppm 8.49 (s, 1H), 8.39 (d, J=8.8 Hz, 1H), 8.33 (d, J=2.5 Hz, 1H), 8.19 (d, J=9.1 Hz, 1H), 7.29 (dd, J=8.8, 3.0 Hz, 1H), 7.23 (dd, J=6.8, 2.5 Hz, 1H), 7.16 (d, J=2.3 Hz, 1H), 4.65 (spt, J=6.0 Hz, 1H), 3.94 (s, 3H), 1.38 (d, J=6.3 Hz, 6H). Preparation of l-chloro-7-fluoro-4-methoxyisoquinoline

Step 1 :

To a solution of (E)-3-(4-fluorophenyl) acrylic acid (25 g, 150 mmol) in benzene (120 mL) was added triethylamine (30.5 g, 301 mmol) followed by DPPA (41.4 g, 150 mmol) at room temperature. The reaction mass was stirred at the same temperature for 18 h. The solvent was evaporated under reduced pressure and the residue was diluted with water and extracted with dichloromethane. The organic layer was dried over anhydrous sodium sulfate, filtered and evaporated to get crude compound. The crude compound was purified by conventional column

chromatography (silica gel, 60-120 mesh) using 10% ethyl acetate in pet ether as mobile phase to get the desired compound as a white solid (26 g, 90%). 1H NMR (400 MHz, CDCI 3 ): δ ppm 7.73-7.69 (d, J= 16 Hz, 1H), 7.55-7.51 (m, 2H), 7.11- 7.07 (m, 2H), 6.36-6.32 (d, J= 16 Hz, 1H).

Step 2:

To a hot (125 °C) diphenyl ether (25 ml) was added (E)-3-(4-fluorophenyl) acryloyl azide (5g, 26.2 mmol) portion wise. The reaction was heated at 250°C for 4 h. The reaction mass was cooled to room temperature and diluted with pet ether. The precipitated solid was filtered washed with pet ether to get crude compound (2.45 g, 57%). The crude compound was taken to the next step without further purification. 1H NMR (400 MHz, CD 3 OD): δ ppm 7.96-7.93 (m, 1H), 7.76-7.72 (m, 1H), 7.56- 7.51 (m, 1H), 7.18-7.16 (m, 1H), 6.72-6.70 (m, 1H); MS: MS m/z 164.1 (M + +l).

Step 3 :

To a solution of 7-fluoroisoquinolin-l(2H)-one (1 1 g, 67.4 mmol) in methanol was added iodozobenzenediacetate (21.7 g, 67.4 mmol) followed by methane sulphonic acid (7.78 g, 81 mmol) at room temperature. The reaction mass was heated at reflux for 3 h. The solvent was evaporated and the residue was diluted with cold water. The precipitated solid was filtered and washed with water to get crude compound (1 1 g, 84%) as light red color solid. 1H NMR (400 MHz, CD 3 OD): δ ppm 8.06-8.04 (m, 1H), 7.96-7.93 (m, 1H), 7.62-7.54 (m, 2H), 6.74 (s, 1H), 3.89 (s, 3H); MS: MS m/z 194.1 (M + +l).

Step 4:

A solution of 7-fluoro-4-methoxyisoquinolin-l(2H)-one (1 1 g, 56.9 mmol) in POCl 3 (100 ml) was refluxed for 18 h. The solvent was evaporated under reduced pressure and the residue was diluted with cold water. The aqueous solution was basified by solid sodium carbonate and extracted with ethyl acetate. The combine organic layer was dried over anhydrous sodium sulfate, filtered and evaporated under reduced pressure to get crude compound. The crude compound was purified by silica gel chromatography (20% ethyl acetate in pet ether) to get desired compound (2.9 g, 24%) as off-white solid. 1H NMR (400 MHz, CDC1 3 ): δ ppm 8.36-8.32 (m, 1H), 7.93-7.90 (m, 1H), 7.88 (s, 1H), 7.70-7.65 (m, 1H), 4.1 1 (s, 3H); MS: MS m/z 212.1 (M + +l).

Preparation of l ,7-difluoro-4-methoxyisoquinoline To a solution of l-chloro-7-fluoro-4-methoxyisoquinoline (3.7 g, 17.48 mmol) in DMSO was added cesium fluoride (10.26 g, 69.9 mmol) at room temperature. The reaction vessel (pressure tube) was sealed and heated at 145°C for 18 h. The reaction mass was diluted with water and extracted with ethyl acetate. The combined organic layer was dried over anhydrous Na 2 S0 4 and evaporated under reduced pressure to get crude compound. The crude compound was purified by silica gel chromatography to get desired compound (1.7 g, 49%) as white solid. 1H NMR (400 MHz, CD 3 OD): δ ppm 8.20-8.18 (m, 1H), 7.69-7.66 (m, 1H), 7.54-7.47 (m, 1H), 7.46 (s, 1 H), 4.04 (s, 3H); 19 F NMR: δ ppm 109.65 (IF), -78.53 (IF); MS: MS m/z 196.1 (M + +l) -difluoro-5-methoxyisoquinoline

Step 1 :

A solution of 4-fluoro-2-methoxybenzaldehyde (1.0 g, 6.49 mmol) and malonic acid (1.350 g, 12.98 mmol) in pyridine (10 mL) was refluxed for 16 h. After

concentration the residue was taken into water. The solid was filtered, washed with water, then dried to give (E)-3-(4-fluoro-2-methoxyphenyl)acrylic acid (1.22 g, 96%> yield). 1H NMR (400MHz, CHLOROFORM-d) δ ppm 8.03 (d, J=16.1 Hz, 1H), 7.53 (dd, J=8.5, 6.8 Hz, 1H), 6.75 - 6.65 (m, 2H), 6.52 (d, J=16.1 Hz, 1H), 3.92 (s, 3H). Step 2:

(E)-3-(4-fluoro-2-methoxyphenyl)acrylic acid (1.22 g, 6.22 mmol),

diphenylphosphinyl azide (1.273 mL, 5.91 mmol), and Et3N (1.734 mL, 12.44 mmol) were dissolved in benzene and stirred for 16 h. The solution was concentrated under vacuum and the residue was purified by silica gel chromatography using 20% EtOAc/hexanes to give (E)-3-(4-fluoro-2-methoxyphenyl)acryloyl azide (1.0 g, 73% yield).

Step 3:

A mixture of (E)-3-(4-fluoro-2-methoxyphenyl)acryloyl azide (lg, 4.52 mmol) in PhCH 2 Ph (5 mL) was slowly heated to 80 °C for 1 h and then to reflux for 3 h. After cooling to RT, the solid was collected and washed with benzene to give 7- fluoro-5-methoxyisoquinolin-l-ol (383 mg, 46% yield). MS m/z 194.2 (M + +l). Step 4:

A solution of 7-fluoro-5-methoxyisoquinolin-l-ol (383 mg, 1.983 mmol) in POCI 3 (2772 μΐ, 29.7 mmol) was refluxed (90 °C) for 4 h. The reaction mixture was concentrated. The residue was dissolved in DCM and the pH was adjusted to 7 with 4N NaOH. The organic phase was collected and dried over sodium sulfate, filtered, then concentrated under vacuum to give l-chloro-7-fluoro-5-methoxyisoquinoline (399 mg, 95% yield).

Step 5:

To a solution of l-chloro-7-fluoro-5-methoxyisoquinoline (350 mg, 1.654 mmol) in DMSO (3 mL), was added CsF (502 mg, 3.31 mmol) and the mixture was heated to 140 °C for 4 h. The reaction was diluted with EtOAc and washed with water, and brine. The organic phase was collected, dried over sodium sulfate, and concentrated under vacuum to give l,7-difluoro-5-methoxyisoquinoline (340 mg, 105%) yield) which was not purified further. MS m/z 196.1 (M + +l). Preparation of l-fluoro-4-ethoxyisoquinoline

Step 1 :

To a solution of l-chloroisoquinolin-4-ol (1.0 g, 5.5 mmol) in acetonitrile (10 mL) was added K 2 CO 3 (2.3 g, 16.7 mmol) followed by ethyl iodide (0.87 ml, 11.0 mmol) at room temperature. The reaction mixture was stirred at room temperature for overnight. The solvent was evaporated under reduced pressure and the residue was diluted with water and extracted with ethyl acetate. The combined organic layer was dried over anhydrous Na 2 S0 4 and evaporated under reduced pressure to get crude compound. The crude compound was purified by silica gel chromatography to get 1- chloro-4-ethoxyisoquinoline (0.7 g, 62%) as off-white solid. 1H NMR (400 MHz, CD 3 OD): δ ppm 8.26-8.24 (m, 2H), 7.79 (s, 1H), 7.76-7.26 (m, 2H), 4.29-4.24 (q, J= 6.8 Hz, 2H), 1.58-1.54 (t, J= 6.8 Hz, 3H); MS: MS m/z 207.7 (M + +l).

Step 2:

To a solution of l-chloro-4-ethoxyisoquinolin (4.8 g, 23.12 mmol) in DMSO was added cesium fluoride (6.9 g, 46.24 mmol) at room temperature. The reaction vessel (pressure tube) was sealed and heated at 145°C for 18 h. The reaction mass was diluted with water and extracted with ethyl acetate. The organic layer was dried over anhydrous Na 2 S0 4 and evaporated under reduced pressure to get crude compound. The crude compound was purified by silica gel chromatography to get desired compound (2.6 g, 58.8%) as white solid. MS: MS m/z 192.3 (M + +l). Preparation of 6-chloro-2,3-dihydro-lH-pyrano[2,3-c]isoquinoline

Step 1 :

A mixture of methyl 2-(cyanomethyl)benzoate (3.50 g, 20 mmol) and sodium methoxide (10 mL, 25% wt in methanol) in 35 mL MeOH was heated to reflux under nitrogen for 3 h. While still hot, the solution was acidified with IN HCl solution until the green solution turned to yellow color and a lot of white solid precipitated out. After cooling, the precipitated product was collected by filtration, washed with water and dried to yield the desired product 3-methoxyisoquinolin- 1 (2H)-one as a white solid (2.8 g, 80%). MS: MS m/z 176.1 (M + +l).

Step 2:

3-methoxyisoquinolin-l(2H)-one (2.8 g, 16.0 mmol) in POCI3 (10 mL) was heated to reflux for 3 h then evaporated in vacuo. The residue was poured into iced NaHC0 3 solution (50 mL). The product was extracted with EtOAc for 2 times. The organic layer was washed with brine, dried over MgS0 4 , filtered, evaporated. The residue was purified by flash chromatography with 20% then 40% of EtOAc/ hexane to afford 1.36 g (44%) of the desired product l-chloro-3-methoxyisoquinoline as a white solid. 1H NMR (400MHz, CHLOROFORM-d) δ 8.29 - 8.16 (d, J=8.3 Hz,

IH), 7.72 (d, J=8.3 Hz, IH), 7.63 (ddd, J=8.3, 6.8, 1.1 Hz, IH), 7.47 (ddd, J=8.5, 7.0, 1.1 Hz, IH), 6.98 (s, IH), 4.05 (s, 3H); MS: MS m/z 194.0 (M + +l).

Step 3:

Dissolve l-chloro-3-methoxyisoquinoline (850 mg, 4.39 mmol) in DCM (5 mL) and cool down the solution to -78°C. BBr 3 (17.6 mL, 1M, 17.6 mmol) in DCM solution was then added to the reaction. The reaction was warmed up to r.t slowly and run for 5 h. Cool down the reaction mixture to -78°C and quench with IN NaOH until PH=7. The product was extracted with EtOAc. EtOAc layer was washed with brine, dried and concentrated. The crude material l-chloroisoquinolin-3-ol (800 mg, 100%) was used directly for the next step reaction. 1H NMR (400MHz, CHLOROFORM-d) δ 8.23 (d, J=9.5 Hz, 1H), 7.73 (d, J=8.5 Hz, 1H), 7.66 (ddd, J=8.3, 6.8, 1.1 Hz, 1H), 7.47 (ddd, J=8.4, 6.9, 1.3 Hz, 1H), 7.04 (s, 1H).

Step 4:

To a stirring solution of NaH (0.267 g, 6.68 mmol) in DMF (10 mL) at 0°C was added l-chloroisoquinolin-3-ol (0.80 g, 4.45 mmol). The mixture was stirred at 0°C for 10 mins before the addition of allyl bromide (0.65 g, 5.35 mmol) dropwise. The reaction mixture was stirred at r.t for 1 h. The reaction mixture was diluted with ethyl acetate and then quenched with IN HCl solution. The organic layer was washed with brine, dried over MgS0 4 , filtered and evaporated to get the crude material. The material was purified by flash chromatography with 10%-20% of EtOAc/ hexane to afford 3-(allyloxy)-l-chloroisoquinoline (670 mg, 69 % yield) as a solid. MS: MS m/z 220.1 (M + +l).

Step 5:

3-(allyloxy)-l-chloroisoquinoline (670 mg, 3.05 mmol) was dissolved in diglyme (5 mL) and heated to 180°C for 2 h. The reaction was cooled down to rt before adding EtOAc and water. Washed EtOAc layer with water then brine solution. The organic layer was then dried and concentrated. The residue was purified by flash

chromatography with 20% of EtOAc/ hexane to afford 4-allyl-l-chloroisoquinolin-3- ol (370 mg, 55%) as product. MS: MS m/z 220.1 (M + +l).

Step 6:

To a stirred solution of 4-allyl-l-chloroisoquinolin-3-ol (370 mg, 1.68 mmol) in dry THF (5 mL) at room temperature was added 0.5 M in THF solution of 9-BBN (10.11 mL, 5.05 mmol) and the mixture was stirred for overnight. 3N NaOH (5.05 mL,

15.16 mmol) and H 2 0 2 (1.56 mL, 16.84 mmol) were the added to the mixture. The mixture was stirred for 45 min before quenching with saturated NaCl solution. 1 N HCl was then added to the solution to adjust PH<7. The reaction was extracted with EtOAc. The organic layer was washed with brine, dried and concentrated to afford a yellow oil that was chromatographed using 20-40%EtOAc/hexane to obtain 1-chloro- 4-(3-hydroxypropyl)isoquinolin-3-ol (300 mg , 75%) as product. MS: MS m/z 238.0 (M + +l).

Step 7:

To a solution of triphenylphosphine (596 mg, 2.27 mmol) and l-chloro-4-(3- hydroxypropyl)isoquinolin-3-ol (300 mg, 1.26 mmol) in THF (5 mL) at 0°C was added diisoprpyl azodicarboxylate (0.50 mL, 2.52 mmol) dropwise. The resulting solution was stirred for 4 h at r.t. After concentration of solvent, the residue was purified by Biotage eluting with 0%-20% ethyl acetate in hexane to give the desired product 6-chloro-2,3-dihydro-lH-pyrano[2,3-c]isoquinoline (280 mg, 100%) as a white solid. 1H NMR (400MHz, CHLOROFORM-d) δ 8.25 (dd, J=17.3, 8.3 Hz, 1H), 7.84 - 7.76 (m, 1H), 7.74 - 7.68 (m, 1H), 7.56 - 7.46 (m, 1H), 4.42 (dd, J=5.6, 4.9 Hz, 2H), 3.05 (q, J=6.4 Hz, 2H), 2.27 - 2.03 (m, 2H); MS: MS m/z 220.0 (M + +l).

Preparation of 6-chloro-9-methoxy-2,3-dihydro-lH-pyrano[2,3-c]isoquinoline

Step 1 :

2-(2-cyano-5-methoxyphenyl)acetic acid (3.8 g, 19.88 mmol), and SOCl 2 (20 mL, 274 mmol) were stirred in dichloromethane (25 mL) at RT. The suspension became a solution over 8 h. The reaction was stirred overnight. The volatile organics were removed under vacuum and the residue was taken up in DCM and filtered. The filtrate was concentrated and then dissolved in 4 N HC1 in dioxane (30 mL) and transferred to a sealed vessel and heated to 60 °C for 3 h. The reaction was cooled and the solid was collected, washed with dioxane, and dried under vacuum to give the product l-chloro-6-methoxyisoquinolin-3-ol(3.3g, 70% yield). 1H NMR (400MHz, CHLOROFORM-d) δ 8.10 (d, J=9.6 Hz, 1H), 7.12 (dd, J=9.3, 2.5 Hz, 1H), 6.95 (s, 2H), 3.98 (s, 3H).

Step 2 - 5:

6-chloro-9-methoxy-2,3-dihydro-lH-pyrano[2,3-c]isoquinoline was then prepared using the similar procedure described for synthesizing 6-chloro-2,3-dihydro-lH- pyrano[2,3-c]isoquinoline. l-chloro-6-methoxyisoquinolin-3-ol was used as starting material in step 2 instead of l-chloroisoquinolin-3-ol. 1H NMR (400MHz,

CHLOROFORM-d) δ 8.16 (d, J=9.0 Hz, 1H), 7.10 (dd, J=9.3, 2.3 Hz, 1H), 6.96 (d, J=2.3 Hz, 1H), 4.46 - 4.29 (m, 2H), 3.98 (s, 3H), 2.97 (t, J=6.5 Hz, 2H), 2.19 (dd, J=5.9, 4.6 Hz, 2H); MS: MS m/z 250.0 (M + +l).

Preparation of intermediate 6-ethoxy-l,7-difluoroisoquinoline

Step 1 :

To a solution of l-chloro-7-fluoro-6-methoxyisoquinoline (1.7 g, 8.03 mmol) in CH 2 CI 2 (5 mL) at room temperature was added BBr 3 (24.10 mL, 24.10 mmol) via syringe. The reaction mixture was stirred at room temperature for 18 h. The reaction mixture was cooled to -78°C and quenched with 1 ml of MeOH. After concentration, the residue was taken into water, the solid was collected washing with water to give 500 mg of the desired product l-chloro-7-fluoroisoquinolin-6-ol. Step 2:

A mixture of l-chloro-7-fluoroisoquinolin-6-ol (1.186 g, 6 mmol), bromoethane (1.308 g, 12.00 mmol), and K 2 C0 3 (2.488 g, 18.00 mmol) in acetone (20 mL) was refluxed for 16 h. After filtration washing with acetone, the filtrate was concentrated and purified by Biotage eluting with 10-20% ethyl acetate in hexane to give 1.1 g of the product l-chloro-6-ethoxy-7-fluoroisoquinoline as a solid. MS: MS m/z 226.03 (M + +l).

Step 3:

To a solution of l-chloro-6-ethoxy-7-fluoroisoquinoline (1.128 g, 5 mmol) in DMSO (10 mL) was added CsF (1.519 g, 10.00 mmol) and heated to 140 °C for 4 hrs. The reaction was diluted with ethylacteate and washed with water, and brine. The organic phase was collected, dried over sodium sulfate, and concentrated under vacuum to give the crude product which was purified by silica gel chromatography using a gradient of 5-25% EtOAc in hexanes. The product fractions were collected and the solvent removed under vacuum to give 1.1 g of the desired product 6-ethoxy-l,7- difluoroisoquinoline. 1H NMR (400MHz, CHLOROFORM-d) δ 8.00 (d, J=5.5 Hz, 1H), 7.77 (dd, J=10.8, 0.5 Hz, 1H), 7.41 (dd, J=5.8, 1.3 Hz, 1H), 7.21 (dd, J=7.8, 1.8 Hz, 1H), 4.28 (q, J=7.0 Hz, 2H), 1.59 (t, J=7.0 Hz, 3H). MS: MS m/z 210.03 (M + +l).

Pre aration of l,l-difluoro-2-methylpropan-2-yl pyridin-2-yl carbonate

Step 1 :

Methylmagnesium bromide (2.53 mL, 7.60 mmol) was added dropwise via syringe to a solution of methyl 2,2-difluoroacetate (0.398 g, 3.62 mmol) in diethyl ether (10 mL) at -20 °C and stirred at this temp for 1 hr before warming up to RT. The reaction was quenched 4N HC1 and extracted with a small portion of Et 2 0. The organic layer was washed with brine, collected, dried over MgS0 4 , filtered and concentrated to give a residue that was used in the next step as it was.

Step 2:

To a suspension of sodium hydride, 60% in mineral oil (0.145 g, 3.62 mmol) in THF (50 mL) was added l,l-difluoro-2-methylpropan-2-ol (0.399 g, 3.62 mmol) in THF (1 mL) from Step 1 at 0°C. After stirring 30 min, to the solution was added with a solution of di(pyridin-2-yl) carbonate (0.783 g, 3.62 mmol) in THF (50 mL) through a cannula. The formed slurry was stirred at 0 °C for 30 min. The slurry was warmed to rt and stirred for 2 h. The reaction was diluted with EtOAc, washed with brine, dried over MgS0 4 , filtered, concentrated to give a residue that was purified by Biotage eluting with 20% EtOAc in hexanes to afford 400 mg of the desired product l,l-difluoro-2-methylpropan-2-yl pyridin-2-yl carbonate as an oil. 1H NMR

(400MHz, CHLOROFORM-d) δ 8.47 - 8.39 (m, 1H), 7.91 - 7.74 (m, 1H), 7.32 - 7.23 (m, 1H), 7.14 (dt, J=8.2, 0.8 Hz, 1H), 6.22 - 5.89 (m, 1H), 1.65 (t, J=1.5 Hz, 6H).

Preparation of Intermediate (2S,3S)-2-((tert-butoxycarbonyl)amino)-3-((R)-hex-5-en-

2-yloxy)butanoic acid

Step 1 :

(2S,3S)-methyl 2-amino-3-hydroxybutanoate, HC1 (2.3 g, 13.56 mmol), trityl chloride (4.16 g, 14.92 mmol) and triethylamine (5.67 mL, 40.7 mmol) were stirred in DCM (50 mL) for 16 h. The reaction was diluted with EtOAc and washed with 10% citric acid solution. The organic layer was collected, dried over sodium sulfate, and concentrated under vacuum. The crude material was purified by silica gel chromatography using a gradient of 20-50% EtOAc in hexanes. The product fractions were collected and the solvent removed under vacuum to give 4.2 g (78%) (2S,3S)-methyl 3-hydroxy-2-(tritylamino)butanoate as a solid. MS: MS m/z 398.0 (M + +Na).

Step 2:

(2S,3S)-methyl 3-hydroxy-2-(tritylamino)butanoate (4.46 g, 11.88 mmol) was dissolved in DCM (29.7 ml) and cooled to 0 °C. Ms-Cl (1.018 ml, 13.07 mmol) was added to the cooled solution followed by the dropwise addition of TEA (2.483 ml, 17.82 mmol). The reaction was allowed to stir for 1 h. The reaction was diluted with DCM and washed with 10% Citric acid solution. The organic layer was collected, dried over sodium sulfate, filtered, and concentrated under vacuum. The crude

(2S,3S)-methyl 3-((methylsulfonyl)oxy)-2-(tritylamino)butanoate 4.8 g was used in the next step without further purification. MS: MS m/z 476.1 (M + +Na).

Step 3:

(2S,3S)-methyl 3-((methylsulfonyl)oxy)-2-(tritylamino)butanoate (4.83 g, 10.65 mmol), and TEA (2.97 ml, 21.30 mmol) were dissolved in THF (7.10 ml) and the mixture was refluxed for 36 h. The reaction was diluted with EtOAc and washed with 10%) citric acid, then saturated sodium bicarbonate solution, followed by brine. The organic layer was collected, dried over sodium sulfate, filtered, and solvent removed under vacuum. The crude material was purified by silica gel chromatography using

20% EtOAc/Hexanes as eluent. The product fractions were collected and the solvent removed under vacuum to give 3.1 g (81%>) (2S,3R)-methyl 3 -methyl- 1- tritylaziridine-2-carboxylate. 1H NMR (400MHz, CHLOROFORM-d) δ 7.58 - 7.46 (m, 6H), 7.35 - 7.17 (m, 9H), 3.72 (s, 3H), 3.00 (qd, J=6.2, 2.3 Hz, 1H), 2.37 (d, J=2.3 Hz, 1H), 0.62 (d, J=6.0 Hz, 3H); MS: MS m/z 380.1 (M + +Na). Step 4:

(2S,3R)-methyl 3-methyl-l-tritylaziridine-2-carboxylate (3.1 g, 8.67 mmol) was stirred in TFA (12 ml, 156 mmol), MeOH (6 mL), and DCM (6 mL) at 0°C for 2 h. The solvent was removed under vacuum and the crude oil was partitioned between Et 2 0 and water. The Et 2 0 layer was discarded and the aquious layer was made alkaline by addition of solid sodium bicarbonate. The solution was then saturated with sodium chloride and extracted with Et 2 0 4 X 50 mL. The combined Et 2 0 layers were dried over sodium sulfate, filtered, and concentrated under vacuum giving caution to the product (2S,3R)-methyl 3-methylaziridine-2-carboxylate being of low molecular weight. The product was carried to the next step without further purification. 1H NMR (400MHz, CHLOROFORM-d) δ 3.78 (s, 3H), 2.36 - 2.29 (m, 2H), 2.10 (br. s, 1H), 1.26 (d, J=5.3 Hz, 3H).

Step 5:

(2S,3R)-methyl 3-methylaziridine-2-carboxylate (1.5 g, 13.03 mmol) and Et 3 N (3.63 mL, 26.1 mmol) were dissolved in DCM (15 mL) and cooled to 0°C. BOC 2 0 (4.54 mL, 19.54 mmol) was added and the reaction was allowed to warm to RT and stirred for 16h. The reaction was diluted with saturated sodium bicarbonate solution and stirred vigorously for 10 min. The organic phase was collected, washed with brine, dried over sodium sulfate, and concentrated under vacuum. The crude material was purified by silica gel chromatography using a gradient of 10-40% EtOAc in hexanes. The product fractions were collected and the solvent removed under vacuum to give 720 mg (25%) (2S,3R)-l-tert-butyl 2-methyl 3-methylaziridine-l,2-dicarboxylate. 1H NMR (400MHz, CHLOROFORM-d) δ 3.77 (s, 3H), 2.87 - 2.81 (m, 1H), 2.80 (d, J=2.5 Hz, 1H), 1.47 (s, 9H), 1.34 (d, J=5.5 Hz, 3H).

Step 6:

(2S,3R)-l-tert-butyl 2-methyl 3-methylaziridine-l,2-dicarboxylate (500 mg, 2.323 mmol) was dissolved in DCM (12 ml) at RT. BF 3 Et 2 (2 drops) was added to the mixture and the reaction was stirred for 16 h. BF 3 Et 2 (another 2 drops) was added to the reaction and it was stirred for an additional 6 h. The reaction was diluted with DCM and washed with saturated sodium bicarbonate solution, followed by brine. The organic layer was collected, dried over sodium sulfate, and concentrated under vacuum. The crude material was purified by silica gel chromatography using a gradient of 10-30% EtOAc in hexanes. The product fractions were collected and dried under vacuum to give 400 mg (55%) (2S,3S)-methyl 2-((tert- butoxycarbonyl)amino)-3-((R)-hex-5-en-2-yloxy)butanoate as an oil. 1H NMR (400MHz, CHLOROFORM-d) δ 5.81 (ddt, J=17.1, 10.3, 6.5 Hz, 1H), 5.24 (d, J=8.5 Hz, 1H), 5.04 (dq, J=17.2, 1.6 Hz, 1H), 4.99 - 4.94 (m, 1H), 4.39 (dd, J=8.5, 4.0 Hz, 1H), 3.89 - 3.81 (m, 1H), 3.76 (s, 3H), 3.53 (sxt, J=6.1 Hz, 1H), 2.20 - 2.00 (m, 2H), 1.65 - 1.54 (m, 1H), 1.50 - 1.39 (m, 10H), 1.19 (d, J=6.5 Hz, 3H), 1.14 (d, J=6.0 Hz, 3H); MS: MS m/z 338.2 (M + +Na).

Step 7:

(2S,3S)-methyl 2-((tert-butoxycarbonyl)amino)-3-((R)-hex-5-en-2-yloxy)butan oate (2000 mg, 6.34 mmol) was dissolved in THF (8 mL), and MeOH (8 mL). Water (8 mL) and LiOH (607 mg, 25.4 mmol) were added to the solution which was then stirred for 16 h. The volatiles were removed under vacumm and the resulting aqueous residue was diluted with water and EtOAc. The aqueous layer acidified with 1 N HC1 and was extracted with ethyl acetate (2 X). The combined organic layesr were washed with brine solution, dried over Na 2 S0 4 , and concentrated to give 1.74 g (91 %>) (2S,3 S)-2-((tert-butoxycarbonyl)amino)-3-((R)-hex-5-en-2-yloxy)bu tanoic acid as an oil. 1H NMR (400MHz, CHLOROFORM-d) δ 5.81 (ddt, J=17.0, 10.3, 6.6 Hz, 1H), 5.26 (d, J=7.3 Hz, 1H), 5.04 (dq, J=17.3, 1.6 Hz, 1H), 4.97 (dq, J=10.2, 1.5 Hz, 1H), 4.40 (br. s., 1H), 3.91 (br. s., 1H), 3.57 (sxt, J=6.3 Hz, 1H), 2.20 - 2.02 (m, 2H), 1.69 - 1.57 (m, 1H), 1.50 - 1.43 (m, 10H), 1.26 (d, J=6.5 Hz, 3H), 1.16 (d, J=6.0 Hz, 3H); MS: MS m/z 347.2 (M + +2Na).

Preparation oftert-butyl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-hydroxy-7,9-dimethyl-

14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrol o[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate

Step 1 :

HATU (2.63 g, 6.93 mmol) was added to a solution of (2S,4R)-methyl 4- hydroxypyrrolidine-2-carboxylate, HCl (1.153 g, 6.35 mmol), (2S,3S)-2-((tert- butoxycarbonyl)amino)-3-((R)-hex-5-en-2-yloxy)butanoic acid (1.74 g, 5.77 mmol), and Et 3 N (3.22 mL, 23.09 mmol) in DCM (30 mL) and was stirred at RT for 16 h. The reaction was diluted with DCM and washed with 1 N HCl (3X), followed by brine. The organic layer was collected, dried over sodium sulfate, and concentrated under vacuum to give (2S,4R)-methyl l-((2S,3S)-2-((tert-butoxycarbonyl)amino)-3- ((R)-hex-5 -en-2-yloxy)butanoyl)-4-hydroxypyrrolidine-2-carboxylate which was used in the next step without further purification. MS: MS m/z 429.2 (M + +l).

Step 2:

(2S,4R)-methyl l-((2S,3S)-2-((tert-butoxycarbonyl)amino)-3-((R)-hex-5-en- 2-yloxy)butanoyl)-4-hydroxypyrrolidine-2-carboxylate (2.5 g, 5.83 mmol) was dissolved in THF (8 mL), and MeOH (8 mL). Water (8 mL) and LiOH (0.559 g, 23.34 mmol) were added to the solution which was then stirred for 16 h. The volitiles were removed under vacumm and the resulting aqueous residue was diluted with water, and extracted with EtOAc. The aqueous layer was acidified with 1 N HCl and extracted with ethyl acetate. Combined organic layers were washed with brine solution, dried over Na 2 S0 4 , concentrated to give 2.29 g (95%) (2S,4R)-l-((2S,3S)-2- ((tert-butoxycarbonyl)amino)-3-((R)-hex-5-en-2-yloxy)butanoy l)-4- hydroxypyrrolidine-2-carboxylic acid. MS: MS m/z 415.2 (M + +l).

Step 3:

HATU (1.761 g, 4.63 mmol) was added to a solution of (2S,4R)-l-((2S,3S)-2- ((tert-butoxycarbonyl)amino)-3-((R)-hex-5-en-2-yloxy)butanoy l)-4- hydroxypyrrolidine-2-carboxylic acid (1.6 g, 3.86 mmol), (lR,2S)-l-amino-N-((l- methylcyclopropyl)sulfonyl)-2-vinylcyclopropanecarboxamide, HCl (1.192 g, 4.25 mmol), and Hunig'sBase (2.70 mL, 15.44 mmol) in DCM (20 mL) and was stirred at RT for overnight. The reaction was diluted with DCM, washed with IN HCl (3X) and then brine. The organic phase was collected and dried over sodium sulfate, anc concentrated under vacuum. The crude material was purified by silica gel chromatography using 20-60% Acetone in hexanes. The product fractions were collected and the solvent removed under vacuum to give 2.0 g (81 >) tert-butyl ((2S,3S)-3-((R)-hex-5-en-2-yloxy)-l-((2S,4R)-4-hydroxy-2-((( lR,2S)-l-(((l- methylcyclopropyl)sulfonyl)carbamoyl)-2-vinylcyclopropyl)car bamoyl)pyrrolidin-l- yl)-l-oxobutan-2-yl)carbamate. MS: MS m/z 641.4 (M + +l).

Step 4:

A solution of tert-butyl ((2S,3S)-3-((R)-hex-5-en-2-yloxy)-l-((2S,4R)-4- hydroxy-2-((( 1 R,2S)- 1 -((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-2- vinylcyclopropyl)carbamoyl)pyrrolidin-l-yl)-l-oxobutan-2-yl) carbamate (2 g, 3.12 mmol) in DCE (390 ml) was sparged with nitrogen for 30 min. and then Hoveyda-

Grubbs catalyst 2nd generation (0.098 g, 0.156 mmol) was added and the reaction heated to 80°C for 2 h. The reaction was concentrated and purified by flash chromatography on the Biotage (20-60% Acetone in hexanes) to give 450 mg (24%) tert-butyl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-hydroxy-7,9-dimethyl- 14a-(((l - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (400MHz, DMSO-d 6 ) δ 11.26 (s, 1H), 9.08 (s, 1H), 7.25 (d, J=9.8 Hz, 1H), 5.62 - 5.48 (m, 1H), 5.19 (d, J=3.3 Hz, 1H), 4.94 (t, J=10.3 Hz, 1H), 4.44 (br. s., 1H), 4.26 (t, J=7.9 Hz, 1H), 4.00 (t, J=9.8 Hz, 1H), 3.88 - 3.65 (m, 2H), 3.61 - 3.40 (m, 2H), 2.76 - 2.64 (m, 1H), 2.11 - 0.82 (m, 30H); MS: MS m/z 613.3 (M + +l).

Preparation of Intermediate tert-butyl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-14a-(((l- (fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-2-hydroxy-7,9- dimethyl-5 , 16-dioxo- 2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrol o[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate

Intermediate tert-butyl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-14a-(((l- (fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-2-hydroxy-7,9- dimethyl-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate was prepared according to the method descibed for the synthesis of intermediate tert-butyl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-hydroxy-7,9-dimethyl-14 a-(((l- methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate except that (lR,2S)-l-amino-N- ((1 -(fluoromethyl)cyclopropyl)sulfonyl)-2-vinylcyclopropanecarb oxamide, HCl was used instead of (lR,2S)-l-amino-N-((l-methylcyclopropyl)sulfonyl)-2- vinylcyclopropanecarboxamide, HCl in step 3.

Step 3:

Modifications: 1.26g (lR,2S)-l-amino-N-((l- (fluoromethyl)cyclopropyl)sulfonyl)-2-vinylcyclopropanecarbo xamide, HCl used, 2.0 g product tert-butyl ((2S,3S)-l-((2S,4R)-2-(((lR,2S)-l-(((l- (fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-2-vinylcyclopr opyl)carbamoyl)-4- hydroxypyrrolidin- 1 -yl)-3 -((R)-hex-5 -en-2-yloxy)- 1 -oxobutan-2-yl)carbamate was obtained (78% yield).; MS: MS m/z 659.4 (M + +l).

Step 4:

Modifications: 1.50 g tert-butyl ((2S,3S)-l-((2S,4R)-2-(((lR,2S)-l-(((l- (fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-2-vinylcyclopr opyl)carbamoyl)-4- hydroxypyrrolidin- l-yl)-3-((R)-hex-5-en-2-yloxy)-l-oxobutan-2-yl)carbamate used, 782 mg tert-butyl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-14a-(((l- (fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-2-hydroxy-7,9- dimethyl-5 , 16-dioxo- 2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[i]pyrrol o[l,2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate was obtained (55% yield). 1H NMR (400MHz, DMSO-d 6 ) δ 11.46 (s, IH), 9.02 (s, IH), 7.24 (d, J=9.8 Hz, IH), 5.63 - 5.47 (m, IH), 5.17 (d, J=3.3 Hz, IH), 4.96 (t, J=10.3 Hz, IH), 4.89 - 4.69 (m, IH), 4.64 - 4.39 (m, 2H), 4.24 (t, J=7.8 Hz, IH), 3.99 (t, J=9.7 Hz, IH), 3.87 - 3.65 (m, 2H), 3.59 - 3.39 (m, 2H), 2.76 - 2.61 (m, IH), 2.48 - 2.33 (m, IH), 2.09 - 1.91 (m, 2H), 1.89 - 1.73 (m, IH), 1.63 - 0.99 (m, 23H); MS: MS m/z 631.3 (M + +l).

Preparation of Compound 2001

Compound 2001 Scheme:

ompoun

Step 1 :

A mixture of (2S,4R)-l-(tert-butoxycarbonyl)-4-hydroxypyrrolidine-2-carbo xylic acid (11.94 g, 51.6 mmol), l-chloro-6-methoxyisoquinoline (10 g, 51.6 mmol), and potassium 2-methylpropan-2-olate (17.37 g, 154.8 mmol) in DMSO (10 mL) where stirred overnight. The reaction was quenched with water, diluted with EtOAc and acidified with IN HC1. The organic phase was collected and dried over sodium sulfate, then concentrated under vacuum to give 15 g (75%) (2S,4R)-l-(tert- butoxycarbonyl)-4-((4-methoxyisoquinolin- 1 -yl)oxy)pyrrolidine-2-carboxylic acid as a solid. MS: MS m/z 389.2 (M + +H).

Step 2:

HATU (295 mg, 0.777 mmol) was added to a solution of (2S,4R)-l-(tert- butoxycarbonyl)-4-((6-methoxyisoquinolin- 1 -yl)oxy)pyrrolidine-2-carboxylic acid (252 mg, 0.648 mmol), (lR,2S)-l-amino-N-((l-methylcyclopropyl)sulfonyl)-2- vinylcyclopropanecarboxamide, HC1 (200 mg, 0.712 mmol), and Et 3 N (0.361 mL, 2.59 mmol) in DCM (5 mL) and was stirred at RT for 16 h. The reaction mixture was concentrated under vacuum and purified by silica gel chromatography using 20%> Acetone in hexanes. The product fractions were collected and the solvent removed under vacuum to give 350 mg (88%>) (2S,4R)-tert-butyl 4-((6-methoxyisoquinolin-l- yl)oxy)-2-((( 1 R,2S)- 1 -((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-2- vinylcyclopropyl)carbamoyl)pyrrolidine-l-carboxylate. MS: MS m/z 389.2 (M + +H). Step 3:

A solution of (2S,4R)-tert-butyl 4-((6-methoxyisoquinolin-l-yl)oxy)-2- ((( 1 R,2S)- 1 -((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-2- vinylcyclopropyl)carbamoyl)pyrrolidine-l-carboxylate (204 mg, 0.332 mmol) and hydrogen chloride, 4 M in dioxane ( 4 mL) was stirred for 2 h. Concentration under vacuum gave 183 mg crude (2S,4R)-4-((6-methoxyisoquinolin-l-yl)oxy)-N- (( 1 R,2S)- 1 -((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-2- vinylcyclopropyl)pyrrolidine-2-carboxamide as the HC1 salt which was used in the next step as is.

Step 4:

HATU (151 mg, 0.398 mmol) was added to a solution of (2S,4R)-4-((6- methoxyisoquinolin- 1 -yl)oxy)-N-(( 1R,2S)- 1 -(((1 - methylcyclopropyl)sulfonyl)carbamoyl)-2-vinylcyclopropyl)pyr rolidine-2- carboxamide, HC1 (183 mg, 0.332 mmol), (2S,3S)-2-((tert-butoxycarbonyl)amino)- 3-((R)-hex-5-en-2-yloxy)butanoic acid (100 mg, 0.332 mmol), and Et 3 N (0.185 mL, 1.327 mmol) in DCM (220 mL) and was stirred at RT for 16 h. The reaction mixture was concentrated under vacuum and purified by silica gel chromatography using 20% Acetone in hexanes. The product fractions were collected and the solvent removed under vacuum to give 231 mg (87%>) tert-butyl ((3S)-3-((R)-hex-5-en-2-yloxy)-l- ((2S,4R)-4-((6-methoxyisoquinolin- 1 -yl)oxy)-2-((( 1 R,2S)- 1 -(((1 - methylcyclopropyl)sulfonyl)carbamoyl)-2-vinylcyclopropyl)car bamoyl)pyrrolidin-l- yl)-l-oxobutan-2-yl)carbamate. MS: MS m/z 798.4 (M + +H). Step 5:

A solution of tert-butyl ((3S)-3-((R)-hex-5-en-2-yloxy)-l-((2S,4R)-4-((6- methoxyisoquinolin- 1 -yl)oxy)-2-((( 1 R,2S)- 1 -((( 1 - methylcyclopropyl)sulfonyl)carbamoyl)-2-vinylcyclopropyl)car bamoyl)pyrrolidin-l- yl)-l-oxobutan-2-yl)carbamate (231 mg, 0.289 mmol) in DCE (40 mL) was sparged with nitrogen for 30 min. and then Hoveyda-Grubbs Catalyst 2nd Generation (9.10 mg, 0.014 mmol) was added and the reaction heated to 80°C for 2 h. The reaction was concentrated and The crude material was purified via preparative LC/MS with the following conditions: Column: Waters X Bridge CI 8, 19 x 200 mm, 5-um particles; Guard Column: Waters X Bridge CI 8, 19 x 10 mm, 5-μιη particles; Mobile Phase A: water with 20-mM ammonium acetate; Mobile Phase B: 95:5

acetonitrile: water with 20-mM ammonium acetate; Gradient: 50-100% B over 13 minutes, then a 4-minute hold at 100% B; Flow: 20 mL/min. Fractions containing the desired product were combined and dried via centrifugal evaporation to give 64 mg (30%) tert-butyl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((6-methoxyisoquinolin- 1 - yl)oxy)-7,9-dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 11.19 (s, 1H), 9.05 (br. s., 1H), 8.02 (d, J=9.2 Hz, 1H), 7.96 (d, J=5.8 Hz, 1H), 7.36 - 7.26 (m, 2H), 7.15 (dd, J=9.2, 2.4 Hz, 1H), 5.84 (br. s., 1H), 5.60 - 5.50 (m, 1H), 5.02 (t, J=10.2 Hz, 1H), 4.43 (t, J=8.2 Hz, 1H), 4.31 (d, J=11.3 Hz, 1H), 4.02 - 3.94 (m, 2H), 3.91 (s, 3H), 3.85 - 3.77 (m, 1H), 3.51 - 3.42 (m, 1H), 2.85 - 2.71 (m, 1H), 2.62 - 2.55 (m, J=7.0 Hz, 1H), 2.34 - 2.23 (m, 1H), 1.89 - 1.78 (m, 1H), 1.66 - 1.58 (m, 1H), 1.57 - 1.23 (m, 9H), 1.18 - 1.03 (m, 16H), 0.96 - 0.85 (m, 2H) ; MS: MS m/z 770.5 (M + +l).

Preparation of Compound 2002

Scheme

Compound 2002

Step 1 :

tert-butyl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((6-methoxyisoquinolin-l - yl)oxy)-7,9-dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate (54 mg, 0.070 mmol) was dissolved in DCM and TFA (1 ml, 12.98 mmol) was added. The reaction was stirred for 1 h. The solvent was removed under vacuum to give 55 mg (100%) crude (2R,6S,7S,9R,13aS,14aR,16aS,Z)-6-amino-2-((6-methoxyisoquino lin-l-yl)oxy)-7,9- dimethyl-N-((l-methylcyclopropyl)sulfonyl)-5,16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecine-14a-carboxamide as the TFA salt which was used in the next step as is. MS: MS m/z 670.3 (M + +l).

Step 2:

A solution of (2R,6S,7S,9R,13aS,14aR,16aS,Z)-6-amino-2-((6- methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl-N-(( 1 -methylcyclopropyl)sulfonyl)- 5,16-dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecine-1 4a-carboxamide, TFA (55 mg, 0.070 mmol) and pyridin-2-yl (l,l,l-trifluoro-2-methylpropan-2-yl) carbonate (20.98 mg, 0.084 mmol) in CH 2 C1 2 (1 mL) was added N-ethyl-N- isopropylpropan-2-amine (0.061 mL, 0.351 mmol). The reaction was stirred for 16 h. The solvent was removed under vacuum and the crude material was purified via preparative LC/MS with the following conditions: Column: Waters X Bridge CI 8, 19 x 200 mm, 5-um particles; Guard Column: Waters X Bridge CI 8, 19 x 10 mm, 5-μιη particles; Mobile Phase A: water with 20-mM ammonium acetate; Mobile Phase B: 95:5 acetonitrile: water with 20-mM ammonium acetate; Gradient: 50-90% B over 12 minutes, then a 5-minute hold at 100% B; Flow: 20 mL/min. Fractions containing the desired product were combined and dried via centrifugal evaporation to give 30 mg (52%) 1,1,1 -trifiuoro-2-methylpropan-2-yl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((6- methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (400MHz,

METHANOLS) δ 8.09 (d, J=9.0 Hz, 1H), 7.92 (d, J=6.0 Hz, 1H), 7.27 (d, J=6.0 Hz, 1H), 7.21 (d, J=2.3 Hz, 1H), 7.12 (dd, J=9.2, 2.4 Hz, 1H), 5.89 (br. s., 1H), 5.70 - 5.57 (m, 1H), 5.16 - 5.02 (m, 1H), 4.68 - 4.49 (m, 2H), 4.18 - 4.10 (m, 2H), 3.95 (s, 3H), 3.93 - 3.83 (m, 1H), 3.62 - 3.52 (m, 1H), 2.89 - 2.58 (m, 3H), 2.52 - 2.39 (m, 1H), 1.97 - 1.85 (m, 1H), 1.78 (dd, J=8.5, 5.5 Hz, 1H), 1.72 - 1.01 (m, 20H), 0.96 - 0.84 (m, 2H) ; MS: MS m/z 824.5 (M + +l).

Preparation of Compound 2003

Compound 2003

Compound 2003 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2002: Compound 2003 : 1,1,1 -trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((4-methoxyisoquinolin-l-y l)oxy)-7,9- dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate; 1H NMR (500MHz, DMSO-d 6 ) δ 11.20 (s, 1H), 9.09 (s, 1H), 8.08 (d, J=3.4 Hz, 1H), 8.07 (d, J=3.7 Hz, 1H), 7.92 (d, J=9.2 Hz, 1H), 7.85 - 7.77 (m, 1H), 7.66 (s, 1H), 7.64 (t, J=8.1 Hz, 1H), 5.80 (br. s., 1H), 5.60 - 5.51 (m, 1H), 5.02 (t, J=10.4 Hz, 1H), 4.48 (t, J=8.4 Hz, 1H), 4.29 (d, J=11.3 Hz, 1H), 4.01 - 3.91 (m, 5H), 3.87 - 3.80 (m, 1H), 3.51 - 3.43 (m, 1H), 2.77 (q, J=9.5 Hz, 1H), 2.61 (dd, J=13.3, 6.6 Hz, 1H), 2.35 - 2.26 (m, 1H), 1.90 - 1.78 (m, 1H), 1.67 - 1.59 (m, 1H), 1.58 - 1.24 (m, 12H), 1.11 - 1.03 (m, 9H), 0.91 (br. s., 2H) ; MS: MS m/z 824.5 (M + +l).

Compound 2004 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2002: Compound 2004 : 1,1,1 -trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((4-ethoxyisoquinolin-l-yl )oxy)-7,9-dimethyl- 14a-((( 1 -methylcy clopropyl)sulfonyl)carbamoy l)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate; 1H NMR (500MHz, DMSO-d 6 ) δ 11.19 (s, 1H), 9.08 (s, 1H), 8.08 (d, J=4.9 Hz, 1H), 8.07 (d, J=4.6 Hz, 1H), 7.92 (d, J=9.2 Hz, 1H), 7.80 (t, J=7.6 Hz, 1H), 7.66 (s, 1H), 7.63 (t, J=8.1 Hz, 1H), 5.79 (br. s., 1H), 5.59 - 5.50 (m, 1H), 5.02 (t, J=10.4 Hz, 1H), 4.53 - 4.44 (m, 1H), 4.29 (d, J=12.2 Hz, 1H), 4.21 (q, J=6.9 Hz, 2H), 4.00 - 3.92 (m, 2H), 3.87 - 3.79 (m, 1H), 3.50 - 3.43 (m, 1H), 2.77 (q, J=9.2 Hz, 1H), 2.60 (dd, J=12.8, 6.7 Hz, 1H), 2.35 - 2.27 (m, 1H), 1.89 - 1.79 (m, 1H), 1.67 - 1.59 (m, 1H), 1.57 - 1.24 (m, 15H), 1.12 1.04 (m, 9H), 0.91 (br. s., 2H) ; MS: MS m/z 824.5 (M + +l).

Preparation of Compound 2005

Step 1 :

To a mixture of tert-butyl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-hydroxy-7,9- dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate (40 mg, 0.065 mmol),l,7-difluoro- 6-methoxyisoquinoline (14.01 mg, 0.072 mmol), and potassium tert-butoxide (36.6 mg, 0.326 mmol) was added DMSO (5 mL) and then sonicated for 15 min. The resulting solution was stirred for 4h. The reaction was quenched with water, acidified with 6 N HC1, extracted with EtOAc, washed with brine, dried over MgSC^, and concentrated to give tert-butyl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((7-fluoro-6- methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate which was used in the next step without further purification. MS: MS m/z 788.6 (M + +l).

Step 2:

tert-butyl ((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-2-((7-fiuoro-6-methoxyisoquinolin- 1 - yl)oxy)-7,9-dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate (70 mg, 0.089 mmol) was dissolved in DCM and TFA (1 ml, 12.98 mmol) was added. The reaction was stirred for 1 h. The solvent was removed under vacuum to give 71 mg (100%)

(2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-6-amino-2-((7-fluoro-6-methoxyisoquinolin- 1 - yl)oxy)-7,9-dimethyl-N-(( 1 -methylcyclopropyl)sulfonyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecine-14a-carboxamide, TFA which was used in the next step as is. MS: MS m/z 788.6 (M + +l).

Step 3:

A solution of (2R,6S,7S,9R,13aS,14aR,16aS,Z)-6-amino-2-((7-fluoro-6- methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl-N-(( 1 -methylcyclopropyl)sulfonyl)- 5,16-dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[i]pyrrolo [ 1 ,2-e] [ 1 ,5 , 8]oxadiazacyclopentadecine- 14a-carboxamide, TFA (35.7 mg, .0445 mmol) and pyridin-2-yl (l,l,l-trifluoro-2-methylpropan-2-yl) carbonate (13.31 mg, 0.053 mmol) in CH 2 CI 2 (1 mL) was added N-ethyl-N- isopropylpropan-2-amine (0.039 mL, 0.223 mmol). The reaction was stirred for 16 h. The solvent was removed under vacuum and the crude material was purified via preparative LC/MS with the following conditions: Column: Waters X Bridge CI 8, 19 x 200 mm, 5-um particles; Guard Column: Waters X Bridge CI 8, 19 x 10 mm, 5-μιη particles; Mobile Phase A: water with 20-mM ammonium acetate; Mobile Phase B: 95:5 acetonitrile: water with 20-mM ammonium acetate; Gradient: 50-100% B over 13 minutes, then a 4-minute hold at 100% B; Flow: 20 mL/min. Fractions containing the desired product were combined and dried via centrifugal evaporation to give 15.4 mg (41%) l,l,l-trifluoro-2-methylpropan-2-yl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2- ((7-fluoro-6-methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 11.17 (br. s, 1H), 9.05 (br. s., 1H), 7.96 (d, J=5.8 Hz, 1H), 7.90 (d, J=8.9 Hz, 1H), 7.71 (d, J=11.3 Hz, 1H), 7.54 (d, J=8.2 Hz, 1H), 7.35 (d, J=5.8 Hz, 1H), 5.81 (br. s., 1H), 5.60 - 5.48 (m, 1H), 5.16 - 4.93 (m, 1H), 4.45 (t, J=8.5 Hz, 1H), 4.25 (d, J=10.4 Hz, 1H), 4.02 - 3.90 (m, 4H), 3.86 - 3.76 (m, 1H), 2.80 - 2.53 (m, 2H), 2.33 - 2.22 (m, 1H), 1.88 - 1.74 (m, 1H), 1.66 - 0.75 (m, 27H); MS: MS m/z 824.5 (M + +l).

Pre aration of Compound 2006

Compound 2006

Compound 2006 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2005 :

Compound 2006: 3,3-difluoro-2-methylbutan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((7-fluoro-6-methoxyisoqui nolin-l-yl)oxy)-7,9- dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 11.16 (br. s., 1H), 9.04 (br. s., 1H), 7.96 (d, J=5.8 Hz, 1H), 7.73 - 7.65 (m, 2H), 7.55 (d, J=8.2 Hz, 1H), 7.35 (d, J=5.8 Hz, 1H), 5.83 (br. s., 1H), 5.59 - 5.47 (m, 1H), 5.02 (br. s., 1H), 4.51 - 4.40 (m, 1H), 4.25 (d, J=11.6 Hz, 1H), 4.03 - 3.89 (m, 5H), 3.79 (dd, J=9.6, 5.6 Hz, 1H), 2.73 (br. s., 1H), 2.63 - 2.53 (m, 1H), 2.36 - 2.22 (m, 1H), 1.88 - 1.76 (m, 1H), 1.66 - 1.19 (m, 17H), 1.07 (dd, J=8.4, 6.3 Hz, 6H), 1.00 (s, 3H), 0.93 - 0.83 (m, 2H); MS: MS m/z 838.8 (M + +l).

Preparation of Compound 2007

Compound 2007

Compound 2007 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2005 :

Compound 2007 : 1,1,1 -trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((7-fluoro-4-methoxyisoqui nolin-l-yl)oxy)-7,9- dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. MS: MS m/z 842.8 (M + +l).

Preparation of Compound 2008

Compound 2008

Compound 2008 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2005 :

Compound 2008: 3,3-difluoro-2-methylbutan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((7-fluoro-4-methoxyisoqui nolin-l-yl)oxy)-7,9- dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 11.18 (br. s., 1H), 9.05 (br. s., 1H), 8.13 (dd, J=9.0, 5.3 Hz, 1H), 7.76 - 7.59 (m, 4H), 5.77 (br. s., 1H), 5.60 - 5.45 (m, 1H), 5.01 (br. s., 1H), 4.52 - 4.42 (m, 1H), 4.26 (d, J=11.6 Hz, 1H), 4.00 - 3.88 (m, 5H), 3.85 - 3.72 (m, 1H), 2.85 - 2.70 (m, 1H), 2.63 - 2.53 (m, 1H), 2.34 - 2.22 (m, 1H), 1.88 - 1.77 (m, 1H), 1.67 - 1.57 (m, 1H), 1.56 - 1.18 (m, 16H), 1.06 (d, J=4.9 Hz, 6H), 0.97 - 0.80 (m, 5H); MS: MS m/z 838.8

(M + +l).

Preparation of Compound 2009

Compound 2009

Compound 2009 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2005 :

Compound 2009 : 1,1,1 -trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2 (2,3-dihydro-[l,4]dioxino[2,3-f]isoquinolin-7- yl)oxy)-7,9-dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. MS: MS m/z 852.7 (M + +l).

Preparation of Compound 2010

Compound 2010 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2005 : Compound 2010: 3 ,3 -difluoro-2-methylbutan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2 (2,3-dihydro-[l,4]dioxino[2,3-ilisoquinolin-7- yl)oxy)-7,9-dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 7.94 (d, J=5.8 Hz, IH), 7.66 (d, J=9.2 Hz, IH), 7.59 (d, J=8.9 Hz, IH), 7.37 (d, J=6.1 Hz, IH), 7.11 (d, J=9.2 Hz, IH), 5.82 (br. s., IH), 5.52 (d, J=6.1 Hz, IH), 5.10 (br. s., IH), 4.49 - 4.32 (m, 5H), 4.21 (d, J=11.6 Hz, IH), 4.03 - 3.88 (m, 2H), 3.83 - 3.74 (m, IH), 2.69 - 2.53 (m, 2H), 2.34 - 2.24 (m, IH), 1.88 - 1.76 (m, IH), 1.65 - 1.17 (m, 17H), 1.06 (d, J=3.4 Hz, 6H), 0.95 (s, 3H), 0.88 - 0.77 (m, 2H); MS: MS m/z 848.7 (M + +l).

Pre aration of Compound 2011

Compound 2011

Compound 2011 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2005 :

Compound 2011 : 1,1,1 -trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((3-(5-isopropoxypyridi n-2-yl)-6- methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 8.40 - 8.30 (m, 2H), 8.23 (s, IH), 7.99 (d, J=9.2 Hz, IH), 7.91 (d, J=8.9 Hz, IH), 7.55 (d, J=9.2 Hz, IH), 7.43 (br. s., IH), 7.12 (d, J=9.2 Hz, IH), 6.00 (br. s., IH), 5.59 - 5.49 (m, IH), 5.09 (br. s., IH), 4.85 - 4.71 (m, IH), 4.47 (t, J=7.9 Hz, IH), 4.31 (d, J=10.4 Hz, IH), 4.07 (d, J=8.9 Hz, IH), 4.00 - 3.93 (m, IH), 3.90 (s, 3H), 3.85 - 3.78 (m, IH), 2.71 - 2.61 (m, IH), 2.45 - 2.32 (m, IH), 1.88 - 1.76 (m, IH), 1.64 - 1.20 (m, 20H), 1.13 - 1.01 (m, 10H), 0.84 (br. s., 2H); MS: MS m/z 959.8 (M + +l).

Pre aration of Compound 2012

Compound 2012

Compound 2012 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2005 :

Compound 2012: 3 ,3 -difluoro-2-methylbutan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((3-(5-isopropoxypyridi n-2-yl)-6- methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 8.36 (d, J=8.9 Hz, IH), 8.33 (br. s, IH), 8.23 (s, IH), 7.99 (d, J=9.2 Hz, IH), 7.70 (d, J=9.5 Hz, IH), 7.56 (d, J=8.2 Hz, IH), 7.44 (s, IH), 7.12 (d, J=8.9 Hz, IH), 6.00 (br. s., IH), 5.59 - 5.46 (m, J=6.7 Hz, IH), 5.10 (br. s., IH), 4.78 (dt, J=12.1, 6.2 Hz, IH), 4.47 (t, J=8.1 Hz, 1H), 4.29 (d, J=11.3 Hz, IH), 4.08 (d, J=7.9 Hz, IH), 4.02 - 3.95 (m, IH), 3.90 (s, 3H), 3.84 - 3.76 (m, IH), 2.71 - 2.61 (m, IH), 2.43 - 2.33 (m, IH), 1.88 - 1.77 (m, 1H), 1.63 - 1.21 (m, 23H), 1.10 - 1.03 (m, 7H), 0.99 (s, 3H), 0.84 (br. s., 2H); MS: MS m/z 955.8 (M + +l).

Pre aration of Compound 2013

Compound 2013

Compound 2013 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2005 :

Compound 2013: 1,1,1 -trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-14a-(((l-

(fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-2-((6-metho xyisoquinolin-l-yl)oxy)- 7,9-dimethyl-5,16-dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecin-6- yl)carbamate. 1H NMR (500MHz, DMSO-de) δ 11.38 (br. s, 1H), 8.95 (br. s, 1H), 7.98 (d, J=9.2 Hz, 1H), 7.93 (d, J=5.5 Hz, 1H), 7.86 (d, J=8.9 Hz, 1H), 7.33 - 7.27 (m, 2H), 7.13 (dd, J=9.3, 2.0 Hz, 1H), 5.80 (br. s., 1H), 5.56 - 5.45 (m, 1H), 5.10 (br. s., 1H), 4.86 - 4.67 (m, 1H), 4.63 - 4.47 (m, 1H), 4.41 (t, J=8.1 Hz, 1H), 4.21 (d, J=10.4 Hz, 1H), 4.01 - 3.91 (m, 2H), 3.88 (s, 3H), 3.83 - 3.77 (m, 1H), 2.59 - 2.53 (m, 1H), 2.45 - 2.34 (m, 1H), 2.34 - 2.25 (m, 1H), 1.85 - 1.74 (m, 1H), 1.65 - 0.96 (m, 22H); MS: MS m/z 842.7 (M + +l). Preparation of Compound 2014

Compound 2014

Compound 2014 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2005 :

Compound 2014: 3 ,3 -difluoro-2-methylbutan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-14a-(((l-

(fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-2-((6-metho xyisoquinolin-l-yl)oxy)- 7,9-dimethyl-5,16-dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecin-6- yl)carbamate. MS: MS m/z 838.7 (M + +l).

Preparation of Compound 2015

Compound 2015

Compound 2015 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2005 : Compound 2015: 1,1,1 -trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-14a-(((l-

(fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-2-((4-metho xyisoquinolin-l-yl)oxy)- 7,9-dimethyl-5,16-dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecin-6- yl)carbamate. MS: MS m/z 842.7 (M + +l).

Preparation of Compound 2016

Compound 2016

Compound 2016 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2005 :

Compound 2016: 1,1 -difluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-14a-(((l-

(fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-2-((4-metho xyisoquinolin-l-yl)oxy)- 7,9-dimethyl-5,16-dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecin-6- yl)carbamate. MS: MS m/z 824.8 (M + +l). Preparation of Compound 2017

Compound 2017 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2005 :

Compound 2017: 1,1,1 -trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((7-fluoro-5-methoxyisoqui nolin-l-yl)oxy)-7,9- dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 11.16 (br. s., IH), 9.06 (br. s, IH), 8.00 (d, J=5.8 Hz, IH), 7.92 (d, J=8.5 Hz, IH), 7.51 (d, J=6.1 Hz, IH), 7.26 (d, J=8.9 Hz, IH), 7.22 (d, J=11.3 Hz, IH), 5.87 - 5.81 (m, IH), 5.58 - 5.48 (m, IH), 5.05 (br. s., IH), 4.46 (t, J=8.4 Hz, IH), 4.30 (d, J=11.3 Hz, IH), 4.00 (s, 3H), 3.98 - 3.90 (m, 2H), 3.86 - 3.78 (m, IH), 2.64 - 2.56 (m, IH), 2.35 - 2.26 (m, IH), 1.86 - 1.75 (m, IH), 1.64 - 1.22 (m, 14H), 1.12 (s, 3H), 1.10 - 1.04 (m, 7H), 0.87 (br. s., 2H); MS: MS m/z 842.7 (M + +l). Preparation of Compound 2018

Compound 201 Compound 2018 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2005 :

Compound 2018: 3 ,3 -difluoro-2-methylbutan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((7-fluoro-5-methoxyisoqui nolin-l-yl)oxy)-7,9- dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 8.94 (br. s., 1H), 8.00 (d, J=6.1 Hz, 1H), 7.69 (d, J=9.2 Hz, 1H), 7.51 (d, J=5.8 Hz, 1H), 7.26 (d, J=9.5 Hz, 1H), 7.22 (d, J=11.0 Hz, 1H), 5.84 (br. s., 1H), 5.61 - 5.45 (m, 1H), 5.08 (br. s., 1H), 4.51 - 4.40 (m, 1H), 4.27 (d, J=11.3 Hz, 1H), 4.06 - 3.90

(m, 5H), 3.85 - 3.74 (m, 1H), 2.65 - 2.54 (m, 1H), 2.36 - 2.26 (m, 1H), 1.88 - 1.74 (m, 1H), 1.65 - 1.20 (m, 17H), 1.11 - 1.03 (m, 7H), 0.98 (s, 3H), 0.84 (br. s., 2H); MS: MS m/z 838.7 (M + +l). Preparation of Compound 2019

Compound 2019 Compound 2019 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2005 :

Compound 2019: 1,1 -difluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((7-fluoro-5-methoxyisoqui nolin-l-yl)oxy)-7,9- dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 8.87 (br. s., 1H), 8.00 (d, J=5.8 Hz, 1H), 7.76 (d, J=8.9 Hz, 1H), 7.51 (d, J=6.1 Hz, 1H), 7.27 (d, J=7.6 Hz, 1H), 7.22 (d, J=11.0 Hz, 1H), 6.00 - 5.70 (m, 2H), 5.58 - 5.47 (m, 1H), 5.12 (br. s., 1H), 4.45 (t, J=8.1 Hz, 1H), 4.33 (d, J=11.3 Hz, 1H), 4.00 (s, 3H), 3.97 - 3.88 (m, 2H), 3.85 - 3.75 (m, 1H), 2.70 - 2.54 (m, 2H), 2.36 - 2.27 (m, 1H), 1.85 - 1.72 (m, J=4.0 Hz, 1H), 1.64 - 1.02 (m, 22H), 0.82 (br. s., 2H); MS: MS m/z 824.6 (M + +l). Preparation of Compound 2020

Compound Compound 2020 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2005 :

Compound 2020 : 1,1,1 -trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((7-fluoro-5-methoxyisoqui nolin-l-yl)oxy)- 14a-((( 1 -(fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-7,9-dimethyl- 5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 11.38 (br. s, 1H), 8.93 (br. s, 1H), 8.00 (d, J=6.1 Hz, 1H), 7.91 (d, J=8.9 Hz, 1H), 7.51 (d, J=5.8 Hz, 1H), 7.26 (d, J=8.2 Hz, 1H), 7.22 (d, J=11.3 Hz, 1H), 5.82 (br. s., 1H), 5.56 - 5.46 (m, 1H), 5.10 (br. s., 1H), 4.87 - 4.69 (m, 1H), 4.65 - 4.48 (m, 1H), 4.42 (t, J=8.2 Hz, 1H), 4.27 (d, J=10.1 Hz, 1H), 4.00 (s, 3H), 3.98 - 3.89 (m, 2H), 3.85 - 3.77 (m, 1H), 2.61 - 2.53 (m, 1H), 2.47 - 2.39 (m, 1H), 2.36 - 2.26 (m, 1H), 1.79 (br. s., 1H), 1.62 - 1.00 (m, 22H); MS: MS m/z 860.7 (M + +l). Preparation of Compound 2021

Compound Compound 2021 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2005 :

Compound 2021 : 3 ,3 -difluoro-2-methylbutan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((7-fluoro-5-methoxyisoqui nolin-l-yl)oxy)- 14a-((( 1 -(fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-7,9-dimethyl- 5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. MS: MS m/z 856.7 (M + +l).

Preparation of Compound 2022

Compound 2022 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2005 : Compound 2022 : 1,1 -difluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((7-fluoro-5-methoxyisoqui nolin-l-yl)oxy)- 14a-((( 1 -(fluoromethyl)cyclopropyl)sulfonyl)carbamoyl)-7,9-dimethyl- 5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. MS: MS m/z 824.6 (M + +l).

Preparation of Compound 2301

Compound 2301 Compound 2301 was prepared using 6-chloro-2,3-dihydro-lH-pyrano[2,3- c]isoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compounds 2301 : 3,3-difluoro-2-methylbutan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((2,3-dihydro-lH-pyrano [2,3-c]isoquinolin-6- yl)oxy)-7-ethyl-9-methyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 ,16- dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecin-6- yl)carbamate. 1H NMR (500MHz, DMSO-de) δ 11.27 (br. s., 1H), 9.06 (br. s., 1H), 7.99 (d, J=7.9 Hz, 1H), 7.79 - 7.64 (m, 3H), 7.39 - 7.25 (m, 1H), 5.73 (br. s., 1H), 5.58 - 5.46 (m, 1H), 5.04 (m, 1H), 4.47 - 4.37 (m, 1H), 4.32 (d, J=11.0 Hz, 1H), 4.27 (d, J=5.2 Hz, 2H), 4.16 (t, J=9.6 Hz, 1H), 3.96 (d, J=7.9 Hz, 1H), 3.84 (m, 1H), 2.93 - 2.81 (m, 2H), 2.79 - 2.70 (m, 1H), 2.64 - 2.40 (m, 5H), 2.27 (t, J=9.8 Hz, 1H), 2.09 - 1.99 (m, 2H), 1.86 (m, 1H), 1.70 (m, 1H), 1.59 (d, J=6.7 Hz, 1H), 1.56 - 1.44 (m, 4H), 1.44 - 1.36 (m, 4H), 1.30 (m, 4H), 1.07 (d, J=6.1 Hz, 4H), 0.94 (m, 4H), 0.88 - 0.71 (m, 4H); MS: MS m/z 860.8 (M + +l).

Preparation of Compound 2302

Compound 2302

Compound 2302 was prepared using 6-chloro-2,3-dihydro-lH-pyrano[2,3- c]isoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001. Compounds 2302 : 1,1,1 -trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((2,3-dihydro-lH-pyrano[2, 3-c]isoquinolin-6- yl)oxy)-7-ethyl-9-methyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 ,16- dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecin-6- yl)carbamate. 1H NMR (500MHz, DMSO-de) δ 11.27 (br. s., IH), 9.06 (br. s., IH), 7.99 (d, J=8.2 Hz, IH), 7.88 (d, J=9.2 Hz, IH), 7.80 - 7.63 (m, 2H), 7.33 (t, J=7.5 Hz, IH), 5.72 (br. s., IH), 5.60 - 5.44 (m, IH), 5.13 (m, IH), 4.42 (t, J=8.2 Hz, IH), 4.32 (d, J=11.0 Hz, IH), 4.29 - 4.20 (m, 2H), 4.14 (t, J=9.8 Hz, IH), 3.95 (d, J=8.5 Hz, IH), 3.84 (d, J=8.5 Hz, 2H), 2.92 - 2.79 (m, 2H), 2.68 - 2.53 (m, 2H), 2.37 (m, IH), 2.28 (t, J=9.5 Hz, IH), 2.03 (d, J=5.5 Hz, 2H), 1.83 (m, IH), 1.68 (m, IH), 1.58 (m, 2H), 1.52 (dd, J=9.0, 5.0 Hz, 2H), 1.40 (m, 6H), 1.36 - 1.21 (m, 3H), 1.06 (m, 3H), 1.00 (s, 3H), 0.86 - 0.66 (m, 5H); MS: MS m/z 864.7 (M + +l). Preparation of Compound 2303

Compound 2303

Compound 2303 was prepared using 6-chloro-2,3-dihydro-lH-pyrano[2,3- c]isoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 2005.

Compounds 2303: 3,3-difluoro-2-methylbutan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((2,3-dihydro-lH-pyrano[2, 3-c]isoquinolin-6- yl)oxy)-7,9-dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 11.27 (br. s., 1H), 9.06 (br. s., 1H), 8.00 (d, J=7.9 Hz, 1H), 7.77 - 7.57 (m, 3H), 7.33 (t, J=7.2 Hz, 1H), 5.72 (br. s., 1H), 5.58 - 5.47 (m, 1H), 5.12 (m, 1H), 4.42 (t, J=8.4 Hz, 1H), 4.31 - 4.16 (m, 3H), 4.01 - 3.87 (m, 2H), 3.84 - 3.71 (m, 1H), 2.93 - 2.78

(m, 2H), 2.61 - 2.49 (m, 4H), 2.46 (m, 1H), 2.28 (t, J=9.6 Hz, 1H), 2.03 (d, J=4.9 Hz, 2H), 1.82 (m, 1H), 1.58 (m, 1H), 1.54 - 1.35 (m, 9H), 1.30 (m, 2H), 1.25 (d, J=10.7 Hz, 2H), 1.06 (d, J=5.8 Hz, 3H), 1.06 (d, J=5.2 Hz, 3H), 0.94 (s, 3H), 0.82 (m, 2H); MS: MS m/z 846.8 (M + +l). Preparation of Compound 2304

Compound 2304

Compound 2304 was prepared using 6-chloro-2,3-dihydro-lH-pyrano[2,3- c]isoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 2005.

Compounds 2304: l,l-difluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((2,3-dihydro-lH-pyrano[2, 3-c]isoquinolin-6- yl)oxy)-7,9-dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 11.27 (br. s., 1H), 9.06 (br. s., 1H), 8.01 (d, J=8.2 Hz, 1H), 7.80 - 7.65 (m, 3H), 7.34 (t, J=7.3 Hz, 1H), 5.68 (m, 2H), 5.57 - 5.46 (m, 1H), 5.14 (br. s., 1H), 4.47 - 4.36 (m, 1H), 4.35 - 4.21 (m, 3H), 4.01 - 3.87 (m, 2H), 3.79 (dd, J=9.6, 6.0 Hz, 1H), 2.92 - 2.78 (m, 2H), 2.55 (m, 3H), 2.33 - 2.25 (m, 2H), 2.09 - 2.00 (m, 2H), 1.57 (m, 1H), 1.55 - 1.47 (m, 2H), 1.39 (s, 3H), 1.31 (d, J=12.8 Hz, 2H), 1.23 (m, 2H), 1.12 - 1.02 (m, 12H), 0.80 (m, 2H); MS: MS m/z 832.8 (M + +l). Preparation of Compound 2305

Compound 2305

Compound 2305 was prepared using 6-chloro-2,3-dihydro-lH-pyrano[2,3- c]isoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 2005.

Compounds 2305: 1,1,1 -trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((2,3-dihydro-lH-pyrano[2, 3-c]isoquinolin-6- yl)oxy)-7,9-dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 11.27 (br. s., 1H), 9.06 (br. s., 1H), 7.76 (d, J=8.2 Hz, 1H), 7.63 (d, J=8.9 Hz, 1H), 7.53 - 7.38 (m, 2H), 7.09 (t, J=7.3 Hz, 1H), 5.46 (br. s., 1H), 5.28 (d, J=7.0 Hz, 1H), 4.84 (m, 1H), 4.18 (t, J=8.1 Hz, 1H), 4.01 (s, 3H), 3.78 - 3.65 (m, 2H), 3.56 (m, 1H), 2.60 (d, J=6.7 Hz, 2H), 2.40-2.19 (m, 4H),2.03 (m, 1H), , 1.78 (d, J=5.2 Hz, 2H), 1.56 (m, 1H), 1.34-0.96 (m, 10H), 0.81-0.78 (m, 10H), 0.59 (m, 3H); MS: MS m/z 850.8 (M + +l). Preparation of Compound 2306

Compound 2306

Compound 2306 was prepared using 6-chloro-9-methoxy-2,3-dihydro-lH- pyrano[2,3-c]isoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 2005.

Compounds 2306: l,l,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((9-methoxy-2,3-dihydro-lH -pyrano[2,3- c]isoquinolin-6-yl)oxy)-7,9-dimethyl-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 11.27 (br. s., 1H), 9.06 (br. s., 1H), 7.90 (dd, J=8.5, 3.7 Hz, 2H), 7.00 (br. s., 1H), 6.92 (d, J=8.9 Hz, 1H), 5.69 (br. s., 1H), 5.52 (d, J=5.8 Hz, 1H), 5.08 (br. s., 1H), 4.45 - 4.35 (m, 1H), 4.24 (d, J=4.6 Hz, 3H), 3.99 - 3.86 (m, 6H), 3.82 (d, J=6.1 Hz, 1H), 2.81 (d, J=5.5 Hz, 2H), 2.67 (m, 1H), 2.55 (s, 3H), 2.26 (t, J=9.9 Hz, 1H), 2.03 (d, J=5.2 Hz, 2H), 1.81 (m, 1H), 1.59 (m, 1H), 1.51 (m, 2H), 1.40 (m, 3H), 1.43 (s, 3H), 1.35 - 1.21 (m, 2H), 1.12 (s, 3H), 1.06 (t, J=6.1 Hz, 6H), 0.85 (m, 2H); MS: MS m/z 880.8 (M + +l). Preparation of Compound 2307

Compound 2307

Compound 2307 was prepared using 6-chloro-9-methoxy-2,3-dihydro-lH- pyrano[2,3-c]isoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compounds 2307: l,l,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-((9-methoxy-2,3 -dihydro-lH- pyrano[2,3-c]isoquinolin-6-yl)oxy)-9-methyl-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (400MHz,

METHANOLS) δ 7.97 (d, J=9.0 Hz, IH), 6.99 (d, J=2.3 Hz, IH), 6.90 (dd, J=9.2, 2.4 Hz, IH), 5.83 - 5.74 (m, IH), 5.61 (td, J=9.9, 6.5 Hz, IH), 5.11 (t, J=10.0 Hz,

IH), 4.68 - 4.50 (m, 2H), 4.39 - 4.28 (m, 3H), 4.08 (dd, J=11.8, 3.5 Hz, IH), 3.93 (m, 4H), 3.54 (td, J=6.4, 3.0 Hz, IH), 2.87 (td, J=6.4, 3.3 Hz, 2H), 2.83 - 2.74 (m, IH), 2.71 - 2.63 (m, 2H), 2.61 - 2.51 (m, IH), 2.41 (ddd, J=13.9, 9.9, 4.3 Hz, 1H), 2.18 - 2.08 (m, 2H), 1.91 - 1.79 (m, 2H), 1.78 - 1.55 (m, 4H), 1.51 (s, 4H), 1.43 (s, 5H), 1.16 (d, J=6.3 Hz, 3H), 1.04 (s, 3H), 0.94 - 0.84 (m, 4H); MS: MS m/z 895.0 (M + +l). Preparation of Compound 2308

Compound 2308

Compound 2308 was prepared using 6-chloro-9-methoxy-2,3-dihydro-lH- pyrano[2,3-c]isoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compounds 2308: 3,3-difluoro-2-methylbutan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-((9-methoxy-2,3-di hydro-lH- pyrano[2,3-c]isoquinolin-6-yl)oxy)-9-methyl-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 11.27 (br. s., IH), 9.06 (br. s., IH), 7.88 (d, J=8.9 Hz, IH), 7.70 (d, J=9.8 Hz, IH), 7.00 (s, IH), 6.94 - 6.87 (m, IH), 5.70 (br. s., IH), 5.57 - 5.49 (m, IH), 5.10 (m, IH), 4.40 (t, J=8.2 Hz, IH), 4.25 (m, 3H), 4.16 (t, J=9.8 Hz, IH), 3.95 (d, J=7.9 Hz, IH), 3.90 (m, 4H), 3.83 (d, J=9.2 Hz, IH), 3.48 (d, J=4.3 Hz, IH), 2.81 (d, J=5.2 Hz, 2H), 2.73 (m, IH), 2.55 (m, IH), 2.43 (m, IH), 2.27 (t, J=9.5 Hz, IH), 2.03 (m, 2H), 1.85 (m, IH), 1.69 (m, IH), 1.58 (m, 2H), 1.55 - 1.43 (m, 4H), 1.40 (m, 3H), 1.33 (m, 3H), 1.27 (m, 2H), 1.07 (d, J=6.1 Hz, 3H), 1.02 (s, 3H), 0.82 (t, J=7.2 Hz, 6H); MS: MS m/z 890.9 (M + +l). Preparation of Compound 2309

Compound 2309

Compound 2309 was prepared using 6-chloro-9-methoxy-2,3-dihydro-lH- pyrano[2,3-c]isoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 2005.

Compounds 2309: 3,3-difluoro-2-methylbutan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((9-methoxy-2,3-dihydro-lH -pyrano[2,3- c]isoquinolin-6-yl)oxy)-7,9-dimethyl-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 11.27 (br. s., 1H), 9.06 (br. s., 1H), 7.90 (d, J=8.9 Hz, 1H), 7.69 (d, J=9.2 Hz, 1H), 7.01 (d, J=1.8 Hz, 1H), 6.92 (d, J=9.2 Hz, 1H), 5.70 (br. s., 1H), 5.52 (d, J=7.3 Hz, 1H), 5.09 (br. s., 1H), 4.40 (t, J=8.4 Hz, 1H), 4.29 - 4.11 (m, 3H), 3.99 - 3.85 (m,

5H), 3.79 (dd, J=9.6, 6.0 Hz, 1H), 3.45 (m, 1H), 2.81 (d, J=6.1 Hz, 2H), 2.55 (s, 1H), 2.27 (t, J=9.5 Hz, 1H), 2.03 (d, J=3.7 Hz, 2H), 1.83 (br. s., 1H), 1.63 - 1.55 (m, 1H), 1.53 - 1.37 (m, 9H), 1.34 (s, 3H), 1.26 (d, J=9.8 Hz, 2H), 1.18 (d, J=l 1.9 Hz, 1H), 1.09 - 0.99 (m, 9H), 0.83 (br. s., 3H); MS: MS m/z 876.9 (M + +l). Preparation of Compound 2310

Compound 2310

Compound 2310 was prepared using 6-chloro-9-methoxy-2,3-dihydro-lH- pyrano[2,3-c]isoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 2005.

Compounds 2310: l,l-difluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((9-methoxy-2,3-dihydro-lH -pyrano[2,3- c]isoquinolin-6-yl)oxy)-7,9-dimethyl-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 11.27 (br. s., 1H), 9.06 (br. s., 1H), 7.90 (d, J=8.9 Hz, 1H), 7.76 (d, J=9.2 Hz, 1H), 7.00 (br. s., 1H), 6.93 (d, J=9.2 Hz, 1H), 6.02-5.66(m, 4H), 5.57 - 5.43 (m, 1H), 5.13 (br. s., 1H), 4.40 (t, J=8.2 Hz, 1H), 4.25 (m, 3H), 3.99 - 3.86 (m, 5H), 3.83 - 3.74 (m, 1H), 3.45 (m, 1H), 2.81 (d, J=6.4 Hz, 2H), 2.56-2.48 (m, 2H), 2.28 (d, J=10.4 Hz, 1H), 2.03 (d, J=5.5 Hz, 2H), 1.80 (m, 1H), 1.57 (m, 1H), 1.55 - 1.46 (m, 2H), 1.39 (s, 3H), 1.35 - 1.19 (m, 4H), 1.15 (m, 2H), 1.12 - 1.03 (m, 8H), 0.81 (m, 2H); MS: MS m/z 862.9 (M + +l). Preparation of Compound 2311

Compound 2311

Compound 2311 was prepared using 6-chloro-9-methoxy-2,3-dihydro-lH- pyrano[2,3-c]isoquinoline and the intermediates described above and by following the general procedure described for the synthesis of Compound 1001.

Compounds 2311 : 1,1 -difluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-((9-methoxy-2,3 -dihydro-lH- pyrano[2,3-c]isoquinolin-6-yl)oxy)-9-methyl-14a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (400MHz,

METHANOLS) δ 7.97 (d, J=9.0 Hz, IH), 6.99 (d, J=2.3 Hz, IH), 6.91 (dd, J=9.0, 2.3 Hz, IH), 5.87-5.55 (m, 2H), 5.10 (m, IH), 4.71 - 4.51 (m, 3H), 4.38 - 4.25 (m,

3H), 4.07 (dd, J=12.0, 3.3 Hz, IH), 3.96 - 3.77 (m, 4H), 3.53 (d, J=2.8 Hz, IH), 2.90 - 2.73 (m, 3H), 2.73 - 2.54 (m, 2H), 2.41 (ddd, J=14.0, 10.1, 4.3 Hz, IH), 2.19 - 2.04 (m, 2H), 1.88 - 1.79 (m, IH), 1.78 - 1.54 (m, 4H), 1.51 (s, 3H), 1.44 - 1.26 (m, 4H), 1.16 (d, J=6.3 Hz, 3H), 1.10 (m, 6H), 0.97 - 0.77 (m, 5H); MS: MS m/z 876.9 (M + +l). Preparation of Compound 2601

Compound 2601

Step 1 :

To a solution of (2S,4S)-4-(allyloxy)pentan-2-ol (2, 13.87 mmol) and Tosyl- CI (5.29 g, 27.7 mmol) in CH 2 C1 2 (20 mL) was added Tosyl-Cl (5.29 g, 27.7 mmol) at 0 °C. The formed was stirred at rt overnight. Quenched with iced water, extracted with EtOAc. The organic layer was washed with 1M HCl, saturated NaHC0 3 , and brine, dried over MgSC^, filtered, evaporated. 2.4 g of the product (2S,4S)-4- (allyloxy)pentan-2-yl 4-methylbenzenesulfonate was obtained as a lightly colored oil. 1H NMR (400MHz, CHLOROFORM-d) δ 7.82 (d, J=8.3 Hz, 2H), 7.38 - 7.33 (m, 2H), 5.94 - 5.82 (m, IH), 5.25 (dq, J=17.1 , 1.7 Hz, IH), 5.15 (dq, J=10.3, 1.4 Hz, IH), 4.93 (dqd, J=9.4, 6.3, 2.9 Hz, IH), 3.95 (ddt, J=12.2, 6.0, 1.3 Hz, IH), 3.63 (ddt, J=12.2, 5.4, 1.5 Hz, IH), 3.46 (dqd, J=9.4, 6.2, 2.9 Hz, IH), 2.46 (s, 3H), 1.78 - 1.69 (m, IH), 1.67 - 1.58 (m, IH), 1.26 (d, J=6.5 Hz, 3H), 1.1 1 (d, J=6.3 Hz, 3H).

Step 2:

To a light yellow solution of (2S,4S)-4-(allyloxy)pentan-2-yl 4- methylbenzenesulfonate (2.4 g, 8.04 mmol) and ethyl 2- ((diphenylmethylene)amino)acetate (2.150 g, 8.04 mmol) in PI1CH 3 (20 mL) was added LiHMDS (9.65 mL, 9.65 mmol) dropwise. It was slightly exothemic. The formed dark brown solution was heated to gentle reflux for overnight. The formed light brown slurry was queched with water, extracted with EtOAc. The organic layer was washed with water and brine, dried over MgS0 4 , filtrated, and concentrated in vacuo to afford 3.17 g of the desired product (with impurities) (3R,5S)-ethyl 5- (allyloxy)-2-((diphenylmethylene)amino)-3-methylhexanoate as a dark red oil. Crude material was taken directly onto next step. Step 3:

A mixture of (3R,5S)-ethyl 5-(allyloxy)-2-((diphenylmethylene)amino)-3- methylhexanoate (3.17 g, 8.06 mmol) in diethyl ether (50 mL) and 1M HC1 (16.11 mL, 16.11 mmol) was stirred at rt for 16h. The reaction was extracted with ether. The aqueous layer was adjusted to pH=8 using sat. sodium bicarbonate addition carefully. It was then extracted with EtOAc, dried over MgS0 4 , filtered and concentrated to obtaina red oil (1.1 g) that will be used in the next step as it is.

Step 4:

A mixture of (3R,5S)-ethyl 5-(allyloxy)-2-amino-3-methylhexanoate (1.1 g, 4.80 mmol), di-tert-butyl dicarbonate (1.256 g, 5.76 mmol) and Et 3 N (1.337 mL, 9.59 mmol) in DCM (10 mL) was stirred at rt for overnight. It was washed with 1M HC1(2X) and then brine. The organic layer was collected, dried over MgS0 4 , filtered and evaporated to give the product (3R,5S)-ethyl 5-(allyloxy)-2-((tert- butoxycarbonyl)amino)-3-methylhexanoate (1.5 g, 4.55 mmol, 95 % yield) as a red oil that will be used in the next step as it is.

Step 5:

NaOH (0.767 g, 19.18 mmol) was added to a solution of (3R,5S)-ethyl 5-(allyloxy)- 2-((tert-butoxycarbonyl)amino)-3-methylhexanoate (1.58 g, 4.80 mmol) in water (10 mL) and MeOH (20 mL) and was stirred at RT for overnight. The reaction was partially evaporated to remove most of the solvent then diluted with 2M HC1 and extracted with EtOAc (2X). The organic layer was washed with brine, collected, dried over MgS0 4 , filtered and evaporated to give the final product (3R,5S)-5- (allyloxy)-2-((tert-butoxycarbonyl)amino)-3-methylhexanoic acid (1.4 g, 4.65 mmol, 97 % yield) as an orange viscous oil. MS: MS m/z 324.15 (M + +23).

Step 6:

HATU (6.36 g, 16.72 mmol) was added to a solution of (2S,4R)-methyl 4- hydroxypyrrolidine-2-carboxylate, HCl (2.78 g, 15.33 mmol), (3R,5S)-5-(allyloxy)- 2-((tert-butoxycarbonyl)amino)-3-methylhexanoic acid (4.2 g, 13.94 mmol) and Hunig'sBase (7.30 mL, 41.8 mmol) in DCM (100 mL) and stirred at r.t. overnight. The reaction was washed with IN HCl (3X) and then brine. The organics were dried with magnesium sulfate, filtered and evaporated to give the crude product. The crude material was purified by flash chromatography on the Biotage (25-75% EtOAc in hexane) to give 5.1 of a mixture of thediastereomers (2S,4R)-methyl l-((3R,5S)-5- (allyloxy)-2-((tert-butoxycarbonyl)amino)-3-methylhexanoyl)- 4-hydroxypyrrolidine- 2-carboxylate. MS: MS m/z 451.15 (M + +23).

Step 7:

To solution of (2S,4R)-methyl l-((3R,5S)-5-(allyloxy)-2-((tert- butoxycarbonyl)amino)-3-methylhexanoyl)-4-hydroxypyrrolidine -2-carboxylate (5.14 g, 12 mmol) in MeOH (10 mL) and THF (10.00 mL) was added the solution of LiOH (1.150 g, 48.0 mmol) in water (10.00 mL). The reaction mixture was stirred at rt for overnight. After removing the volatiles under vacumm, the aqueous layer was neutralised with 1.5 N HCl, and extracted with ethyl acetate. Combined organic layers was washed with brine solution, dried over Na 2 S0 4 , concentrated to give 4.2 g of the product (2S,4R)-l-((3R,5S)-5-(allyloxy)-2-((tert-butoxycarbonyl)amin o)-3- methylhexanoyl)-4-hydroxypyrrolidine-2-carboxylic acid as yellow solid. MS: MS m/z 437.14 (M + +23).

Step 8;

HATU (1.825 g, 4.80 mmol) was added to a solution of (2S,4R)-l-((3R,5S)-5- (allyloxy)-2-((tert-butoxycarbonyl)amino)-3 -methylhexanoyl)-4-hydroxypyrrolidine-

2-carboxylic acid (1.658 g, 4 mmol), (lR,2S)-l-amino-N-((l- methylcyclopropyl)sulfonyl)-2-vinylcyclopropanecarboxamide, HCl (1.348 g, 4.80 mmol), and Hunig'sBase (2.79 ml, 16.00 mmol) in DCM (20 ml) and was stirred at RT for overnight. The reaction was washed with IN HC1 (3X) and then brine and evaporated on rotovap to get the product tert-butyl ((3R,5S)-5-(allyloxy)-l-((2S,4R)- 4-hydroxy-2-((( 1 R,2S)- 1 -((( 1 -methylcyclopropyl)sulfonyl)carbamoy l)-2- vinylcyclopropyl)carbamoyl)pyrrolidin- 1 -yl)-3 -methyl- 1 -oxohexan-2-yl)carbamate (1.5 g, 2.341 mmol, 58.5 % yield) as a light orange foam. MS: MS m/z 663.24 (M + +23).

Step 9:

A solution of tert-butyl ((3R,5S)-5-(allyloxy)-l-((2S,4R)-4-hydroxy-2-(((lR,2S)-l- (((1 -methylcyclopropyl)sulfonyl)carbamoyl)-2- vinylcyclopropyl)carbamoyl)pyrrolidin- 1 -yl)-3 -methyl- 1 -oxohexan-2-yl)carbamate (32.0 mg, 0.05 mmol) in DCE (10 mL) was sparged with nitrogen for 30 min. and then Hoveyda-Grubbs Catalyst 2nd Generation (1.886 mg, 3.00 μιηοΐ) was added and the reaction heated to 80°C for 2 hrs then cooled down to 45°C and stirred at this temp for over the weekend. The reaction was concentrated and purified by flash chromatography on the Biotage (20-60% Acetone in hexanes) to give the final product contaminated with ruthenium catalyst as a light brown crispy foam that was purified by prep HPLC to give 12 mg of the desired product tert-butyl

((2R,6S,7R,9S,13aS,14aR,16aS,Z)-2-hydroxy-7,9-dimethyl-14 a-(((l- methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,8,9,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[e]pyrrol o[2,l- i][l,7,10]oxadiazacyclopentadecin-6-yl)carbamate as a sloid. MS: MS m/z 613.34 (M + +l). Step 10:

To a mixture of tert-butyl ((2R,6S,7R,9S,13aS,14aR,16aS,Z)-2-hydroxy-7,9- dimethyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,8,9,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[e]pyrrol o[2,l- i][l,7,10]oxadiazacyclopentadecin-6-yl)carbamate (30.6 mg, 0.05 mmol),l-fluoro-4- methoxyisoquinoline (10.63 mg, 0.060 mmol), and tert-BuOK(28.1 mg, 0.250 mmol) was added DMSO (5 mL) and then sonicated for 15 min. The resulting solution was stirred for 4h. The reaction was quenched with water, acidified with 6 N HC1, extracted with EtOAc, washed with brine, dried over MgS0 4 . After concentration, the residue was purified by prep HPLC to give 10.3 mg of compound 2601.

Compound 2601 : tert-butyl ((2R,6S,7R,9S,13aS,14aR,16aS,Z)-2-((4- methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,8,9,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[e]pyrrolo[2, 1- i][l,7,10]oxadiazacyclopentadecin-6-yl)carbamate. MS: MS m/z 770.5 (M + +l).

Pr aration of Compound 2602

Compound 2602

Compound was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2001 :

Compound 2602: tert-butyl ((2R,6S,7R,9S,13aS,14aR,16aS,Z)-2-((6- methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,8,9,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[e]pyrrolo[2, 1- i][l,7,10]oxadiazacyclopentadecin-6-yl)carbamate. MS: MS m/z 770.6 (M + +l).

Preparation of Compound 2603

Compound 2603

Step 1 :

HATU (1.506 g, 3.96 mmol) was added to a solution of (2S,4R)-l-((3R,5S)-5- (allyloxy)-2-((tert-butoxycarbonyl)amino)-3-methylhexanoyl)- 4-hydroxypyrrolidine- 2-carboxylic acid (1.368 g, 3.3 mmol), (lR,2S)-l-amino-N-(cyclopropylsulfonyl)-2- vinylcyclopropanecarboxamide, p-toluenesulfonate salt (1.457 g, 3.63 mmol), and Hunig'sBase (2.305 ml, 13.20 mmol) in DCM (20 ml) and was stirred at RT for overnight. The reaction was washed with IN HC1 (3X) and then brine and evaporated on rotovap to get the final product tert-butyl ((3R,5S)-5-(allyloxy)-l- ((2S,4R)-2-(((lR,2S)-l-((cyclopropylsulfonyl)carbamoyl)-2- vinylcyclopropyl)carbamoyl)-4-hydroxypyrrolidin- 1 -yl)-3 -methyl- 1 -oxohexan-2- yl)carbamate (1.5 g, 2.393 mmol, 72.5 % yield) as a light orange foam. MS: MS m/z 649.2 (M + +23).

Step 2:

A solution of tert-butyl ((2S,3R,5S)-5-(allyloxy)-l-((2S,4R)-2-(((lR,2S)-l- ((cyclopropylsulfonyl)carbamoyl)-2-vinylcyclopropyl)carbamoy l)-4- hydroxypyrrolidin-l-yl)-3 -methyl- l-oxohexan-2-yl)carbamate (31.3 mg, 0.05 mmol) in DCE (500 mL) was sparged with nitrogen for 30 min. and then Hoveyda-Grubbs Catalyst 2nd Generation (1.886 mg, 3.00 μιηοΐ) was added and the reaction heated to 80°C for 2 hrs, then cooled down to 45°C and stirred at this temp for over the weekend. The reaction was concentrated and purified by flash chromatography on the Biotage (20-60% Acetone in hexanes) to give the final product contaminated with ruthenium catalyst as a light brown crispy foam that was purified by prep HPLC to give 24.6 mg of the desired product as a solid. MS: MS m/z 599.4 (M + +l).

Step 3 :

To a mixture of tert-butyl ((2R,6S,7R,9S, 13aS,14aR,16aS,Z)-14a- ((cyclopropylsulfonyl)carbamoyl)-2-hydroxy-7,9-dimethyl-5 , 16-dioxo- 2,3,5,6,7,8,9, l l ,13a, 14,14a,15, 16,16a-tetradecahydro-lH-cyclopropa[e]pyrrolo[2,l- i][l ,7,10]oxadiazacyclopentadecin-6-yl)carbamate (29.9 mg, 0.05 mmol),l-chloro-6- methoxyisoquinoline (1 1.62 mg, 0.060 mmol), and tert-BuOK (28.1 mg, 0.250 mmol) was added DMSO (5 mL) and then sonicated for 15 min. The resulting solution was stirred for 4h. The reaction was quenched with water, acidified with 6 N HC1, extracted with EtOAc, washed with brine, dried over MgS0 4 . After concentration, the residue was purified by prep HPLC to give 13.5 mg of the product as a solid..

Compound 2603 : tert-butyl ((2R,6S,7R,9S,13aS, 14aR,16aS,Z)-14a- ((cyclopropylsulfonyl)carbamoyl)-2-((6-methoxyisoquinolin-l- yl)oxy)-7,9-dimethyl- 5, 16-dioxo-2,3,5,6,7,8,9,l l , 13a, 14,14a, 15,16, 16a-tetradecahydro-lH- cyclopropa[e]pyrrolo[2, 1 -i] [ 1 ,7, 10]oxadiazacyclopentadecin-6-yl)carbamate. MS : MS m/z 756.4 (M + +l).

Preparation of Compound 2604

Compound Compound was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 2002:

Compound 2604 : 1,1,1 -trifluoro-2-methylpropan-2-yl

((2R,6S,7R,9S,13aS,14aR,16aS,Z)-14a-((cyclopropylsulfonyl)ca rbamoyl)-2-((6- methoxyisoquinolin- 1 -yl)oxy)-7,9-dimethyl-5 , 16-dioxo-

2,3,5,6,7,8,9,l l,13a,14,14a,15,16,16a-tetradecahydro-lH-cyclopropa[e]pyrrol o[2,l- i][l,7,10]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (400MHz,

METHANOLS) δ 8.08 (d, J=9.0 Hz, 1H), 7.91 (d, J=6.0 Hz, 1H), 7.25 (d, J=5.8 Hz, 1H), 7.19 (d, J=2.3 Hz, 1H), 7.11 (dd, J=9.2, 2.4 Hz, 1H), 5.84 (br. s., 1H), 5.71 - 5.49 (m, 2H), 4.70 - 4.55 (m, 2H), 4.25 (dd, J=11.4, 6.7 Hz, 1H), 4.07 (dd, J=11.5, 3.0 Hz, 1H), 3.96 - 3.87 (m, 5H), 3.50 (q, J=5.0 Hz, 1H), 2.94 - 2.85 (m, 1H), 2.77 - 2.45 (m, 3H), 1.99 (dd, J=12.9, 5.1 Hz, 2H), 1.78 (dd, J=8.4, 5.1 Hz, 1H), 1.62 (dd, J=9.5, 5.0 Hz, 1H), 1.37 (s, 3H), 1.29 - 1.05 (m, 7H), 1.00 - 0.92 (m, 7H). MS: MS m/z 810.4 (M + +l).

Preparation of Compound 2605

Compound 2605

Compounds 2605 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 1001 :

Compound 2605: 3,3-difluoro-2-methylbutan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-((7-fiuoro-4-metho xyisoquinolin-l- yl)oxy)-9-methyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. MS: MS m/z 852.7 (M + +l). Preparation of Compound 2606

Compound 2606

Compounds 2606 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 1001 :

Compound 2606: l,l,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-((7-fiuoro-4-metho xyisoquinolin-l- yl)oxy)-9-methyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 11.16 (br. s., 1H), 9.05 (br. s., 1H), 8.13 (dd, J=9.2, 5.2 Hz, 1H), 7.92 (d, J=8.9 Hz, 1H), 7.75 - 7.61 (m, 3H), 5.79 (br. s., 1H), 5.54 (d, J=7.0 Hz, 1H), 5.05 (br.s., 1H), 4.46 (br. s., 1H), 4.33 (d, J=11.3 Hz, 1H), 4.14 (t, J=9.6 Hz, 1H), 3.97 (s, 4H), 3.86 (d, J=9.2 Hz, 1H), 3.47 (br. s., 1H), 2.74 (s, 1H), 2.58 (br. s., 1H), 2.29 (br. s., 1H), 1.89 - 1.21 (m, 16H), 1.11 - 1.03 (m, 6H), 0.96 - 0.76 (m, 5H). MS: MS m/z 856.7 (M + +l).

Preparation of Compound 2607

Compound 2607

Compounds 2607 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 1001 :

Compound 2607: 3,3-difluoro-2-methylbutan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((6-ethoxy-7-fiuoroisoquin olin-l-yl)oxy)-7- ethyl-9-methyl- 14a-((( 1 -methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 11.16 (br. s., 1H), 9.03 (br. s., 1H), 7.97 (d, J=5.8 Hz, 1H), 7.74 - 7.66 (m, 2H), 7.54 (d, J=8.5 Hz, 1H), 7.34 (d, J=5.8 Hz, 1H), 5.84 (br. s., 1H), 5.59 - 5.48 (m, 1H), 5.03 (br. s., 1H), 4.49 - 4.42 (m, 1H), 4.33 (d, J=l 1.3 Hz, 1H), 4.26 (q, J=6.9 Hz, 2H), 4.17 (t, J=9.5 Hz, 1H), 3.97 (d, J=8.5 Hz, 1H), 3.84 (d, J=9.5 Hz, 1H), 3.48 (br. s., 1H), 2.79 (br. s., 1H), 2.58 (d, J=6.7 Hz, 1H), 2.29 (t, J=9.6 Hz, 1H), 1.86 (br. s., 1H), 1.78 - 1.21 (m, 21H), 1.08 (d, J=6.1 Hz, 3H), 1.01 (s, 3H), 0.89 (br. s., 2H), 0.82 (t, J=7.3 Hz, 3H). MS: MS m/z 866.7 (M + +l). Preparation of Compound 2608

Compound 2608

Compounds 2608 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 1001 :

Compound 2608: l,l,l-trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((6-ethoxy-7-fiuoroisoquin olin-l-yl)oxy)-7- ethyl-9-methyl- 14a-((( 1 -methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 11.15 (br. s., 1H), 9.03 (br. s., 1H), 8.02 - 7.91 (m, 2H), 7.69 (d, J=11.3 Hz, 1H), 7.54 (d, J=8.2 Hz, 1H), 7.34 (d, J=5.8 Hz, 1H), 5.83 (br. s., 1H), 5.59 - 5.50 (m, 1H), 5.03 (br. s., 1H), 4.50 - 4.42 (m, 1H), 4.33 (d, J=l 1.6 Hz, 1H), 4.26 (q, J=6.8 Hz, 2H), 4.16 (t, J=9.6 Hz, 1H), 3.96 (d, J=8.2 Hz, 1H), 3.87 (d, J=8.9 Hz, 1H), 3.47 (br. s., 1H), 2.74 (s, 1H), 2.62 - 2.55 (m, 1H), 2.29 (t, J=9.9 Hz, 1H), 1.89 - 1.21 (m, 19H), 1.15 (s, 3H), 1.07 (d, J=6.4 Hz, 3H), 0.89 (br. s., 2H), 0.82 (t, J=7.3 Hz, 3H). MS: MS m/z 870.7 (M + +l). Preparation of Compound 2609

Compound 2609

Compounds 2609 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 1001 :

Compound 2609: 3,3-difluoro-2-methylbutan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2 (2,3-dihydro-[l,4]dioxino[2,3-f]isoquinolin-7- yl)oxy)-7-ethyl-9-methyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 ,16- dioxo-2,3,5,6,7,9,10,ll,13a,14,14a,15,16,16a-tetradecahydro- lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecin-6- yl)carbamate.1H NMR (500MHz, DMSO-d 6 ) δ 11.17 (br. s., IH), 9.06 (br. s., IH), 7.96 (d, J=6.1 Hz, IH), 7.68 (d,J=9.5 Hz, IH), 7.59 (d,J=9.2 Hz, IH), 7.38 (d,J=6.1 Hz, IH), 7.12 (d,J=9.2 Hz, IH), 5.84 (br. s., IH), 5.61 - 5.47 (m, IH), 5.02 (br. s., IH), 4.48 - 4.27 (m, 6H), 4.21 -4.12 (m, IH), 4.01 - 3.94 (m, IH), 3.83 (d,J=9.2Hz, IH), 3.46 (d, J=6.7 Hz,

IH), 2.83 -2.71 (m, IH), 2.58 (dd, J=13.1, 7.0 Hz, IH), 2.29 (t, J=9.8 Hz, IH), 1.90- 1.21 (m, 19H), 1.07 (d,J=6.1 Hz, 3H), 0.97 - 0.74 (m, 8H). MS: MS m/z 862.6

(M + +l). Preparation of Compound 2610

Compound 2610

Compounds 2610 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 1001 :

Compound 2610: 1,1,1 -trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2 (2,3-dihydro-[l,4]dioxino[2,3-f]isoquinolin-7- yl)oxy)-7-ethyl-9-methyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 ,16- dioxo-2,3,5,6,7,9,10,l l,13a,14,14a,15,16,16a-tetradecahydro-lH- cyclopropa[i]pyrrolo[l,2-e][l,5,8]oxadiazacyclopentadecin-6- yl)carbamate. 1H NMR (500MHz, DMSO-de) δ 11.16 (br. s., IH), 9.07 (br. s., IH), 7.96 (d, J=6.1 Hz, IH), 7.88 (d, J=9.2 Hz, IH), 7.59 (d, J=8.9 Hz, IH), 7.37 (d, J=5.8 Hz, IH), 7.11 (d, J=8.9 Hz, IH), 5.84 (br. s., IH), 5.59 - 5.48 (m, IH), 5.02 (br. s., IH), 4.49 - 4.27 (m, 7H), 4.13 (t, J=9.5 Hz, IH), 3.97 (d, J=7.9 Hz, IH), 3.85 (d, J=9.2 Hz, IH), 2.73 (s, IH), 2.56 (d, J=6.7 Hz, IH), 2.29 (t, J=9.8 Hz, IH), 1.84 (br. s., IH), 1.76 - 1.21 (m, 15H), 1.10 - 0.99 (m, 6H), 0.92 - 0.74 (m, 5H). MS: MS m/z 866.7 (M + +l).

Preparation of Compound 2611

Compound 2611

Compounds 2611 was prepared using the intermediates described herein and by following the Step 10 general procedure described for the synthesis of Compound 2601 :

Compound 2611 : tert-butyl ((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((2,3-dihydro- [l,4]dioxino[2,3-f]isoquinolin-7-yl)oxy)-7-ethyl-9-methyl-14 a-(((l- methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (400MHz,

METHANOLS) δ 7.90 (d, J=6.0 Hz, IH), 7.69 (d, J=9.0 Hz, IH), 7.43 (d, J=6.0 Hz, IH), 7.04 (d, J=9.0 Hz, IH), 5.88 (br. s., IH), 5.59 (td, J=10.0, 6.4 Hz, IH), 5.13 (br. s., IH), 4.63 - 4.52 (m, 2H), 4.45 - 4.35 (m, 5H), 4.14 (dd, J=11.7, 3.6 Hz, IH), 3.87 (d, J=9.8 Hz, IH), 3.59 - 3.51 (m, IH), 2.82 - 2.38 (m, 3H), 1.92 - 1.34 (m, 13H), 1.21 - 1.09 (m, 12H), 0.96 - 0.83 (m, 5H). MS: MS m/z 812.7 (M + +l).

Preparation of Compound 2612

Compound 2612

Compounds 2612 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 1001 :

Compound 2612: 1 1,1,1 -trifluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-((3-(5-isopropo xypyridin-2-yl)-6- methoxyisoquinolin- 1 -yl)oxy)-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (400MHz,

METHANOLS) δ 8.41 (d, J=8.8 Hz, IH), 8.26 (d, J=2.8 Hz, IH), 8.13 - 8.03 (m, 2H), 7.53 (dd, J=8.9, 2.9 Hz, IH), 7.27 (d, J=2.5 Hz, IH), 7.09 (dd, J=9.2, 2.4 Hz, IH), 6.07 (br. s., IH), 5.66 - 5.57 (m, IH), 5.21 - 5.07 (m, IH), 4.76 (dt, J=12.0, 6.0 Hz, IH), 4.66 - 4.57 (m, IH), 4.40 (d, J=10.0 Hz, IH), 4.26 (dd, J=l 1.5, 4.0 Hz, IH), 3.99 - 3.88 (m, 4H), 3.63 - 3.51 (m, IH), 2.82 - 2.69 (m, 2H), 2.55 (ddd, J=13.7, 9.2, 4.6 Hz, 2H), 1.97 - 1.35 (m, 22H), 1.21 - 1.08 (m, 6H), 0.98 - 0.80 (m, 5H). MS: MS m/z 974.0 (M + +l). Preparation of Compound 2613

Compound 2613

Compounds 2613 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 1001 :

Compound 2613: 3,3-difluoro-2-methylbutan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-((3-(5-isopropoxyp yridin-2-yl)-6- methoxyisoquinolin- 1 -yl)oxy)-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. MS: MS m/z 970.0 (M + +l).

Pre aration of Compound 2614

Compound 2614 Compounds 2614 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 1001 :

Compound 2614: l,l-difluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-((3-(5-isopropo xypyridin-2-yl)-6- methoxyisoquinolin- 1 -yl)oxy)-9-methyl- 14a-((( 1 - methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo-

2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. MS: MS m/z 955.9 (M + +l).

Preparation of Compound 2615

Compound 2615

Compounds 2615 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 1001 :

Compound 2615: 1,1 -difluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-2-((6-ethoxy-7-fiuoroisoquin olin-l-yl)oxy)-7- ethyl-9-methyl- 14a-((( 1 -methylcyclopropyl)sulfony l)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 7.96 (d, J=5.8 Hz, 1H), 7.76 (d, J=9.2 Hz, 1H), 7.69 (d, J=11.6 Hz, 1H), 7.53 (d, J=7.9 Hz, 1H), 7.33 (d, J=5.8 Hz, 1H), 5.99 - 5.73 (m, 2H), 5.57 - 5.48 (m, 1H), 5.06 (br. s., 1H), 4.49 - 4.34 (m, 2H), 4.28 - 4.21 (m, 2H), 4.14 (t, J=9.6 Hz, 1H), 3.95 (d, J=8.5 Hz, 1H), 3.85 (d, J=9.5 Hz, 1H), 2.73 (s, 1H), 2.62 - 2.53 (m, 2H), 2.29 (t, J=9.5 Hz, IH), 1.88 - 1.21 (m, 21H), 1.17 - 1.04 (m, 9H), 0.90 - 0.75 (m, 5H). MS: MS m/z 852.7 (M + +l).

Preparation of Compound 2616

Compound 2616

Compounds 2616 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 1001 : Compound 2616: 1,1 -difluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-((7-fluoro-6-metho xyisoquinolin-l- yl)oxy)-9-methyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 7.98 (d, J=5.8 Hz, IH), 7.76 (d, J=9.2 Hz, IH), 7.70 (d, J=l 1.6 Hz, IH), 7.55 (d,

J=8.5 Hz, IH), 7.35 (d, J=6.1 Hz, IH), 5.99 - 5.72 (m, 2H), 5.57 - 5.47 (m, IH), 5.06 (br. s., 1H), 4.45 (t, J=8.1 Hz, IH), 4.37 (d, J=l 1.0 Hz, IH), 4.13 (t, J=9.5 Hz, IH), 4.00 - 3.91 (m, 4H), 3.84 (d, J=8.5 Hz, IH), 2.73 (s, IH), 2.61 - 2.53 (m, IH), 2.29 (t, J=9.6 Hz, IH), 1.88 - 1.02 (m, 22H), 0.91 - 0.76 (m, 5H). MS: MS m/z 838.8 (M + +l). Preparation of Compound 2617

Compound 2617

Compounds 2617 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 1001 :

Compound 2617: l,l-difluoro-2-methylpropan-2-yl

((2R,6S,7S,9R,13aS,14aR,16aS,Z)-7-ethyl-2-((7-fiuoro-4-metho xyisoquinolin-l- yl)oxy)-9-methyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 8.13 (dd, J=9.0, 5.3 Hz, 1H), 7.77 - 7.62 (m, 4H), 5.99 - 5.72 (m, 2H), 5.59 - 5.48 (m, 1H), 5.06 (br. s., 1H), 4.50 - 4.34 (m, 2H), 4.13 (t, J=9.8 Hz, 1H), 4.01 - 3.92 (m, 4H), 3.84 (d, J=9.5 Hz, 1H), 2.73 (s, 1H), 2.62 - 2.54 (m, 1H), 2.29 (t, J=9.6 Hz, 1H), 1.89 - 0.98 (m, 22H), 0.91 - 0.73 (m, 5H). MS: MS m/z 838.9 (M + +l).

Preparation of Compound 2618

Compound 2618

Compounds 2618 was prepared using the intermediates described herein and by following the general procedure described for the synthesis of Compound 1001 :

Compound 2618: 1,1 -difluoro-2-methylpropan-2-yl

((2R,6S,7S,9R, 13aS, 14aR, 16aS,Z)-7-ethyl-2-((4-methoxyisoquinolin- 1 -yl)oxy)-9- methyl- 14a-((( 1 -methylcyclopropyl)sulfonyl)carbamoyl)-5 , 16-dioxo- 2,3,5,6,7,9,10,11,13a, 14, 14a,15, 16, 16a-tetradecahydro-lH-cyclopropa[i]pyrrolo[l, 2- e][l,5,8]oxadiazacyclopentadecin-6-yl)carbamate. 1H NMR (500MHz, DMSO-d 6 ) δ 8.06 (d, J=8.2 Hz, 2H), 7.82 - 7.71 (m, 2H), 7.67 - 7.59 (m, 2H), 6.01 - 5.72 (m, 2H), 5.57 - 5.47 (m, 1H), 5.16 (br. s., 1H), 4.46 - 4.33 (m, 2H), 4.15 (t, J=9.6 Hz, 1H), 4.01 - 3.93 (m, 4H), 3.84 (d, J=10.1 Hz, 1H), 2.66 - 2.53 (m, 2H), 2.46 - 2.26 (m, 2H), 1.88 - 0.97 (m, 22H), 0.80 (t, J=7.2 Hz, 5H). MS: MS m/z 820.8 (M + +l).

Biological Studies

HCV NS3/4A protease complex enzyme assays and cell-based HCV replicon assays were utilized in the present disclosure, and were prepared, conducted and validated as follows:

Generation of recombinant HCV NS3/4 A protease complex HCV NS3 protease complexes, derived from the BMS strain, H77 strain or J4L6S strain, were generated, as described below. These purified recombinant proteins were generated for use in a homogeneous assay (see below) to provide an indication of how effective compounds of the present disclosure would be in inhibiting HCV NS3 proteolytic activity.

Serum from an HCV-infected patient was obtained from Dr. T. Wright, San Francisco Hospital. An engineered full-length cDNA (compliment deoxyribonucleic acid) template of the HCV genome (BMS strain) was constructed from DNA fragments obtained by reverse transcription-PCR (RT-PCR) of serum RNA

(ribonucleic acid) and using primers selected on the basis of homology between other genotype la strains. From the determination of the entire genome sequence, a genotype la was assigned to the HCV isolate according to the classification of Simmonds et al. (See P Simmonds, KA Rose, S Graham, SW Chan, F McOmish, BC Dow, EA Follett, PL Yap and H Marsden, J. Clin. Microbiol, 31(6), 1493-1503

(1993)). The amino acid sequence of the nonstructural region, NS2-5B, was shown to be >97% identical to HCV genotype la (H77) and 87% identical to genotype lb (J4L6S). The infectious clones, H77 (la genotype) and J4L6S (lb genotype) were obtained from R. Purcell (NIH) and the sequences are published in Genbank

(AAB67036, see Yanagi,M., Purcell,R.H., Emerson,S.U. and Bukh, J. Proc. Natl Acad. Sci. U.S.A. 94(16), 8738-8743 (1997); AF054247, see Yanagi,M., St Claire,M., Shapiro,M., Emerson,S.U., Purcell,R.H. and Bukh, J., Virology 244 (1), 161-172. (1998)).

The H77 and J4L6S strains were used for production of recombinant NS3/4A protease complexes. DNA encoding the recombinant HCV NS3/4A protease complex (amino acids 1027 to 1711) for these strains was manipulated as described by P. Gallinari et al. (see Gallinari P, Paolini C, Brennan D, Nardi C, Steinkuhler C, De Francesco R. Biochemistry. 38(17):5620-32, (1999)). Briefly, a three-lysine solubilizing tail was added at the 3 '-end of the NS4A coding region. The cysteine in the PI position of the NS4A-NS4B cleavage site (amino acid 1711) was changed to a glycine to avoid the proteolytic cleavage of the lysine tag. Furthermore, a cysteine to serine mutation was introduced by PCR at amino acid position 1454 to prevent the autolytic cleavage in the NS3 helicase domain. The variant DNA fragment was cloned in the pET21b bacterial expression vector (Novagen) and the NS3/4A complex was expressed in Escherichia, coli strain BL21 (DE3) (Invitrogen) following the protocol described by P. Gallinari et al. (see Gallinari P, Brennan D, Nardi C, Brunetti M, Tomei L, Steinkuhler C, De Francesco R., J Virol. 72(8):6758-69 (1998)) with modifications. Briefly, the NS3/4A protease complex expression was induced with 0.5 millimolar (mM) Isopropyl β-D-l-thiogalactopyranoside (IPTG) for 22 hours (h) at 20°C. A typical fermentation (1 Liter (L)) yielded approximately 10 grams (g) of wet cell paste. The cells were resuspended in lysis buffer (10 mL/g) consisting of 25 mM N-(2-Hydroxyethyl)Piperazine-N'-(2-Ethane Sulfonic acid) (HEPES), pH 7.5, 20% glycerol, 500 mM Sodium Chloride (NaCl), 0.5% Triton X- 100, 1 microgram/milliliter ('^g/mL") lysozyme, 5 mM Magnesium Chloride (MgCl 2 ), 1 μg/ml Dnasel, 5 mM β-Mercaptoethanol (βΜΕ), Protease inhibitor- Ethylenediamine Tetraacetic acid (EDTA) free (Roche), homogenized and incubated for 20 minutes (min) at 4°C. The homogenate was sonicated and clarified by ultra- centrifugation at 235000 g for 1 hour (h) at 4°C. Imidazole was added to the supernatant to a final concentration of 15 mM and the pH adjusted to 8.0. The crude protein extract was loaded on a Nickel-Nitrilotriacetic acid (Ni-NTA) column pre- equilibrated with buffer B (25 mM HEPES, pH 8.0, 20% glycerol, 500 mM NaCl, 0.5%) Triton X-100, 15 mM imidazole, 5 mM βΜΕ). The sample was loaded at a flow rate of 1 mL/min. The column was washed with 15 column volumes of buffer C (same as buffer B except with 0.2% Triton X-100). The protein was eluted with 5 column volumes of buffer D (same as buffer C except with 200 mM Imidazole).

NS3/4A protease complex-containing fractions were pooled and loaded on a desalting column Superdex-S200 pre-equilibrated with buffer D (25 mM HEPES, pH 7.5, 20% glycerol, 300 mM NaCl, 0.2% Triton X-100, 10 mM βΜΕ). Sample was loaded at a flow rate of 1 mL/min. NS3/4A protease complex-containing fractions were pooled and concentrated to approximately 0.5 mg/ml. The purity of the NS3/4A protease complexes, derived from the BMS, H77 and J4L6S strains, were judged to be greater than 90% by SDS-PAGE and mass spectrometry analyses. The enzyme was stored at -80 °C, thawed on ice and diluted prior to use in assay buffer.

FRET peptide assay to monitor HCV NS3/4 A proteolytic activty The purpose of this in vitro assay was to measure the inhibition of HCV NS3 protease complexes, derived from the BMS strain, H77 strain or J4L6S strain, as described above, by compounds of the present disclosure. This assay provides an indication of how effective compounds of the present disclosure would be in inhibiting HCV NS3 proteolytic activity. In order to monitor HCV NS3/4A protease activity, an NS3/4A peptide substrate was used. The substrate was RET S 1 (Resonance Energy Transfer

Depsipeptide Substrate; AnaSpec, Inc. cat # 22991)(FRET peptide), described by Taliani et al. in Anal. Biochem. 240(2):60-67 (1996). The sequence of this peptide is loosely based on the NS4A/NS4B natural cleavage site for the HCV NS3 protease except there is an ester linkage rather than an amide bond at the cleavage site. The peptide also contains a fluorescence donor, EDANS, near one end of the peptide and an acceptor, DABCYL, near the other end. The fluorescence of the peptide is quenched by intermolecular resonance energy transfer (RET) between the donor and the acceptor, but as the NS3 protease cleaves the peptide the products are released from RET quenching and the fluorescence of the donor becomes apparent.

The peptide substrate was incubated with one of the three recombinant NS3/4A protease complexes, in the absence or presence of a compound of the present disclosure. The inhibitory effects of a compound were determined by monitoring the formation of fluorescent reaction product in real time using a Cytofluor Series 4000.

The reagents were as follow: HEPES and Glycerol (Ultrapure) were obtained from GIBCO-BRL. Dimethyl Sulfoxide (DMSO) was obtained from Sigma, β- Mercaptoethanol was obtained from Bio Rad.

Assay buffer: 50 mM HEPES, pH 7.5; 0.15 M NaCl; 0.1% Triton; 15% Glycerol; 10 mM βΜΕ. Substrate: 2 μΜ final concentration (from a 2 mM stock solution in DMSO stored at -20°C). HCV NS3/4A protease type la (lb), 2-3 nM final concentration (from a 5 μΜ stock solution in 25 mM HEPES, pH 7.5, 20% glycerol, 300 mM NaCl, 0.2% Triton-XlOO, 10 mM βΜΕ). For compounds with potencies approaching the assay limit, the assay was made more sensitive by adding 50 μg/ml Bovine Serum Albumin (Sigma) to the assay buffer and reducing the end protease concentration to 300 pM.

The assay was performed in a 96-well polystyrene black plate from Falcon. Each well contained 25 μΐ NS3/4A protease complex in assay buffer, 50 μΐ of a compound of the present disclosure in 10%> DMSO/assay buffer and 25 μΐ substrate in assay buffer. A control (no compound) was also prepared on the same assay plate.

The enzyme complex was mixed with compound or control solution for 1 min before initiating the enzymatic reaction by the addition of substrate. The assay plate was read immediately using the Cytofluor Series 4000 (Perspective Biosystems). The instrument was set to read an emission of 340 nm and excitation of 490 nm at 25°C. Reactions were generally followed for approximately 15 min.

The percent inhibition was calculated with the following equation:

100-[(5F mh /5F con )xl00]

where 5F is the change in fluorescence over the linear range of the curve. A nonlinear curve fit was applied to the inhibition-concentration data, and the 50% effective concentration (IC 50 ) was calculated by the use of Excel XLfit software using the equation, y=A+((B-A)/(l+((C/x) A D))).

Compounds of the present disclosure, which were tested against more than one type of NS3/4A complex, were found to have similar inhibitory properties though the compounds uniformly demonstrated greater potency against the lb strains as compared to the la strains. Generation of HCV Replicon

An HCV replicon whole cell system was established as described by

Lohmann V, Korner F, Koch J, Herian U, Theilmann L, Bartenschlager R., Science 285(5424): 110-3 (1999) and modified to introduce a luciferase reporter, as first described by Krieger et al (Krieger N, Lohmann V, and Bartenschlager R, J. Virol. 75(10):4614-4624 (2001)). CDNA encoding a humanized form of the Renilla luciferase gene and a linker sequence fused directly to the 3 '-end of the luciferase gene were introduced into the replicon construct using an Ascl restriction site located in core, directly upstream of the neomycin marker gene. The adaptive mutation at position 1179 (serine to isoleucine) was also introduced (Blight KJ, Kolykhalov, AA, Rice, CM, Science 290(5498): 1972- 1974). A stable cell line constitutively expressing this HCV replicon construct was generated by first linearizing plasmid DNAs with Seal. RNA transcripts were synthesized in vitro using the T7 MegaScript transcription kit (Ambion, Austin, TX) according to manufacturer's directions. In vitro transcripts of the cDNA were transfected into the human hepatoma cell line, HUH-7. Selection for cells constitutively expressing the HCV replicon was achieved in the presence of the selectable marker, neomycin (G418). Resulting cell lines were characterized for positive and negative strand RNA production and protein production over time.

A stable HCV replicon luciferase reporter cell line representing the genotype la H77 strain (Yanagi M, Purcell RH, Emerson SU, et al. Transcripts from a single full-length cDNA clone of hepatitis C virus are infectious when directly transfected into the liver of a chimpanzee. Proc Natl Acad Sci USA 1997; 94(16): 8738-8743) was generated as described previously for the genotype lb(Conl) replicon luciferase cell line. The replicon construct was modified by introducing mutations were introduced into the genes encoding the NS3 helicase domain (proline replaced by leucine at position 1496) and NS5A (serine to isoleucine at position 2204) to improve replication in cell culture.

HCV Replicon Luciferase Reporter Assay

HCV replicon luciferase assays were developed to monitor the inhibitory effects of compounds described in the disclosure on HCV genotypes la and lb viral replication. HUH-7 cells, constitutively expressing the HCV replicon, were grown in Dulbecco's Modified Eagle Media (DMEM) (Gibco-BRL) containing 10% Fetal calf serum (FCS) (Sigma) and 1 mg/mL G418 (Gibco-BRL). Compounds were serially diluted 3 folds in DMSO for a twenty-point titration and subsequently transferred to sterile 384-well tissue-culture treated plates (Corning cat # 3571). The plates were then seeded with 50 μΕ of cells at a density of 3.0 x 10 3 cells/well in DMEM containing 4 % FCS (final DMSO concentration at 0.5 %). After 3 days incubation at 37°C, cells were analyzed for Renilla Luciferase activity using the EnduRen as substrate (Promega cat #E6485). The EnduRen substrate was diluted in DMEM and then added to the plates to a final concentration of 7.5 μΜ. The plates were incubated for 2 hrs at 37°C and then read immediately for 30 seconds with Viewlux Imager

(PerkinElmer) using a luminescence program. To assess cytotoxicity of compounds, CC 50 values were generated by multiplexing the EnduRen-containing plates with Cell Titer-Blue (Promega, cat # G8082). Cell-Titer Blue (3 \ ) was added to each well and incubated for 8 hrs at 37°C. The fluorescence signal from each well was read, with an excitation wavelength at 525/10 nm and an emission wavelength of 598/10 nm, using the Viewlux Imager.

The EC 50 values for compounds were calculated by using a four-parameter logistic equation:

y = A+((B-A)/(l+((C/x) A D))),

where A and B denotes minimal and maximal % inhibition, respectively, C is the EC 50 , D is the hill slope and x represents compound concentration.

Table 2 shows the EC50 values of representative compounds of the present disclosure. Ranges are as follows: A = 0.10 nM - 0.50 nM; B = 0.51 nM - 1.00 nM; C = 1.01 nM - 5.00 nM; D = 5.01 nM - 35.00 nM; E = 35.01 nM-620 nM.

Table 2

1005 C B

1006 C A

1007 C B

1008 0.41 A 0.28 A

1009 C C

1010 B C 1011 B B

1012 C C

1013 C C

1014 C C

1015 A A

1016 A A 1017 B B

1018 C B

1019 C A

1020 C B

2001 C C

2002 B A 2003 B A

2004 B A

2005 C A

2006 C A

2007 0.63 B 0.24 A

2008 B A 2009 C B

2010 C B

2011 B A

2012 A A

2013 C B

2014 2.38 C 0.66 B 2015 C B

2016 C B

2017 0.89 B 0.42 A

2018 B A

2019 C B

2020 C B 2021 C B

2022 C C

2301 B A

2302 B A

2303 B B

2304 0.49 A 0.64 B 2305 B B

2306 A A

2307 A A

2308 A A

2309 B B

2310 A A 2311 A A

2601 C C

2602 C C

2603 9.08 D 6.64 D

2604 D C

2605 B A 2606 B A

2607 B A

2608 B A

2609 C B

2610 C B

2611 C B 2618 A A

It will be evident to one skilled in the art that the present disclosure is not limited to the foregoing illustrative examples, and that it can be embodied in other specific forms without departing from the essential attributes thereof. It is therefore desired that the examples be considered in all respects as illustrative and not restrictive, reference being made to the appended claims, rather than to the foregoing examples, and all changes which come within the meaning and range of equivalency of the claims are therefore intended to be embraced therein.