Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SULFONYLPIPERAZINYL COMPOUNDS FOR TREATMENT OF BACTERIAL INFECTIONS
Document Type and Number:
WIPO Patent Application WO/2023/061617
Kind Code:
A1
Abstract:
The present invention relates to compounds of formula (I), wherein R1 to R3, A, Q1, Q2, W and X are as described herein, and their pharmaceutically acceptable salt thereof, and compositions including the compounds and methods of using the compounds.

Inventors:
AMREIN KURT (CH)
DEY FABIAN (CH)
DING XIAO (CN)
HUANG XINYI (CN)
LERNER CHRISTIAN (CH)
SHI HOUGUANG (CN)
TAN XUEFEI (CN)
WU JUN (CN)
ZHENG JIAMIN (CN)
ZHOU MINGWEI (CN)
Application Number:
PCT/EP2021/086690
Publication Date:
April 20, 2023
Filing Date:
December 20, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
HOFFMANN LA ROCHE (US)
HOFFMANN LA ROCHE (US)
International Classes:
C07D401/12; A61K31/496; A61P31/04; C07D401/14; C07D403/12; C07D413/12; C07D487/08; C07D487/10
Domestic Patent References:
WO2004069816A12004-08-19
WO1998001446A11998-01-15
Other References:
BOHL THOMAS E. ET AL: "The substrate-binding cap of the UDP-diacylglucosamine pyrophosphatase LpxH is highly flexible, enabling facile substrate binding and product release", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 293, no. 21, 1 May 2018 (2018-05-01), US, pages 7969 - 7981, XP055894625, ISSN: 0021-9258, Retrieved from the Internet DOI: 10.1074/jbc.RA118.002503
ANN. REV. BIOCHEM, vol. 76, 2007, pages 295 - 329
ANSEL, HOWARD C. ET AL.: "Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems", 2004, LIPPINCOTT, WILLIAMS & WILKINS
GENNARO, ALFONSO R. ET AL.: "Remington: The Science and Practice of Pharmacy", 2000, LIPPINCOTT, WILLIAMS & WILKINS
ROWE, RAYMOND C.: "Handbook of Pharmaceutical Excipients", 2005, PHARMACEUTICAL PRESS
CAS, no. Silica Gel: 63231-67-4
Attorney, Agent or Firm:
SALUD, Carlos (CH)
Download PDF:
Claims:
CLAIMS

1. A compound of formula (I), wherein

R1 is amino, hydroxyC1-6alkyl, C1-6alkylamino, aminoC1-6alkylamino or piperazinyl;

R2 is halogen or C1-6alkyl;

R3 is -Y-C(O)-R4; wherein

Y is C3-10cycloalkyl, phenyl, pyrazinyl or pyridinyl;

R4 is ((aminoC1-6alkyl)2amino)C1-6alkylamino, ((aminoC1-6alkyl)pyrrolidiniumyl)C1- 6alkylamino, ((aminoC1-6alkylcarbonyl)amino)C1-6alkylamino, ((C1-6alkyl)2amino)C1- 6alkylamino, ((C1-6alkyl)3ammonio)C1-6alkylamino, (aminoC1-6alkoxy)C1-6alkylamino, (aminoC1-6alkyl(C1-6alkyl)2ammonio)C1-6alkylamino, (C1-6alkyl(aminoC1- 6alkyl)2ammonio)C1-6alkylamino, ((aminoC1-6alkylcarbonyl)amino)C3- 10cycloalkyl amino, ((aminoC1-6alkyl)2pyrrolidinyl)C3-10cycloalkylamino, (aminoC1- 6alkyl)2amino, (aminoC1-6alkyl)azetidinyl, (aminoC1-6alkyl)C3-10cycloalkylamino, (aminoC1-6alkyl)piperazinyl, (aminoC1-6alkylamino)C1-6alkylamino, (aminoC1- 6alkylcarbonyl)aminocarbamoyl, (aminopyrrolidinyl)C3-10cycloalkylamino, (azetidinylC1-6alkylamino)C1-6alkylamino, (C1-6alkylamino)C1-6alkylamino, (C1- 6alkylpyrrolidiniumyl)C1-6alkylamino, (diaminopiperidinyl)C1-6alkylamino, (diaminopiperidinyl)C3-10cycloalkylamino, (piperidinylcarbonylamino)carbamoyl, 3- azabicyclo[3.1.0]hexanylamino, amino, amino(C1-6alkoxy)C1-6alkylamino, amino(C1- 6alkyl)piperidinyl, (aminoC1-6alkyl)2pyrrolidinyl, amino(hydroxy)C1-6alkylamino, aminoC1-6alkylamino, aminoC3-10cycloalkylamino, aminocarbonylC1-6alkylamino, aminopyrrolidinylamino, amino(C1-6alkyl)pyrrolidinylamino, azetidinylC1-6alkylamino, C1-6alkoxy, C1-6alkyl(aminoC1-6alkyl)amino, C1-6alkylpyrrolidinylamino, C1- 6alkyl(aminoC1-6alkyl)piperaziniumyl, C1-6alkyl-2,6-diazaspiro[3.3]heptanyl, C1- 6alkylamino, diaminoC1-6alkylamino, diaminopiperidinyl, hydroxyC1-6alkylamino, hydroxypyrrolidinylamino, morpholinylC1-6alkylamino or pyrrolidinylC1-6alkylamino;

X is CH2 or O;

A is CH or N; Q1 is CH orN;

Q2 is CH orN;

W is haloC1-6alkyl; or a pharmaceutically acceptable salt thereof.

2. A compound according to claim 1, wherein R1 is amino, C1-6alkylamino or hydroxyC1-6alkyl.

3. A compound according to claim 1 or 2, wherein R1 is amino, methylamino or hydroxymethyl.

4. A compound according to any one of claims 1 to 3, wherein R2 is halogen.

5. A compound according to any one of claims 1 to 4, wherein R2 is chloro.

6. A compound according to any one of claims 1 to 5, wherein R3 is -Y-C(O)-R4; wherein Y is C3-10cycloalkyl, phenyl or pyrazinyl.

7. A compound according to any one of claims 1 to 6, wherein R3 is -Y-C(O)-R4; wherein Y is bicyclo[2.2.2]octanyl, bicyclo[3.1.0]hexanyl, cyclohexanyl, norbomanyl, phenyl or pyrazinyl.

8. A compound according to any one of claims 1 to 7, wherein R3 is -Y-C(O)-R4; wherein R4 is amino(C1-6alkyl)pyrrolidinylamino, ((aminoC1-6alkyl)2amino)C1-6alkylamino, ((aminoC1- 6alkyl)2pyrrolidinyl)C3-10cycloalkylamino, ((aminoC1-6alkyl)pyrrolidiniumyl)C1-6alkylamino, (aminoC1-6alkyl(C1-6alkyl)2ammonio)C1-6alkylamino, (aminoC1-6alkyl^amino, (aminoC1- 6alkyl)2pyrrolidinyl, (aminoC1-6alkyl)C3-10cycloalkylamino, (C1-6alkyl(aminoC1- 6alkyl)2ammonio)C1-6alkylamino, (aminopyrrolidinyl)C3-10cycloalkylamino, (C1-6alkyl(aminoC1- 6alkyl)2ammonio)C1-6alkylamino, (diaminopiperidinyl)C1-6alkylamino, (diaminopiperidinyl)C3- 10cycloalkyl amino, amino, amino(hydroxy)C1-6alkylamino, aminoC1-6alkylamino, aminoC3- 10cycloalkyl amino, C1-6alkylpyrrolidinylamino, diaminoC1-6alkylamino or diaminopiperidinyl.

9. A compound according to any one of claims 1 to 8, wherein R3 is -Y-C(O)-R4; wherein R4 is ((2-aminoethyl)2amino)propylamino, ((2-aminoethyl)pyrrolidin-l-ium-l-yl)ethylamino, (2- aminoethyl(dimethyl)ammonio)ethylamino, (2-aminoethyl)2amino, (3,5-diamino-l- piperidinyl)propylamino, (3-aminocyclopentyl)amino, (3-aminopropyl)2 amino, (4- methylpyrrolidin-3-yl)amino, [3-(3,5-diamino-l-piperidinyl)cyclopentyl]amino, [3-[3,4- bis(aminomethyl)pyrrolidin-l-yl]cyclopentyl]amino, [3-[3-aminopyrrolidin-l- yl]cyclopentyl]amino, l-(aminomethyl)cyclopropylamino, 2-(2-aminoethylamino)ethylamino, 2,3 -diaminopropylamino, 2-aminoethyl amino, 3,4-bis(aminomethyl)pyrrolidin-l-yl, 3,5- diaminopiperidinyl, 3-[3-aminopropyl(dimethyl)ammonio]propylamino, 3-[bis(2-aminoethyl)- methyl-ammonio]propylamino, 3-amino-2-hydroxy-propylamino, 3 -aminopropylamino, 3- hydroxypropylamino, 4-amino-2-methyl-pyrrolidin-l-yl, amino or aminopropylamino.

10. A compound according to any one of claims 1 to 9, wherein A is CH.

11. A compound according to any one of claims 1 to 10, wherein Q1 is N.

12. A compound according to any one of claims 1 to 11, wherein Q2 is CH.

13. A compound according to any one of claims 1 to 12, wherein W is CF2.

14. A compound according to any one of claims 1 to 13, wherein

R1 is amino or hydroxyC1-6alkyl;

R2 is halogen;

R3 is -Y-C(O)-R4; wherein

Y is C3-10cycloalkyl, phenyl or pyrazinyl;

R4 is amino(C1-6alkyl)pyrrolidinylamino, ((aminoC1-6alkyl)2amino)C1-6alkylamino, ((aminoC1-6alkyl)2pyrrolidinyl)C3-10cycloalkylamino, ((aminoC1- 6alkyl)pyrrolidiniumyl)C1-6alkylamino, (aminoC1-6alkyl(C1-6alkyl)2ammonio)C1- 6alkylamino, (aminoC1-6alkyl^amino, (aminoC1-6alkyl^pyrrolidinyl, (aminoC1- 6alkyl)C3-10cycloalkylamino, (C1-6alkyl(aminoC1-6alkyl)2ammonio)C1-6alkylamino, (aminopyrrolidinyl)C3-10cycloalkylamino, (C1-6alkyl(aminoC1-6alkyl)2ammonio)C1- 6alkylamino, (diaminopiperidinyl)C1-6alkylamino, (diaminopiperidinyl)C3- 10cycloalkyl amino, amino, amino(hydroxy)C1-6alkylamino, aminoC1-6alkylamino, aminoC3-10cycloalkylamino, C1-6alkylpyrrolidinylamino, diaminoC1-6alkylamino or diaminopiperidinyl;

X is CH2 or O;

A is CH; Q1 is N;

Q2 is CH;

W is haloC1-6alkyl; or a pharmaceutically acceptable salt thereof.

15. A compound according to any one of claims 1 to 14, wherein

R1 is amino or hydroxymethyl;

R2 is chloro;

R3 is -Y-C(O)-R4; wherein

Y is bicyclo[2.2.2]octanyl, bicyclo[3.1.0]hexanyl, cyclohexanyl, phenyl or pyrazinyl;

R4 is ((2-aminoethyl)2amino)propylamino, ((2-aminoethyl)pyrrolidin-l-ium-l- yl)ethylamino, (2-aminoethyl(dimethyl)ammonio)ethylamino, (2-aminoethyl)2amino, (3,5-diamino-l-piperidinyl)propylamino, (3-aminocyclopentyl)amino, (3- aminopropyl)2 amino, (4-methylpyrrolidin-3-yl)amino, [3-(3,5-diamino-l- piperidinyl)cyclopentyl]amino, [3-[3,4-bis(aminomethyl)pyrrolidin-l- yl]cyclopentyl]amino, [3-[3-aminopyrrolidin-l-yl]cyclopentyl]amino, 1- (aminomethyl)cyclopropylamino, 2-(2-aminoethylamino)ethylamino, 2,3- diaminopropylamino, 2-aminoethylamino, 3,4-bis(aminomethyl)pyrrolidin-l-yl, 3,5- diaminopiperidinyl, 3-[3-aminopropyl(dimethyl)ammonio]propylamino, 3-[bis(2- aminoethyl)-methyl-ammonio]propylamino, 3-amino-2-hydroxy-propylamino, 3- aminopropylamino, 3 -hydroxypropylamino, 4-amino-2-methyl-pyrrolidin-l-yl, amino or aminopropylamino;

X is CH2 or O;

A is CH;

Q1 is N;

Q2 is CH;

W is CF2; or a pharmaceutically acceptable salt thereof.

16. A compound selected from:

7ra//.s-methyl 4-[[2-[4-[4-(4-amino-2-oxo-pyrrolidin-l-yl)phenyl]sulfonylpiperazin-l-yl]- 6-chloro-4-pyridyl]-difluoro-methyl]cyclohexanecarboxylate; Trans-4-[[2-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-6- chloro-4-pyridyl]-difluoro-methyl]-N-(azetidin-3-ylmethyl)cyclohexanecarboxamide;

Trans-4-[[2-[4-[4-(4-amino-2-oxo-pyrrolidin-l-yl)phenyl]sulfonylpiperazin-l-yl]-6-chloro- 4-pyridyl]-difluoro-methyl]-N-(3-hydroxypropyl)cyclohexanecarboxamide;

Trans-(4R)-4-amino-l-[4-[4-[4-[[4-(4-amino-4-methyl-piperidine-l-carbonyl)cyclohexyl]- difluoro-methyl] -6-chloro-2-pyridyl]piperazin-l-yl]sulfonylphenyl]pyrrolidin-2-one;

Trans-(4R)-4-amino-l-[4-[4-[6-chloro-4-[difluoro-[4-(6-methyl-2,6-diazaspiro[3.3]heptane-2- carbonyl)cyclohexyl]methyl]-2-pyridyl]piperazin-l-yl]sulfonylphenyl]pyrrolidin-2-one;

Trans-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin- l-yl]-4-pyridyl]-difluoro-methyl]-N-(4-hydroxypyrrolidin-3-yl)cyclohexanecarboxamide;

Trans-N-(3 -aminocyclobutyl)- 4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide;

Trans-N-(2-amino-l-methyl-ethyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide;

Trans-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin- l-yl]-4-pyridyl]-difluoro-methyl]-N-(2-pyrrolidin-l-ylethyl)cyclohexanecarboxamide;

Trans-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin- l-yl]-4-pyridyl]-difluoro-methyl]-N-[(lS,5R)-3-azabicyclo[3.1.0]hexan-6- yl]cyclohexanecarboxamide;

Trans-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin- l-yl]-4-pyridyl]-difluoro-methyl]-N-[c7.s-4-aminopyrrolidin-3-yl]cyclohexanecarboxamide;

Trans-N-[l-(aminomethyl)cyclopropyl]-4-[[2-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide;

Trans-(4R)-4-amino-l-[4-[4-[4-[[4-[4-(2-aminoethyl)piperazine-l-carbonyl]cyclohexyl]- difluoro-methyl] -6-chloro-2-pyridyl]piperazin-l-yl]sulfonylphenyl]pyrrolidin-2-one;

Trans-4-[[2-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-6- chloro-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide;

4-[[2-[4-[4-(4-amino-2-oxo-pyrrolidin-l-yl)phenyl]sulfonylpiperazin-l-yl]-6-chl oro-4- pyridyl] -difluoro-methyl] -N-(3 -hydroxypropyl )benzamide;

4-[[2-[4-[4-(4-amino-2-oxo-pyrrolidin-l-yl)phenyl]sulfonylpiperazin-l-yl]-6-chl oro-4- pyridyl] -difluoro-methyl] -N-(3 -aminopropyl )benzamide;

N-(2-aminoethyl)-4-[[2-[4-[4-(4-amino-2-oxo-pyrrolidin-l-yl)phenyl]sulfonylpiperazin-l- yl]-6-chloro-4-pyridyl]-difluoro-methyl]benzamide; N-(3-amino-2-hydroxy-propyl)-4-[[2-[4-[4-(4-amino-2-oxo-pyrrolidin-l- yl)phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluoro-methyl]benzamide;

N-(3-amino-2-hydroxy-propyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]benzamide;

4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-

4-pyridyl]-difluoro-methyl]-N-[3-(dimethylamino)propyl]benzamide;

N-(3-aminopropyl)-5-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]pyrazine-2-carboxamide;

N-(3-amino-2-hydroxy-propyl)-5-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]pyrazine-2-carboxamide;

5-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-

4-pyridyl]-difluoro-methyl]-N-[3-(dimethylamino)propyl]pyrazine-2-carboxamide;

N-(2-aminoethyl)-5-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]pyrazine-2-carboxamide;

5-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-

4-pyridyl]-difluoro-methyl]-N-(2-morpholinoethyl)pyrazine-2-carboxamide;

N-(3-aminopropyl)-6-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]pyridine-3-carboxamide;

6-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-

4-pyridyl]-difluoro-methyl]-N-[3-(dimethylamino)propyl]pyridine-3-carboxamide;

N-[2-(2-aminoethylamino)ethyl]-6-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]pyridine-3-carboxamide;

5-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-

4-pyridyl]-difluoro-methyl]-N-[3-(methylamino)propyl]pyridine-2-carboxamide;

Trans-N-(2-aminoethyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]-N-methyl- cyclohexanecarboxamide;

Cis-N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide;

Cis-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l- yl ] -4 -pyri dyl ] -difluoro-methyl ] -N-(3 -hy droxypropyl )cy cl ohexanecarb oxami de; N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]bicyclo[2.2.2]octane-l- carboxamide;

N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]norbornane-l-carboxamide;

N-(3-aminopropyl)-3-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]cyclopentanecarboxamide;

Trans-N-(3-amino-2-hydroxy-propyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro- methyl]cyclohexanecarboxamide;

3-[[4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4- pyridyl]-difluoro-methyl]benzoyl]amino]propyl-trimethyl-ammonium;

4-[[2-Chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-

4-pyri dyl]-difluoro-m ethyl] -N-[3 -(1 -methylpyrrolidin- 1 -ium- 1 -yl)propyl]benzamide;

5-[[2-Chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-

4-pyridyl]-difluoro-methyl]-N-[3-(2-hydroxyethylamino)propyl]pyridine-2-carboxamide;

N-(3-aminopropyl)-3-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl] sulfonylpiperazin- 1 -yl ] -4-pyri dy 1] -difluoro-methyl]bicyclo[ 1.1.1 ]pentane- 1 - carboxamide;

Trans-N-(2-aminoethyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide;

Trans-N-[2-(2-aminoethoxy)ethyl]-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide;

Trans-N,N-bis(2-aminoethyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide;

Trans-N,N-bis(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide; N,N-bis(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]bicyclo[2.2.2]octane-l- carboxamide; N,N-bis(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl] sulfonylpiperazin- 1 -yl] -4-pyri dyl] -difluoro-methyl]norbornane- 1 -carboxamide; Trans-N-[3-[bis(2-aminoefliyl)amino]propyl]-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro- methyl]cyclohexanecarboxamide;

Trans-(4R)-4-amino-l-[4-[4-[6-chloro-4-[[4-cis-(3,5-diaminopiperidine-l- carbonyl)cyclohexyl]-difluoro-methyl]-2-pyridyl]piperazin-l-yl]sulfonylphenyl]pyrrolidin-2-one;

Trans-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin- l-yl]-4-pyridyl]-difluoro-methyl]-N-[3-[cis-3,5-diamino-l- piperidyl]propyl]cyclohexanecarboxamide;

Trans-N'-(2-aminoacetyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]cyclohexanecarbohydrazide;

Trans-N'-[4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]cyclohexanecarbonyl]piperidine-4- carbohydrazide;

Trans-N-(2-amino-3-hydroxy-propyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro- methyl]cyclohexanecarboxamide;

(2R,55)-5-[[2-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-6- chloro-4-pyridyl]-difluoro-methyl]-N-(3-aminopropyl)bicyclo[2.2.2]octane-2-carboxamide;

Zrazz5-N-(3-aminopropyl)-4-[difluoro-[6-methyl-2-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin- l-yl]phenyl]sulfonylpiperazin-l-yl]pyrimidin-4-yl]methyl]cyclohexanecarboxamide;

N-(3-aminopropyl)-6-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]bicyclo[3.1.0]hexane-3- carboxamide;

Trans-N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide;

Trans-(4R)-4-amino-l-[4-[4-[4-[[4-[3-(aminomethyl)azetidine-l-carbonyl]cyclohexyl]- difluoro-methyl] -6-chloro-2-pyridyl]piperazin-l-yl]sulfonylphenyl]pyrrolidin-2-one;

Trans-N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(5S)-5-(hydroxymethyl)-2-oxo-oxazolidin- 3-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide;

Trans-N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(5R)-5-(hydroxymethyl)-2-oxo- oxazolidin-3-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro- mefh yl ] cy cl ohexanecarb oxami de ; Trans-N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-(2-oxo-4-piperazin-l-yl-pyrrolidin-l- yl)phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide;

Trans-N-(3-aminopropyl)-4-[[2-chloro-6-[4-[[6-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]-3- pyridyl]sulfonyl]piperazin-l-yl]-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide;

Trans-N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonyl-l-piperidyl]-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide;

Cis-N-(2-amino-2-oxo-ethyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide; (4R)-4-amino-l-[4-[4-[4-[[4-[(25,45)-4-amino-2-methyl-pyrrolidine-l- carbonyl]cyclohexyl]-difluoro-methyl]-6-chloro-2-pyridyl]piperazin-l- yl]sulfonylphenyl]pyrrolidin-2-one;

Trans-bis(2-aminoethyl)-[3-[[4-[[2-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluoro- methyl]cyclohexanecarbonyl]amino]propyl]-methyl -ammonium;

Trans-N-[(17?,35)-3-aminocyclopentyl]-4-[[2-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide;

Trans-(4R)-4-amino-l-[4-[4-[4-[[4-[cis-3,4-bis(aminomethyl)pyrrolidine-l- carbonyl]cyclohexyl]-difluoro-methyl]-6-chloro-2-pyridyl]piperazin-l- yl]sulfonylphenyl]pyrrolidin-2-one;

Trans-4-[[2-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-6- chloro-4-pyridyl]-difluoro-methyl]-N-[(3R,4S)-4-methylpyrrolidin-3- yl]cyclohexanecarboxamide;

Trans-N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-(methylamino)-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide;

Trans-(4R)-4-amino-l-[4-[4-[4-[[4-[4-(2-aminoethyl)-4-methyl-piperazin-4-ium-l- carbonyl]cyclohexyl]-difluoro-methyl]-6-chloro-2-pyridyl]piperazin-l- yl]sulfonylphenyl]pyrrolidin-2-one;

Trans-N-[2-(2-aminoethylamino)ethyl]-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro- methyl]cyclohexanecarboxamide;

Trans-2-aminoethyl-[2-[[4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro- methyl]cyclohexanecarbonyl]amino]ethyl]-dimethyl-ammonium; Trans-N-[2-[l-(2-aminoethyl)pyrrolidin-l-ium-l-yl]ethyl]-4-[[2-[4-[4-[(4A)-4-amino-2- oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluoro- methyl]cyclohexanecarboxamide;

Trans-N-[2-[(2-aminoacetyl)amino]ethyl]-4-[[2-chloro-6-[4-[4-[(4A)-4-amino-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro- methyl]cyclohexanecarboxamide;

Trans-N-(3-amino-2-methoxy-propyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro- methyl]cyclohexanecarboxamide;

Trans-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin- l-yl]-4-pyridyl]-difluoro-methyl]-N-(2,3-diaminopropyl)cyclohexanecarboxamide;

4-[[2-[4-[4-[4-(3-Aminopropylamino)-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l- yl ] -6 -chi oro-4 -pyri dyl ] -difluoro-methyl ] -N -m ethyl -b enzami de;

Trans-N-[(1R,3S)-3-[(2-aminoacetyl)amino]cyclopentyl]-4-[[2-[4-[4-[(4A)-4-amino-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluoro- methyl]cyclohexanecarboxamide;

Trans-4-[[2-[4-[4-[(4A)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-6- chloro-4-pyridyl]-difluoro-methyl]-N-[(1S,3R)-3-[cis-3,4-bis(aminomethyl)pyrrolidin-l- yl ] cy cl opentyl ] cy cl ohexanecarb oxami de;

Trans-4-[[2-chloro-6-[4-[4-[(4A)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin- l-yl]-4-pyridyl]-difluoro-methyl]-N-[(1S)-3-[cis-3,5-diamino-l- piperidyl]cyclopentyl]cyclohexanecarboxamide;

Trans-4-[[2-[4-[4-[(4A)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-6- chloro-4-pyridyl]-difluoro-methyl]-N-[(15,3A)-3-[(3A)-3-aminopyrrolidin-l- yl ] cy cl opentyl ] cy cl ohexanecarb oxami de; and

Trans-3-[[4-[[2-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]- 6-chloro-4-pyridyl]-difluoro-methyl]cyclohexanecarbonyl]amino]propyl-(3-aminopropyl)- dimethyl-ammonium; or a pharmaceutically acceptable salt thereof.

17. A process for the preparation of a compound according to any one of claims 1 to 16 comprising any of the following steps: a) formation of compound of formula (XIX), via deprotection of compound of formula (XVIII), using an acid, wherein the acid is tri fluoroacetic acid, or a reducing reagent, wherein the reducing agent is palladium on carbon in the presence of H2; b) formation of compound of formula (XXXV), via methylation of compound of formula (XXXIII), using a suitable reagent, wherein the agent is Mel, followed by deprotection with an acid, wherein the acid is trifluoroacetic acid, or a reducing reagent, wherein the reducing agent is palladium on carbon in the presence of H2; c) formation of compound of formula (XXXIV), via deprotection of compound of formula (XXXIII), using an acid, wherein the acid is trifluoroacetic acid, or a reducing reagent, wherein the reducing agent is palladium on carbon in the presence of H2; d) formation of compound of formula (XXXVIII), via deprotection of the compound of formula (XXXVII),

acid, wherein the acid is trifluoroacetic acid, or a reducing reagent, wherein the reducing agent is palladium on carbon in the presence of H2; wherein R4’ is R4 or R4-PG3; L1 is C1-6alkyl or C3-10cycloalkyl; R7 is C1-6alkyl, aminoC1- 6alkyl or aminoC1-iocycloalkyl; R7 is R7-PG5; PG2, PG3 and PG5 are independently selected from tert-butoxycarbonyl or benzyl oxy carbonyl; R1, R2, Y, A, Q1, Q2 and X are defined as in any one of claims 1 to 15.

18. A compound or pharmaceutically acceptable salt according to any one of claims 1 to 16 for use as therapeutically active substance.

19. A pharmaceutical composition comprising a compound in accordance with any one of claims 1 to 16 and a therapeutically inert carrier.

20. The use of a compound according to any one of claims 1 to 16 for the inhibition of LpxH.

21. The use of a compound according to any one of claims 1 to 16 for the treatment or prophylaxis of bacterial infection, particularly the bacteria is gram-negative bacteria.

22. The use of a compound according to any one of claims 1 to 16 for the preparation of a medicament for the treatment or prophylaxis of bacterial infection, particularly the bacteria is gram-negative bacteria.

23. The use of a compound according to claim 21 or 22, wherein the gram-negative bacteria is selected from Enter obacleriaceae. Neisseria gonorrhoeae, Haemophilus influenzae, Helicobacter pylorus, Acinetobacter baumannii and Pseudomonas aeruginosa.

24. The use of a compound according to claim 23, wherein the gram-negative bacteria is

Enter obacleriaceae. wherein Enterobacteriaceae is Klebsiella pneumoniae or Escherichia coli.

25. A compound or pharmaceutically acceptable salt according to any one of claims 1 to 16 for the treatment or prophylaxis of bacterial infection, particularly the bacteria is gram -negative bacteria.

26. A compound according to claim 25, wherein the gram-negative bacteria is selected from Enterobacteriaceae, Neisseria gonorrhoeae, Haemophilus influenzae, Helicobacter pylorus, Acinetobacter baumannii and Pseudomonas aeruginosa.

27. A compound according to claim 26, wherein the gram-negative bacteria is Enterobacteriaceae, wherein Enterobacteriaceae is Klebsiella pneumoniae or Escherichia coli.

28. A compound or pharmaceutically acceptable salt according to any one of claims 1 to 16, when manufactured according to a process of claim 17.

29. A method for the treatment or prophylaxis of bacterial infection, particularly the bacteria is gram-negative bacteria, which method comprises administering a therapeutically effective amount of a compound as defined in any one of claims 1 to 16.

Description:
Sulfonylpiperazinyl compounds for treatment of bacterial infections

The present invention relates to organic compounds useful for the treatment and/or prevention of bacterial infections in a mammal. Specifically these molecules can inhibit the LPS synthesis pathway, in particular to inhibit LpxH, and are useful for treating bacterial infections. BACKGROUND OF THE INVENTION

The intensive use of antibiotics has exerted a selective evolutionary pressure on microorganisms to produce genetically based resistance mechanisms. Modern medicine and socio-economic behaviour exacerbate the problem of resistance development by creating slow growth situations for pathogenic microbes, e.g. in artificial joints, and by supporting long-term host reservoirs, e.g. in immune-compromised patients. In hospital settings, an increasing number of strains of Staphylococcus aureus, Streptococcus pneumoniae, Enterococcus spp., Enter obacteriaceae such as Klebsiella pneumonia, Acinetobacter baumannii and Pseudomonas aeruginosa, major sources of infections, are becoming multi-drug resistant and therefore difficult to treat. This is particularly the case for Gram-negative organisms where the situation is getting worrisome since no novel agents with a differentiated mechanism of action have been approved for decades. Therefore, there is an important medical need for new antibacterial compounds addressing Gram-negative resistant bacteria, in particular third generation cephalosporins- and carbapenem-resistant Enter obacteriaceae and multi-drug-resistant Pseudomonas aeruginosa and Acinetobacter baumannii. One way to tackle the problem of cross-resistance to established classes of antibiotics is to inhibit an essential protein or function not targeted by current antibiotics.

Gram-negative bacteria are unique in that their outer membrane contains Lipopolysaccharide (LPS), which is crucial for maintaining membrane integrity, and is essential for bacterial viability (reviewed in Ann. Rev. Biochem 76: 295-329, 2007). The major lipid component of LPS is Lipid A, and inhibition of Lipid A biosynthesis is lethal to bacteria. Lipid A is synthesized on the cytoplasmic surface of the bacterial inner membrane via a pathway that consists of nine different enzymes. These enzymes are highly conserved in most Gram-negative bacteria. LpxH, a calcineurin-like phosphatase (CLP), catalyzes the hydrolysis of UDP-2,3- diacyl-glucosamine (UDP-DAGn) to yield Lipid X and UMP (22, 24, 25). LpxH has no mammalian homologue, making it a good target for the development of novel antibiotics targeting Gram-negative bacteria.

SUMMARY OF THE INVENTION

The present invention relates to novel compounds of formula (I), wherein

R 1 is amino, hydroxyC 1-6 alkyl, C 1-6 alkylamino, aminoC 1-6 alkylamino or piperazinyl;

R 2 is halogen or C 1-6 alkyl;

R 3 is -Y-C(O)-R 4 ; wherein

Y is C 3-10 cycloalkyl, phenyl, pyrazinyl or pyridinyl;

R 4 is ((aminoC 1-6 alkyl) 2 amino)C 1-6 alkylamino, ((aminoC 1-6 alkyl)pyrrolidiniumyl)C 1- 6 alkylamino, ((aminoC 1-6 alkylcarbonyl)amino)C 1-6 alkylamino, ((C 1-6 alkyl) 2 amino)C 1- 6 alkylamino, ((C 1-6 alkyl) 3 ammonio)C 1-6 alkylamino, (aminoC 1-6 alkoxy)C 1-6 alkylamino, (aminoC 1-6 alkyl(C 1-6 alkyl) 2 ammonio)C 1-6 alkylamino, (C 1-6 alkyl(aminoC 1- 6 alkyl) 2 ammonio)C 1-6 alkylamino, ((aminoC 1-6 alkylcarbonyl)amino)C3- 1 0 cycloalkyl amino, ((aminoC 1-6 alkyl) 2 pyrrolidinyl)C 3-10 cycloalkylamino, (aminoC 1- 6 alkyl) 2 amino, (aminoC 1-6 alkyl)azetidinyl, (aminoC 1-6 alkyl)C 3-10 cycloalkylamino, (aminoC 1-6 alkyl)piperazinyl, (aminoC 1-6 alkylamino)C 1-6 alkylamino, (aminoC 1- 6 alkylcarbonyl)aminocarbamoyl, (aminopyrrolidinyl) C 3-10 cycloalkylamino, (azetidinylC 1-6 alkylamino)C 1-6 alkylamino, (C 1-6 alkylamino)C 1-6 alkylamino, (C 1- 6 alkylpyrrolidiniumyl)C 1-6 alkylamino, (diaminopiperidinyl)C 1-6 alkylamino, (diaminopiperidinyl)C 3-10 cycloalkylamino, (piperidinylcarbonylamino)carbamoyl, 3- azabicyclo[3.1.0]hexanylamino, amino, amino(C 1-6 alkoxy)C 1-6 alkylamino, amino(C 1- 6 alkyl)piperidinyl, (aminoC 1-6 alkyl) 2 pyrrolidinyl, amino(hydroxy)C 1-6 alkylamino, aminoC 1-6 alkylamino, aminoC 3-10 cycloalkylamino, aminocarbonylC 1-6 alkylamino, aminopyrrolidinylamino, amino(C 1-6 alkyl)pyrrolidinylamino, azetidinylC 1-6 alkylamino, C 1-6 alkoxy, C 1-6 alkyl(aminoC 1-6 alkyl)amino, C 1-6 alkylpyrrolidinylamino, C 1- 6 alkyl(aminoC 1-6 alkyl)piperaziniumyl, C 1-6 alkyl-2,6-diazaspiro[3.3]heptanyl, C 1- 6 alkylamino, diaminoC 1-6 alkylamino, diaminopiperidinyl, hydroxyC 1-6 alkylamino, hydroxypyrrolidinylamino, morpholinylC 1-6 alkylamino or pyrrolidinylC 1-6 alkylamino; X is CH 2 or O;

A is CH or N;

Q 1 is CH or N;

Q 2 is CH or N;

W is haloC 1-6 alkyl; or a pharmaceutically acceptable salt thereof.

DETAILED DESCRIPTION OF THE INVENTION

DEFINITIONS

The term “C 1-6 alkyl” denotes a saturated, linear or branched chain alkyl group containing 1 to 6, particularly 1 to 4 carbon atoms, for example methyl, ethyl, //-propyl, isopropyl, n-butyl, isobutyl, tert-butyl and the like. Particular “C 1-6 alkyl” groups are methyl, ethyl and propyl.

The term “C 1-6 alkoxy” denotes C 1-6 alkyl-O-.

The term “amino”, alone or in combination, signifies the primary amino group, the secondary amino group, or the tertiary amino group.

The term “halogen” and “halo” are used interchangeably herein and denote fluoro, chloro, bromo, or iodo.

The term “haloC 1-6 alkyl” denotes a C 1-6 alkyl group wherein at least one of the hydrogen atoms of the C 1-6 alkyl group has been replaced by same or different halogen atoms, particularly fluoro atoms. Examples of haloC 1-6 alkyl include monofluoro-, difluoro- or trifluoro-methyl, - ethyl or -propyl, for example 3,3,3-trifluoropropyl, 2-fluoroethyl, trifluoroethyl, fluoromethyl, difluoromethyl, difluoroethyl or trifluoromethyl.

The term “ C 3-10 cycloalkyl” denotes saturated carbon monocyclic, bicyclic or tricyclic ring containing from 3 to 10 carbon atoms, particularly from 3 to 8 carbon atoms. C 3-10 cycloalkyl includes, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, bicyclo[l. l.l]pentanyl, bicyclo[2.2.2]octanyl, bicyclo[3.1.0]hexanyl, norbomanyl, adamantanyl and the like. Particular “ C 3-10 cycloalkyl” groups are bicyclo[l. l.l]pentanyl, bicyclo[2.2.2]octanyl, bicyclo[3.1.0]hexanyl, cyclohexanyl, cyclopentanyl and norbomanyl.

The term “pharmaceutically acceptable salts” denotes salts which are not biologically or otherwise undesirable. Pharmaceutically acceptable salts include both acid and base addition salts.

The term “pharmaceutically acceptable acid addition salt” denotes those pharmaceutically acceptable salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, carbonic acid, phosphoric acid, and organic acids selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, gluconic acid, lactic acid, pyruvic acid, oxalic acid, malic acid, maleic acid, maloneic acid, succinic acid, fumaric acid, tartaric acid, citric acid, aspartic acid, ascorbic acid, glutamic acid, anthranilic acid, benzoic acid, cinnamic acid, mandelic acid, embonic acid, phenylacetic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, and salicyclic acid.

The term “pharmaceutically acceptable base addition salt” denotes those pharmaceutically acceptable salts formed with an organic or inorganic base. Examples of acceptable inorganic bases include sodium, potassium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, and aluminum salts. Salts derived from pharmaceutically acceptable organic nontoxic bases includes salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-diethylaminoethanol, trimethamine, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperizine, piperidine, A-ethylpiperidine, and polyamine resins.

The term “A pharmaceutically active metabolite” denotes a pharmacologically active product produced through metabolism in the body of a specified compound or salt thereof. After entry into the body, most drugs are substrates for chemical reactions that may change their physical properties and biologic effects. These metabolic conversions, which usually affect the polarity of the compounds of the invention, alter the way in which drugs are distributed in and excreted from the body. However, in some cases, metabolism of a drug is required for therapeutic effect.

The term “therapeutically effective amount” denotes an amount of a compound or molecule of the present invention that, when administered to a subject, (i) treats or prevents the particular disease, condition or disorder, (ii) attenuates, ameliorates or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition or disorder described herein. The therapeutically effective amount will vary depending on the compound, the disease state being treated, the severity of the disease treated, the age and relative health of the subject, the route and form of administration, the judgement of the attending medical or veterinary practitioner, and other factors. The term “pharmaceutical composition” denotes a mixture or solution comprising a therapeutically effective amount of an active pharmaceutical ingredient together with pharmaceutically acceptable excipients to be administered to a mammal, e.g., a human in need thereof.

INHIBITOR OF UDP-2,3-DIACYLGLUCOSAMINE HYDROLASE (LpxH)

The present invention relates to (i) a compound of formula (I), wherein

R 1 is amino, hydroxyC 1-6 alkyl, C 1-6 alkylamino, aminoC 1-6 alkylamino or piperazinyl;

R 2 is halogen or C 1-6 alkyl;

R 3 is -Y-C(O)-R 4 ; wherein

Y is C 3-10 cycloalkyl, phenyl, pyrazinyl or pyridinyl;

R 4 is ((aminoC 1-6 alkyl) 2 amino)C 1-6 alkylamino, ((aminoC 1-6 alkyl)pyrrolidiniumyl)C 1- 6 alkylamino, ((aminoC 1-6 alkylcarbonyl)amino)C 1-6 alkylamino, ((C 1-6 alkyl) 2 amino)C 1- 6 alkylamino, ((C 1-6 alkyl)3ammonio)C 1-6 alkylamino, (aminoC 1-6 alkoxy)C 1-6 alkylamino, (aminoC 1-6 alkyl(C 1-6 alkyl) 2 ammonio)C 1-6 alkylamino, (C 1-6 alkyl(aminoC 1- 6 alkyl) 2 ammonio)C 1-6 alkylamino, ((aminoC 1-6 alkylcarbonyl)amino)C3- 1 0 cycloalkyl amino, ((aminoC 1-6 alkyl) 2 pyrrolidinyl)C 3-10 cycloalkylamino, (aminoC 1- 6 alkyl) 2 amino, (aminoC 1-6 alkyl)azetidinyl, (aminoC 1-6 alkyl)C 3-10 cycloalkylamino, (aminoC 1-6 alkyl)piperazinyl, (aminoC 1-6 alkylamino)C 1-6 alkylamino, (aminoC 1- 6 alkylcarbonyl)aminocarbamoyl, (aminopyrrolidinyl)C 3-10 cycloalkylamino, (azetidinylC 1-6 alkylamino)C 1-6 alkylamino, (C 1-6 alkylamino)C 1-6 alkylamino, (C 1- 6 alkylpyrrolidiniumyl)C 1-6 alkylamino, (diaminopiperidinyl)C 1-6 alkylamino, (diaminopiperidinyl)C 3-10 cycloalkylamino, (piperidinylcarbonylamino)carbamoyl, 3- azabicyclo[3.1.0]hexanylamino, amino, amino(C 1-6 alkoxy)C 1-6 alkylamino, amino(C 1- 6 alkyl)piperidinyl, (aminoC 1-6 alkyl^pyrrolidinyl, amino(hydroxy)C 1-6 alkylamino, aminoC 1-6 alkylamino, aminoC 3-10 cycloalkylamino, aminocarbonylC 1-6 alkylamino, aminopyrrolidinylamino, amino(C 1-6 alkyl)pyrrolidinylamino, azetidinylC 1-6 alkylamino, C 1-6 alkoxy, C 1-6 alkyl(aminoC 1-6 alkyl)amino, C 1-6 alkylpyrrolidinylamino, C 1- 6 alkyl(aminoC 1-6 alkyl)piperaziniumyl, C 1-6 alkyl-2,6-diazaspiro[3.3]heptanyl, C 1- 6 alkylamino, diaminoC 1-6 alkylamino, diaminopiperidinyl, hydroxyC 1-6 alkylamino, hydroxypyrrolidinylamino, morpholinylC 1-6 alkylamino or pyrrolidinylC 1-6 alkylamino;

X is CH 2 or O;

A is CH or N;

Q 1 is CH orN;

Q 2 is CH orN;

W is haloC 1-6 alkyl; or a pharmaceutically acceptable salt thereof.

The present invention relates to (i’) a compound of formula (I), wherein

R 1 is amino, hydroxyC 1-6 alkyl, aminoC 1-6 alkylamino or piperazinyl;

R 2 is halogen or C 1-6 alkyl;

R 3 is -Y-C(O)-R 4 ; wherein

Y is C 3-10 cycloalkyl, phenyl, pyrazinyl or pyridinyl;

R 4 is ((aminoC 1-6 alkyl) 2 amino)C 1-6 alkylamino, ((aminoC 1-6 alkyl)pyrrolidiniumyl)C 1- 6 alkylamino, ((aminoC 1-6 alkylcarbonyl)amino)C 1-6 alkylamino, ((C 1-6 alkyl) 2 amino)C 1- 6 alkylamino, ((C 1-6 alkyl)3ammonio)C 1-6 alkylamino, (aminoC 1-6 alkoxy)C 1-6 alkylamino, (aminoC 1-6 alkyl(C 1-6 alkyl) 2 ammonio)C 1-6 alkylamino, (aminoC 1-6 alkyl^amino, (aminoC 1-6 alkyl)azetidinyl, (aminoC 1-6 alkyl)C 3-10 cycloalkylamino, (aminoC 1- 6 alkyl)piperazinyl, (aminoC 1-6 alkylamino)C 1-6 alkylamino, (aminoC 1- 6 alkylcarbonyl)aminocarbamoyl, (azetidinylC 1-6 alkylamino)C 1-6 alkylamino, (C 1- 6 alkylamino)C 1-6 alkylamino, (C 1-6 alkylpyrrolidiniumyl)C 1-6 alkylamino, (diaminopiperidinyl)C 1-6 alkylamino, (piperidinylcarbonylamino)carbamoyl, 3- azabicyclo[3.1.0]hexanylamino, amino, amino(C 1-6 alkoxy)C 1-6 alkylamino, amino(C 1- 6 alkyl)piperidinyl, amino(hydroxy)C 1-6 alkylamino, aminoC 1-6 alkylamino, aminoC 3- 10 cycloalkyl amino, aminocarbonylC 1-6 alkylamino, aminopyrrolidinylamino, azetidinylC 1-6 alkylamino, C 1-6 alkoxy, C 1-6 alkyl(aminoC 1-6 alkyl)amino, C 1- 6 alkyl(aminoC 1-6 alkyl)piperaziniumyl, C 1-6 alkyl-2,6-diazaspiro[3.3]heptanyl, C 1- 6 alkylamino, diaminoC 1-6 alkylamino, diaminopiperidinyl, hydroxyC 1-6 alkylamino, hydroxypyrrolidinylamino, morpholinylC 1-6 alkylamino or pyrrolidinylC 1-6 alkylamino;

X is CH 2 or O;

A is CH or N;

Q 1 is CH or N;

Q 2 is CH or N;

W is haloC 1-6 alkyl; or a pharmaceutically acceptable salt thereof.

A further embodiment of present invention is (ii’) a compound of formula (I) according to (i), or a pharmaceutically acceptable salt thereof, wherein R 1 is amino or hydroxyC 1-6 alkyl.

A further embodiment of present invention is (ii) a compound of formula (I) according to (i), or a pharmaceutically acceptable salt thereof, wherein R 1 is amino, C 1-6 alkylamino or hydroxyC 1-6 alkyl.

A further embodiment of present invention is (iii’) a compound of formula (I) according to (i) or (ii), or a pharmaceutically acceptable salt thereof, wherein R 1 is amino or hydroxymethyl.

A further embodiment of present invention is (iii) a compound of formula (I) according to (i) or (ii), or a pharmaceutically acceptable salt thereof, wherein R 1 is amino, methylamino or hydroxymethyl.

A further embodiment of present invention is (iv) a compound of formula (I), according to any one of (i) to (iii), or a pharmaceutically acceptable salt thereof, wherein R 2 is halogen.

A further embodiment of present invention is (v) a compound of formula (I) according to any one of (i) to (iv), wherein R 2 is chloro.

A further embodiment of present invention is (vi) a compound of formula (I), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (v), wherein R 3 is -Y- C(O)-R 4 ; wherein Y is C 3-10 cycloalkyl, phenyl or pyrazinyl.

A further embodiment of present invention is (vii’) a compound of formula (I), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (vi), wherein R 3 is -Y- C(O)-R 4 ; wherein Y is bicyclo[2.2.2]octanyl, bicyclo[3.1.0]hexanyl, cyclohexanyl, phenyl or pyrazinyl.

A further embodiment of present invention is (vii) a compound of formula (I), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (vi), wherein R 3 is -Y- C(O)-R 4 ; wherein Y is bicyclo[2.2.2]octanyl, bicyclo[3.1.0]hexanyl, cyclohexanyl, norbornanyl, phenyl or pyrazinyl.

A further embodiment of present invention is (viii ’ ) a compound of formula (I), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (vii), wherein R 3 is -Y- C(O)-R 4 ; wherein R 4 is ((aminoC 1-6 alkyl) 2 amino)C 1-6 alkylamino, ((aminoC 1- 6 alkyl)pyrrolidiniumyl)C 1-6 alkylamino, (aminoC 1-6 alkyl(C 1-6 alkyl) 2 ammonio)C 1-6 alkylamino, (aminoC 1-6 alkyl) 2 amino, (diaminopiperidinyl)C 1-6 alkylamino, amino, amino(hydroxy)C 1- 6 alkylamino or aminoC 1-6 alkylamino.

A further embodiment of present invention is (viii) a compound of formula (I), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (vii), wherein R 3 is -Y- C(O)-R 4 ; wherein R 4 is amino(C 1-6 alkyl)pyrrolidinylamino, ((aminoC 1-6 alkyl) 2 amino)C 1- 6 alkylamino, ((aminoC 1-6 alkyl) 2 pyrrolidinyl)C 3-10 cycloalkylamino, ((aminoC 1- 6 alkyl)pyrrolidiniumyl)C 1-6 alkylamino, (aminoC 1-6 alkyl(C 1-6 alkyl) 2 ammonio)C 1-6 alkylamino, (aminoC 1-6 alkyl) 2 amino, (aminoC 1-6 alkyl^pyrrolidinyl, (aminoC 1-6 alkyl)C 3-10 cycloalkylamino, (C 1-6 alkyl(aminoC 1-6 alkyl) 2 ammonio)C 1-6 alkylamino, (aminopyrrolidinyl)C 3-10 cycloalkylamino, (C 1-6 alkyl(aminoC 1-6 alkyl) 2 ammonio)C 1-6 alkylamino, (diaminopiperidinyl)C 1-6 alkylamino, (diaminopiperidinyl)C 3-10 cycloalkylamino, amino, amino(hydroxy)C 1-6 alkylamino, aminoC 1- 6 alkylamino, aminoC 3-10 cycloalkylamino, C 1-6 alkylpyrrolidinylamino, diaminoC 1-6 alkylamino or diaminopiperidinyl .

A further embodiment of present invention is (ix’) a compound of formula (I), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (viii), wherein R 4 is ((2- aminoethyl) 2 amino)propylamino, ((2-aminoethyl)pyrrolidin-l-ium-l-yl)ethylamino, (2- aminoethyl(dimethyl)ammonio)ethylamino, (3 -aminopropyl ) 2 amino, (3,5-diamino-l- piperidinyl)propylamino, amino, 3-amino-2-hydroxy-propylamino, 3 -aminopropylamino, 3- aminopropylamino or 2-aminoethylamino.

A further embodiment of present invention is (ix) a compound of formula (I), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (viii), wherein R 4 is ((2- aminoethyl) 2 amino)propylamino, ((2-aminoethyl)pyrrolidin-l-ium-l-yl)ethylamino, (2- aminoethyl(dimethyl)ammonio)ethylamino, (2-aminoethyl) 2 amino, (3,5-diamino-l- piperidinyl)propylamino, (3-aminocyclopentyl)amino, (3-aminopropyl) 2 amino, (4- methylpyrrolidin-3-yl)amino, [3-(3,5-diamino-l-piperidinyl)cyclopentyl]amino, [3-[3,4- bis(aminomethyl)pyrrolidin-l-yl]cyclopentyl]amino, [3-[3-aminopyrrolidin-l- yl]cyclopentyl]amino, l-(aminomethyl)cyclopropylamino, 2-(2-aminoethylamino)ethylamino, 2,3 -diaminopropylamino, 2-aminoethyl amino, 3,4-bis(aminomethyl)pyrrolidin-l-yl, 3,5- diaminopiperidinyl, 3-[3-aminopropyl(dimethyl)ammonio]propylamino, 3-[bis(2-aminoethyl)- methyl-ammonio]propylamino, 3-amino-2-hydroxy-propylamino, 3 -aminopropylamino, 3- hydroxypropylamino, 4-amino-2-methyl-pyrrolidin-l-yl, amino or aminopropylamino.

A further embodiment of present invention is (x) a compound of formula (I), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (ix), wherein A is CH.

A further embodiment of present invention is (xi) a compound of formula (I), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (x), wherein Q 1 is N.

A further embodiment of present invention is (xii) a compound of formula (I), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (xi), wherein Q 2 is CH.

A further embodiment of present invention is (xiii) a compound of formula (I), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (xii), wherein W is CF2.

A further embodiment of present invention is (xiv’) a compound of formula (I), according to any one of (i) to (xiii), wherein

R 1 is amino or hydroxyC 1-6 alkyl;

R 2 is halogen;

R 3 is -Y-C(O)-R 4 ; wherein

Y is C 3-10 cycloalkyl, phenyl or pyrazinyl;

R 4 is ((aminoC 1-6 alkyl) 2 amino)C 1-6 alkylamino, ((aminoC 1-6 alkyl)pyrrolidiniumyl)C 1- 6 alkylamino, (aminoC 1-6 alkyl(C 1-6 alkyl) 2 ammonio)C 1-6 alkylamino, (aminoC 1- 6 alkyl) 2 amino, (diaminopiperidinyl)C 1-6 alkylamino, amino, amino(hydroxy)C 1- 6 alkylamino or aminoC 1-6 alkylamino;

X is CH 2 or O;

A is CH;

Q 1 is N;

Q 2 is CH;

W is haloC 1-6 alkyl; or a pharmaceutically acceptable salt thereof.

A further embodiment of present invention is (xiv) a compound of formula (I), according to any one of (i) to (xiii), wherein

R 1 is amino or hydroxyC 1-6 alkyl;

R 2 is halogen;

R 3 is -Y-C(O)-R 4 ; wherein Y is C 3-10 cycloalkyl, phenyl or pyrazinyl;

R 4 is amino(C 1-6 alkyl)pyrrolidinylamino, ((aminoC 1-6 alkyl) 2 amino)C 1-6 alkylamino, ((aminoC 1-6 alkyl) 2 pyrrolidinyl)C 3-10 cycloalkylamino, ((aminoC 1- 6 alkyl)pyrrolidiniumyl)C 1-6 alkylamino, (aminoC 1-6 alkyl(C 1-6 alkyl) 2 ammonio)C 1- 6 alkylamino, (aminoC 1-6 alkyl^amino, (aminoC 1-6 alkyl^pyrrolidinyl, (aminoC 1- 6 alkyl)C 3-10 cycloalkylamino, (C 1-6 alkyl(aminoC 1-6 alkyl) 2 ammonio)C 1-6 alkylamino, (aminopyrrolidinyl)C 3-10 cycloalkylamino, (C 1-6 alkyl(aminoC 1-6 alkyl) 2 ammonio)C 1- 6 alkylamino, (diaminopiperidinyl)C 1-6 alkylamino, (diaminopiperidinyl)C3- 1 0 cycloalkyl amino, amino, amino(hydroxy)C 1-6 alkylamino, aminoC 1-6 alkylamino, aminoC 3-10 cycloalkylamino, C 1-6 alkylpyrrolidinylamino, diaminoC 1-6 alkylamino or diaminopiperidinyl;

X is CH 2 or O;

A is CH;

Q 1 is N;

Q 2 is CH;

W is haloC 1-6 alkyl; or a pharmaceutically acceptable salt thereof.

A further embodiment of present invention is (xv’) a compound of formula (I), according to any one of (i) to (xiv), wherein

R 1 is amino or hydroxymethyl;

R 2 is chloro;

R 3 is -Y-C(O)-R 4 ; wherein

Y is bicyclo[2.2.2]octanyl, bicyclo[3.1.0]hexanyl, cyclohexanyl, phenyl or pyrazinyl;

R 4 is ((2-aminoethyl) 2 amino)propylamino, ((2-aminoethyl)pyrrolidin-l-ium-l- yl)ethylamino, (2-aminoethyl(dimethyl)ammonio)ethylamino, (3 -aminopropyl) 2 amino, (3,5-diamino-l-piperidinyl)propylamino, amino, 3-amino-2-hydroxy-propylamino, 3- aminopropylamino, 3 -aminopropylamino or 2-aminoethylamino;

X is CH 2 or O;

A is CH;

Q 1 is N;

Q 2 is CH;

W is CF 2; or a pharmaceutically acceptable salt thereof. A further embodiment of present invention is (xv) a compound of formula (I), according to any one of (i) to (xiv), wherein R 1 is amino or hydroxymethyl;

R 2 is chloro;

R 3 is -Y-C(O)-R 4 ; wherein

Y is bicyclo[2.2.2]octanyl, bicyclo[3.1.0]hexanyl, cyclohexanyl, phenyl or pyrazinyl;

R 4 is ((2-aminoethyl) 2 amino)propylamino, ((2-aminoethyl)pyrrolidin-l-ium-l- yl)ethylamino, (2-aminoethyl(dimethyl)ammonio)ethylamino, (2-aminoethyl) 2 amino, (3,5-diamino-l-piperidinyl)propylamino, (3-aminocyclopentyl)amino, (3- aminopropyl) 2 amino, (4-methylpyrrolidin-3-yl)amino, [3-(3,5-diamino-l- piperidinyl)cyclopentyl]amino, [3-[3,4-bis(aminomethyl)pyrrolidin-l- yl]cyclopentyl]amino, [3-[3-aminopyrrolidin-l-yl]cyclopentyl]amino, 1- (aminomethyl)cyclopropylamino, 2-(2-aminoethylamino)ethylamino, 2,3- diaminopropylamino, 2-aminoethylamino, 3,4-bis(aminomethyl)pyrrolidin-l-yl, 3,5- diaminopiperidinyl, 3-[3-aminopropyl(dimethyl)ammonio]propylamino, 3-[bis(2- aminoethyl)-methyl-ammonio]propylamino, 3-amino-2-hydroxy-propylamino, 3- aminopropylamino, 3 -hydroxypropylamino, 4-amino-2-methyl-pyrrolidin-l-yl, amino or aminopropylamino;

X is CH 2 or O;

A is CH;

Q 1 is N;

Q 2 is CH;

W is CF 2; or a pharmaceutically acceptable salt thereof.

Another embodiment of present invention is a compound of formula (I) selected from the following:

Trans-methyl 4-[[2-[4-[4-(4-amino-2-oxo-pyrrolidin-l-yl)phenyl]sulfonylpi perazin-l-yl]- 6-chloro-4-pyridyl]-difluoro-methyl]cyclohexanecarboxylate;

Trans-4-[[2-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phe nyl]sulfonylpiperazin-l-yl]-6- chloro-4-pyridyl]-difluoro-methyl]-N-(azetidin-3-ylmethyl)cy clohexanecarboxamide;

Trans-4-[[2-[4-[4-(4-amino-2-oxo-pyrrolidin-l-yl)phenyl]s ulfonylpiperazin-l-yl]-6-chloro- 4-pyridyl]-difluoro-methyl]-N-(3-hydroxypropyl)cyclohexaneca rboxamide; Trans-(4R)-4-amino-l-[4-[4-[4-[[4-(4-amino-4-methyl-piperidi ne-l-carbonyl)cyclohexyl]- difluoro-methyl]-6-chloro-2-pyridyl]piperazin-l-yl]sulfonylp henyl]pyrrolidin-2-one;

Trans-(4R)-4-amino-l-[4-[4-[6-chloro-4-[difluoro-[4-(6-me thyl-2,6-diazaspiro[3.3]heptane-2- carbonyl)cyclohexyl]methyl]-2-pyridyl]piperazin-l-yl]sulfony lphenyl]pyrrolidin-2-one;

Trans-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin -l-yl]phenyl]sulfonylpiperazin- l-yl]-4-pyridyl]-difluoro-methyl]-N-(4-hydroxypyrrolidin-3-y l)cyclohexanecarboxamide;

7ra//.s-N-(3-aminocyclobutyl)- 4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]cyclohexanecarboxamide;

Trans-N-(2-amino-l-methyl-ethyl)-4-[[2-chloro-6-[4-[4-[(4 R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]cyclohexanecarboxamide;

Trans-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin -l-yl]phenyl]sulfonylpiperazin- l-yl]-4-pyridyl]-difluoro-methyl]-N-(2-pyrrolidin-l-ylethyl) cyclohexanecarboxamide;

Trans-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin -l-yl]phenyl]sulfonylpiperazin- l-yl]-4-pyridyl]-difluoro-methyl]-N-[(lS,5R)-3-azabicyclo[3. 1.0]hexan-6- yl]cyclohexanecarboxamide;

Trans-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin -l-yl]phenyl]sulfonylpiperazin- l-yl]-4-pyridyl]-difluoro-methyl]-N-[ci.s-4-aminopyrrolidin- 3-yl]cyclohexanecarboxamide;

Trans-N-[l-(aminomethyl)cyclopropyl]-4-[[2-[4-[4-[(4R)-4- amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluo ro-methyl]cyclohexanecarboxamide;

Trans-(4R)-4-amino-l-[4-[4-[4-[[4-[4-(2-aminoethyl)pipera zine-l-carbonyl]cyclohexyl]- difluoro-methyl] -6-chloro-2-pyridyl]piperazin-l-yl]sulfonylphenyl]pyrrolidin -2-one;

Trans-4-[[2-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phe nyl]sulfonylpiperazin-l-yl]-6- chloro-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide;

4-[[2-[4-[4-(4-amino-2-oxo-pyrrolidin-l-yl)phenyl]sulfony lpiperazin-l-yl]-6-chl oro-4- pyridyl] -difluoro-methyl] -N-(3 -hydroxypropyl )benzamide;

4-[[2-[4-[4-(4-amino-2-oxo-pyrrolidin-l-yl)phenyl]sulfony lpiperazin-l-yl]-6-chl oro-4- pyridyl] -difluoro-methyl] -N-(3 -aminopropyl )benzamide;

N-(2-aminoethyl)-4-[[2-[4-[4-(4-amino-2-oxo-pyrrolidin-l- yl)phenyl]sulfonylpiperazin-l- yl]-6-chloro-4-pyridyl]-difluoro-methyl]benzamide;

N-(3-amino-2-hydroxy-propyl)-4-[[2-[4-[4-(4-amino-2-oxo-p yrrolidin-l- yl)phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluo ro-methyl]benzamide;

N-(3-amino-2-hydroxy-propyl)-4-[[2-chloro-6-[4-[4-[(4R)-4 -amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]benzamide; 4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phe nyl]sulfonylpiperazin-l-yl]- 4-pyridyl]-difluoro-methyl]-N-[3-(dimethylamino)propyl]benza mide;

N-(3-aminopropyl)-5-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-ox o-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]pyrazine-2-carboxamide;

N-(3-amino-2-hydroxy-propyl)-5-[[2-chloro-6-[4-[4-[(4R)-4 -amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]pyrazine-2-carboxamide;

5-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl] phenyl]sulfonylpiperazin-l-yl]- 4-pyridyl]-difluoro-methyl]-N-[3-(dimethylamino)propyl]pyraz ine-2-carboxamide;

N-(2-aminoethyl)-5-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo -pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]pyrazine-2-carboxamide;

5-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl] phenyl]sulfonylpiperazin-l-yl]- 4-pyridyl]-difluoro-methyl]-N-(2-morpholinoethyl)pyrazine-2- carboxamide;

N-(3-aminopropyl)-6-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-ox o-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]pyridine-3-carboxamide;

6-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl] phenyl]sulfonylpiperazin-l-yl]-

4-pyridyl]-difluoro-methyl]-N-[3-(dimethylamino)propyl]py ridine-3-carboxamide;

N-[2-(2-aminoethylamino)ethyl]-6-[[2-chloro-6-[4-[4-[(4R) -4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]pyridine-3-carboxamide;

5-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl] phenyl]sulfonylpiperazin-l-yl]-

4-pyridyl]-difluoro-methyl]-N-[3-(methylamino)propyl]pyri dine-2-carboxamide;

Trans-N-(2-aminoethyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino -2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]-N-methyl- cyclohexanecarboxamide;

C/s-N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino- 2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]cyclohexanecarboxamide;

Cis-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l -yl]phenyl]sulfonylpiperazin-l- yl ] -4 -pyri dyl ] -difluoro-methyl ] -N-(3 -hy droxypropyl )cy cl ohexanecarb oxami de;

N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-ox o-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]bicyclo[2.2.2]octane-l- carboxamide;

N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-ox o-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]norbornane-l-carboxamide; N-(3-aminopropyl)-3-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-p yrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]cyclopentanecarboxamide;

Trans-N-(3-amino-2-hydroxy-propyl)-4-[[2-chloro-6-[4-[4-[ (4R)-4-amino-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-di fluoro- methyl]cyclohexanecarboxamide;

3-[[4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l -yl]phenyl]sulfonylpiperazin-l-yl]-4- pyridyl]-difluoro-methyl]benzoyl]amino]propyl-trimethyl-ammo nium;

4-[[2-Chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl] phenyl]sulfonylpiperazin-l-yl]-

4-pyridyl]-difluoro-m ethyl] -N-[3 -(1 -methylpyrrolidin- 1 -ium- 1 -yl)propyl]benzamide;

5-[[2-Chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl] phenyl]sulfonylpiperazin-l-yl]- 4-pyridyl]-difluoro-methyl]-N-[3-(2-hydroxyethylamino)propyl ]pyridine-2-carboxamide;

N-(3-aminopropyl)-3-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-ox o-pyrrolidin-l- yl]phenyl] sulfonylpiperazin- 1 -yl ] -4-pyri dy 1] -difluoro-methyl]bicyclo[ 1.1.1 ]pentane- 1 - carboxamide;

Trans-N-(2-aminoethyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino -2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]cyclohexanecarboxamide;

Trans-N-[2-(2-aminoethoxy)ethyl]-4-[[2-chloro-6-[4-[4-[(4 R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]cyclohexanecarboxamide;

Trans-N,N-bis(2-aminoethyl)-4-[[2-chloro-6-[4-[4-[(4R)-4- amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]cyclohexanecarboxamide;

Trans-N,N-bis(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4 -amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]cyclohexanecarboxamide; N,N-bis(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2- oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]bicyclo[2.2.2]octane-l- carboxamide; N,N-bis(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2- oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]norbornane-l-carboxamide;

Trans-N-[3-[bis(2-aminoethyl)amino]propyl]-4-[[2-chloro-6 -[4-[4-[(4R)-4-amino-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-di fluoro- methyl]cyclohexanecarboxamide;

Trans-(4R)-4-amino-l-[4-[4-[6-chloro-4-[[4-cis-(3,5-diami nopiperidine-l- carbonyl)cyclohexyl]-difluoro-methyl]-2-pyridyl]piperazin-l- yl]sulfonylphenyl]pyrrolidin-2-one; Trans-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin- l-yl]-4-pyridyl]-difluoro-methyl]-N-[3-[cis-3,5-diamino-l- piperidyl]propyl]cyclohexanecarboxamide;

Trans-N'-(2-aminoacetyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-ami no-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]cyclohexanecarbohydrazide;

Trans-N'-[4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrol idin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]cyclohexanecarbonyl]piperidine-4- carbohydrazide;

Trans-N-(2-amino-3-hydroxy-propyl)-4-[[2-chloro-6-[4-[4-[ (4R)-4-amino-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-di fluoro- methyl]cyclohexanecarboxamide;

(2R,5S)-5-[[2-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]p henyl]sulfonylpiperazin-l-yl]-6- chloro-4-pyridyl]-difluoro-methyl]-N-(3-aminopropyl)bicyclo[ 2.2.2]octane-2-carboxamide;

Trans-N-(3-aminopropyl)-4-[difluoro-[6-methyl-2-[4-[4-[(4 R)-4-amino-2-oxo-pyrrolidin- l-yl]phenyl]sulfonylpiperazin-l-yl]pyrimidin-4-yl]methyl]cyc lohexanecarboxamide;

N-(3-aminopropyl)-6-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-ox o-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]bicyclo[3.1.0]hexane-3- carboxamide;

Trans-N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amin o-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]cyclohexanecarboxamide;

Trans-(4R)-4-amino-l-[4-[4-[4-[[4-[3-(aminomethyl)azetidi ne-l-carbonyl]cyclohexyl]- difluoro-methyl] -6-chloro-2-pyridyl]piperazin-l-yl]sulfonylphenyl]pyrrolidin -2-one;

Trans-N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(5S)-5-(hyd roxymethyl)-2-oxo-oxazolidin- 3-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-meth yl]cyclohexanecarboxamide;

Trans-N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(5R)-5-(hyd roxymethyl)-2-oxo- oxazolidin-3-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-di fluoro- methyl]cyclohexanecarboxamide;

Trans-N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-(2-oxo-4-pip erazin-l-yl-pyrrolidin-l- yl)phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]cyclohexanecarboxamide;

Trans-N-(3-aminopropyl)-4-[[2-chloro-6-[4-[[6-[(4R)-4-ami no-2-oxo-pyrrolidin-l-yl]-3- pyridyl]sulfonyl]piperazin-l-yl]-4-pyridyl]-difluoro-methyl] cyclohexanecarboxamide;

Trans-N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amin o-2-oxo-pyrrolidin-l- yl]phenyl]sulfonyl-l-piperidyl]-4-pyridyl]-difluoro-methyl]c yclohexanecarboxamide; Cis-N-(2-amino-2-oxo-ethyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-ami no-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]cyclohexanecarboxamide; (4R)-4-amino-l-[4-[4-[4-[[4-[(2S,4S)-4-amino-2-methyl-pyrrol idine-l- carbonyl]cyclohexyl]-difluoro-methyl]-6-chloro-2-pyridyl]pip erazin-l- yl]sulfonylphenyl]pyrrolidin-2-one;

Trans-bis(2-aminoethyl)-[3-[[4-[[2-[4-[4-[(4R)-4-amino-2- oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluo ro- methyl]cyclohexanecarbonyl]amino]propyl]-methyl -ammonium;

Trans-N-[(17?,35)-3-aminocyclopentyl]-4-[[2-[4-[4-[(4R)-4 -amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluo ro-methyl]cyclohexanecarboxamide;

Trans-(4R)-4-amino-l-[4-[4-[4-[[4-[cis-3,4-bis(aminomethy l)pyrrolidine-l- carbonyl]cyclohexyl]-difluoro-methyl]-6-chloro-2-pyridyl]pip erazin-l- yl]sulfonylphenyl]pyrrolidin-2-one;

Trans-4-[[2-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phe nyl]sulfonylpiperazin-l-yl]-6- chloro-4-pyridyl]-difluoro-methyl]-N-[(37?,45)-4-methylpyrro lidin-3- yl]cyclohexanecarboxamide;

Trans-N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-(met hylamino)-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]cyclohexanecarboxamide;

Trans-(4R)-4-amino-l-[4-[4-[4-[[4-[4-(2-aminoethyl)-4-met hyl-piperazin-4-ium-l- carbonyl]cyclohexyl]-difluoro-methyl]-6-chloro-2-pyridyl]pip erazin-l- yl]sulfonylphenyl]pyrrolidin-2-one;

Trans-N-[2-(2-aminoethylamino)ethyl]-4-[[2-chloro-6-[4-[4 -[(4R)-4-amino-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-di fluoro- methyl]cyclohexanecarboxamide;

Trans-2-aminoethyl-[2-[[4-[[2-chloro-6-[4-[4-[(4R)-4-amin o-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro- methyl]cyclohexanecarbonyl]amino]ethyl]-dimethyl-ammonium;

Trans-N-[2-[l-(2-aminoethyl)pyrrolidin-l-ium-l-yl]ethyl]- 4-[[2-[4-[4-[(4R)-4-amino-2- oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro- 4-pyridyl]-difluoro- methyl]cyclohexanecarboxamide;

Trans-N-[2-[(2-aminoacetyl)amino]ethyl]-4-[[2-chloro-6-[4 -[4-[(4R)-4-amino-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-di fluoro- methyl]cyclohexanecarboxamide; Trans-N-(3-amino-2-methoxy-propyl)-4-[[2-chloro-6-[4-[4-[(4A )-4-amino-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-di fluoro- methyl]cyclohexanecarboxamide;

Trans-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin -l-yl]phenyl]sulfonylpiperazin- l-yl]-4-pyridyl]-difluoro-methyl]-N-(2,3-diaminopropyl)cyclo hexanecarboxamide;

4-[[2-[4-[4-[4-(3-Aminopropylamino)-2-oxo-pyrrolidin-l-yl ]phenyl]sulfonylpiperazin-l- yl ] -6 -chi oro-4 -pyri dyl ] -difluoro-methyl ] -N -m ethyl -b enzami de;

Trans-N-[(1R,3S)-3-[(2-aminoacetyl)amino]cyclopentyl]-4-[ [2-[4-[4-[(4A)-4-amino-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-py ridyl]-difluoro- methyl]cyclohexanecarboxamide;

Trans-4-[[2-[4-[4-[(4A)-4-amino-2-oxo-pyrrolidin-l-yl]phe nyl]sulfonylpiperazin-l-yl]-6- chloro-4-pyridyl]-difluoro-rnethyl]-N-[(LS',3A > )-3-[c7.s-3,4-bis(arninornethyl)pyrrolidin- l- yl ] cy cl opentyl ] cy cl ohexanecarb oxami de;

Trans-4-[[2-chloro-6-[4-[4-[(4A)-4-amino-2-oxo-pyrrolidin -l-yl]phenyl]sulfonylpiperazin- l-yl]-4-pyridyl]-difluoro-methyl]-N-[(15)-3-[cis-3,5-diamino -l- piperidyl]cyclopentyl]cyclohexanecarboxamide;

Trans-4-[[2-[4-[4-[(4A)-4-amino-2-oxo-pyrrolidin-l-yl]phe nyl]sulfonylpiperazin-l-yl]-6- chloro-4-pyridyl]-difluoro-methyl]-N-[(15,3A)-3-[(3A)-3-amin opyrrolidin-l- yl ] cy cl opentyl ] cy cl ohexanecarb oxami de; and

Trans-3-[[4-[[2-[4-[4-[(4A)-4-amino-2-oxo-pyrrolidin-l-yl ]phenyl]sulfonylpiperazin-l-yl]- 6-chloro-4-pyridyl]-difluoro-methyl]cyclohexanecarbonyl]amin o]propyl-(3-aminopropyl)- dimethyl-ammonium; or a pharmaceutically acceptable salt thereof.

A further embodiment of present invention is (xviii) a compound of formula (I), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (xvi), for use as therapeutically active substance.

A further embodiment of present invention is (xix) a pharmaceutical composition comprising a compound according to any one of (i) to (xvi), and a therapeutically inert carrier.

A further embodiment of present invention is (xx) the use of a compound according to any one of (i) to (xvi), for the inhibition of LpxH. A further embodiment of present invention is (xxi) the use of a compound according to any one of (i) to (xvi), for the treatment or prophylaxis of bacterial infection, particularly the bacteria is gram-negative bacteria.

A further embodiment of present invention is (xxii) the use of a compound according to any one of (i) to (xvi), for the preparation of a medicament for the treatment or prophylaxis of bacterial infection, particularly the bacteria is gram-negative bacteria.

A further embodiment of present invention is (xxiii) the use of a compound according to (xxi) or (xxii), wherein the gram-negative bacteria is selected from Enter obacleriaceae. Neisseria gonorrhoeae, Haemophilus influenzae, Helicobacter pylorus, Acinetobacter baumannii and Pseudomonas aeruginosa.

A further embodiment of present invention is (xxiv) the use of a compound according to (xxiii) wherein the gram-negative bacteria is Enter obacteriaceae, wherein Enterobacteriaceae is Klebsiella pneumoniae or Escherichia coli.

A further embodiment of present invention is (xxv) a compound according to any one of (i) to (xvi), for the treatment or prophylaxis of bacterial infection, particularly the bacteria is gramnegative bacteria.

A further embodiment of present invention is (xxvi) a compound according to (xxv), wherein the gram-negative bacteria is selected from Enterobacteriaceae, Neisseria gonorrhoeae, Haemophilus influenzae, Helicobacter pylorus, Acinetobacter baumannii and Pseudomonas aeruginosa.

A further embodiment of present invention is (xxvii) a compound according to (xxvi), wherein the gram-negative bacteria is Enterobacteriaceae, wherein Enterobacteriaceae is Klebsiella pneumoniae or Escherichia coli.

A further embodiment of present invention is (xxviii) a method for the treatment or prophylaxis of bacterial infection, particularly the bacteria is gram-negative bacteria, which method comprises administering a therapeutically effective amount of a compound according to any one of (i) to (xvi).

PHARMACEUTICAL COMPOSITIONS AND ADMINISTRATION

Another embodiment provides pharmaceutical compositions or medicaments containing the compounds of the invention and a therapeutically inert carrier, diluent or excipient, as well as methods of using the compounds of the invention to prepare such compositions and medicaments. In one example, compounds of formula (I) may be formulated by mixing at ambient temperature at the appropriate pH, and at the desired degree of purity, with physiologically acceptable carriers, i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed into a galenical administration form. The pH of the formulation depends mainly on the particular use and the concentration of compound, but preferably ranges anywhere from about 3 to about 8. In one example, a compound of formula (I) is formulated in an acetate buffer, at pH 5. In another embodiment, the compounds of formula (I) are sterile. The compound may be stored, for example, as a solid or amorphous composition, as a lyophilized formulation or as an aqueous solution.

Compositions are formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners. The “effective amount” of the compound to be administered will be governed by such considerations, and is the minimum amount necessary to reduced bacterial load or improve host survival through the inhibition of Lipid A biosynthesis by targeting LpxH enzyme. For example, such amount may be below the amount that is toxic to normal cells, or the mammal as a whole.

In one example, the pharmaceutically effective amount of the compound of the invention administered parenterally per dose will be in the range of about 0.1 to 1000 mg/kg, alternatively about 1 to 100 mg/kg of patient body weight per day, with the typical initial range of compound used being 0.3 to 15 mg/kg/day. In another embodiment, oral unit dosage forms, such as tablets and capsules, preferably contain from about 5 to about 5000 mg of the compound of the invention.

The compounds of the invention may be administered by any suitable means, including oral, topical (including buccal and sublingual), rectal, vaginal, transdermal, parenteral, subcutaneous, intraperitoneal, intrapulmonary, intradermal, intrathecal and epidural and intranasal, and, if desired for local treatment, intralesional administration. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration.

The compounds of the present invention may be administered in any convenient administrative form, e.g., tablets, powders, capsules, solutions, dispersions, suspensions, syrups, sprays, suppositories, gels, emulsions, patches, etc. Such compositions may contain components conventional in pharmaceutical preparations, e.g., diluents, carriers, pH modifiers, sweeteners, bulking agents, and further active agents. A typical formulation is prepared by mixing a compound of the present invention and a carrier or excipient. Suitable carriers and excipients are well known to those skilled in the art and are described in detail in, e.g., Ansel, Howard C., et al., Ansel’s Pharmaceutical Dosage

Forms and Delivery Systems. Philadelphia: Lippincott, Williams & Wilkins, 2004;

Gennaro, Alfonso R., et al. Remington: The Science and Practice of Pharmacy. Philadelphia: Lippincott, Williams & Wilkins, 2000; and Rowe, Raymond C. Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005. The formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents, diluents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).

An example of a suitable oral dosage form is a tablet containing about 10 to 500 mg of the compound of the invention compounded with about 40 to 400mg anhydrous lactose, about 5 to 50 mg sodium croscarmellose, about 5 to 50 mg polyvinylpyrrolidone (PVP) K30, and about 1 to 10 mg magnesium stearate. The powdered ingredients are first mixed together and then mixed with a solution of the PVP. The resulting composition can be dried, granulated, mixed with the magnesium stearate and compressed to tablet form using conventional equipment. An example of an aerosol formulation can be prepared by dissolving the compound, for example 5 to 1000 mg) of the invention in a suitable buffer solution, e.g. a phosphate buffer, adding a tonicifier, e.g. a salt such sodium chloride, if desired. The solution may be filtered, e.g., using a 0.2 micron filter, to remove impurities and contaminants.

An embodiment, therefore, includes a pharmaceutical composition comprising a compound of Formula (I), or a stereoisomer or pharmaceutically acceptable salt thereof. In a further embodiment includes a pharmaceutical composition comprising a compound of Formula (I), or a stereoisomer or pharmaceutically acceptable salt thereof, together with a pharmaceutically acceptable carrier or excipient.

Another embodiment includes a pharmaceutical composition comprising a compound of formula (I) for use in the treatment and/or prevention of bacterial infections.

The following composition A and B illustrate typical compositions of the present invention, but serve merely as representative thereof. Composition A

A compound of the present invention can be used in a manner known per se as the active ingredient for the production of tablets of the following composition:

Per tablet

Active ingredient 200 mg

Microcrystalline cellulose 155 mg

Corn starch 25 mg

Talc 25 mg

Hydroxypropylmethylcellulose 20 mg

425 mg

Composition B

A compound of the present invention can be used in a manner known per se as the active ingredient for the production of capsules of the following composition:

Per capsule

Active ingredient 100.0 mg

Corn starch 20.0 mg

Lactose 95.0 mg

Talc 4.5 mg

Magnesium stearate 0.5 mg

220.0 mg

INDICATIONS AND METHODS OF TREATMENT

The compounds of the invention are inhibitors of the LpxH enzyme, a key enzyme of the LPS synthesis pathway that is essential in most gram-negative bacteria. Accordingly, the compounds of the invention can prevent bacterial growth of susceptible organisms and are useful for: preventing or treating a bacterial infection, preferably a Gram-negative bacterial infection (all claimed) e.g. nosocomial pneumonia, urinary tract infections, systemic infections (bacteraemia and sepsis), skin and soft tissue infections, surgical infections, eye infections, intraabdominal infections, lung infections and diabetic foot infections caused by Gram-negative bacteria e.g. third generation cephalosporins- and carbapenem- resistant Enterobacteriaeceae (e.g. Klebsiella pneumoniae, Escherichia coli) and multi-drug-resistant Pseudomonas aeruginosa and Acinetobacter baumannii or Acinetobacter spp., e.g. Neisseria gonorrhoeae, Haemophilus influenzae, Helicobacter pylorus e.g. Bacteroides spp. e.g. Bacteroides fragilis, Bacteroides thetaiotaomicron, Bacteroides dislasonis, Campylobacter jejuni, Campylobacter fetus or Campylobacter coil, Francisella tularensis and Providencia spp. e.g. Providencia stuartii, Providencia rettgeri or Providencia alcalifaciens and Pseudomonas spp.,' and for cleaning purposes e.g. to remove pathogenic microbes and bacteria from surgical instruments, catheters and artificial implants or to make a room or an area aseptic.

The products of the invention can be administered, for example, parenterally e.g. by injection, or administered orally, perorally, such as in the form of tablets, coated tablets, dragees, hard and soft gelatin capsules, solutions, emulsions or suspensions, or rectally, such as in the form of suppositories. Pharmaceutical compositions containing these compounds can be prepared using conventional procedures familiar to those skilled in the art, such as by combining the ingredients into a dosage form together with suitable, non-toxic, inert, therapeutically compatible solid or liquid carrier materials and, if desired, the usual pharmaceutical adjuvants. It is contemplated that the compounds are ultimately embodied into compositions of suitable oral, parenteral or topical dosage forms. The compositions of this invention can contain, as optional ingredients, any of the various adjuvants, which are used ordinarily in the production of pharmaceutical preparations. Thus, for example, in formulating the present compositions into the desired oral dosage forms, one may use, as optional ingredients, fillers, such as co-precipitated aluminum hydroxide-calcium carbonate, di-calcium phosphate or lactose; disintegrating agents such as maize starch; and lubricating agents, such as talc, calcium stearate, and the like. It should be fully understood, however, that the optional ingredients herein named are given by way of example only and that the invention is not restricted to the use hereof. Other such adjuvants, which are well known in the art, can be employed in carrying out this invention. Suitable as such carrier materials are not only inorganic, but also organic carrier materials. Thus, for tablets, coated tablets, dragees and hard gelatin capsules there can be used, for example, lactose, maize starch or derivatives thereof, talc, stearic acid or its salts. Suitable carriers for soft gelatin capsules are, for example, vegetable oils, waxes, fats and semi-solid and liquid polyols (depending on the nature of the active substance; no carriers are, however, required in the case of soft gelatin capsules). Suitable carrier materials for the preparation of solutions and syrups are, for example, water, polyols, saccharose, invert sugar and glucose. Suitable carrier materials for suppositories are, for example, natural or hardened oils, waxes, fats and semi-liquid or liquid polyols. As pharmaceutical adjuvants there are contemplated the usual preservatives, solubilizers, stabilizers, wetting agents, emulsifiers, sweeteners, colorants, flavorings, salts for varying the osmotic pressure, buffers, coating agents and antioxidants.

SYNTHESIS

The compounds of the present invention can be prepared by any conventional means.

Suitable processes for synthesizing these compounds as well as their starting materials are provided in the schemes below and in the subsequent examples. All substituents, in particular, R 1 to R 4 , Y, A, Q 1 , Q 2 , W and X are defined as below unless otherwise indicated. Furthermore, and unless explicitly otherwise stated, all reactions, reaction conditions, abbreviations and symbols have the meanings well known to a person of ordinary skill in the art.

Scheme 1

Wherein Q 1 is N; B 1 is halogen; PG 1 and PG 2 independently are protecting groups, such as /c/V-butoxy carbonyl and benzyl oxy carbonyl group.

Compound of formula (VI) could be obtained from a sulfonating reaction between halogenated phenyl sulfonyl chlorides (IV) and compound of formula (V). The coupling reaction of compound of formula (VI) with a cyclic amide such as lactam or carbamate (VII), using catalysts, such as Cui, in the presence of a base, such as C S2 CO 3 , affords the compound of formula (VIII). Compound of formula (IX) can be obtained by the deprotection of compound of formula (VIII) with a suitable acid such as trifluoroacetic acid, or a reducing reagent such as palladium on carbon in the presence of H 2 .

Scheme 2

Wherein Q 1 is CH.

Alternatively compound of formula (IX) can also be prepared in the process illustrated in scheme 2. The reaction between compound of formula (XXIII) and compound of formula (XXII) will give compound of formula (XXIV) in the presence of suitable Mitsunobu reagent, such as N,N,N-N-tetramethylazodicarboxamide and tributylphosphine. Compound of formula (XXV) can be obtained from the oxidation of compound of formula (XXIV) using a suitable oxidizing reagent, such as m-CPBA. The coupling reaction of compound of formula (XXV) with a cyclic amide such as lactam or carbamate (VII), using catalysts such as Cui, in the presence of a base, such as C S2 CO 3 , affords sulfoxide (XXVI). The oxidation of sulfoxide (XXVI) with a suitable oxidizing reagent, such as m-CPBA, affords compound of formula (XXVII). Compound of formula (IX) can be obtained by the deprotection of compound of formula (VIII) with a suitable acid such as trifluoroacetic acid, or a reducing reagent such as palladium on carbon in the presence of H 2 .

Scheme 3

Wherein B 3 is halogen; B 2 is halogen, or methylsulfonyl; R 5 is aldehyde, acyl chloride, or ester; R 6 is C 1-6 alkyl.

A magnesium-halogen exchange of compound of formula (X) with a suitable Grignard reagent, such as isopropylmagnesium chloride lithium chloride complex, followed by addition of compound of formula (XI) affords the compound of formula (XII). Compound of formula (XIII) can be obtained from the oxidation of compound of formula (XII) in the presence of a suitable oxidant such as Dess-Martin reagent. Fluorination of compound of formula (XIII) in the presence of suitable reagent, such as DAST, can give the compound of formula (XIV).

Scheme 4

Wherein PG 2 is a protecting group, such as Zc/'Z-butoxy carbonyl or benzyl oxy carbonyl; R 4 ’ is R 4 or R 4 -PG 3 , wherein PG 3 is a protecting group, such as tert-butoxycarbonyl or b enzyl oxy carb onyl . The nucleophilic substitution between compound of formula (IX) and compound of formula (XIV) affords ester (XV). Hydrolysis of ester (XV) under suitable basic condition, such as LiOH, or suitable acid condition such as HC1, affords acid (XVI). The coupling of acid (XVI) with an amine or alcohol of compound of formula (XVII) using a suitable reagent such as HATU, gives the compound of formula (XVIII). The final compound of formula (XIX) could be obtained from deprotection of compound of formula (XVIII) using a suitable acid such as trifluoroacetic acid, or a reducing reagent such as palladium on carbon in the presence of H 2 .

Scheme 5

Alternatively, the compound of formula (XIX) can also be prepared according to the scheme 5. Hydrolysis of compound of formula (XIV) under suitable basic condition such as LiOH, or acid condition such as HC1, affords compound of formula (XX). The coupling of compound of formula (XX) with an amine or alcohol of compound of formula (XVII) using a suitable reagent such as HATU, gives the compound of formula (XXI). The nucleophilic substitution of compound of formula (XXI) with compound of formula (IX) in the presence of a suitable base, such as K 2 CO 3 , affords compound of formula (XVIII). The final compound of formula (XIX) could be obtained from the deprotection of compound of formula (XVIII) using a suitable acid such as trifluoroacetic acid, or a reducing reagent such as palladium on carbon in the presence of H 2 .

Scheme 6 Wherein PG 4 and PG 5 are protecting groups, such as tert-butoxy carbonyl and benzyloxycarbonyl; L 1 is C 1-6 alkyl or C 3-10 cycloalkyl; R 7 is C 1-6 alkyl, aminoC 1-6 alkyl or aminoC 3-10 cycloalkyl; R 7 is R 7 -PG 5 .

Compound of formula (XVI) can react with compound of formula (XXVIII-1) using a suitable coupling reagent such as HATU to give compound of formula (XXIX). The oxidation of alcohol of the compound of formula (XXIX) with a suitable oxidant, such as Dess-Martin reagent, affords compound of formula (XXX). Compound of formula (XXXIII) can be obtained from the reductive amination of compound of formula (XXX) with compound of formula (XXVIII-3) in the presence of a suitable reducing reagent, such as sodium tri acetoxyb orohydri de.

Compound of formula (XXXIII) can also be pursued from the alternative route in the scheme 6. Compound of formula (XXXI) can be prepared from the coupling of compound of formula (XVI) with amine (XXVIII-2) using a suitable coupling reagent such as HATU. The deprotection of compound of formula (XXXI) in the presence of a suitable acid, such as trifluoroacetic acid, gives the compound of formula (XXXII). Compound of formula (XXXIII) can be obtained from the substitution of compound of formula (XXXII) with compound of formula (XXVIII-4) under basic condition such as DIPEA, or through substitution reaction with coupling reagents such as HATU.

The final compound of formula (XXXIV) can be obtained from the deprotection of compound of formula (XXXIII) using a suitable acid, such as trifluoroacetic acid, or a reducing reagent such as palladium on carbon in the presence of H 2 . Another final compound of formula (XXXV) can be obtained from the methylation of compound of formula (XXXIII) using a suitable reagent, such as Mel, followed by deprotection with a suitable acid, such as trifluoroacetic acid, or a reducing reagent such as palladium on carbon in the presence of H 2 .

Scheme 7

Wherein L 1 is C 1-6 alkyl or C 3-10 cycloalkyl; R 7 is C 1-6 alkyl, aminoC 1-6 alkyl or aminoC 3- 10 cycloalkyl; R 7 is R 7 -PG 5 .

Compound of formula (XXXVI) can be obtained from the coupling of compound of formula (XVI) with compound of formula (XXVIII-5) using a suitable coupling reagent, such as HATU. The substitution of compound of formula (XXXVI) with compound of formula (XXVIII-4) gives the compound of formula (XXXVII) in the presence of a suitable base, such as DIPEA. The final compound of formula (XXXVIII) can be obtained from the deprotection of the compound of formula (XXXVII) with a suitable acid, such as trifluoroacetic acid, or a reducing reagent such as palladium on carbon in the presence of H 2 .

Compounds of this invention can be obtained as mixtures of diastereomers or enantiomers, which can be separated by methods well known in the art, e.g. (chiral) HPLC or SFC. This invention also relates to a process for the preparation of a compound of formula (I) comprising following step: a) formation of compound of formula (XIX), via deprotection of compound of formula (XVIII), using a suitable acid, such as trifluoroacetic acid, or a reducing reagent, such as palladium on carbon in the presence of H 2 ; b) formation of compound of formula (XXXV), via methylation of compound of formula (XXXIII),

Mel, followed by deprotection with a suitable acid, such as trifluoroacetic acid, or a reducing reagent such as palladium on carbon in the presence of H 2 ; c) formation of compound of formula (XXXIV), via deprotection of compound of formula (XXXIII), using a suitable acid, such as trifluoroacetic acid, or a reducing reagent such as palladium on carbon in the presence of H 2 ; d) formation of compound of formula (XXXVIII), via deprotection of the compound of formula (XXXVII), suitable acid, such as trifluoroacetic acid, or a reducing reagent such as palladium on carbon in the presence of H 2 ; wherein R 4 ’ is R 4 or R 4 -PG 3 ; L 1 is C 1-6 alkyl or C 3-10 cycloalkyl; R 7 is C 1-6 alkyl, aminoC 1- 6 alkyl or aminoC 3-10 cycloalkyl; R 7 is R 7 -PG 5 ; PG 2 , PG 3 and PG 5 are independently protecting groups, such as tert-butoxycarbonyl or benzyl oxy carbonyl;

A compound of formula (I) when manufactured according to the above process is also an object of the invention.

EXAMPLES

The invention will be more fully understood by reference to the following examples. They should not, however, be construed as limiting the scope of the invention.

ABBREVIATIONS

The invention will be more fully understood by reference to the following examples. They should not, however, be construed as limiting the scope of the invention.

Abbreviations used herein are as follows:

MeCN: acetonitrile aq.: aqueous

DAST: diethylaminosulfur trifluoride DBU: l,8-diazabicyclo[5.4.0]undec-7-ene

DMAP: 4-dimethylaminopyridine

DMED A: 1 ,2-dimethyl ethylenediamine

DMP: Dess-Martin periodinane

FA: formic acid

IC 50 : the molar concentration of an inhibitor, which produces 50% of the maximum possible response for that inhibitor.

FBS: fetal bovine serum

HPLC: high performance liquid chromatography

MS (ESI): mass spectroscopy (electron spray ionization)

Ms: methyl sulfonyl

MTBE: methyl tert-butyl ether obsd.: observed

PE: petroleum ether

DCM: dichloromethane

TFA: trifluoroacetic acid

DIPEA: A A-Diisopropylethylamine

TEA: triethylamine

HATU: l-[bis(dimethylamino)methylene]-lH-l,2,3-triazolo[4,5-b]pyri dinium 3- oxid hexafluorophosphate δ: chemical shift

SFC: supercritical fluid chromatography

TLC : thi n 1 ay er chromatography

GENERAL EXPERIMENTAL CONDITIONS

Intermediates and final compounds were purified by flash chromatography using one of the following instruments: i) Biotage SP1 system and the Quad 12/25 Cartridge module, ii) ISCO combi-flash chromatography instrument. Silica gel brand and pore size: i) KP-SIL 60 A, particle size: 40-60 pm; ii) CAS registry NO: Silica Gel: 63231-67-4, particle size: 47-60 micron silica gel; iii) ZCX from Qingdao Haiyang Chemical Co., Ltd, pore: 200-300 or 300-400.

Intermediates and final compounds were purified by preparative HPLC on reversed phase column using XB ridge™ Prep-C18 (5 μm, OBDTM 30 x 100 mm) column, SunFire™ Prep-C18 (5 μm, OBD™ 30 x 100 mm) column, Phenomenex Synergi-C18 (10 μm, 25 x 150 mm) or Phenomenex Gemini-C18 (10 μm, 25 x 150 mm). Waters AutoP purification System (Sample Manager 2767, Pump 2525, Detector: Micromass ZQ and UV 2487, solvent system: acetonitrile and 0.1% ammonium hydroxide in water; acetonitrile and 0.1% FA in water or acetonitrile and 0.1% TFA in water). Or Gilson-281 purification System (Pump 322, Detector: UV 156, solvent system: acetonitrile and 0.05% ammonium hydroxide in water; acetonitrile and 0.225% FA in water; acetonitrile and 0.05% HC1 in water; acetonitrile and 0.075% TFA in water; or acetonitrile and water).

For SFC chiral separation, intermediates were separated by chiral column (Daicel chiralpak IC, 5 μm, 30 x 250 mm), AS (10 μm, 30 x 250 mm) or AD (10 μm, 30 x 250 mm) using Mettler Toledo Multigram III system SFC, Waters 80Q preparative SFC or Thar 80 preparative SFC, solvent system: CO 2 and IPA (0.5% TEA in IP A) or CO 2 and MeOH (0.1% NH 3 H 2 O in MeOH), back pressure lOObar, detection UV@ 254 or 220 nm.

LC/MS spectra of compounds were obtained using a LC/MS (Waters™ Alliance 2795- Micromass ZQ, Shimadzu Alliance 2020-Micromass ZQ or Agilent Alliance 6110-Micromass ZQ), LC/MS conditions were as follows (running time 3 or 1.5 mins):

Acidic condition I: A: 0.1% TFA in H 2 O; B: 0.1% TFA in acetonitrile;

Acidic condition II: A: 0.0375% TFA in H 2 O; B: 0.01875% TFA in acetonitrile;

Basic condition I: A: 0.1% NH 3 H 2 O in H 2 O; B: acetonitrile;

Basic condition II: A: 0.025% NH 3 H 2 O in H 2 O; B: acetonitrile;

Neutral condition: A: H 2 O; B: acetonitrile.

Mass spectra (MS): generally only ions which indicate the parent mass are reported, and unless otherwise stated the mass ion quoted is the positive mass ion (MH) + .

NMR Spectra were obtained using Bruker Avance 400 MHz.

The microwave assisted reactions were carried out in a Biotage Initiator Sixty microwave synthesizer. All reactions involving air-sensitive reagents were performed under an argon or nitrogen atmosphere. Reagents were used as received from commercial suppliers without further purification unless otherwise noted.

PREPARATIVE EXAMPLES

The following examples are intended to illustrate the meaning of the present invention but should by no means represent a limitation within the meaning of the present invention: Intermediate 1: Tert-butyl N-[5-oxo-l-(4-piperazin-l-ylsulfonylphenyl)-pyrrolidin-3- yl] carbamate

Step 1: Benzyl 4-(4-iodophenyl) sulfonylpiperazine-l-carboxylate

To a solution of 1 -Cbz-piperazine (8.01 g, 36.4 mmol), TEA (6.45 mL, 46.3 mmol) in DCM (200 mL) was added 4-iodobenzenesulfonyl chloride (10.0 g, 33.06 mmol) at 0°C and the mixture was stirred at room temperature for 2 hours. The mixture was concentrated in vacuo to give the residue, which was triturated in PE and washed by water to give the desired compound Int-la (18.0 g, 37.0 mmol, 95.2% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 487.0. NMR (400 MHz, CDC1 3 ) δ ppm 7.94 (br d, J = 8.8Hz, 2H), 7.89 (m, 2H), 7.43 (m, 1H), 7.32- 7.33 (m, 4H), 5.01 (s, 2H), 3.12-3.14 (m, 8H).

Step 2: Benzyl 4-((4-(4-((tert-butoxycarbonyl)amino)-2-oxopyrrolidin-l-yl)p henyl)sulfonyl) piperazine-l-carboxylate

To a mixture of compound Int-la (18.0 g, 37.0 mmol), tert-butyl 7V-(5-oxopyrrolidin-3- yl)carbamate (8.89 g, 44.4 mmol) and K 2 CO 3 (15.4 g, 111 mmol) in NMP (180 mL) was added Cui (7.05 g, 37.0 mmol) and N1,N2-dimethyl cyclohexane- 1,2-diamine (5.26 g, 37.0 mmol). The mixture was stirred at 90°C for 2 hours under nitrogen. The mixture was diluted with EtOAc (200 mL) and then filtered through fritted glass funnel. The filtrate was washed with NH 3 .H 2 O (2% wt, 200 mL), aq. CaCL solution (150 mL, 2 N) and brine (150 mL x 2). The filtrate was collected and dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give yellow oil. The oil was purified by prep-HPLC to give the desired compound Int-lb (13.5 g, 24.2 mmol, 65.3% yield) as a gray solid. MS obsd. (ESI + ) [(M+H) + ]: 559.3.

Step 3: Tert-butyl N-[5-oxo-l-(4-piperazin-l-ylsulfonylphenyl)pyrrolidin-3-yl]c arbamate

To a solution of compound Int-lb (500 mg, 0.90 mmol) in methanol (10 mL) was added Pd/C (10% wt, 100.0 mg). The mixture was stirred under H 2 ( 15psi) at room temperature for 2 hours. The mixture was diluted with MeOH (100 mL), filtered through a pad of Celite and concentrated in vacuo to give the desired compound Int-1 (300 mg, 0.710 mmol, 78.96% yield) as a grey solid. MS obsd. (ESI + ) [(M+H) + ]: 425.3.

Intermediate 2: Tert-butyl N-[(3R)-5-oxo-l-(4-piperazin-l-ylsulfonylphenyl)pyrrolidin-3 - yl] carbamate

The title compound was prepared in analogy to the preparation of compound Int-1 by using tert-butyl M[(3R)-5-oxopyrrolidin-3-yl]carbamate instead of tert-butyl N-(5- oxopyrrolidin-3-yl)carbamate. Compound Int-2 was obtained as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 425.2.

Intermediate 3A and 3B: (5S)-5-(hydroxymethyl)-3-(4-piperazin-l- ylsulfonylphenyl)oxazolidin-2-one and (5R)-5-(hydroxymethyl)-3-(4-piperazin-l- ylsulfonylphenyl)oxazolidin-2-one

Step 1: Tert-butyl 4-(4-iodophenyl)sulfonylpiperazine-l-carboxylate

To a solution of 4-iodobenzenesulfonyl chloride (1.0 g, 3.31 mmol) and tert-butyl piperazine- 1 -carboxylate (739 mg, 3.97 mmol) in DCM (100 mL) was added TEA (668 mg, 6.61 mmol) dropwise at room temperature. The mixture was washed with aq. NaHCO 3 , solution (100 mL), extracted with DCM (50 mL x 3), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give compound Int-3a (1.5g, 3.32 mmol, 100 % yield), which was used in the next step directly. MS obsd. (ESI + ) [(M+H) + ]: 452.98.

Step 2: Tert-butyl 4-[4-[(55)-5-(hydroxymethyl)-2-oxo-oxazolidin-3- yl]phenyl]sulfonylpiperazine-l-carboxylate (Int-5bA) and tert-butyl 4- [4-[(5R)-5- (hydroxymethyl)-2-oxo-oxazolidin-3-yl]phenyl]sulfonylpiperaz ine-l-carboxylate (Int-5bB)

A degassed mixture of compound Int-3a (0.430 g, 951 pmol), 5- (hydroxymethyl)oxazolidin-2-one (111 mg, 951 pmol), A, A'-dimethyl ethane- 1,2-diamine (29.3 mg, 333 pmol), Cui (63.4 mg, 333 pmol) and K3PO4 (404 mg, 1.9 mmol) in DMF (4 ml) was heated at 75°C in a microwave reactor for 1 hour. After completion, the mixture was poured into ice water, extracted with DCM (25 mL x 3), dried over anhydrous sodium sulfate, and concentrated in vacuo to give crude product, which was purified by silica gel chromatography (eluent with EtOAc : PE = 0-80%) to afford the enantiomeric mixture of Int-3bA and Int-3bB (310 mg, 702 pmol, 73.9 % yield). MS obsd. (ESI + ) [(M+H) + ]: 442.1. The resulting solid was then separated by SFC to give the two enantiomers Int-3bA (fast eluted) and Int-3bB (slow eluted). SFC condition: Column: IA (250mmx30mm, 5 um); 0.1% NH 3 H 2 O EtOH; begin B 50%, end B 50%; Flow Rate (ml/min): 50.

Step 3: (55)-5-(hydroxymethyl)-3-(4-piperazin-l-ylsulfonylphenyl)oxa zolidin-2-one (Int-3A) and (5R)-5-(hydroxymethyl)-3-(4-piperazin-l-ylsulfonylphenyl)oxa zolidin-2-one (Int-3B)

Compound Int-3bA (60 mg, 0.136 mmol) and TFA (0.5 mL, 6.49 mmol) were mixed in the DCM (5 mL). The reaction mixture was stirred at room temperature for 1 hour. After completion, the residue was concentrated in vacuo to give crude compound Int-3A (45 mg, 97 %) as light yellow oil. MS obsd. (ESI + ) [(M+H) + ]: 342.1. Int-3B was prepared in analogy to the procedure described for the preparation of compound Int-3A by using compound Int-3bB as the starting material instead of compound Int- 3bA. MS obsd. (ESI + ) [(M+H) + ]: 342.1.

Intermediate 4: Tert-butyl 4-[5-oxo-l-(4-piperazin-l-ylsulfonylphenyl)pyrrolidin-3- yl] piperazine- 1-carboxylate

Step 1: Benzyl 4-[4-(4-hydroxy-2-oxo-pyrrolidin-l-yl)phenyl]sulfonylpiperaz ine-l- carboxylate

To a solution of compound Int-3a (5.5 g, 11.31 mmol), 4-hydroxypyrrolidin-2-one (2.29 g, 22.62 mmol) and potassium carbonate (4.69 g, 33.93 mmol) in NMP (25 mL) was added copper iodide (1.51 g, 7.92 mmol) and Trans-A,A-dimethylcyclohexane (1.13 g, 7.92 mmol) under nitrogen. The mixture was stirred at 90°C. After 2 hours, the mixture was diluted with EtOAc (600 mL) and filtered. The filtrate was washed with 2% aq. NH 3 .H 2 O solution (100 mL x 2), aq. CaCL (100 mL x 3) and brine (100 mL x 2). Then, the organic layer was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give crude product, which was triturated in PE (100 mL) to give the compound Int-4a (4.5 g, 9.81 mmol, 86.78% yield) as a brown solid. MS obsd. (ESI + ) [(M+H) + ]: 459.9.

Step 2: Benzyl 4-[4-(5-oxo-2H-pyrrol-l-yl)phenyl]sulfonylpiperazine-l-carbo xylate

To a solution of compound Int-4a (4.5 g, 9.79 mmol) and DIPEA (5.05 g, 39.17 mmol) in DCM (80 mL) was added methanesulfonyl chloride (1.58 mL, 20.35 mmol) slowly at 0°C. Then, the mixture was stirred at room temperature for 3 hours. After completion, the reaction mixture was poured into water (100 mL) at 0°C and stirred at room temperature for 30 minutes. Then, the solution was extracted with DCM (200 mL x 3) and washed with aq. NaOH solution (0.5 N, 100 mL x 3) and brine (100 mL x 3). The organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo to give crude product, which was triturated in PE/MTBE (100 mL, v/v = 1 : 1) to give compound Int-4b (3.5 g, 7.95 mmol, 81.13% yield) as a grey solid. MS obsd. (ESI + ) [(M+H) + ]: 441.9.

Step 3: Tert-butyl 4-[l-[4-(4-benzyloxycarbonylpiperazin-l-yl)sulfonylphenyl]-5 -oxo- pyrrolidin-3-yl]piperazine-l-carboxylate

To a solution of compoundlnt-4b (2.0 g, 4.53 mmol) in NMP (0.5 mL) was added 1- BOC-piperazine (10.0 g, 53.69 mmol). The mixture was stirred at 90°C for 2 hours. After completion, the mixture was diluted with water (100 mL), extracted with EtOAc (100 mL x 2) and washed with brine (100 mL). The combined organic layer was dried over anhydrous sodium sulfate, and concentrated in vacuo to give the crude residue, which was purified by silica gel chromatography (eluent with EtOAc : PE = 0-100%) to afford compound Int-4c (1.6 g, 2.55 mmol, 56.26% yield) as a yellow solid. MS obsd. (ESI + ) [(M+H) + ]: 628.3.

Step 4: Tert-butyl 4-[5-oxo-l-(4-piperazin-l-ylsulfonylphenyl)pyrrolidin-3-yl]p iperazine-l- carboxylate

To a solution of compound Int-4c (1.5 g, 2.39 mmol) in THF (4 mL) was added Pd/C (800 mg, 2.39 mmol). The mixture was stirred at 25°C for 2 hours under H 2 ( 15psi). After completion, the mixture was filtered through a Celite pad, and the filtrate was concentrated in vacuo to give the crude product, which was purified by prep-HPLC to give compound Int-4 (550 mg, 1.11 mmol, 38.18% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 494.3. Intermediate 5: Trans-4-[[2-[4-[4-[(4R)-4-(terCbutoxycarbonylamino)-2-oxo-py rrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluo ro- m ethyl] cyclohexanecarboxylic acid

Step 1: Trans-methyl 4-(hydroxymethyl)cyclohexanecarboxylate

To a solution of Trans-4-methoxycarbonylcyclohexanecarboxylic acid (50.0 g, 268.51 mmol) in THF (150 mL) was added BH 3 .Me 2 S (53.7 mL, 537.03 mmol) dropwise at 0 ~ 10°C for 1 hour under nitrogen. The mixture was stirred at 25°C for 12 hours. After completion, the reaction was quenched by adding methanol (100 mL) dropwise at 0°C. The mixture was concentrated in vacuo at 40°C to give crude residue, which was purified by silica gel chromatography (eluent with EtOAc : PE = 0-50%) to give compound Int-5a (28 g, 162.58 mmol, 60.55% yield) as colorless oil.

Step 2: Trans-methyl 4-formylcyclohexanecarboxylate

To a solution of oxalyl chloride (30.95 g, 243.87 mmol) in DCM (100 mL) was added dimethyl sulfoxide (25.4 g, 325.17 mmol) at -60 °C and stirred at -60 °C for 0.5 hour under nitrogen. Then the solution of compound Int-5a (28.0 g, 162.58 mmol) in DCM (100 mL) was added into the reaction at -60 °C and kept stirring for additional 1 hour. Then, DIPEA (84.05 g, 650.33 mmol) was added at -60 °C. The reaction was allowed to warm to 0°C and kept stirring for 2 hours. After completion, the reaction mixture was diluted with EtOAc (600 mL) and washed with aq. CaC1 2 (100 mL x 3) and brine (50 mL x 2). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give crude residue, which was purified by silica gel chromatography (eluent with EtOAc : PE = 3-50%) to give compound Int- 5b (24 g, 141 mmol, 86.73% yield) as light yellow oil.

Step 3: Trans-methyl 4- [(2,6-dichloro-4-pyridyl)-hydroxy-methyl] cyclohexanecarboxylate

To a solution of 2,6-dichloro-4-iodo-pyridine (42.48 g, 155.1 mmol) in THF (50.0 mL) was added isopropylmagnesium chloride lithium chloride complex in THF (130.16 mL, 169.2 mmol) at -40°C under nitrogen. The mixture was stirred at 25°C for 1 hour. The reaction mixture was then added into a solution of compound Int-5b (24.0 g, 141 mmol) in THF (100 mL) dropwise at -40°C. The mixture was stirred at 25°C for 3 hours. After completion, the reaction mixture was poured into sat. aq. NH4CI solution (200 mL) and extracted with EtOAc (300 mL x 3). The organic layer was washed with brine (100 mL x 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give crude residue, which was purified by silica gel chromatography (eluent with EtOAc : PE = 0-30%) to give compound Int-5c (33 g, 103.71 mmol, 73.55% yield) as a white solid. MS obsd. (ESI + ) [(M+C1) + ]: 317.8.

Step 4: Trans-methyl 4-(2,6-dichloropyridine-4-carbonyl)cyclohexanecarboxylate

To a solution of compound Int-5c (33.0 g, 103.71 mmol) in THF (150 mL) was added DMP (48.39 g, 114.08 mmol) slowly at 0°C under nitrogen. The reaction was stirred at 25 °C for 3 hours. After completion, the mixture was diluted with EtOAc (500 mL), then poured into aq. K2CO3 solution (200 mL) and kept stirring at room temperature for 0.5 hour. The organic layer was separated and washed with brine (100 mL x 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give crude compound Int-5d (30 g, 94.88 mmol, 91.49% yield) as a white solid, which was used in the next step directly. MS obsd. (ESI + ) [(M+H) + ]: 315.8.

Step 5: Trans-methyl 4-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]cyclohexanecarbo xylate

A solution of compound Int-5d (29.5 g, 93.3 mmol) in DCM (20 mL) was added DAST (135.35 g, 839.71 mmol) and stirred at 45°C for 12 hours. After completion, the reaction mixture was poured into ice-water and extracted with EtOAc (200 mL x 3). The combined organic layer was washed with aq. K 2 CO 3 (100 mL x 2) and brine (100 mL x 2), dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo to give a crude residue. The residue was recrystallized with MeOH (150 mL) to give compound Int-5e (25.6 g, 75.7 mmol, 81.13% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 337.8.

Step 6: Trans-methyl 4-[[2-chloro-6-[4-[4-[(4R)-4-(tert-butoxycarbonylamino)-2-ox o- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-di fluoro- m ethyl] cyclohexanecarboxylate

To a solution of compound Int-5e (3.58 g, 10.6 mmol) and compound Int-2 (4.5 g, 10.6 mmol) in DMSO (10 mL) was added DIPEA (5.55 mL, 31.8 mmol). The mixture was stirred at 115°C for 18 hours. After completion, the reaction mixture was concentrated in vacuo and purified by prep-HPLC to give compound Int-5f (7 g, 9.64 mmol, 90.93% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 670.1.

Step 7: Trans- 4-[[2-chloro-6-[4-[4-[(4R)-4-(tert-butoxycarbonylamino)-2-ox o-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]cyclohexanecarboxylic acid To a solution of compound Int-5e (7.0 g, 9.64 mmol) in MeCN (100 mL) and water (10 mL) were added TEA (13.43 mL, 96.39 mmol), lithium bromide (16.74 g, 192.78 mmol) in one portion. The reaction was stirred at 40°C for 12 hours. After completion, the reaction was diluted with 2-methyltetrahydrofuran (500 mL) and adjusted to pH 5 with 2N aq. HC1 solution. The resulting organic layer was washed with 0.2 N aq. HC1 solution (100.0 mL x 2) and brine (100 mL x 2 ), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give crude product. The crude product was recrystallized with MeCN (100 mL) to give compound Int-5 (6.7 g, 9.41 mmol, 97.6% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 712.1.

Intermediate 6: Trans-4-[[2-chloro-6-[4-[4-(4-methyl-2-oxo-pyrrolidin-l- yl)phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]cyclohexanecarboxylic acid

Compound Int-6 was prepared in analogy to the procedure described for the preparation of compound Int-5 by using compound Int-2 as the coupling reagent instead of compound Int-1 in Step 6. MS obsd. (ESI + ) [(M+H) + ]: 656.1.

Intermediate 7: Cis-4-[[2-chloro-6-[4-[4-[(4R)-4-(tert-butoxycarbonylamino)- 2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-di fluoro- m ethyl] cyclohexanecarboxylic acid

Compound Int-7 was prepared in analogy to the procedure described for the preparation of compound Int-5 by using cis-4-methoxycarbonylcyclohexanecarboxylic acid instead of trans- 4-methoxycarbonylcyclohexanecarboxylic acid in Step 1. MS obsd. (ESI + ) [(M+H) + ]: 712.2.

Intermediate 8 : 3- [ [2-Chloro-6- [4- [4- [(4R)-4-(tert-butoxycarbonylamino)-2-oxo-pyr rolidin- l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-meth yl]cyclopentanecarboxylic acid

The title compound was prepared in analogy to the preparation of compound Int-5 by using 3-methoxycarbonylcyclopentanecarboxylic acid instead of trans-4- methoxycarbonylcyclohexanecarboxylic acid in Step 1. Compound Int-8 (20.0 mg) was obtained as light yellow oil. MS obsd. (ESI + ) [(M+C1) + ]: 698.1.

Intermediate 9 : 4- [ [2-Chloro-6- [4- [4- [(4R)-4-(tert-butoxycarbonylamino)-2-oxo-pyr rolidin- l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-meth yl]norbornane-l-carboxylic acid

The title compound was prepared in analogy to the preparation of compound Int-5 by using 4-(methoxycarbonyl)bicyclo[2.2.1]heptane-l -carboxylic acid instead of trans-4- methoxycarbonylcyclohexanecarboxylic acid in Step 1. Compound Int-9 (150.0 mg) was obtained as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 724.4.

Intermediate 10: 4- [ [2-Chloro-6- [4- [4- [(4R)-4-(tert-butoxycarbonylamino)-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-di fluoro- methyl]bicyclo[2.2.2]octane-l-carboxylic acid

The title compound was prepared in analogy to the preparation of compound Int-5 by using 4-(methoxycarbonyl)bicyclo[2.2.2]octane-l -carboxylic acid instead of trans-4- methoxycarbonyl cyclohexanecarboxylic acid in Step 1. Compound Int-10 (250.0 mg) was obtained as a light yellow solid. MS obsd. (ESI + ) [(M+H) + ]: 738.4. Intermediate 11: 3- [ [2-Chloro-6- [4- [4- |(4R)-4-(tert-biitoxyc:irbonyl:imino)-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-di fluoro- methyl] bicyclo [1.1.1] pentane- 1-carboxylic acid

The title compound was prepared in analogy to the preparation of compound Int-5 by using 3 -(m ethoxy carbonyl )bicyclo[l. l.l]pentane- 1-carboxylic acid instead of trans-4- methoxycarbonylcyclohexanecarboxylic acid in Step 1. Compound Int-11 (150.0 mg) was obtained as a light yellow oil. MS obsd. (ESI + ) [(M+H) + ]: 696.0.

Intermediate 12 : 4- [ [2-Chloro-6- [4- [4- [(4R)-4-(tert-butoxycarbonylamino)-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-di fluoro-methyl]benzoic acid

Step 1: Methyl 4- [(2,6-dichloro-4-pyridyl)-hydroxy-methyl] benzoate

To a mixture of 2,6-dichloro-4-iodo-pyridine (36.71 g, 134.02 mmol) in THF (180 mL) was added isopropylmagnesium chloride lithium chloride complex solution (1.3 M in THF, 121.83 mL) under nitrogen at -40°C. The mixture was stirred at 25°C for 2 hours. Then to the mixture was added methyl 4-formylbenzoate (20 g, 121.83 mmol) in THF (200 mL) at -40 °C. The mixture was stirred at 25°C for 2 hours. After completion, the mixture was added to sat. aq NH4CI solution (1 L) in ice-water bath and extracted with EtOAc (500 mL x 2). The combined organic layer was washed with brine (500 mL x 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give crude compound Int-12a (30 g, 96.11 mmol, 78.88% yield) as a yellow solid. MS obsd. (ESI + ) [(M+H) + ]: 312.1.

Step 2: Methyl 4-(2,6-dichloropyridine-4-carbonyl)benzoate

To a solution of compound Int-12a (25 g, 80.09 mmol) in THF (250 mL) was added DMP (37.37 g, 88.10 mmol) at 0°C. The mixture was stirred at 0°C for 1 hour. After completion, the reaction mixture was filtered, and the filtrate was diluted with water (200 mL) and extracted with EtOAc (200 mL x 2). The combined organic layer was washed with brine (300 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give a residue. The residue was suspended in PE (50 mL) and the resulting precipitate was collected by filtration. Compound Int-12b (20.3 g, 65.46 mmol, 81.73% yield) was obtained as a white solid. NMR (400 MHz, CHLOROFORM-d) δ ppm 8.21 (d, J = 8.44Hz, 2H) 7.86 (d, J= 8.44Hz, 2H) 7.54 (s, 2H) 3.99 (s, 3H).

Step 3: Methyl 4- [(2, 6-dichloro-4-pyridyl)-difluoro-methyl] benzoate

To a mixture of compound Int-12b (20.3 g, 65.46 mmol) in DCM (150 mL) was added DAST (84.41 g, 523.65 mmol) and stirred at 40°C for 12 hours. After completion, the mixture was added into aq NaHCCL solution (500 mL) in ice-water bath and extracted with DCM (200 mL x 2). The combined organic layer was washed with brine (200 mL x 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give crude residue, which was purified by silica gel chromatography (EtOAc : PE = 2-5%) to give compound Int-12c (20.2 g, 60.82 mmol, 92.92% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 332.0.

Step 4: Methyl 4-[[2-chloro-6-[4-[4-[(4R)-4-(tert-butoxycarbonylamino)-2-ox o-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]benzoate

To a solution of compound Int-2 (100.0 mg, 0.240 mmol) and compound Int-12c (78.24 mg, 0.240 mmol) in DMSO (1 mL) was added DIPEA (0.2 mL, 0.710 mmol). The mixture was stirred at 110°C for 12 hours. After completion, the mixture was diluted with EtOAc (50 mL) and water (30 mL). The organic phase was washed with brine (50 mL), dried over anhydrous sodium sulfate, concentrated in vacuo to give the crude residue compound Int-12d (120 mg, 0.170 mmol, 70.73% yield) as a yellow solid, which was used directly in the next step. MS obsd. (ESI + ) [(M+H) + ]: 720.3.

Step 5 : 4- [ [2-Chloro-6- [4- [4- [(4R)-4-(tert-butoxycarbonylamino)-2-oxo-pyrrolidin- 1- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]benzoic acid

To a solution of compound Int-12d (116.74 mg, 0.160 mmol) in MeCN (5 mL) and water (1 mL) was added LiBr (69.71 mg, 0.810 mmol) and TEA (100.5 mg, 0.810 mmol). The mixture was stirred at 60°C for 12 hours. After completion, the mixture was diluted with EtOAc (50 mL) and water (30 mL). The organic phase was washed with aq. HC1 (1 M, 50 mL) and brine (30 mL), dried over anhydrous sodium sulfate, filtered concentrated in vacuo to give a residue compound Int-12 (105 mg, 0.150 mmol, 91.73% yield) as a yellow solid, which was used directly in the next step. MS obsd. (ESI + ) [(M+H) + ]: 706.4.

Intermediate 13: 4-[[2-[4-[4-[4-(Tert-butoxycarbonylamino)-2-oxo-pyrrolidin-l - yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluo ro-methyl]benzoic acid The title compound was prepared in analogy to the preparation of compound Int-12 by using compound Int-1 instead of compound Int-2 in Step 4. Compound Int-13 (280.0 mg) was obtained as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 706.0.

Intermediate 14: 5-[[2-Chloro-6-[4-[4-[(4R)-4-methyl-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]pyridine-2-carboxylic acid

The title compound was prepared in analogy to the preparation of compound Int-12 by using methyl 5-formylpyridine-2-carboxylate instead of methyl 4-formylbenzoate in Step 1. Compound Int-14 (105 mg, 0.150 mmol, 91.73% yield) was obtained as a yellow solid. MS obsd. (ESI + ) [(M+H) + ]: 707.4.

Intermediate 15: 6- [ [2-Chloro-6- [4- [4- [(4R)-4-methyl-2-oxo-pyrrolidin- 1 - yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]pyridine-3-carboxylic acid

The title compound was prepared in analogy to the preparation of compound Int-12 by using methyl 6-formylpyridine-3 -carboxylate instead of methyl 4-formylbenzoate in Step 1. Compound Int-15 (886 mg, 1.25 mmol, 80% yield) was obtained as a yellow solid. MS obsd. (ESI + ) [(M+H) + ]: 707.4.

Intermediate 16: 5- [ [2-Chloro-6- [4- [4- [(4R)-4-methyl-2-oxo-pyrrolidin- 1- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]pyrazine-2-carboxylic acid

Step 1: Pyrazine-2,5-dicarbonyl chloride

To a solution of pyrazine-2,5-dicarboxylic acid (25.0 g, 148.71 mmol) in thionyl chloride (106.15 g, 892.27 mmol) was added DMF (0.5 mL, 148.71 mmol). The reaction was stirred at 60°C for 3 hours. After completion, the mixture was concentrated in vacuo and give crude Compound Int-16a (27.8 g, 135.61 mmol, 91.19% yield) as a brown solid, which was used in the next step directly. MS obsd. (ESI + ) [(M+H) + ]: 197.2 (quenched with MeOH).

Step 2: Dimethyl pyrazine-2,5-dicarboxylate

To a solution of methanol (100 mL, 2469 mmol) and TEA (53.03 mL, 380.49 mmol) in THF (100 mL) was added compound Int-16a (26.0 g, 126.83 mmol) in THF (150 mL) dropwise at 0°C. Then the reaction was stirred at 20°C for 3 hours. After completion, the mixture was diluted with EtOAc (600 mL) and water (50 mL). The organic layer was washed with brine (100 mL x 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give crude product, which was recrystallized with MTBE (200 mL) to give Compound Int-16b (23.5 g, 119.8 mmol, 94.46% yield) as a light yellow solid. MS obsd. (ESI + ) [(M+H) + ]: 197.2 ([M+H] + ).

Step 3: Methyl 5-(2,6-dichloropyridine-4-carbonyl)pyrazine-2-carboxylate

To a solution of 2,6-dichloro-4-iodopyridine (20.0 g, 73.02 mmol) in THF (60 mL) was added isopropylmagnesium chloride lithium chloride complex (59 mL, 76.67 mmol) under nitrogen. The reaction mixture was stirred at 20°C for 1 hour. Then the reaction mixture was added into the solution of compound Int-16b (23.5 g, 119.8 mmol) in THF (360 mL) at 70°C dropwise for 1 hour. After addition, the reaction was quenched with sat. aq. NH4CI solution (100 mL) and extracted with EtOAc (300 mL x 3). The combined organic layer was washed with brine (100 mL x 3), dried over anhydrous sodium sulfate, filtered and the concentrated in vacuo to give a crude residue, which was purified by silica gel chromatography (eluent with EtOAc : PE = 5-50%) to give compound Int-16c (10.8 g, 34.6 mmol, 47.39% yield) as a light yellow solid. MS obsd. (ESI + ) [(M+H) + ]: 312.1.

Step 4: Methyl 5-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]pyrazine-2-carbo xylate

To a solution of compound Int-16c (10.8 g, 34.6 mmol) in DCM (40 mL) was added DAST (55.78 g, 346.03 mmol) in one portion. The reaction was stirred at 50°C for 32 hours. After completion, the reaction was poured into ice water and then adjusted pH to 7-8 with sat. aq. NaHCCE solution. The aqueous phase was extracted with EtOAc (300 mL x 3). The combined organic layer was washed with brine (100 mL x 3), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give crude product, which was purified by silica gel chromatography (eluent with EtOAc : PE = 5 to 25%) to givelnt-16d (10.2 g, 30.53 mmol, 86.46% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 334.1. 1 H NMR (400 MHz, DMSO- d 6 ) δ ppm: 9.38-9.43 (m, 1H), 9.30-9.35 (m, 1H), 7.95-8.01 (m, 2H), 3.98-4.02 (m, 3H).

Step 5: Methyl 5-[[2-chloro-6-[4-[4-[(4R)-4-methyl-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]pyrazine-2-carboxylate

A mixture of compound Int-2 (639 mg, 1.51 mmol), compound Int-16d (504 mg, 1.51 mmol), DIPEA (584 mg, 4.52 mmol) in DMSO (3 mL) was stirred at 120°C for 12 hours. After completion, the mixture was poured into water, extracted with EtOAc (200 mL x 3). The combined organic layer was dried over sodium sulfate, filtered and concentrated in vacuo. The crude was purified by silica gel chromatography (eluent with EtOAc : PE = 10-25%) to give compound Int-16e (762 mg, 70% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 722.2.

Step 6: 5-[[2-Chloro-6-[4-[4-[(4R)-4-methyl-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]pyrazine-2-carboxylic acid

A mixture of compound Int-16e (150 mg, 208 pmol), lithium hydroxide hydrate (85.2 mg, 2.08 mmol) in MeOH (3 mL) and water (0.5 mL) was heated at 60°C for 1 hour. After removal of the solvent, the resulting residue was treated with 5 N aq. HC1 solution and adjusted to pH = 5. The mixture was extracted with EtOAc (100 mL), and the organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo to afford compound Int-16 (132.4 mg, 0.187 mmol, 90% yield) as a yellow solid. MS obsd. (ESI + ) [(M+H) + ]: 708.2.

Intermediate 17: Tert-butyl N-[l-(3-aminopropyl)-5-(tert-butoxycarbonylamino)-3- piperidyl] carbamate

Step 1: Tert-butyl N-[5-(tert-butoxycarbonylamino)-3-pyridyl] carbamate

To a solution of pyridine-3,5-diamine (200 mg, 1.74 mmol) in 1,4-dioxane (10 mL) was added B OC2 O (1.9 g, 8.71 mmol) and DMAP (638.10 mg, 5.22 mmol) at 25°C. The mixture was stirred at 110°C for 12 hours. After completion, the reaction mixture was poured into water (50 mL) and extracted by EtOAc (50 mL x 3). The organic layer was washed by brine (50 ml x 2), dried over anhydrous sodium sulfate and concentrated in vacuo to give a residue. To the resulting residue was added MeOH (20 mL) and K 2 CO 3 , and the mixture was stirred at 25°C for 1 hour. Then, the mixture was filtered and concentrated in vacuo to give the crude compound, which was purified by silica gel chromatography (eluent with EtOAc : PE = 0.5-50%) to give compound Int-17a (500 mg, yield: 92.8%) as a yellow solid. MS obsd. (ESI + ) [(M+H) + ]: 309.9. Step 2: Tert-butyl N-[5-(tert-butoxycarbonylamino)-3-piperidyl]carbamate

To a solution of compound Int-17a (6.2 g, 20.04 mmol) in THF (1 mL) and acetic acid (4.0 mL, 0.320 mmol) was added PtCO 2 (100.0 mg, 0.320 mmol). The mixture was stirred at 60°C for 12 hours under H 2 . Then, the mixture was filtered through a Celite pad, and the filtrate was diluted with water (100 mL) and extracted with EtOAc (100 mL x 2). The organic layer was washed with brine (100 mL), dried over anhydrous sodium sulfate and concentrated in vacuo to give compound Int-17b (1.6 g, 5.07 mmol, 25.31% yield). MS obsd. (ESI + ) [(M+H) + ]: 316.2.

Step 3: Tert-butyl N-[l-[3-(benzyloxycarbonylamino)propyl]-5-(tert-butoxycarbon ylamino)- 3-piperidyl]carbamate

To a solution of benzyl N-(3-oxopropyl)carbamate (105 mg, 0.510 mmol) and compound Int-17b (160.0 mg, 0.510 mmol) in methanol (3 mL) was stirred for 1 hour. Then sodium cyanoborohydride (95.63 mg, 1.52 mmol) was added and the reaction was stirred at 20°C for 2 hours. After completion, the reaction was added with aq. LiOH solution (1 M, 20 mL) and diluted with EtOAc (20 mL). The organic layer was separated and the aqueous phase was extracted with EtOAc (10 mL x 2). The combined organic layer was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo to afford compound Int-17c (77 mg, 0.150 mmol, 29.96% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 507.4.

Step 4: Tert-butyl N-[l-(3-aminopropyl)-5-(tert-butoxycarbonylamino)-3- piperidyl] carbamate

To a solution of compound Int-17c (67.0 mg, 0.130 mmol) in methanol (1 mL) were added Pd/C (280.36 mg, 2.64 mmol) and formic acid (3043.35 mg, 66.12 mmol) in one portion. The reaction was stirred at 25 °C for 1 hour. After completion, the mixture was filtered and the filtrate was concentrated in vacuo to give the resulting residue, which was re-suspended in the sat. aq. Na 2 CO 3 solution (20 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo to afford compound Int-17 (49 mg, 0.130 mmol, 99.47% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 373.3.

Intermediate 18: Tert-butyl N-[2-[3-aminopropyl-[2-(tert-butoxycarbonyl- amino)ethyl] amino] ethyl] carbamate

Step 1: Tert-butyl N-[2-[3-(benzyloxycarbonylamino)propyl-[2-(tert- butoxycarbonylamino)ethyl] amino] ethyl] carbamate

To a solution of tert-butyl N-[2-[2-(tert-butoxycarbonylamino)ethylamino] ethylcarbamate (500.0 mg, 1.65 mmol) in THF (5 mL) and MeOH (5 mL) was added benzyl N- (3-oxopropyl)carbamate (512.27 mg, 2.47 mmol) and NaBH 3 CN (1035.6 mg, 16.48 mmol).

After stirring at 45°C for 3 hours, the reaction was concentrated in vacuo. The resulting residue was taken up in EtOAc (20 ml) and washed with water (30 mL) and brine (20 mL). The combined organic layer was dried over anhydrous sodium sulfate and concentrated in vacuo to give the crude compound, which was purified by prep-HPLC to give compound Int-18a (350 mg, 0.710 mmol, 42.94% yield) as colorless oil. MS obsd. (ESI + ) [(M+H) + ]: 495.0.

Step 2: Tert-butyl N-[2-[3-aminopropyl-[2-(tert-butoxycarbonylamino)ethyl] amino] ethyl] carbamate

To a solution of compound Int-18a (350 mg, 0.710 mmol) in MeOH (2 mL) was added Pd/C (1000 mg, 0.710 mmol) under nitrogen, then degassed by EL (1.0 L X 33 and stirred at 20°C for 2 hours. After completion, the reaction mixture was filtered through a Celite pad. The filtrate was then washed with MeOH (20 mL) and concentrated in vacuo to give compound Int-18 (200 mg, 0.550 mmol, 78.4% yield) as colorless oil. MS obsd. (ESI + ) [(M+H) + ]: 361.1.

Intermediate 19: Tert-butyl 6-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]bicyclo[3.1.0]- hexane-3-carboxylate

Step 1: Tert-butyl 6-formylbicyclo[3.1.0]hexane-3-carboxylate

To a solution of tert-butyl 6-(hydroxym ethyl )bicyclo[3.1.0]hexane-3 -carboxylate (4.5 g, 21.2 mmol) and TEMPO (0.33 g, 2.12 mmol) in EtOAc (23.55 mL) was added tri chloroisocyanuric acid (4.93 g, 21.2 mmol) at 0°C under nitrogen. The mixture was stirred at 0°C for 2 hours. After completion, the mixture was diluted with water (300 mL), extracted with EtOAc (500 mL x 2) and washed with aq. HC1 (1 N, 100 mL), aq. K 2 CO 3 (300 ml) and brine (200 ml). The combined organic layer was dried by anhydrous sodium sulfate, filtered and concentrated in vacuo to give compound Int-19a (3.1 g, 14.74 mmol, 69.55% yield) as yellow oil.

Step 2: Tert-butyl 6-[(2,6-dichloro-4-pyridyl)-hydroxy-methyl]bicyclo[3.1.0]-he xane-3- carboxylate

To a solution of 2,6-dichloro-4-iodopyridine (4.84 g, 17.69 mmol) in THF (10 mL) was added isopropylmagnesium chloride lithium chloride complex solution (1.3 M in THF, 17.01 mL, 22.11 mmol) at -40°C. The mixture was stirred at 25°C for 1 hour. Then compound Int-19a (3.1 g, 14.74 mmol) in THF (10 mL) was added into the reaction at -40 °C, and the mixture was stirred at 25°C for 1 hour. After completion, the mixture was quenched with sat. aq. NH4CI solution (300 mL), extracted with EtOAc (500 mL x 2) and washed with brine (300 mL). The combined organic layer was dried by anhydrous sodium sulfate, filtered, and concentrated in vacuo to give a crude residue, which was purified by silica gel column (eluent with EtOAc : PE = 0-100%). Compound Int-19b (4.3 g, 12 mmol, 81.41% yield) was obtained as yellow oil. MS obsd. (ESI + ) [(M+H) + ]: 358.1.

Step 3: Tert-butyl 6-(2,6-dichloropyridine-4-carbonyl)bicyclo[3.1.0]hexane-3-ca rboxylate

To a solution of DMP (5.82 g, 13.73 mmol) in THF (30 mL) and was added compound Int-19b (4.1 g, 11.44 mmol). The reaction mixture was stirred at 25°C for 1 hour. After completion, the mixture was diluted with EtOAc (300 mL) and adjusted to pH = 9 with aq. K 2 CO 3 solution. Then the resulting mixture was filtered through fritted glass funnel. The combined organic layer was washed with brine (200 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give compound Int-19c (3.9 g, 10.95 mmol, 95.66% yield) as yellow oil. MS obsd. (ESI + ) [(M+H) + ]: 356.1.

Step 4: Tert-butyl 6-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]bicyclo[3.1.0]-h exane-3- carboxylate

To a solution of compound Int-19c (3.8 g, 10.67 mmol) in DCM (5 mL) was added DAST (30 mL, 10.67 mmol), and the mixture was stirred at 50°C for 60 hours. After completion, the mixture was quenched with ice water and adjusted to pH = 9 with 1 M aq. NaHCO 3 solution. The mixture was extracted with EtOAc (500 mL x 2) and washed with brine (500 mL). The combined organic layer was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give a crude residue, which was purified by silica gel column (eluent with EtOAc : PE = 0-10%) and prep-HPLC to give compound Int-19 (200 mg, 0.260 mmol, 2.48% yield). MS obsd. (ESI + ) [(M+H) + ]: 378.3.

Intermediate 20: Tert-butyl N-[(3R)-5-oxo-l-(5-piperazin-l-ylsulfonyl-2-pyridyl)pyrrolid in- 3-yl] carbamate

Step 1: 6-Bromopyridine-3-sulfonyl chloride

To a solution of 6-bromopyri din-3 -amine (5.0 g, 28.9 mmol) in HC1 (85 mL, 1020 mmol) was added NaNCO 2 (1.79 g, 26.01 mmol) in water (25 mL) slowly at 0°C. The mixture was stirred at 0°C for 2 hours. SO 2 was bubbled through a stirred solution of CuC1 2 (1976.76 mg, 11.56 mmol) in acetic acid (50 mL) at 0°C for 0.5 hour. Then the diazotized reaction mixture prepared above was added. The resulting mixture was slowly warmed to room temperature and stirred for 30 minutes. After completion, the mixture was added EtOAc (200 mL) and water (200 mL). The organic phase was washed with aq. Na 2 CO 3 , dried over anhydrous sodium sulfate, concentrated in vacuo to give crude compound Int-20a (5 g, 23.58 mmol, 81.59% yield) as yellow oil, which was used directly in the next step.

Step 2: Benzyl 4-[(6-chloro-3-pyridyl)sulfonyl]piperazine-l-carboxylate

To a solution of compound Int-20a (4.13 g, 19.49 mmol) in DCM (1 mL) was added DIPEA (12.1 g, 97.47 mmol) and 1 -Cbz-piperazine (4.29 g, 19.49 mmol). The mixture was stirred at 25°C for 30 minutes. Then, the mixture was concentrated in vacuo to give a crude residue, which was triturated in MeOH to afford compound Int-20b (4.5 g, 11.37 mmol, 52.43% yield) as a white solid. MS obsd. (ESI + ) [(M+H)] + : 395.8.

Step 3: Benzyl 4-[(6-iodo-3-pyridyl)sulfonyl]piperazine-l-carboxylate

To a solution of compound Int-20c (4.0 g, 10.1 mmol) in acetone (100 mL) was added Nal (4.5 g, 30.31 mmol) and TFA (2.0 mL, 10.1 mmol). The mixture was stirred at 25°C for 12 hours. After completion, the mixture was concentrated in vacuo to give a crude residue, which was triturated in MeOH to afford compound Int-20c (2.5 g, 5.13 mmol, 50.77% yield) as a white solid. MS obsd. (ESI + ) [(M+H)] + : 487.7.

Step 4: Benzyl 4-[[6-[(4R)-4-(tert-butoxycarbonylamino)-2-oxo-pyrrolidin-l- yl]-3- pyridyl]sulfonyl]piperazine-l-carboxylate

To a solution of tert-butyl N-[(3A)-5-oxopyrrolidin-3-yl]carbamate (123.27 mg, 0.620 mmol) and compound Int-20c (200.0 mg, 0.410 mmol) in DMF (1 mL) was added Cui (62.38 mg, 0.330 mmol), K2CO3 (169.91 mg, 1.23 mmol) and DMEDA (28.89 mg, 0.330 mmol). The mixture was stirred under nitrogen at 70°C for 2 hours. After completion, the mixture was added EtOAc (50 mL) and filtered. The organic phase was added NEE* H 2 O (5%, 30 mL). The combined organic layer was washed with brine (50 mL), dried over anhydrous sodium sulfate, concentrated in vacuo to give a residue, which was purified by prep-HPLC to give compound Int-20d (160 mg, 0.290 mmol, 69.66% yield) as a white solid. MS obsd. (ESI+) [(M+H)] + : 560.0.

Step 5: Tert-butyl N-[(3R)-5-oxo-l-(5-piperazin-l-ylsulfonyl-2-pyridyl)pyrrolid in-3- yl] carbamate

To a solution of compound Int-20d (160.0 mg, 0.290 mmol) in methanol (20 mL) was added Pd/C (80.0 mg, 0.060 mmol). The mixture was stirred under H 2 (15psi) at 25°C for 2 hours. After completion, the mixture was filtered through Celite. The filtrate was concentrated in vacuo to give the crude compound Int-20 (100 mg, 0.240 mmol, 82.2% yield) as a white solid, which was used directly in the next step. MS obsd. (ESI+) [(M+H)] + : 426.2.

Intermediate 21: Tert-butyl N-[(3R)-5-oxo-l-[4-(4-piperidylsulfonyl)phenyl] pyrrolidin-3- yl] carbamate

Step 1: Benzyl 4-(4-iodophenyl)sulfanylpiperidine-l-carboxylate

To a solution of 4-bromobenzenethiol (5.0 g, 26.45 mmol), benzyl 4-hydroxypiperidine-l- carboxylate (4.4 mL, 29.09 mmol) and tributylphosphine (8.03 g, 39.67 mmol) in THF (150 mL) was added N,N,N,N,-tetramethylazodicarboxamide (6.83 g, 39.67 mmol) at 0°C. The mixture was stirred at 45°C for 2 hours. After completion, the reaction mixture was diluted with water (400 mL), extracted with EtOAc (400 mL x 2) and washed with brine (500 mL). The combined organic layer was dried by anhydrous sodium sulfate, filtered and concentrated in vacuo to give a crude residue, which was purified by silica gel column (eluent with EtOAc : PE = 0-10%) to give compound Int-21a (7.5 g, 18.46 mmol, 69.79% yield) as colorless oil. MS obsd. (ESI+) [(M+H)] + : 407.2.

Step 2: Benzyl 4-(4-iodophenyl)sulfinylpiperidine-l-carboxylate

To a solution of compound Int-21a (7.3 g, 17.97 mmol) in THF (100 mL) was added m- CPBA (4.38 g, 21.56 mmol). The reaction was stirred at 25°C for 2 hours. After completion, the reaction mixture was quenched with sat. aq. ISfeSCE solution and extracted with EtOAc (200 mL x 2). The combined organic layer was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give a crude product, which was purified by silica gel column to give compound Int-21b (5.7 g, 13.5 mmol, 75.12% yield) as a white solid. MS obsd. (ESI+) [(M+H)] + : 424.1.

Step 3: Benzyl 4-[4-[(4R)-4-(tert-butoxycarbonylamino)-2-oxo-pyrrolidin-l- yl]phenyl]sulfinylpiperidine-l-carboxylate

To a solution of compound Int-21c (237.04 mg, 1.18 mmol) in DMF (5 mL) was added tert-butyl N-[(3A)-5-oxopyrrolidin-3-yl]carbamate (355.57 mg, 1.78 mmol), Cui (157.74 mg, 0.830 mmol), DMEDA (72.93 mg, 0.830 mmol) and K 2 CO 3 (490.13 mg, 3.55 mmol) at 30°C. The reaction mixture was stirred at 90°C under nitrogen for 2 hours. After completion, the reaction mixture was filtered and the filtrate was dissolved in EtOAc (200 mL). The organic layer was washed with brine (50 mL x 3), and concentrated in vacuo to give a crude residue. The residue was purified by prep-HPLC to afford compound Int-21c (450 mg, 0.830 mmol, 70.17% yield) as colorless oil. MS obsd. (ESI+) [(M+H)] + : 542.2.

Step 4: Benzyl 4-[4-[(4R)-4-(tert-butoxycarbonylamino)-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperidine-l-carboxylate

To a solution of compound Int-21c (220.0 mg, 0.410 mmol) in THF (5 mL) was added m- CPBA (164.92 mg, 0.810 mmol) at 30°C. The reaction mixture was stirred at 30°C for 2 hours. After completion, the reaction mixture was quenched with sat. aq. NazSOs solution (20 mL), and extracted with EtOAc (100 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give a crude product, which was purified by prep-HPLC to give compound Int-21d (180 mg, 0.320 mmol, 79.47% yield) as a white solid. MS obsd. (ESI+) [(M+H)] + : 558.3.

Step 5: Tert-butyl N-[(3R)-5-oxo-l-[4-(4-piperidylsulfonyl)phenyl] pyrrolidin-3- yl] carbamate

To a solution of compound Int-21d (180.0 mg, 0.320 mmol) in methanol (5 mL) was added formic acid (5 mL) and Pd/C (100 mg) at 30°C under nitrogen. After stirring at 30°C for 16 hours, the reaction mixture was filtered and the filtrate was concentrated in vacuo to give a crude residue, which was purified by prep-HPLC to give compound Int-21 (100 mg, 0.240 mmol, 73.15% yield) as a white solid. MS obsd. (ESI+) [(M+H)] + : 424.2.

Intermediate 22: Tert-butyl N-[3-(tert-butoxycarbonylamino)propyl]-N-[5-oxo-l-(4- piperazin-l-ylsulfonylphenyl)pyrrolidin-3-yl]carbamate

Step 1: Benzyl 4-[4-[4-(3-hydroxypropylamino)-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazine-l-carboxylate To a solution of compound Int-4b (5.0 g, 11.33 mmol) in NMP (50 mL) was added 3- amino-1 -propanol (5.2 mL, 67.95 mmol). The mixture was stirred at 90°C for 2 hours. After completion, the mixture was diluted with water (150 mL) and extracted with EtOAc (80 mL x 3). The combined organic layer was washed with brine (100 mL x 3), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give a crude residue compound Int-22a, which was used in the next step without further purification. MS obsd. (ESI + )[(M+H) + ]: 517.2.

Step 2: Benzyl 4-[4-[4-[tert-butoxycarbonyl(3-hydroxypropyl)amino]-2-oxo-py rrolidin-l- yl]phenyl]sulfonylpiperazine-l-carboxylate

To a solution of compound Int-22a (5.2 g, 10.07 mmol) in DCM (50 mL) was added DIPEA (7.01 mL, 40.26 mmol) and di-t -butyl di carb onate (4.63 mL, 20.13 mmol) in icewater bath, then the reaction was stirred at 25°C for 2 hours. After completion, the mixture was concentrated in vacuo and purified by silica gel chromatography (eluent with MeOH : DCM = 0%-10%) to afford compound Int-22b (5.8 g, 9.4 mmol, 93.43% yield) as a light yellow solid. MS obsd. (ESI + )[(M+H) + ]: 617.3.

Step 3: Benzyl 4-[4-[4-[3-bromopropyl(tert-butoxycarbonyl)amino]-2-oxo-pyrr olidin-l- yl]phenyl]sulfonylpiperazine-l-carboxylate

To a solution of compound Int-22b (5.8 g, 9.4 mmol) in DCM (60 mL) was added carbon tetrabromide (6.24 g, 18.81 mmol) at 0°C under nitrogen then triphenylphosphine (6.17 g, 23.51 mmol, 2.5 eq) was added into the solution. The mixture was stirred at 0°C for 2 hours. After completion, the mixture was washed with brine (60 mL x 3). The organic layer was concentrated in vacuo to give a crude, which was purified by reverse-phase chromatography to give compound Int-22c (5.4 g, 7.95 mmol, 84.49% yield) as a light yellow solid. MS obsd. (ESI + )[(M+H) + ]: 679.2. Step 4: Benzyl 4-[4-[4-[3-aminopropyl(tert-butoxycarbonyl)amino]-2-oxo-pyrr olidin-l- yl]phenyl]sulfonylpiperazine-l-carboxylate

To a solution of compound Int-22c (5.0 g, 7.36 mmol) in ethanol (50 mL) was added ammonium hydroxide (10.0 mL, 30%). The mixture was stirred at 50°C for 4 hours. After completion, the mixture was concentrated in vacuo to afford compound Int-22d (3.2 g, 5.2 mmol, 64.99% yield) as a yellow solid. MS obsd. (ESI + )[(M+H) + ]: 616.3.

Step 5: Benzyl 4-[4-[4-[tert-butoxycarbonyl-[3-(tert-butoxycarbonylamino)-p ropyl]amino]- 2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazine-l-carboxylat e

To a solution of compound Int-22d (2.0 g, 3.25 mmol) in DCM (20 mL) was added DIPEA (1.7 mL, 9.74 mmol) and di-t -butyl di carbonate (1.49 mL, 6.5 mmol) in ice- water bath, then the reaction was stirred at 25°C for 2 hours. After completion, the mixture was concentrated in vacuo and purified by silica gel chromatography (eluent with MeOH : DCM = 0%-10%) to afford compound Int-22e (2.1 g, 2.93 mmol, 90.31% yield) as a white solid. MS obsd. (ESI + )[(M+Na) + ]: 738.3.

Step 6: Tert-butyl N-[3-(tert-butoxycarbonylamino)propyl]-N-[5-oxo-l-(4-piperaz in-l- ylsulfonylphenyl)pyrrolidin-3-yl]carbamate

To a solution of compound Int-22e (1.0 g, 1.4 mmol) in THF (30 mL) was added Pd/C (100 mg). The mixture was stirred at 50°C under H 2 for 16 hours. After completion, the reaction mixture was filtered and the filtrate was concentrated in vacuo to give a crude product, which was purified by silica gel column (eluent with MeOH : DCM= 0%-10%) to afford compound Int-22 (620 mg, 1.07 mmol, 72.48% yield) as a white solid. MS obsd. (ESI + )[(M+H) + ]: 582.3. 1 H NMR (400 MHz, DMSO) δ 7.99 (d, J= 8.7 Hz, 2H), 7.71 (d, J= 8.8 Hz, 2H), 6.81 (s, 1H), 4.36 (s, 1H), 4.14 (t, J= 9.2 Hz, 1H), 3.84 (dd, J= 9.6, 5.3 Hz, 1H), 3.23 (d, J= 5.7 Hz, 2H), 2.93-2.88 (m, 2H), 2.76-2.72 (m, 10H), 1.65-1.54 (m, 2H), 1.36 (d, J= 11.1 Hz, 18H).

Int-23: 7rans-4-[[2-chloro-6-[4-[4-[(4R)-4-(tert-butoxycarbonylamino )-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]sulfanyl]cyclohe xanecarboxylic acid

Step 1: 4-Benzylsulfanyl-2,6-dichloro-pyridine

To a solution of benzyl mercaptan (4.58 g, 36.86 mmol), 4-bromo-2,6-dichloro-pyridine (8.2 g, 36.14 mmol) in DMF (50 mL) were added TEA (15.11 mL, 108.42 mmol), tris(dibenzylideneacetone)dipalladium(0) (1.32 g, 1.45 mmol) and 9,9-dimethyl-4,5- bis(diphenylphosphino)xanthene (2.09 g, 3.61 mmol) in one portion under nitrogen. The reaction was stirred at 80°C for 2 hours. After completion, the reaction was diluted with water (100 ml) and extracted with EtOAc (200 mL x 3). The combined organic layer was washed with sat. aq CaCL (200 mL x 3) and brine (200 mL x 3). The organic layer was dried over anhydrous sodium sulfate and concentrated in vacuo to give a crude residue, which was purified by silica gel chromatography (eluent with EtOAc : PE = 0-100%) to give compound Int-23a (8.8 g, 32.57 mmol, 90.12% yield) as a light yellow solid. MS obsd. (ESI + )[(M+H) + ]: 269.8.

Step 2: 2,6-Dichloropyridine-4-thiol

To a solution of compound Int-23a (4.0 g, 14.8 mmol) in DCM (50 mL) was added aluminum trichloride (9.87 g, 74.02 mmol). The reaction was stirred at 25°C for 2 hours. After completion, the reaction mixture was poured into water (300 mL) in ice-water bath and extracted by EtOAc (300 mL x 3). The combined organic layer was washed with brine (200 mL x 2, dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo to give a crude product, which was purified by prep-HPLC to afford compound Int-23b (1.5 g, 8.33 mmol, 56.27% yield) as a yellow solid. MS obsd. (ESI + )[(M+H) + ]: 180.4.

Step 3: Trans-methyl 4- [(2, 6-dichloro-4-pyridyl)sulfanyl] cyclohexanecarboxylate

To a solution of compound Int-23c (500.0 mg, 2.78 mmol), triphenylphosphine (3637.68 mg, 13.88 mmol) and methyl 4-hydroxycyclohexanecarboxylate (900.0 mg, 5.69 mmol) in THF (5 mL) was added diisopropyl azodi carboxyl ate (1683.61 mg, 8.33 mmol) dropwise at 0°C. The mixture was stirred at 70°C for 16 hours. After completion, the mixture was diluted with water (100 mL) and extracted with EtOAc (200 mL x 2). The combined organic layer was washed with brine (200 mL), dried over anhydrous sodium sulfate, concentrated in vacuo to give a crude residue, which was purified by prep-TLC (PE: EA = 5:1, Rf = 0.35) and prep-HPLC. The compound Int-23c (260 mg, 0.810 mmol, 29.33% yield) was obtained as colorless oil. MS obsd. (ESI + )[(M+H) + ]: 320.2.

Step 4 : Trans-4- [ [2-chloro-6- [4- [4- [(4R)-4-(tert-butoxycarbonylamino)-2-oxo-pyrrolidin- 1 - yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]sulfanyl]cyclohe xanecarboxylic acid

The title compound was prepared in analogy to the preparation of compound Int-5 by replacing compound Int-23c instead oflnt-5e in Step 6. Compound Int-23 (470 mg) was obtained as a light yellow solid. MS obsd. (ESI + ) [(M+H) + ]: 694.2. Int-24: 7rans-4-[difluoro-[6-methyl-2-[4-[4-[(4R)-4-(tert-butoxycarb onylamino)-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]pyrimidin-4- yl] methyl] cyclohexanecarboxylic acid

Step 1: Chlorolithium; chloro-(2,2,6,6-tetramethyl-l-piperidyl)magnesium

A solution of isopropylmagnesium chloride lithium chloride complex solution (1.3 M in THF, 50.0 mL, 65 mmol) was added 2,2,6,6-tetramethylpiperidine (12.07 mL, 71.5 mmol) dropwise at -40°C. The reaction was stirred at 25°C under nitrogen for 1 day. The compound Int-24a (60 g, 247.52 mmol, 86.55% yield) in THF (60.0 mL) was obtained and used in the next step directly.

Step 2: Trans-methyl 4-[(6-chloro-2-methylsulfanyl-pyrimidin-4-yl)-hydroxy- m ethyl] cyclohexanecarboxylate

To a solution of 4-chloro-2-methylsulfanyl-pyrimidine (3.0 g, 18.68 mmol) in THF (8 mL) was added compound Int-24a (18.68 mL, 20.55 mmol) at -40°C. The mixture was stirred at 0°C for 40 minutes and then added into the solution of trans-methyl 4-formylcyclohexanecarboxylate (3.5 g, 20.55 mmol) in THF (12 mL) at -40°C. The reaction mixture was stirred at 25°C for 1 hour. After the starting material was consumed, the mixture was poured into water (200 mL) and extracted with EtOAc (200 mL x 3). The combined organic layer was washed with brine (100 mL x 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give a crude residue, which was purified by silica gel chromatography on (eluent with EtOAc : PE = 0-100%) to give compound Int-24b (1.8 g, 5.44 mmol, 29.13% yield) as light yellow oil. MS obsd. (ESI + ) [(M+H) + ]: 331.3.

Step 3: Trans-methyl 4-(6-chloro-2-methylsulfanyl-pyrimidine-4- carbonyl)cyclohexanecarboxylate

To a solution of compound Int-24b (1.7 g, 5.14 mmol) in THF (10 mL) was added DMP (2.62 g, 6.17 mmol) at 0°C, and the mixture was stirred at 25°C for 2 hours. After completion, the mixture was quenched with ice water and adjusted to pH = 9 with aq. NaHCCE solution. The mixture was extracted with EtOAc (100 mL x 2) and washed with brine (100 ml). The combined organic layer was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give a crude residue, which was purified by silica gel column (eluent with EtOAc : PE = 0-50%) to give compound Int-24c (1.6 g, 4.87 mmol, 94.7% yield) as a yellow solid. MS obsd. (ESI + ) [(M+H) + ]: 329.3.

Step 4: Trans-methyl 4-[(6-chloro-2-methylsulfanyl-pyrimidin-4-yl)-difluoro- m ethyl] cyclohexanecarboxylate

To a solution of compound Int-24c (1.6 g, 4.87 mmol) in THF (10 mL) was added DAST (10.0 mL, 4.87 mmol). The mixture was stirred at 25°C for 2 hours. After completion, the mixture was quenched with ice water, adjusted to pH = 9 with aq. NaHCOs solution, and extracted with EtOAc (200 mL x 2). The combined organic layer was washed with brine (100 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give a crude residue, which was purified by silica gel column (eluent with EtOAc : PE = 0-20%) to give compound Int-24d (1.5 g, 4.28 mmol, 87.87% yield) as a yellow solid. MS obsd. (ESI + ) [(M+H) + ]: 351.3.

Step 5: Trans-methyl 4-[difluoro-(6-methyl-2-methylsulfanyl-pyrimidin-4- yl)methyl]cyclohexanecarboxylate

To a solution of compound Int-24d (900.0 mg, 2.57 mmol), chloro(2- dicyclohexylphosphino-2',4',6'-triisopropyl-l, 1 '-biphenyl)[2-(2'-amino-l, 1 biphenyl)]palladium(II) (201.85 mg, 0.260 mmol) and potassium phosphate (1.6 g, 7.7 mmol) in 1,4-dioxane (5 mL) and water (2 mL) was added trimethylboroxine (9.66 mL, 30.79 mmol). The mixture was stirred at 80°C under nitrogen for 12 hours. After completion, the mixture was diluted with water (100 mL), and extracted with EtOAc (100 mL x 2). The combined organic layer was washed with brine (100 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give a crude residue, which was purified by silica gel column (eluent with EtOAc : PE = 10-30%) to afford compound Int-24e (420 mg, 1.27 mmol, 34.69% yield) as yellow oil. MS obsd. (ESI + ) [(M+H) + ]: 331.3.

Step 6: Trans-methyl 4-[difluoro-(6-methyl-2-methylsulfonyl-pyrimidin-4- yl)methyl]cyclohexanecarboxylate

To a solution of compound Int-24e (400.0 mg, 0.850 mmol) in THF (5 mL) was added m- CPBA (516.18 mg, 2.54 mmol), and the mixture was stirred at 25°C for 1 hour. After completion, the reaction mixture was quenched by sat. aq. Na 2 SO 3 solution and extracted with EtOAc (100 mL x 2). The combined organic layer was washed with brine (200 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give a residue, which was purified by silica gel column (eluent with EtOAc : PE = 5-50%) to afford compound Int-24f (355 mg, 0.980 mmol, 87.85% yield) as yellow oil. MS obsd. (ESI + ) [(M+H) + ]: 363.2.

Step 7 : Trans-4- [difluoro- 16-m el hy 1-2- [4- [4- [(4R)-4-(tert-bntoxycarbonylamino)-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]pyrimidin-4- yl] methyl] cyclohexanecarboxylic acid

The title compound was prepared in analogy to the preparation of compound Int-5 by replacing compound Int-24f instead of compound Int-5e in Step 6. Compound Int-24 (260 mg) was obtained as a light yellow solid. MS obsd. (ESI + ) [(M+H) + ]: 693.4.

Intermediate 25: Tert-butyl N-methyl-N -[(3R)-5-oxo-l-(4-piperazin-l- ylsulfonylphenyl)pyrrolidin-3-yl]carbamate

Step 1: Benzyl 4-[4-[(4R)-4-(tert-butoxycarbonylamino)-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazine-l-carboxylate

To a solution of Int-la (412 mg, 2.06 mmol) in DMF (10 mL) was added benzyl 4-(4- iodophenyl)sulfonylpiperazine-l-carboxylate ( 1 g, 2.06 mmol), K2CO3 (853 mg, 6.17 mmol), Cui (195.81 mg, 1.03 mmol) and DMEDA (72.42 mg, 0.820 mmol). The suspension was stirred at 80 °C for 2 hours under N2. After completion, the reaction was diluted with water (50 mL), extracted with EtOAc (50 mL x 3). The combined organic layer was washed with brine (30 mL * 2), dried over anhydrous sodium sulfate and concentrated. The crude product was purified by trituration in EtOAc (20 mL), which was filtered and the filtrate was concentrated to give Int- 25a (660 mg, 1.18 mmol, 57.45% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 559.1.

Step 2: Benzyl 4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpipe razine-l- carboxylate

A mixture of Int-25a (660.0 mg, 1.18 mmol) and TFA (6.0 mL) in the DCM (6 mL) was stirred at room temperature for 2 hours. After the starting material was consumed, the mixture was concentrated in vacuo to afford Int-25b (660 mg, 1.15 mmol, 97.57% yield) as a light yellow gum. MS obsd. (ESI + ) [(M+H) + ]: 459.2.

Step 3: Benzyl 4-[4-[(4R)-4-[(3,4-dimethoxyphenyl)methyl-methyl-amino]-2-ox o-pyrrolidin- l-yl]phenyl]sulfonylpiperazine-l-carboxylate

To a solution of Int-25b (660.0 mg, 1.44 mmol) in THF (1 mL) was added Ti(z-PrO)4 (4.1 g, 14.39 mmol) and 2,4-dimethoxybenzaldehyde (239.18 mg, 1.44 mmol) at 15 °C and then warmed to 25 °C. The mixture was stirred at 25 °C for 2 hours. The mixture was cooled to 15°C and then added methanol (1 mL) and NaBH 3 CN (452.25 mg, 7.2 mmol). After stirring at 25 °C for 12 hours, paraformaldehyde (705.9 mg, 7.2 mmol, 5 eq) was added and the reaction was kept stirring at 25°C for additional 12 hours. After completion, the mixture was diluted with DCM (50 mL) and water (50 mL). The aqueous phase was extracted with DCM (50 mL x 2). The combined organic layer was washed with brine (50 mL), dried over anhydrous MgSO 4 , filtered, and concentrated in vacuo to give the crude product, which was purified by silica gel chromatography (eluent with EtOAc : PE = 0-50%) to afford Int-25c (210 mg, 0.340 mmol, 23.43% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 623.3. Step 4: Tert-butyl N-methyl-/V-[(31?)-5-oxo-l-(4-piperazin-l-ylsulfonylphenyl)p yrrolidin-3- yl] carbamate

To a solution of Int-25c (100.0 mg, 0.160 mmol) in the mixture of THF (0.1 mL) and IPA (1 mL) was added Pd/C (10.0 mg, 5%, wet) in one portion under N2. The mixture was degassed under vacuum and purged with H 2 three times. The reaction mixture was stirred at 40 °C under H 2 balloon for 12 hours. After completion, the mixture was filtered and the filtration was concentrated in vacuo to afford Int-25 (75 mg, 0.150 mmol, 95.59% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 489.3.

Intermediate 26: 5-[[2-chloro-6-[4-[4-[(4R)-4-methyl-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]bicyclo[2.2.2]octane-2- carboxylic acid

The title compound was prepared in analogy to the preparation of compound Int-5 by using 5-methoxycarbonylbicyclo[2.2.2]octane-2-carboxylic acid instead of Irans-4- methoxycarbonyl cyclohexanecarboxylic acid in Step 1. Compound Int-26 (450 mg) was obtained as a grey solid. MS obsd. (ESI + ) [(M+H) + ]: 738.3.

Example 001: Trans-methyl 4-[[2-[4-[4-(4-amino-2-oxo-pyrrolidin-l- yl)phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluo ro- m ethyl] cyclohexanecarboxylate

Step 1: Trans-methyl 4-[[2-[4-[4-(4-amino-2-oxo-pyrrolidin-l-yl)phenyl]sulfonylpi perazin- l-yl]-6-chloro-4-pyridyl]-difluoro-methyl]cyclohexanecarboxy late

Compound Int-5e (250 mg, 344 pmol) was dissolved in the DCM (10 mL), followed by adding TFA (265 pL, 3.44 mmol). The reaction was stirred at room temperature for 1 hour. Then, the reaction mixture was concentrated in vacuo and purified by prep-HPLC to give product Example 001 (35 mg, 0.056 mmol, 15.4% yield) as a white solid. MS obsd. (ESI + ) [(M+H)] + : 626.2.

Example 001: 1 H NMR (400 MHz, DMSO-d 6 ) δ ppm 7.91 (m, J=8.93Hz, 2H), 7.75 (m, J=8.93Hz, 2H), 6.76 (s, 1H), 6.69 (s, 1H), 4.00 (dd, 7=9.90, 6.11Hz, 1H), 3.60-3.70 (m, 6H), 3.45-3.54 (m, 1H), 3.32 (br s, 3H), 2.96 (br t, 7=4.65Hz, 4H), 2.78 (dd, J=16.87, 6.97Hz, 1H), 2.52-2.53 (m, 1H), 2.11-2.38 (m, 3H), 1.91 (br d, 7=10.64Hz, 2H), 1.66 (br d, 7=10.64Hz, 2H), 1.11-1.35 (m, 4H). 19 F NMR (376 MHz, DMSO-7 6 ) δ ppm -103.88 (br d, 7=14.98Hz, IF).

Example 002 : Trans-4- [ [2- [4- [4- [(4R)-4-amino-2-oxo-pyrrolidin- 1- yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluo ro-methyl]-N-(azetidin-3- ylmethyl)cyclohexanecarboxamide Step 1: Trans-4-[[2-chloro-6-[4-[4-[(4R)-4-methyl-2-oxo-pyrrolidin-l - yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]-N-

(cyclobutylmethyl)cyclohexanecarboxamide

A mixture of azeti din-3 -ylmethanamine (24.52 mg, 0.132 mmol), compound Int-5 (75 mg, 0.105 mmol), HATU (57 mg, 0.15 mmol) and DIPEA (55.12 pL, 0.316 mmol) in DCM (15 mL) was stirred at room temperature overnight. After completion, the reaction mixture was diluted with EtOAc (50 mL x 3), washed with brine (50 mL). Then, the combined organic layer was dried over anhydrous sodium sulfate, filtered, concentrated in vacuo to give crude compound 002a (80 mg, 86.28% yield) as a light yellow solid, which was used directly in the next step. MS obsd. (ESI + ) [(M+H)] + : 880.5.

Step 2 : Trans-4- [ [2- [4- [4- [(4R)-4-amino-2-oxo-pyrrolidin-l-yl] phenyl] sulf onylpiperazin- 1- yl]-6-chloro-4-pyridyl]-difluoro-methyl]-N-(azetidin-3-ylmet hyl)cyclohexanecarboxamide

Compound 002a (80 mg, 91 pmol) was dissolved in the DCM (5 mL), followed by adding TFA (265 pL, 3.44 mmol). The reaction was stirred at room temperature for 1 hour and went completed. Then, the reaction was concentrated in vacuo and purified by prep-HPLC to give product Example 002 (19 mg, 30.7% yield) as white solid. MS obsd. (ESI + ) [(M+H)] + : 680.2.

Example 002: 1 H NMR (400 MHz, METHANOL-d 4 ) δ ppm 7.95 (m, 7=8.93Hz, 2H), 7.86 (m, 7=8.93Hz, 2H), 6.67 (d, 7=9.41Hz, 2H), 5.27-5.46 (m, 1H), 4.95-5.16 (m, 2H), 4.42 (dd, 7=11.43, 7.03Hz, 1H), 4.14-4.27 (m, 1H), 4.02-4.11 (m, 2H), 3.84-4.00 (m, 3H), 3.64-3.77 (m, 4H), 3.36- 3.44 (m, 2H), 3.21 (dd, 7=18.16, 8.25Hz, 1H), 3.03-3.13 (m, 5H), 2.71 (dd, 7=18.03, 3.00Hz, 1H), 2.50 (br s, 1H), 2.00-2.07 (m, 2H), 1.57 (br d, 7=5.75Hz, 6H), 1.33 (br d, 7=18.10Hz, 1H). 19 F NMR (376 MHz, METHANOL-7 4 ) δ ppm -111.94 -101.94 (m, IF). The following Examples 003 to 052 were prepared in analogy to the procedure described for the preparation of Example 002, replacing compound Int-5 with “LINKER” in Step 1, and azeti din-3 -ylmethanamine with “TAIL” in Step 1, by the reagents indicated in Table 1.

Table 1 : Compound synthesis and characterization

Example 055: N-(3-aminopropyl)-6-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-p yrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]bicyclo[3.1.0]hexane-3-

Step 1: 6-[(2,6-Dichloro-4-pyridyl)-difluoro-methyl]bicyclo[3.1.0]he xane-3-carboxylic acid To a solution of compound Int-19 (200.0 mg, 0.530 mmol) in DCM (2 mL) was added TFA (2.0 mL, 25.96 mmol), and the mixture was stirred at 25°C for 2 hours. After completion, the mixture was concentrated in vacuo to give a crude residue, which was purified by prep- HPLC. The compound 055a (120 mg, 0.370 mmol, 70.45% yield) was obtained as yellow oil. MS obsd. (ESI+) [(M+H) + ]: 322.2. Step 2: Tert-butyl N-[3-[[6-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]bicyclo[3 .1.0]hexane-

3-carbonyl] amino] propyl] carbamate

To a solution of compound 055a (120.0 mg, 0.370 mmol), N-Boc- 1,3 -diaminopropane (194.72 mg, 1.12 mmol) and DIPEA (0.19 mL, 1.12 mmol) in DMF (0.5 mL) was added HATU (212.47 mg, 0.560 mmol). The mixture was stirred at 25°C for 2 hours. After completion, the mixture was diluted with water (100 mL), extracted with EtOAc (100 mL x 2) and washed with brine (200 mL). The combined organic layer was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give a crude residue, which was purified by prep-HPLC to afford compound 055b (120 mg, 0.250 mmol, 67.34% yield) as a yellow solid. MS obsd. (ESI+) [(M+H) + ]: 421.9.

Step 3: Tert-butyl N-[3-[[6-[[2-chloro-6-[4-[4-[(4R)-4-(tert-butoxycarbonylamin o)-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-di fluoro- methyl]bicyclo[3.1.0]hexane-3-carbonyl]amino]propyl]carbamat e

To a solution of compound 055b (120 mg, 0.250 mmol) and compound Int-5 (117.14 mg, 0.280 mmol) in DMSO (2 mL) was added DIPEA (1.0 mL, 5.74 mmol). The mixture was stirred at 100°C for 12 hours. After completion, the mixture was diluted with water (100 mL), extracted with EtOAc (100 mL x 2) and washed with brine (100 mL). The combined organic layer was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give a residue, which was purified by prep-TLC (eluent with EtOAc : PE = 33%) and prep-HPLC. Compound 054c (40 mg, 0.050 mmol, 18.4% yield) was obtained as a yellow solid. MS obsd. (ESI+) [(M+H) + ]: 866.3. Step 4: N-(3-aminopropyl)-6-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-p yrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]bicyclo[3.1.0]hexane-3-

To a solution of compound 055c (40.0 mg, 0.050 mmol) in DCM (2 mL) was added TFA (2.0 mL, 25.96 mmol). The mixture was stirred at 25°C for 1 hour. After completion, the mixture was concentrated in vacuo to give a crude residue, which was purified by prep-HPLC to give Example 055 (10.6 mg, 0.020 mmol, 29.24% yield) as a white solid. MS obsd. (ESI+) [(M+H) + ]: 666.5.

Example 055: 1 H NMR (400 MHz, DMSO-7 6 ) δ ppm 8.61 (br s, 3H), 8.02-8.27 (m, 1H), 7.94- 8.02 (m, 3H), 7.91 (d, 7=8.93 Hz, 2H), 7.76-7.81 (m, 2H), 6.65-6.88 (m, 2H), 4.26 (br dd, 7=11.07, 6.91 Hz, 1H), 4.05 (br s, 1H), 3.90 (br d, 7=11.00 Hz, 1H), 3.66 (br s, 4H), 3.01-3.16 (m, 3H), 2.96 (br s, 4H), 2.58-2.79 (m, 3H), 1.94-2.39 (m, 2H), 1.28-2.45 (m, 3H). 19 F NMR (376 MHz, DMSO-7 6 ) δ ppm -96.02 (br s, 2F).

Example 056: Trans-N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2 -oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-di fluoro- methylIcyclohexanecarboxamide

Step 1: Trans-4-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]cyclohexan ecarboxylic acid To a solution of compound Int-5e (2.0 g, 5.91 mmol) and lithium bromide (5.1 g, 60 mmol) in MeCN (10 mL) and water (3 mL) was added TEA (3.0 mL, 21.52 mmol). The mixture was stirred at 50°C for 12 hours. After completion, the mixture was adjusted to pH = 5 with 1 M HC1 solution, extracted with EtOAc (300 mL x 2) and washed with brine (300 mL). The combined organic layer was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give compound 056a (1.8 g, 5.55 mmol, 93.9% yield) as a white solid. MS obsd. (ESI+) [(M+H) + ]: 323.8.

Step 2: Trans-N-butyl-4-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]cy clohexanecarboxamide

To a solution of compound 056a (150 mg, 463 pmol), HATU (264 mg, 694 pmol,) and DIPEA (242 pL, 1.39 mmol) in DMF (5 mL) was added tert-butyl N-(3-aminopropyl)carbamate (96.8 mg, 555 pmol). The mixture was stirred at room temperature for 2 hours. After completion, the reaction mixture was diluted with EtOAc (10 mL) and washed with brine (50 mL). The organic layer was dried over anhydrous sodium sulfate and concentrated in vacuo to give crude product, which was purified by silica gel chromatography (eluent with EtOAc : PE = 0 to 40%) to give compound 056b (180 mg, 375 pmol, 81 % yield) as yellow oil. MS obsd. (ESI+) [(M+H) + ]: 480.6.

Step 3: Trans-tert-butyl N-[3-[[4-[[2-chloro-6-[4-[4-[(4R)-4-(tert-butoxycarbonylamin o)-2- oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl ]-difluoro- methyl]cyclohexanecarbonyl]amino]propyl]carbamate

A solution of compound Int-5 (100 mg, 236 pmol), compound 056b (147 mg, 306 pmol) and DIPEA (152 mg, 1.18 mmol) in DMSO (2 mL) was stirred at 100°C overnight. After completion, the mixture was diluted with EtOAc (10 mL) and washed with brine (10 mL). The organic layer was dried over anhydrous sodium sulfate and concentrated in vacuo to give the desired product compound 056c (205 mg, 236 pmol, 100 % yield) as yellow oil, which was used in the next step directly. MS obsd. (ESI+) [(M+H) + ]: 868.8.

Step 4: Trans-N-(3-aminopropyl)-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2 -oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]cyclohexanecarboxamide

A solution of compound 056c (205 mg, 236 pmol) and TFA (364 pL, 4.72 mmol) was stirred at room temperature for 3 hours. The mixture was then concentrated in vacuo to give the crude product, which was purified by prep-HPLC to give the desired product Example 056 (89 mg, 124 pmol, 52.5 % yield) as a yellow solid. MS obsd. (ESI + ) [(M+H)] + : 668.2.

Example 056: 1 H NMR (400 MHz, METHANOL-d 4 ) ppm 8 7.84 (br d, J= 8.8Hz, 2H), 7.72 (d, J= 8.7Hz, 2H), 6.58 (s, 1H), 6.55 (s, 1H), 4.32 (br dd, J= 11.2, 6.9Hz, 1H), 4.12 (br s, 1H), 3.98-3.84 (m, 1H), 3.59 (br s, 4H), 3.19-2.98 (m, 4H), 2.96 (br s, 4H), 2.91-2.78 (m, 2H), 2.64 (dd, J= 18.1, 2.8Hz, 1H), 2.10-1.89 (m, 3H), 1.82-1.65 (m, 7H), 1.41-1.26 (m, 2H), 1.24-1.04 (m, 3H). 19 F NMR (376 MHz, METHANOL-d 4 ) 6 ppm -106.88 (s, IF), -106.92 (s, IF).

The following Examples 057 to 062 were prepared in analogy to the procedure described for the preparation of Example 056, replacing compound Int-5 with “LINKER” in Step 3, and replacing tert-butyl N-(3-aminopropyl)carbamate with “TAIL” in Step 2 by the reagents indicated in Table 2.

Table 2: Compound synthesis and characterization

Example 071: Trans- (4R)-4-amino- 1- [4- [4- [4- [ [4- [4-(2-aminoethyl)-4-methyl-piperazin-4- ium-l-carbonyl]cyclohexyl]-difluoro-methyl]-6-chloro-2-pyrid yl]piperazin-l- yl]sulfonylphenyl]pyrrolidin-2-one

Step 1: Trans-tert-butyl N-| (3R)~ 1 - [4- [ 4-[ 4-[ [ 4- [4-[ 2-(tert- butoxycarbonylamino)ethyl]piperazine-l-carbonyl]cyclohexyl]- difluoro-methyl]-6-chloro-

2-pyridyl]piperazin-l-yl]sulfonylphenyl]-5-oxo-pyrrolidin -3-yl]carbamate A mixture of N-(2-piperazinoethyl)carbamic acid tert-butyl ester (72.45 mg, 0.316 mmol), compound Int-5 (150 mg, 0.211 mmol), HATU (160.07 mg, 0.421 mmol) and DIPEA (110.23 pL, 0.632 mmol) in DCM (15 mL) was stirred at room temperature for 2 hours. After completion, the mixture was diluted with EtOAc (50 mL), and washed with brine (50 mL). Then, the combined organic layer was dried over anhydrous sodium sulfate, filtered, concentrated in vacuo to give crude compound 071a (190 mg, 97.68%) as light yellow viscous oil. MS obsd. (ESI+) [(M+H) + ]: 923.6.

Step 2: Trans-tert-butyl N-[2-[4-[4-[[2-chloro-6-[4-[4-[(4R)-4-(tert-butoxycarbonylam ino)-2- oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl ]-difluoro- methyl]cyclohexanecarbonyl]-l-methyl-piperazin-l-ium-l-yl]et hyl]carbamate

Compound 071a (100 mg, 0.108 mmol) and DIPEA (28.37 pL, 0.162 mmol) were dissolved in the DCM (5.15 mL). Into the stirring solution was added iodomethane (6.77 pL, 0.108 mmol). The reaction was stirred at room temperature overnight. Then, the reaction was concentrated in vacuo to give the desired product compound 071b (90 mg, 96 pmol, 88.56% yield) as light yellow oil, which was used directly in the next step. MS obsd. (ESI+) [(M+H) + ]: 939.7.

Step 3: Trans- (4R)-4-amino-l-[4-[4-[4-[[4-[4-(2-aminoethyl)-4-methyl-piper azin-4-ium-l- carbonyl] cyclohexyl] -difluoro-methyl] -6-chloro-2-pyridyl] piperazin- 1- yl]sulfonylphenyl]pyrrolidin-2-one

Compound 071b (90 mg, 0.096 mmol) and TFA (0.5 mL, 6.5 mmol) were mixed in the DCM (5 mL). The reaction mixture was stirred at room temperature for 1 hour and went completed. After completion, the residue was concentrated in vacuo to give crude product, which was purified by prep-HPLC to give the desired product Example 071 (35 mg, 46.96%) as a white solid. MS obsd. (ESI+) [(M+H) + ]: 737.4.

Example 071: 1 H NMR (400 MHz, METHANOL-^) δ ppm 8.43 (br s, 1H), 7.92 (m, 7=9.05Hz, 2H), 7.82 (m, 7=9.05Hz, 2H), 6.67 (d, 7=7.34Hz, 2H), 4.91-4.95 (m, 1H), 4.34 (dd, 7=11.13, 6.85Hz, 1H), 3.94-4.17 (m, 3H), 3.89 (br dd, 7=11.07, 2.63Hz, 2H), 3.79 (br d, 7=15.16Hz, 1H), 3.65-3.73 (m, 4H), 3.41-3.62 (m, 6H), 3.22-3.29 (m, 5H), 3.01-3.16 (m, 5H), 2.57-2.69 (m, 2H), 1.99-2.15 (m, 1H), 1.81 (br t, 7=11.62Hz, 4H), 1.45 (dt, 7=6.69, 1.60Hz, 2H), 1.25-1.39 (m, 2H). 19 F NMR (376 MHz, METHANOL-d 4 ) δ ppm -106.86 (s, IF), -106.89 (s, IF).

Example 072 : Trans-N- [2-(2-aminoethylamino)ethyl] -4- [ [2-chloro-6- [4- [4- [(4R)-4-amino-2- oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl ]-difluoro- methylIcyclohexanecarboxamide

Step 1: Trans-tert-butyl N-[(3R)-l-[4-[4-[6-chloro-4-[difluoro-[4-(2- hydroxyethylcarbamoyl)cyclohexyl]methyl]-2-pyridyl]piperazin -l-yl]sulfonylphenyl]-5- oxo-pyrrolidin-3-yl] carbamate

To a solution of compound Int-5 (300.0 mg, 0.420 mmol) in DMF (3 mL) were added HATU (208.21 mg, 0.550 mmol), DIPEA (0.22 mL, 1.26 mmol), 2-hydroxyethylamine (0.03 mL, 0.460 mmol) in one portion under nitrogen. The reaction was stirred at 25°C for 16 hours. After completion, the reaction was diluted with water (20 mL), extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine (50 mL), dried over anhydrous sodium sulfate and then filtered. The filtrate was concentrated in vacuo to give a crude residue, which was purified by prep-HPLC to afford compound 072a (220 mg, 0.290 mmol, 69.15% yield) as a white solid. MS obsd. (ESI + ) [(M+H)] + : 755.6. Step 2: Trans-tert-butyl N-[(5R)-l-[4-[4-[6-chloro-4-[difluoro-[4-(2- oxoethylcarbamoyl)cyclohexyl]methyl]-2-pyridyl]piperazin-l-y l]sulfonylphenyl]-5-oxo- pyrrolidin-3-yl] carbamate

To a solution of compound 072a (180.0 mg, 0.240 mmol) in DCM (3 mL) was added DMP (111.19 mg, 0.260 mmol) in one portion under nitrogen. The reaction was stirred at 25°C for 1 hour. After completion, the reaction was quenched with sat. aq. ISfeCCL solution (100 mL) and extracted with EtOAc (200 mL). The organic layer was separated and dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo to afford compound 072b (150 mg, 0.200 mmol, 83.56% yield) as a yellow solid, which was used directly for the next step. MS obsd. (ESI + ) [(M+H)] + : 755.6.

Step 3: Trans-tert-butyl N-[2-[2-[[4-[[2-chloro-6-[4-[4-[(4R)-4-(tert-butoxycarbonyla mino)- 2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyrid yl]-difluoro- m ethyl] cyclohexanecarbonyl] amino] ethylamino] ethyl] carbamate

To a solution of compound 072b (130.0 mg, 0.170 mmol), N-Boc-ethylenediamine (55.3 mg, 0.350 mmol) in methanol (1 mL) was stirred at 25°C for 1 hour. Then sodium cynaoborohydride (54.23 mg, 0.860 mmol) was added and the reaction was stirred at 25°C for additional 1 hour. After completion, the reaction was diluted with water (10 mL) and extracted with EtOAc (20 mL x 2). The combined organic layer was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo to give a residue, which was purified by prep-HPLC to afford compound 072c (85 mg, 0.090 mmol, 54.88% yield) as colorless oil. MS obsd. (ESI + ) [(M+H)] + : 897.0. Step 4 : Trans-N- [2-(2-aminoethylamino)ethyl] -4- [[2-chloro-6- [4- [4- [(4R)-4-amino-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-di fluoro- methyljcyclohexanecarboxamide

To a solution of compound 072c (35.0 mg, 0.040 mmol) in DCM (1 mL) was added TFA (0.5 mL, 6.5 mmol) in one portion. The reaction was stirred at 25°C for 1 hour. After completion, the reaction was concentrated in vacuo to give a residue, which was purified by prep-HPLC to afford Example 072 (20 mg, 0.029 mmol, 71% yield) as a white solid. MS obsd. (ESI + ) [(M+H)] + : 697.0.

Example 072: 1 H NMR (400 MHz, DMSO-d 6 ) δ ppm 8.34-8.26 (m, 3H), 8.21-8.12 (m, 3H), 8.10-8.03 (m, 1H), 7.94-7.90 (m, 2H), 7.82-7.78 (m, 2H), 6.81-6.77 (m, 1H), 6.73-6.69 (m, 1H),

4.28-4.22 (m, 1H), 4.10-4.04 (m, 1H), 3.87-3.82 (m, 1H), 3.68-3.63 (m, 4H), 3.21-3.14 (m, 4H),

3.14-3.05 (m, 2H), 3.05-3.00 (m, 2H), 2.97 (br d, J= 2.8 Hz, 4H), 2.61 (br s, 1H), 2.21-1.98 (m,

2H), 1.87-1.78 (m, 2H), 1.70-1.63 (m, 2H), 1.39-1.25 (m, 2H), 1.19-1.08 (m, 2H). 19 F NMR (376

MHz, DMSO-d 6 ) δ ppm -104.086 (br s, 2F).

Example 073 : Trans-2-aminoethyl- [2- [ [4- [ [2-chloro-6- [4- [4- [(4R)-4-amino-2-oxo-pyrrolidin- l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro- methyl]cyclohexanecarbonyl]amino]ethyl]-dimethyl-ammonium

Step 1 : Trans-2-(tert-butoxycarbonylamino)ethyl- [2- [ [4- [ [2-chloro-6- [4- [4- {(4R)-A-(tert- butoxycarbonylamino)-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpi perazin-l-yl]-4-pyridyl]- difluoro-methyl] cyclohexanecarbonyl] amino] ethyl] -dimethyl-ammonium

To a solution of compound 072c (35.0 mg, 0.040 mmol) in DCM (1 mL) were added sodium hydrogen carbonate (32.76 mg, 0.390 mmol) and methyl trifluoromethane-sulfonate (19.2 mg, 0.120 mmol) in one portion. The reaction was stirred at 25°C for 1 hour. After completion, the reaction was filtered and the filtrate was added with NEh’FbO. The reaction was concentrated in vacuo to give compound 073a (28 mg, 0.030 mmol, 77.49% yield) as a white solid. MS obsd. (ESI + ) [(M+H)] + : 925.4.

Step 2 : Trans-2-aminoethyl- [2- [ [4- [ [2-chloro-6- [4- [4- [(4R)-4-amino-2-oxo-pyrrolidin- 1- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro- methyl]cyclohexanecarbonyl]amino]ethyl]-dimethyl-ammonium

To a solution of compound 073a (41.0 mg, 0.040 mmol) in DCM (1 mL) was added TFA (0.17 mL, 2.21 mmol) in one portion. The reaction was stirred at 25°C for 1 hour. After completion, the reaction was concentrated in vacuo to give a residue, which was purified by prep-HPLC to give Example 073 (20 mg, 0.030 mmol, 58.33% yield) as a white solid. MS obsd. (ESI + ) [(M+H)] + : 725.1.

Example 073: 1 H NMR (400 MHz, DMSO-d 6 ) δ ppm 8.52-8.41 (m, 5H), 8.33-8.24 (m, 1H), 7.96-7.88 (m, 2H), 7.79 (d, J = 9.0 Hz, 2H), 6.78 (s, 1H), 6.71 (s, 1H), 4.31-4.22 (m, 1H), 4.11- 4.02 (m, 1H), 3.90-3.85 (m, 1H), 3.69-3.57 (m, 6H), 3.48-3.41 (m, 2H), 3.38 (br s, 2H), 3.15 (s, 6H), 3.10-3.04 (m, 1H), 2.97 (br s, 4H), 2.66-2.59 (m, 1H), 2.24-2.05 (m, 2H), 1.82-1.73 (m, 2H), 1.67 (br d, J= 10.1 Hz, 2H), 1.39-1.26 (m, 2H), 1.14 (br s, 2H). 19 F NMR (376 MHz, DMSO-tL) 6 ppm -104.004 (br s, 2F).

Example 074: Trans-N-[2-[l-(2-aminoethyl)pyrrolidin-l-ium-l-yl]ethyl]-4-[ [2-[4-[4-[(4R)-4- amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-6 -chloro-4-pyridyl]-difluoro- methyljcyclohexanecarboxamide

Step 1: Trans-tert-butyl N-[(51?)-l-[4-[4-[6-chloro-4-[difluoro-[4-(2-pyrrolidin-l- ylethylcarbamoyl)cyclohexyl]methyl]-2-pyridyl]piperazin-l-yl ]sulfonylphenyl]-5-oxo- pyrrolidin-3-yl] carbamate

A mixture of 2-pyrrolidinoethylamine (24.05 mg, 0.211 mmol), compound Int-5 (100 mg, 0.140 mmol), HATU (106.71 mg, 0.281 mmol) and DIPEA (73.49 pL, 0.421 mmol) in DCM (10 mL) was stirred at room temperature for 2 hours. After completion, the mixture was diluted with EtOAc (10 mL), washed with water (10 mL) and brine (10 mL). Then, the combined organic layer was dried over anhydrous sodium sulfate, filtered, concentrated in vacuo to give crude 074a (100 mg, 88.1% yield) as yellow viscous oil. MS obsd. (ESI+) [(M+H) + ]: 808.7.

Step 2: Trans-tert-butyl N-[2-[l-[2-[[4-[[2-chloro-6-[4-[4-[(4R)-4-(tert- butoxycarbonylamino)-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpi perazin-l-yl]-4-pyridyl]- difluoro-methyljcyclohexanecarbonyl] amino] ethyl] pyrrolidin-l-ium-l-yl] ethyl] carbamate

Compound 074a (100 mg, 0.124 mmol) and N-(2-bromoethyl)carbamic acid tert-butyl ester (138.61 mg, 0.619 mmol) were dissolved in the anhydrous MeCN (8 mL). Into the stirring solution was added DIPEA (64.81 μL, 0.371 mmol) and 0.1 mg DMAP. The reaction mixture was stirred at 90°C overnight. LCMS showed half of the staring material was converted. Then, the reaction was cooled to room temperature and concentrated in vacuo to give crude compound 074b (57 mg, 49%) as yellow oil, which was used directly in the next step. MS obsd. (ESI + ) [(M+H) + ]: 951.5.

Step 3: Trans-N-[2-[l-(2-aminoethyl)pyrrolidin-l-ium-l-yl]ethyl]-4-[ [2-[4-[4-[(4R)-4-amino- 2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-6-chlor o-4-pyridyl]-difluoro- methylIcyclohexanecarboxamide

Compound 074b (100 mg, 0.105 mmol) and TFA (1 mL, 13 mmol) were dissolved in the DCM (10 mL). The reaction was stirred at room temperature for 2 hours. After completion, the reaction was concentrated in vacuo and purified by prep-HPLC to give the desired product Example 074 (18.1 mg, 20.63%) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 751.8.

Example 074: 1 H NMR (400 MHz, METHANOL-d4) δ ppm 7.83 (m, 7=8.93Hz, 2H), 7.73 (m, 7=8.93Hz, 2H), 6.57 (d, 7=7.70Hz, 2H), 4.30 (dd, 7=11.43, 7.03Hz, 1H), 4.06-4.13 (m, 1H), 3.86 (dd, 7=11.43, 2.51Hz, 1H), 3.37-3.64 (m, 14H), 3.29-3.35 (m, 2H), 3.05-3.17 (m, 1H), 2.98 (br t, 7=4.89Hz, 4H), 2.60 (dd, 7=18.10, 3.06Hz, 1H), 1.89-2.17 (m, 7H), 1.78 (br d, 7=11.13Hz, 2H), 1.70 (br d, 7=11.00Hz, 2H), 1.09-1.37 (m, 8H). 19 F NMR (376 MHz, METHANOL-d 4 ) δ ppm - 106.97 (s, IF), -107.01 (s, IF).

Example 075 : Trans-N- [2- [(2-aminoacetyl)amino] ethyl] -4- [ [2-chloro-6- [4- [4- [(4R)-4-amino- 2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyrid yl]-difluoro- methylIcyclohexanecarboxamide

Step 1: Trans-tert-butyl N-[(31?)-l-[4-[4-[4-[[4-(2-aminoethylcarbamoyl)cyclohexyl]- difluoro-methyl]-6-chloro-2-pyridyl]piperazin-l-yl]sulfonylp henyl]-5-oxo-pyrrolidin-3- yl] carbamate

To a solution of compound Int-5 (100.0 mg, 0.140 mmol) in DMF was added DIPEA (52.23 mg, 0.420 mmol) and HATU (80.09 mg, 0.210 mmol). The mixture was stirred at 25°C for 1 hour. Then the mixture was added ethylenediamine (337 mg, 5.6 mmol) in DMF (3 mL). After stirring at 25°C for 1 hour, the mixture was diluted with EtOAc (50 mL) and water (30 mL). The organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give crude compound 075a (100 mg, 0.130 mmol, 94.42% yield) as a white solid. The residue was used directly in the next step. MS obsd. (ESI + ) [(M+H) + ]: 754.3.

Step 2: Trans-tert-butyl N-[2-[2-[[4-[[2-chloro-6-[4-[4-[(4R)-4-(tert-butoxycarbonyla mino)-

2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-py ridyl]-difluoro- methyl] cyclohexanecarbonyl] amino] ethylamino] -2-oxo-ethyl] carbamate

To a solution of compound 075a (100.0 mg, 0.130 mmol) and BOC-glycine (23.22 mg, 0.130 mmol) in DMF (1 mL) was added DIPEA (49.32 mg, 0.400 mmol,) and HATU (46.79 mg, 0.200 mmol). After stirring at 25°C for 1 hour, the mixture was diluted with EtOAc (50 mL) and water (30 mL). The organic phase was washed with brine (50 mL), dried over sodium sulfate, filtered and concentrated in vacuo to give a crude residue, which was purified by prep-TLC (PE:EA=L 1) to give compound 075b (95 mg, 0.100 mmol, 78.62% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 911.5.

Step 3 : Trans-N- [2- [(2-aminoacetyl)amino] ethyl] -4- [ [2-chloro-6- [4- [4- [(4R)-4-amino-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-di fluoro- methyljcyclohexanecarboxamide

To a solution of compound 075b (90.0 mg, 0.100 mmol) in DCM (5 mL) was added TFA (56.28 mg, 0.490 mmol). The mixture was stirred at 25°C for 2 hours. After completion, the mixture was concentrated in vacuo to give a crude residue, which was purified by prep-HPLC to afford Example 075 (22.7 mg, 0.030 mmol, 30.8% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 711.2.

Example 075: 1 H NMR (400 MHz, DMSO-d 6 ) δ ppm 8.50 (d, 7=12.47Hz, 4H), 8.11 (s, 3H), 7.91 (d, 7=8.93Hz, 3H), 7.80 (d, 7=8.93Hz, 2H), 6.78 (s, 1H), 6.71 (s, 1H), 4.21-4.32 (m, 1H), 4.07 (s, 1H), 3.89 ( d, 7=9.29Hz, 1H), 3.66 (s, 5H), 2.91-3.31 (m, 8H), 2.64 (dd, 7=17.91, 2.75Hz, 1H), 2.17 (t, 7=11.98Hz, 1H), 1.97-2.08 (m, 1H), 1.59-1.83 (m, 4H), 1.22-1.39 (m, 2H), 1.02- 1.19 (m, 2H). 19 F NMR (376 MHz, DMSO-d 6 ) δ ppm -105.48-102.95 (m, 2F).

Example 076: 7rans-N-(3-amino-2-methoxy-propyl)-4-[[2-chloro-6-[4-[4-[(4R )-4-amino-2- oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl ]-difluoro- methyljcyclohexanecarboxamide

Step 1: Trans-tert-butyl N-[3-[[4-[[2-chloro-6-[4-[4-[(4R)-4-(tert-butoxycarbonylamin o)-2- oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl ]-difluoro- m ethyl] cyclohexanecarbonyl] amino] -2-hydroxy-propyl] carbamate To a solution of compound Int-5 (250.0 mg, 0.350 mmol) in DMF (4 mL) was added tertbutyl N-(3-amino-2-hydroxy-propyl)carbamate (133.56 mg, 0.700 mmol), HATU (200.21 mg, 0.530 mmol) and DIPEA (136.1 mg, 1.05 mmol). The reaction mixture was stirred at 20°C for 1 hour. After completion, the reaction mixture was diluted with EtOAc (200 mL) and the organic layer was washed with brine (30 mL x 3), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give a crude residue, which was purified by reverse-phase column to give compound 076a (260 mg, 0.290 mmol, 83.75% yield) as a light yellow solid. MS obsd. (ESI + ) [(M+H) + ]: 886.5.

Step 2: Trans-tert-butyl N-|3-||4-||2-chloro-6-|4-|4-|(4R)-4-(tert-biitoxycarbonylain ino)-2- oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl ]-difluoro- methyl]cyclohexanecarbonyl]amino]-2-methoxy-propyl]carbamate

To a solution of compound 076a (150.0 mg, 0.170 mmol) in MeCN (2 mL) was added Ag2O (589.52 mg, 2.54 mmol) and Mel (240.73 mg, 1.7 mmol) at 20°C. The reaction was stirred at 20°C for 16 hours. Then, the reaction mixture was concentrated in vacuo and purified by prep- TLC (PE: EA=1 :3) to give compound 076b (40 mg, 0.040 mmol, 26.25% yield) as a light yellow solid. MS obsd. (ESI + ) [(M+H) + ]: 900.2.

Step 3: Trans-N-(3-amino-2-methoxy-propyl)-4-[[2-chloro-6-[4-[4-[(4R )-4-amino-2-oxo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-di fluoro- methyljcyclohexanecarboxamide

To a solution of compound 076 (60.0 mg, 0.070 mmol) in DCM (2 mL) was added TFA (761.44 mg, 6.68 mmol) at 30°C. The reaction mixture was stirred at 30°C for 1 hour. After completion, the reaction mixture was concentrated in vacuo to give a crude residue, which was purified by prep-HPLC to give Example 076 (23.9 mg, 0.030 mmol, 49.77% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 700.2.

Example 076: 1 H NMR (400 MHz, DEUTERIUM OXIDE) δ ppm 7.69 (br d, I = 8.6 Hz, 2H), 7.56 (br d, I = 8.6 Hz, 2H), 6.56 (s, 2H), 4.39-4.11 (m, 2H), 4.10-3.95 (m, 1H), 3.57 (br s, 1H), 3.47 (br s, 4H), 3.35 (s, 3H), 3.29 (br d, I = 4.2 Hz, 2H), 3.22-3.01 (m, 2H), 2.88-2.68 (m, 6H), 2.07-1.74 (m, 2H), 1.69-1.41 (m, 4H), 1.19-0.86 (m, 4H). 19 F NMR (400 MHz, DEUTERIUM OXIDE) δ ppm: -105.02 (br s, 2F).

Example 077: 7rans-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]-N-(2,3- diaminopropyl)cyclohexanecarboxamide

Step 1: Trans-tert-butyl N-[3-[[4-[[2-chloro-6-[4-[4-[(4R)-4-(tert-butoxycarbonylamin o)-2- oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl ]-difluoro- methyl]cyclohexanecarbonyl]amino]-2-oxo-propyl]carbamate

To a solution of compound 076a (100.0 mg, 0.110 mmol) in THF (2 mL) was added DMP (143.87 mg, 0.340 mmol) at 20°C. The reaction was stirred at 20°C for 2 hours. After completion, the reaction mixture was purified by prep-TLC (PE : EtOAc = 1 :3) to give 077a (50 mg, 0.060 mmol, 50.11% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 882.4.

Step 2: Trans-tert-butyl N-[(3R)-l-[4-[4-[6-chloro-4-[difluoro-[4-[[(2E)-3-(tert- butoxycarbonylamino)-2-hydroxyimino-propyl]carbamoyl]cyclohe xyl]methyl]-2- pyridyl]piperazin-l-yl]sulfonylphenyl]-5-oxo-pyrrolidin-3-yl ]carbamate

To a solution of compound 077a (120.0 mg, 0.140 mmol) in ethanol (2 mL) was added hydroxylamine hydrochloride (18.9 mg, 0.270 mmol) at 30°C. The reaction was stirred at 30°C for 2 hours. After completion, the mixture was diluted with EtOAc (200 mL) and washed with brine (50 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give compound 077b (122 mg, 0.140 mmol, 99.97% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 897.0.

Step 3: Trans-tert-butyl N-[2-amino-3-[[4-[[2-chloro-6-[4-[4-[(4R)-4-(tert- butoxycarbonylamino)-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpi perazin-l-yl]-4-pyridyl]- difluoro-methyl]cyclohexanecarbonyl]amino]propyl]carbamate

To a solution of compound 077b (120.0 mg, 0.130 mmol) in ethanol (10 mL) was added Pt/C (500.0 mg, 7.69 mmol) at 30°C. The reaction was stirred at 30°C for 24 hours under H 2 . After completion, the reaction mixture was filtered and the filtrate was concentrated in vacuo to give a crude residue, which was purified by prep-TLC to afford compound 077c (30 mg, 0.030 mmol, 25.4% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 883.0.

Step 4 : Trans-4- [ [2-chloro-6- [4- [4- [(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]-N-(2,3- diaminopropyl)cyclohexanecarboxamide To a solution of compound 077c (30.0 mg, 0.030 mmol) in DCM (1 mL) was added TFA (1.0 mL, 12.98 mmol) in one portion. After addition, the mixture was stirred at 25°C for 30 minutes. After completion, the reaction was concentrated in vacuo and then purified by prep- HPLC to afford Example 077 (10.0 mg, 0.010 mmol, 37.67% yield) as a white powder. MS obsd. (ESI + ) [(M+H) + ]: 683.2.

Example 077: 1 H NMR (400 MHz, DMSO-d 6 ) δ ppm 8.45-8.03(m, 7H), 7.95-7.85 (d, 2H), 7.80-7.60 (d, 2H), 6.78(s, 1H), 6.71 (s, 1H), 4.30-4.20 (m, 2H), 3.80-3.73 (m, 1H), 3.70-3.62 (m, 4H), 3.20-2.95 (m, 9H), 2.65-2.52 (m, 2H), 2.25-2.05 (m, 2H), 1.90-1.75 (m, 2H), 1.74-1.60 (m, 2H), 1.40-1.26 (m, 2H), 1.35-1.08 (m, 2H). 19 F NMR (376 MHz, DMSO-d 6 ) δ ppm -104.025 (br s, 2F).

Example 078: 4-[[2-[4-[4-[4-(3-Aminopropylamino)-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluo ro-methyl]-N-methyl- benzamide

Step 1: 4-[[2-[4-[4-[4-[Tert-butoxycarbonyl-[3-(tert-butoxycarbonyla mino)propyl] -amino]-

2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-6-ch loro-4-pyridyl]-difluoro- m ethyl] benzoic acid

Compound 078a was prepared in analogy to the preparation of compound Int-12 by using compound Int-22 instead of compound Int-2 in Step 4. Compound Int-078a (235.3 mg) was obtained as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 863.3. Step 2: Tert-butyl N-[3-(tert-butoxycarbonylamino)propyl]-N-[l-[4-[4-[6-chloro- 4-[difluoro- [4-(methylcarbamoyl)phenyl]methyl]-2-pyridyl]piperazin-l-yl] sulfonylphenyl]-5-oxo- pyrrolidin-3-yl] carbamate

A mixture of compound 078a (61 mg, 70.8 pmol), methanamine hydrochloride (14.2 mg, 212 pmol), HATU (40.4 mg, 106 pmol), and TEA (21.5 mg, 212 pmol) in DCM (3 mL) was stirred at room temperature for 12 hours. After completion, the residue was washed with water (50 mL). The organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo to give a crude product compound 078b (50 mg, 0.571 mmol, 80% yield) as yellow oil. MS obsd. (ESI + ) [(M+H) + ]: 876.3.

Step 4: 4-[[2-[4-[4-[4-(3-aminopropylamino)-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluo ro-methyl]-N-methyl- benzamide

A mixture of compound 078b (34.5 mg, 39.4 pmol) and TFA (89.8 mg, 787 pmol) in DCM (3 mL) was stirred at room temperature for 1 hour. After completion, the reaction mixture was concentrated in vacuo to give a crude residue, which was purified by prep-HPLC to give Example 078 (6.8 mg, 0.01 mmol, 11% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 676.1. Example 078: 1 H NMR (400 MHz, METHANOL-d 4 ) δ ppm 1.64-1.85 (m, 2H), 2.54-2.60 (m, 1H), 2.67 (br d, J=1.83 Hz, 1H), 2.72-2.89 (m, 7H), 2.96 (m, 4H), 3.67 (m, 4H), 4.09 (m, 1H), 6.79 (s, 1H), 6.96 (s, 1H), 7.68 (d, J=8.44 Hz, 2H), 7.77 (d, J=8.93 Hz, 2H), 7.91 (t, J=8.50 Hz, 4H), 8.53-8.58 (m, 1H). 19 F NMR (376 MHz, DMSO-d 6 ) δ ppm -92.70 (s, 2F). Example 079 : Trans-N- | ( 1 R.3.S)-3- [(2-aminoacetyl)amino] cyclopentyl] -4- [ [2- [4- [4- [(4R)- 4- amino-2-oxo-pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-6 -chloro-4-pyridyl]-difluoro- methylIcyclohexanecarboxamide

The title compound was prepared in analogy to the preparation of Example 075 by using (15, 3R)-cyclopentane- 1,3 -diamine instead of ethylenediamine in Step 1. Example 079 (24 mg) was obtained as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 751.4. 1 H NMR (500 MHz, DMSO-d 6 ) δ ppm 8.35-8.45 (m, 1H), 8.16-8.28 (m, 2H), 7.87-8.00 (m, 2H), 7.73-7.85 (m, 2H), 6.77-6.86 (m, 1H), 6.61-6.76 (m, 1H), 4.20-4.33 (m, 1H), 4.05-4.15 (m, 1H), 3.87-4.05 (m, 3H), 3.74-3.87 (m,

2H), 3.60-3.74 (m, 5H), 3.02-3.16 (m, 2H), 2.92-3.02 (m, 4H), 2.13-2.27 (m, 2H), 1.93-2.03 (m,

1H), 1.78-1.93 (m, 3H), 1.59-1.78 (m, 5H), 1.40-1.59 (m, 3H), 1.17-1.40 (m, 4H), 0.94-1.17 (m,

3H). 19 F NMR (471 MHz, DMSO-d 6 ) δ ppm -104.30 (br s, IF).

Example 080 : Trans-4- [ [2- [4- [4- [(4R)-4-amino-2-oxo-pyrrolidin- 1- yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluo ro-methyl]-N-[(l»S',3R)-3-[cis- 3,4-bis(aminomethyl)pyrrolidin-l-yl]cyclopentyl]cyclohexanec arboxamide

Step 1: Benzyl N-[(15)-3-oxocyclopentyl] carbamate To a solution of oxalyl chloride (0.81 g, 6.38 mmol) in DCM (10 mL) was added a solution of DMSO (0.73 g, 9.35 mmol) in DCM (10 mL) at -78° C under N2. The reaction was stirred at -78° C° C for 1 hour, and then the solution of benzyl N-[( LS')-3-hydroxycyclopentyl]- carbamate (1.0 g, 4.25 mmol) in DCM (10 mL) was added. After stirring at -78 ° C for 1 hour, DIEA (1.72 g, 17 mmol) was added at -78 ° C and then the reaction was allowed to warm to 20 °C. After the starting material was consumed, the reaction mixture was quenched with water (100 mL) and extracted with DCM (100 mL x 3). The combined organic layer was washed with brine (200 mL), dried over anhydrous sodium sulfate, and concentrated in vacuo to give the crude product, which was purified by silica gel column (elute with EtOAc: PE = 30%) to afford 080a (820 mg, 3.52 mmol, 82.71% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 234.0. Step 2: Tert-butyl N-[[cis-1-[(1R,3S)-3-(benzyloxycarbonylamino)cyclopentyl]-4- [(tert- butoxycarbonylamino)methyl]pyrrolidin-3-yl]methyl]carbamate

To a solution of 080a (30.0 mg, 0.130 mmol) and tert-butyl N-[[cA-4-[(tert- butoxycarbonylamino)methyl]pyrrolidin-3-yl]methyl]carbamate (42.37 mg, 0.130 mmol) in 1,2- di chloroethane (0.5 mL) was added AcOH (46.3 mg, 0.770 mmol) and sodium tri acetoxyb or ohydri de (41 mg, 0.2 mmol). The mixture was stirred at 25 °C for 4 hours. After completion, the reaction mixture was diluted with water (150 mL) and extracted with EtOAc (200mL x 3). The combined organic layer was washed with brine (100 mL x 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give crude residue, which was purified by prep-HPLC and then prep-TLC to give 080ba (80 mg, 0.150 mmol, 7.11% yield) as a white solid and 080bb (100 mg, 0.180 mmol, 9% yield). 080ba: MS obsd. (ESI + ) [(M+H) + ]: 547.4. The cis and trans conformations were determined by 2D NMR.

Step 3: Tert-butyl N-[[cis-l-[(1R,3S)-3-aminocyclopentyl]-4-[(tert- butoxycarbonylamino)methyl]pyrrolidin-3-yl]methyl]carbamate

To a solution of Pd/C (110.0 mg) powder in the mixture of THF (1 mL) and IPA (1 mL) was added 080ba (100.0 mg, 0.180 mmol). The mixture was stirred at 45 °C for 3 hours under H 2 atmosphere. After the starting material was consumed, the mixture was filtrated through celite and concentrated in vacuo to give 080c (100.0 mg, 0.180 m mol) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 412.4.

Step 4: Tert-butyl N-[(3R)-l-[4-[4-[4-[[4-[[(1S,3R)-3-[cis-3,4-bis[(tert- butoxycarbonylamino)methyl]pyrrolidin-l-yl]cyclopentyl]carba moyl]cyclohexyl]-difluoro- methyl]-6-chloro-2-pyridyl]piperazin-l-yl]sulfonylphenyl]-5- oxo-pyrrolidin-3-yl]carbamate

To the solution of Int-5 (120 mg, 0.170 mmol) in DMF (1 mL) was added HATU (96.04 mg, 0.250 mmol) and DIEA (108.68 mg, 0.840 mmol). The reaction mixture was stirred at 25 °C for 15 minutes. Then 080c (70 mg, 0.170 mmol) was added and the mixture was stirred at 25°C for 2 hours. After the starting material was consumed, the reaction mixture was quenched by adding water (100 mL) and diluted with EtOAc (100 mL). The aqueous layer was extracted with EtOAc (100 mL x 2). The combined organic layer was washed with brine (100 mL), dried over drying anhydrous sodium sulfate, filtered and concentrated in vacuo to give the crude product, which was purified by prep-HPLC and prep-TLC to give 080d (40 mg, 0.040 mmol, 21.5% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 1106.6. Step 5 : Trans-4- [ [2- [4- [4- [(4R)-4-amino-2-oxo-pyrrolidin-l-yl] phenyl] sulf onylpiperazin- 1- yl]-6-chloro-4-pyridyl]-difluoro-methyl]-N-[(1S,3R)-3-[cis-3 ,4-bis(aminomethyl)pyrrolidin- l-ylIcyclopentylIcyclohexanecarboxamide

To a solution of 080d (40.0 mg, 0.040 mmol) in DCM (1 mL) was added TFA (1 mL) in one portion. The mixture was stirred at 25°C for 1 hour. After completion, the reaction mixture was concentrated in vacuo. The crude product was purified by prep-HPLC to give Example 080 (25.5 mg, 0.030 mmol, 83.37% yield) as a yellow solid. MS obsd. (ESI + ) [(M+H) + ]: 806.3. Example 080: 1 H NMR (400 MHz, DMSO-d 6 ) δ ppm 8.30 - 8.49 (m, 3 H), 8.04 - 8.27 (m, 6 H), 7.92 (d, J = 9.01 Hz, 2 H), 7.79 (d, J = 8.88 Hz, 2 H), 6.78 (s, 1 H), 6.71 (s, 1 H), 4.26 (dd, J = 11.26, 7.00 Hz, 1 H), 3.98 - 4.10 (m, 2 H), 3.82 - 3.89 (m, 1 H), 3.57 - 3.76 (m, 7 H), 3.02-3.16 (m, 5 H), 2.90 - 3.01 (m, 6 H), 2.63 (br s, 2 H), 2.12 - 2.30 (m, 2 H), 2.00 - 2.09 (m, 1 H), 1.89 - 1.98 (m, 2 H), 1.69 - 1.86 (m, 4 H), 1.55 - 1.69 (m, 4 H), 1.27 - 1.40 (m, 2 H), 1.01 - 1.18 (m, 2 H). 19 F NMR (376 MHz, DMSO-d 6 ) δ ppm -105.40 - -102.96 (m, 2 F).

Example 081: Trans-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-4-pyridyl]-difluoro-methyl ]-N-[(15)-3-[cis-3,5-diamino-l- piperidyl] cyclopentyl] cyclohexanecarboxamide

The title compound was prepared in analogy to the preparation of Example 080 by using tert-butyl N-[cis-5-(tert-butoxycarbonylamino)-3-piperidyl]carbamate instead of tert-butyl N- [[cz5-4-[(tert-butoxycarbonylamino)methyl]pyrrolidin-3-yl]me thyl]carbamate in Step 4. Example 081 (50 mg) was obtained as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 792.5. Example 081: 1 H NMR (400 MHz, DMSO-d 6 ) δ ppm 7.84-7.92 (m, 2H), 7.75-7.83 (m, 2H), 6.70 (br d, J = 18.51Hz, 2H), 4.23-4.34 (m, 1H), 4.05 (br s, 1H), 3.82 (br d, J = 10.63Hz, 1H), 3.50-3.70 (m, 9H), 3.08 (br dd, J = 18.07, 8.32Hz, 1H), 2.94 (br s, 4H), 2.75-2.84 (m, 2H), 2.61 (br d, J = 2.13Hz, 1H), 2.30-2.47 (m, 2H), 1.96-2.14 (m, 4H), 1.52-1.92 (m, 9H), 1.29 (br d, J = 11.38Hz, 2H), 1.10 (br d, J = 12.76Hz, 2H). 19 F NMR (376 MHz, DMSO-d 6 ) δ ppm -106.09- 102.01 (m, 2F).

Example 082 : Trans-4- [ [2- [4- [4- [(4R)-4-amino-2-oxo-pyrrolidin- 1- yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluo ro-methyl]-N-[(l»S,3^)-3-[(3R)- 3-aminopyrrolidin-l-yl]cyclopentyl]cyclohexanecarboxamide

The title compound was prepared in analogy to the preparation of Example 080 by using tert-butyl N-[(3R)-pyrrolidin-3-yl]carbamate instead of tert-butyl N-[[cis-4-[(tert- butoxycarbonylamino)methyl]pyrrolidin-3-yl]methyl]carbamate in Step 4. Example 082 (60 mg) was obtained as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 763.3.

Example 082: 1 H NMR (400 MHz, DMSO-d 6 ) δ ppm 7.94 (d, J = 9.2Hz, 2H), 7.83 (d, J = 8.8Hz, 2H), 6.67 (s, 1H), 4.65 (s, 1H), 4.38-4.43 (m, 1H), 4.15-4.20 (m, 2H), 4.07-4.11 (m, 1H), 3.91-3.98 (m, 2H), 3.76-3.81 (m, 1H), 3.69-3.71 (m, 4H), 3.57-3.63 (m, 1H), 3.17-3.23 (m, 2H), 3.07 (t, J = 4.8Hz, 2H), 2.71 (dd, J = 3.2Hz, J = 18.0Hz ,2H), 2.51-2.58 (m, 1H), 2.14-2.25 (m, 3H), 1.95-2.10 (m, 3H), 1.76-1.86 (m, 6H), 1.42-1.45 (m, 2H), 1.21-1.28 (m, 2H). 19 F NMR (376 MHz, DMSO-d 6 ) δ ppm -104.55-102.79 (m, 2F).

Example 084: Trans-3-[[4-[[2-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-l- yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluo ro- methyl]cyclohexanecarbonyl]amino]propyl-(3-aminopropyl)-dime thyl-ammonium

Step 1: Trans-tert-butyl N-[3-[3-[[4-[[2-[4-[4-[(4R)-4-(tert-butoxycarbonylamino)-2-o xo- pyrrolidin-l-yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-py ridyl]-difluoro- methyl]cyclohexanecarbonyl]amino]propyl-methyl-amino]propyl] carbamate

To a mixture of Int-5 (110.0 mg, 0.150 mmol) and A,A-diisopropylethylamine (70.0 mg, 0.540 mmol) in anhydrous DMF (4 mL) was added HATU (75.0 mg, 0.2 mmol) in one portion. The reaction mixture was stirred for 1 hour at 20 °C. Then N'-(3-aminopropyl)-N'- m ethylpropane- 1,3 -diamine (60.0 mg, 0.410 mmol) was added into the reaction mixture and stirred for 11 hours. After the starting material was consumed, the reaction mixture was filtered and purified by prep-HPLC. The resulting residue was then dissolved in DCM (3 mL) and into the solution was added with BOC2O (100 mg). The reaction was stirred at 25 °C for 12 hours. After completion, the mixture was concentrated in vacuo to give the crude product, which was purified by prep-HPLC to give 084a (101 mg, 0.110 mmol, 79.14% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 938.3.

Step 2 : Trans-3- [ [4- [ [2- [4- [4- [(4R)-4-(tert-butoxycarbonylamino)-2-oxo-pyrrolidin- 1- yl]phenyl]sulfonylpiperazin-l-yl]-6-chloro-4-pyridyl]-difluo ro- methyl]cyclohexanecarbonyl]amino]propyl-[3-(tert-butoxycarbo nylamino)propyl]- dimethyl-ammonium

To a solution of 084a (60.0 mg, 0.060 mmol) in DMSO (1 mL) was added sodium hydrogen carbonate (160.9 mg, 1.90 mmol) and iodomethane (0.3 mL, 0.12 mmol). The mixture was stirred at 40 °C for 12 hours. After completion, the reaction mixture was purified by prep- HPLC to give 084b (50 mg, 0.050 mmol, 82.02% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 953.1.

Step 3 : Trans-3- [ [4- [ [2- [4- [4- [(4R)-4-amino-2-oxo-pyrrolidin-l-yl] phenyl] sulfonylpiperazin- l-yl]-6-chloro-4-pyridyl]-difluoro-methyl]cyclohexanecarbony l]amino]propyl-(3- aminopropyl)-dimethyl-ammonium

To a solution of 084b (30.0 mg, 0.030 mmol) in DCM (0.5 mL) was added TFA (0.5 mL, 0.040 mmol). The mixture was stirred at 20 °C for 2 hours. Then, the reaction mixture was filtered and concentrated in vacuo to give a residue, which was purified by prep-HPLC to give Example 084 (10.2 mg, 0.010 mmol, 41.89% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 753.4.

Example 084: 1 H NMR (400 MHz, METHANOL-d 4 ) δ ppm 7.94 (br d, 7=8.56Hz, 2H), 7.82 (br d, 7=8.44Hz, 2H), 6.66-6.70 (m, 1H), 6.65 (s, 1H), 4.42 (br d, 7=4.16Hz, 1H), 4.22 (br s, 1H), 4.00 (br d, 7=10.51Hz, 1H), 3.69 (br s, 4H), 3.48 (br dd, 7=2.63, 1.41Hz, 2H), 3.35-3.43 (m, 2H), 3.24-3.29 (m, 2H), 3.21 (br d, J=10.03Hz, 1H), 3.14 (s, 6H), 3.06 (br s, 6H), 2.74 (dd, 7=18.03, 2.26Hz, 1H), 2.18 (br s, 3H), 1.96-2.11 (m, 3H), 1.88 (br d, 7=10.15Hz, 2H), 1.78 (br d, 7=11.74Hz, 2H), 1.43 (br d, 7=12.10Hz, 2H), 1.24 (br d, 7=11.74Hz, 2H). 19 F NMR (376 MHz, METHANOL-d 4 ) δ ppm -107.09-106.74 (m, 2F). Compound JH-LPH-33 disclosed in the literature (Bioorganic Chemistry (2020), 102, 104055) and reported having LPS synthesis pathway inhibition activity and displaying antibiotic activity against efflux-deficient Escherichia coli strains, was chosen as reference compound in present invention.

BIOLOGICAL EXAMPLES

Example 085: 50% Inhibitory Concentration (ICso) of Escherichia coli UDP-2,3- diacylglucosamine hydrolase (LpxH)

The inhibitory potency of compounds of Escherichia coli UDP-2,3 -di acyl glucosamine hydrolase (LpxH) was determined in an enzymatic assay. UDP-2,3 -diacylglucosamine (UDP- DAG) was purified from the Caulobacter crescentus Lpxl D225 A mutant protein that contained a tightly bound UDP-DAG molecule. The enzyme was diluted using assay buffer containing 50 mM NaCl, 20 mM Tris-HCl pH7.5, 2.5 mM MnCl 2 , 0.01% Triton X-100,lmg/mL BSA, and the final concentration is 2 nM. The compounds were diluted by Agilent liquid handler Bravo, the compound's serial dilution dose ranges from 100 pM to 1.7 nM. Then the enzyme and compounds mixture were incubated at room temperature for 10 mins. The enzymatic assay was started by adding UDP-DAG (FAC is 5 pM) and incubated for 20mins at room temperature. The plate was then heated to 95°C for 15mins on a water batch to stop the reaction. The hydrolysis of UDP-DAG by LpxH yielded 2,3 -diacylglucosamine 1 -phosphate (lipid X) and UMP, which was converted to ATP and quantified by the luciferase reaction using the UMP/CMP- Glo™ Glycosyltransferase Assay kit from Promega. The compound’s inhibitory effect of LpxH is determined by measuring the light change using a luminometer (Envision).

Table 3: Enzymatic ICso values of the compounds of this invention against Escherichia coli LpxH

Example 086: Minimal Inhibitory Concentration Protocol (MIC) Assay:

The antibacterial activity of molecules was evaluated against the commonly used quality control strain Escherichia coli BW 25113, Escherichia coli ATCC 25922 and the rifampin- resistant mutant strain Klebsiella pneumonia ATCC 43816. Escherichia coli BW 25113 was obtained from the Coli Genetic Stock Center (CGSC) (strain 7636). Both Escherichia coli ATCC 25922 and Klebsiella pneumonia ATCC 43816 were originally derived from human clinical samples and are available from ATCC (American Type Culture Collection). The in vitro potency of compounds to inhibit Escherichia coli (BW 25113, ATCC 25922) and Klebsiella pneumonia (ATCC 43816) growth was assessed by the MIC (Minimal Inhibitory Concentration) broth dilution method. Specifically compound dilutions were prepared from 10 mM DMSO stock solutions as follows: i) serial 2-fold dilution in 20 μL DMSO were prepared in a master plate (Greiner, Cat No: 651201), ii) 180 pL sterile distilled water was added to each aliquot and iii) 10 pL diluted compounds were transferred into a new assay plate (Costar, 3599).

Vials of each test microorganisms were maintained frozen in the vapor phase of a liquid nitrogen freezer. Single-use frozen vials of the three strains Escherichia coli BW 25113, Escherichia coli ATCC 25922 (KWIKSTIK, 0335K) and Klebsiella pneumonia ATCC 43816, with predetermined CFU/mL, were taken out from the freezer, thawed at room temperature, and diluted in Cation-Adjusted Mueller Hinton Broth (CAMHB) to achieve a final inoculum of 5 * 10 5 CFU/ mL. 90 pL bacteria containing broth was dispensed to the assay plate containing the pre-dispensed compound dilutions and mixed by pipetting 5 times. Then the assay plates were incubated for 20 hours at 35°C in ambient air. Following incubation, the MIC (pg/mL), the lowest concentration of drug that inhibits visible growth of the microorganism was read with the help of a magnification mirror and recorded.

Table 4: MIC values of the compounds of this invention against E. coli and K. pneumonia

Example 087: Human microsome stability assay

The human microsomal stability assay is used for early assessment of metabolic stability of a test compound in human liver microsomes. Human liver microsomes (Cat.NO. : 452117, Corning, USA) were preincubated with test compound for 10 minutes at 37°C in 100 mM potassium phosphate buffer, pH 7.4. The reactions were initiated by adding NADPH regenerating system. The final incubation mixtures contained 1 pM test compound, 0.5 mg/mL liver microsomal protein, 1 mM MgCL, 1 mMNADP, 1 unit/mL isocitric dehydrogenase and 6 mM isocitric acid in 100 mM potassium phosphate buffer, pH 7.4. After incubation times of 0, 3, 6, 9, 15 and 30 minutes at 37°C, 300 μL of cold acetonitrile (including internal standard) was added to 100 pL incubation mixture to terminate the reaction. Following precipitation and centrifugation, the amount of compound remaining in the samples were determined by LC-MS/MS. Controls of no NADPH regenerating system at zero and 30 minutes were also prepared and analyzed. The compounds of present invention showed good human liver microsome stability determined in the above assay, results are shown in Table 5 below. (Human microsome clearance: <7 mL/min/kg: low; 7-16.2 mL/min/kg: medium; >16.2 mL/min/kg: high)

Table 5: Human microsome clearance values of the compounds of this invention