Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PIPERAZINYLSULFONYLARYL COMPOUNDS FOR TREATMENT OF BACTERIAL INFECTIONS
Document Type and Number:
WIPO Patent Application WO/2023/072794
Kind Code:
A1
Abstract:
The present invention relates to compounds of formula (I), wherein R1, R2, R3, R4, Y, Q1, Q2, Q3, Q4 and Q5 are as described herein, and their pharmaceutically acceptable salt thereof, and compositions including the compounds and methods of using the compounds.

Inventors:
DEY FABIAN (CH)
DING XIAO (CN)
SHI HOUGUANG (CN)
TAN XUEFEI (CN)
WU JUN (CN)
ZHENG JIAMIN (CN)
ZHOU MINGWEI (CN)
Application Number:
PCT/EP2022/079506
Publication Date:
May 04, 2023
Filing Date:
October 24, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
HOFFMANN LA ROCHE (US)
HOFFMANN LA ROCHE (US)
International Classes:
A61P31/04; C07D401/14; C07D403/14; C07D413/14; C07D471/10; C07D487/04; C07D487/10; C07D498/04; C07D498/10
Domestic Patent References:
WO2003032962A22003-04-24
Other References:
KUMAR G. V. ET AL: "DESIGN, SYNTHESIS AND BIOLOGICAL EVALUATION OF NOVEL BROMO-PYRIMIDINE ANALOGS AS ANTICANCER AND ANTIMICROBIAL AGENTS", IJPCBS, vol. 9, no. 1, 1 January 2019 (2019-01-01), pages 27 - 41, XP093016039
ANN. REV. BIOCHEM, vol. 76, 2007, pages 295 - 329
ANSELHOWARD C. ET AL.: "Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems", 2004, WILLIAMS & WILKINS
GENNAROALFONSO R. ET AL.: "Remington: The Science and Practice of Pharmacy", 2000, WILLIAMS & WILKINS
ROWERAYMOND C: "Handbook of Pharmaceutical Excipients", 2005, PHARMACEUTICAL PRESS
Attorney, Agent or Firm:
SALUD, Carlos (CH)
Download PDF:
Claims:
CLAIMS 1. A compound of formula (I), wherein R1 is 2-oxo-1-oxa-3,8-diazaspiro[4.5]decanyl unsubstituted or substituted by aminoC1-6alkyl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridinyl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[5,4-c]pyridinyl, 2-oxo-3a,4,6,7,8,8a-hexahydrooxazolo[4,5-c]azepinyl substituted by aminoC1-6alkyl or C1- 6alkylaminoC1-6alkyl, 2-oxo-3a,5,6,6a-tetrahydrofuro[2,3-d]oxazolyl substituted twice by substituents independently selected from hydroxy and hydroxyC1-6alkyl, 2-oxo-4,5,6,6a-tetrahydro-3aH-pyrrolo[3,4-d]oxazolyl, 2-oxo-4,5,6,7,8,8a-hexahydro-3aH-oxazolo[4,5-c]azepinyl, 2-oxo-5,6,7,7a-tetrahydro-3aH-pyrano[2,3-d]oxazolyl substituted three times by substituents independently selected from hydroxy and hydroxyC1-6alkyl, 3-oxo-2,6,9-triazaspiro[4.5]decanyl, 3-oxo-2,6-diazaspiro[4.5]decanyl, 3-oxo-2,8-diazaspiro[4.5]decanyl, 5-oxo-1,2,3,3a,6,6a-hexahydropyrrolo[3,2-b]pyrrolyl, 6-oxo-5-oxa-2,7-diazaspiro[3.4]octanyl, 7-oxo-2,6-diazaspiro[3.4]octanyl, oxoazetidinyl substituted by amino, oxooxazolidinyl substituted once or twice by substituents independently selected from ((aminoC1-6alkyl)azetidinyl)C1-6alkyl, (C1-6alkyl)2aminoC1-6alkyl, (dihydroxypyrrolidinyl)C1-6alkyl, (pyrrolidinylamino)C1-6alkyl, 2,6- diazaspiro[3.3]heptanylC1-6alkyl, dihydroxy(C1-6alkoxy)tetrahydrofuranyl, aminoazetidinylC1-6alkyl, aminoC1-6alkyl, C1-6alkyl, C1-6alkylaminoC1-6alkyl, C1- 6alkylcarbonylaminoC1-6alkyl, hydroxyC1-6alkyl and piperazinylC1-6alkyl, or oxopyrrolidinyl substituted once, twice or three times by substituents independently selected from (((C1-6alkyl)2aminoC1-6alkyl)azetidinyl)C1-6alkyl, ((aminoC1- 6alkyl)azetidinyl)C1-6alkyl, ((aminoC3-7cycloalkyl)azetidinyl)C1-6alkyl, ((C1- 6alkylamino)azetidinyl)C1-6alkyl, (aminoazetidinyl)C1-6alkyl, (aminoC1- 6alkyl)azetidinyl, (azetidinylC1-6alkylamino)C1-6alkyl, (C1-6alkylpiperazinyl)C1-6alkyl, (hydroxyC1-6alkyl)piperazinyl, 2,5-diazabicyclo[2.2.1]heptanyl, 2,6- diazaspiro[3.3]heptanyl, amino, amino(hydroxy)piperidinyl, amino(hydroxy)pyrrolidinyl, aminoazetidinyl, aminoC1-6alkyl, (aminoC1- 6alkyl)amino, aminopyrrolidinyl, azetidinylamino, C1-6alkyl, C1-6alkylamino, dihydroxypyrrolidinyl, (hydroxyC1-6alkyl)amino, piperazinyl, piperazinylC1-6alkyl, piperidinylamino and pyrrolidinylamino; R2 is H or cyano; R3 is benzyl, C3-7cycloalkylsulfonyl, cyanoC3-7cycloalkyl, phenoxy, phenylC3-7cycloalkyl, tetrahydropyranylsulfonyl, or C1-6alkyl substituted twice or three times by substituents independently selected from halogen, (aminoC1-6alkyl)C3-7cycloalkyl, (hydroxyC1-6alkyl)C3-7cycloalkyl, 1,2,3,6- tetrahydropyridinyl, 1,4-dioxanyl, 3,6-dihydro-2H-pyranyl, 3- azabicyclo[3.1.0]hexanyl, 3-oxabicyclo[3.1.0]hexanyl, 8-oxabicyclo[3.2.1]octanyl, azetidinyl, C1-6alkylhalopyrazinyl, C1-6alkylmorpholinyl, C1-6alkylpyrazinyl, carbamoylC3-7cycloalkyl, hydroxyC3-7cycloalkyl, morpholinyl, oxetanyl, piperidinyl, pyrazinyl, pyridinyl, pyrimidinyl, tetrahydrofuranyl, tetrahydropyranyl, phenyl and pyridinyl; R4 is halogen, C1-6alkyl, C3-7cycloalkyl, C1-6alkoxy or C2-6alkynyl; Q1 is N or CRa, wherein Ra is H or halogen; Q2 is N or CRb, wherein Rb is H or halogen; Q3 is N or CH; Q4 is N or CH; Q5 is N or CH; Y is O or NH; or a pharmaceutically acceptable salt thereof. 2. A compound of formula (Ia), (Ia), wherein R1 is 2-oxo-1-oxa-3,8-diazaspiro[4.5]decanyl unsubstituted or substituted by aminoC1-6alkyl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridinyl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[5,4-c]pyridinyl, 2-oxo-3a,4,6,7,8,8a-hexahydrooxazolo[4,5-c]azepinyl substituted by aminoC1-6alkyl or C1- 6alkylaminoC1-6alkyl, 2-oxo-3a,5,6,6a-tetrahydrofuro[2,3-d]oxazolyl substituted twice by substituents independently selected from hydroxy and hydroxyC1-6alkyl, 2-oxo-4,5,6,6a-tetrahydro-3aH-pyrrolo[3,4-d]oxazolyl, 2-oxo-4,5,6,7,8,8a-hexahydro-3aH-oxazolo[4,5-c]azepinyl, 2-oxo-5,6,7,7a-tetrahydro-3aH-pyrano[2,3-d]oxazolyl substituted three times by substituents independently selected from hydroxy and hydroxyC1-6alkyl, 3-oxo-2,6,9-triazaspiro[4.5]decanyl, 3-oxo-2,6-diazaspiro[4.5]decanyl, 3-oxo-2,8-diazaspiro[4.5]decanyl, 5-oxo-1,2,3,3a,6,6a-hexahydropyrrolo[3,2-b]pyrrolyl, 6-oxo-5-oxa-2,7-diazaspiro[3.4]octanyl, 7-oxo-2,6-diazaspiro[3.4]octanyl, oxoazetidinyl substituted by amino, oxooxazolidinyl substituted once or twice by substituents independently selected from ((aminoC1-6alkyl)azetidinyl)C1-6alkyl, (C1-6alkyl)2aminoC1-6alkyl, (dihydroxypyrrolidinyl)C1-6alkyl, (pyrrolidinylamino)C1-6alkyl, 2,6- diazaspiro[3.3]heptanylC1-6alkyl, dihydroxy(C1-6alkoxy)tetrahydrofuranyl, aminoazetidinylC1-6alkyl, aminoC1-6alkyl, C1-6alkyl, C1-6alkylaminoC1-6alkyl, C1- 6alkylcarbonylaminoC1-6alkyl, hydroxyC1-6alkyl and piperazinylC1-6alkyl, or oxopyrrolidinyl substituted once, twice or three times by substituents independently selected from (((C1-6alkyl)2aminoC1-6alkyl)azetidinyl)C1-6alkyl, ((aminoC1- 6alkyl)azetidinyl)C1-6alkyl, ((aminoC3-7cycloalkyl)azetidinyl)C1-6alkyl, ((C1- 6alkylamino)azetidinyl)C1-6alkyl, (aminoazetidinyl)C1-6alkyl, (aminoC1- 6alkyl)azetidinyl, (azetidinylC1-6alkylamino)C1-6alkyl, (C1-6alkylpiperazinyl)C1-6alkyl, (hydroxyC1-6alkyl)piperazinyl, 2,5-diazabicyclo[2.2.1]heptanyl, 2,6- diazaspiro[3.3]heptanyl, amino, amino(hydroxy)piperidinyl, amino(hydroxy)pyrrolidinyl, aminoazetidinyl, aminoC1-6alkyl, (aminoC1- 6alkyl)amino, aminopyrrolidinyl, azetidinylamino, C1-6alkyl, C1-6alkylamino, dihydroxypyrrolidinyl, (hydroxyC1-6alkyl)amino, piperazinyl, piperazinylC1-6alkyl, piperidinylamino and pyrrolidinylamino; R2 is H or cyano; R3 is benzyl, C3-7cycloalkylsulfonyl, cyanoC3-7cycloalkyl, phenoxy, phenylC3-7cycloalkyl, tetrahydropyranylsulfonyl, or C1-6alkyl substituted twice or three times by substituents independently selected from halogen, (aminoC1-6alkyl)C3-7cycloalkyl, (hydroxyC1-6alkyl)C3-7cycloalkyl, 1,2,3,6- tetrahydropyridinyl, 1,4-dioxanyl, 3,6-dihydro-2H-pyranyl, 3- azabicyclo[3.1.0]hexanyl, 3-oxabicyclo[3.1.0]hexanyl, 8-oxabicyclo[3.2.1]octanyl, azetidinyl, C1-6alkylhalopyrazinyl, C1-6alkylmorpholinyl, C1-6alkylpyrazinyl, carbamoylC3-7cycloalkyl, hydroxyC3-7cycloalkyl, morpholinyl, oxetanyl, piperidinyl, pyrazinyl, pyridinyl, pyrimidinyl, tetrahydrofuranyl, tetrahydropyranyl, phenyl and pyridinyl; R4 is halogen, C1-6alkyl, C3-7cycloalkyl, C1-6alkoxy or C2-6alkynyl; Q1 is N or CRa, wherein Ra is H or halogen; Q2 is CRb, wherein Rb is H or halogen; Q3 is N; Q4 is N or CH; Q5 is N or CH; Y is O; or a pharmaceutically acceptable salt thereof. 3. A compound according to claim 1 or 2, wherein R1 is 2-oxo-1-oxa-3,8-diazaspiro[4.5]decanyl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridinyl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[5,4-c]pyridinyl, 2-oxo-3a,5,6,6a-tetrahydrofuro[2,3-d]oxazolyl substituted twice by substituents independently selected from hydroxy and hydroxyC1-6alkyl, 2-oxo-4,5,6,6a-tetrahydro-3aH-pyrrolo[3,4-d]oxazolyl, 2-oxo-4,5,6,7,8,8a-hexahydro-3aH-oxazolo[4,5-c]azepinyl, 3-oxo-2,6-diazaspiro[4.5]decanyl, 5-oxo-1,2,3,3a,6,6a-hexahydropyrrolo[3,2-b]pyrrolyl, 6-oxo-5-oxa-2,7-diazaspiro[3.4]octanyl, oxooxazolidinyl substituted once or twice by substituents independently selected from ((aminoC1-6alkyl)azetidinyl)C1-6alkyl, (C1-6alkyl)2aminoC1-6alkyl, dihydroxy(C1- 6alkoxy)tetrahydrofuranyl, aminoC1-6alkyl, C1-6alkyl, C1-6alkylaminoC1-6alkyl, hydroxyC1-6alkyl and piperazinylC1-6alkyl, or oxopyrrolidinyl substituted by substituent selected from ((aminoC1-6alkyl)azetidinyl)C1- 6alkyl, ((aminoC3-7cycloalkyl)azetidinyl)C1-6alkyl, (aminoazetidinyl)C1-6alkyl, (C1- 6alkylpiperazinyl)C1-6alkyl, (hydroxyC1-6alkyl)piperazinyl, amino, amino(hydroxy)piperidinyl, amino(hydroxy)pyrrolidinyl, aminoC1-6alkyl, aminopyrrolidinyl, C1-6alkylamino, dihydroxypyrrolidinyl, (hydroxyC1-6alkyl)amino, piperazinyl, piperazinylC1-6alkyl and piperidinylamino. 4. A compound according to claim 3, wherein R1 is 2-oxo-1-oxa-3,8-diazaspiro[4.5]decanyl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridinyl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[5,4-c]pyridinyl, 2-oxo-3a,5,6,6a-tetrahydrofuro[2,3-d]oxazolyl substituted twice by substituents independently selected from hydroxy and hydroxymethyl, 2-oxo-4,5,6,6a-tetrahydro-3aH-pyrrolo[3,4-d]oxazolyl, 2-oxo-4,5,6,7,8,8a-hexahydro-3aH-oxazolo[4,5-c]azepinyl, 3-oxo-2,6-diazaspiro[4.5]decanyl, 5-oxo-1,2,3,3a,6,6a-hexahydropyrrolo[3,2-b]pyrrolyl, 6-oxo-5-oxa-2,7-diazaspiro[3.4]octanyl, oxooxazolidinyl substituted once or twice by substituents independently selected from (3- (aminomethyl)azetidin-1-yl)methyl, (dimethylamino)methyl, 3,4-dihydroxy-5- methoxy-tetrahydrofuran-2-yl, aminomethyl, aminomethyl, hydroxymethyl, methyl, (methylamino)methyl and piperazin-1-ylmethyl, or oxopyrrolidinyl substituted by substituent selected from (3-(1-aminocyclopropyl)azetidin- 1-yl)methyl, (3-(aminomethyl)azetidin-1-yl)methyl, (3-aminoazetidin-1-yl)methyl, (4-methylpiperazin-1-yl)methyl, 3-(hydroxymethyl)piperazin-1-yl, 3,4- dihydroxypyrrolidin-1-yl, 3-amino-4-hydroxypiperidin-1-yl, 3-amino-4-hydroxy- pyrrolidin-1-yl, 3-aminopyrrolidin-1-yl, 4-piperidinylamino, amino, aminomethyl, (hydroxyethyl)amino, methylamino, piperazin-1-yl and piperazin-1-ylmethyl. 5. A compound according to claim 4, wherein R1 is 2-oxo-1-oxa-3,8-diazaspiro[4.5]decan-3-yl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridin-3-yl, 2-oxo-3a,4,5,6,7,7a- hexahydrooxazolo[5,4-c]pyridin-1-yl, 2-oxo-4,5,6,6a-tetrahydro-3aH-pyrrolo[3,4-d]oxazol-3-yl, 2-oxo-4,5,6,7,8,8a-hexahydro-3aH-oxazolo[4,5-c]azepin-3-yl, 3-oxo-2,6-diazaspiro[4.5]decan-2- yl, 4-((3-(1-aminocyclopropyl)azetidin-1-yl)methyl)-2-oxo-pyrrolidin-1-yl, 4-((3-aminoazetidin- 1-yl)methyl)-2-oxo-pyrrolidin-1-yl, 4-((4-methylpiperazin-1-yl)methyl)-2-oxo-pyrrolidin-1-yl, 4-(2-hydroxyethylamino)-2-oxo-pyrrolidin-1-yl, 4-(3-(aminomethyl)azetidin-1-yl)methyl)-2- oxo-pyrrolidin-1-yl, 4-(3-(hydroxymethyl)piperazin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(3,4- dihydroxypyrrolidin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(3-amino-4-hydroxypiperidin-1-yl)-2-oxo- pyrrolidin-1-yl, 4-(3-amino-4-hydroxy-pyrrolidin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(3- aminopyrrolidin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(4-piperidinylamino)-2-oxo-pyrrolidin-1-yl, 4- (methylamino)-2-oxo-pyrrolidin-1-yl, 4-(piperazin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(piperazin-1- ylmethyl)-2-oxo-pyrrolidin-1-yl, 4-amino-2-oxo-pyrrolidin-1-yl, 4-aminomethyl-2-oxo- oxazolidin-3-yl, 4-aminomethyl-2-oxo-pyrrolidin-1-yl, 4-methyl-2-oxo-5-(3,4-dihydroxy-5- methoxy-tetrahydrofuran-2-yl)oxazolidin-3-yl, 5-((3-(aminomethyl)azetidin-1-yl)methyl)-2-oxo- oxazolidin-3-yl, 5-((dimethylamino)methyl)-2-oxo-oxazolidin-3-yl, 5-(2-aminoethyl)-2-oxo- oxazolidin-3-yl, 5-(methylaminomethyl)-2-oxo-oxazolidin-3-yl, 5-(piperazin-1-ylmethyl)-2-oxo- oxazolidin-3-yl, 5-aminomethyl-2-oxo-oxazolidin-3-yl, 5-hydroxymethyl-2-oxo-oxazolidin-3-yl, 5-oxo-1,2,3,3a,6,6a-hexahydropyrrolo[3,2-b]pyrrol-4-yl, 6-hydroxy-5-hydroxymethyl-2-oxo- 3a,5,6,6a-tetrahydrofuro[2,3-d]oxazol-3-yl or 6-oxo-5-oxa-2,7-diazaspiro[3.4]octan-7-yl.

6. A compound according to claim 3, wherein R1 is oxopyrrolidinyl substituted by substituent selected from ((aminoC1-6alkyl)azetidinyl)C1-6alkyl, ((aminoC3-7cycloalkyl)azetidinyl)C1-6alkyl, (aminoazetidinyl)C1-6alkyl, (C1-6alkylpiperazinyl)C1-6alkyl, (hydroxyC1-6alkyl)piperazinyl, amino, amino(hydroxy)piperidinyl, amino(hydroxy)pyrrolidinyl, aminoC1-6alkyl, aminopyrrolidinyl, C1-6alkylamino, dihydroxypyrrolidinyl, (hydroxyC1-6alkyl)amino, piperazinyl, piperazinylC1-6alkyl and piperidinylamino. 7. A compound according to claim 6, wherein R1 is 4-((3-(1-aminocyclopropyl)azetidin-1- yl)methyl)-2-oxo-pyrrolidin-1-yl, 4-((3-aminoazetidin-1-yl)methyl)-2-oxo-pyrrolidin-1-yl, 4-((4- methylpiperazin-1-yl)methyl)-2-oxo-pyrrolidin-1-yl, 4-(2-hydroxyethylamino)-2-oxo-pyrrolidin- 1-yl, 4-(3-(aminomethyl)azetidin-1-yl)methyl)-2-oxo-pyrrolidin-1-yl, 4-(3- (hydroxymethyl)piperazin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(3,4-dihydroxypyrrolidin-1-yl)-2-oxo- pyrrolidin-1-yl, 4-(3-amino-4-hydroxypiperidin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(3-amino-4- hydroxy-pyrrolidin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(3-aminopyrrolidin-1-yl)-2-oxo-pyrrolidin-1- yl, 4-(4-piperidinylamino)-2-oxo-pyrrolidin-1-yl, 4-(methylamino)-2-oxo-pyrrolidin-1-yl, 4- (piperazin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(piperazin-1-ylmethyl)-2-oxo-pyrrolidin-1-yl, 4-amino- 2-oxo-pyrrolidin-1-yl or 4-aminomethyl-2-oxo-pyrrolidin-1-yl. 8. A compound according to claim 1 or 2, wherein R3 is benzyl, phenylC3-7cycloalkyl, or C1- 6alkyl substituted three times by substituents independently selected from halogen, (hydroxyC1- 6alkyl)C3-7cycloalkyl, 1,2,3,6-tetrahydropyridinyl, 1,4-dioxanyl, 3,6-dihydro-2H-pyranyl, 3- azabicyclo[3.1.0]hexanyl, 3-oxabicyclo[3.1.0]hexanyl, 8-oxabicyclo[3.2.1]octanyl, C1- 6alkylhalopyrazinyl, C1-6alkylpyrazinyl, carbamoylC3-7cycloalkyl, hydroxyC3-7cycloalkyl, morpholinyl, oxetanyl, pyrazinyl, pyridinyl, tetrahydropyranyl and phenyl. 9. A compound according to claim 8, wherein R3 is C1-6alkyl substituted three times by substituents independently selected from halogen, (hydroxyC1-6alkyl)C3-7cycloalkyl, 1,4- dioxanyl, 3-azabicyclo[3.1.0]hexanyl, 3-oxabicyclo[3.1.0]hexanyl, C1-6alkylhalopyrazinyl, C1- 6alkylpyrazinyl, hydroxyC3-7cycloalkyl, morpholinyl, oxetanyl, phenyl, pyridinyl and tetrahydropyranyl.

10. A compound according to claim 9, wherein R3 is difluoro(1,4-dioxan-2-yl)methyl, difluoro(2-(hydroxymethyl)cyclopropyl)methyl, difluoro(2-pyridinyl)methyl, difluoro(3- hydroxycyclobutyl)methyl, difluoro(3-oxabicyclo[3.1.0]hexan-6-yl)methyl, difluoro(5-chloro-6- methyl-pyrazin-2-yl)methyl, difluoro(6-methylpyrazin-2-yl)methyl, difluoro(morpholin-2- yl)methyl, difluoro(oxetan-3-yl)methyl, difluoro(phenyl)methyl, difluoro(tetrahydropyran-4- yl)methyl or difluoro[3-azabicyclo[3.1.0]hexan-1-yl]methyl. 11. A compound according to claim 1 or 2, wherein R2 is H. 12. A compound according to claim 1 or 2, wherein R4 is halogen or C1-6alkyl. 13. A compound according to claim 12, wherein R4 is chloro or methyl. 14. A compound according to claim 1 or 2, wherein Q1 is CRa, wherein Ra is H or halogen. 15. A compound according to claim 14, wherein Q1 is CRa, wherein Ra is H or fluoro. 16. A compound according to claim 1 or 2, wherein Q2 is CH. 17. A compound according to claim 1 or 2, wherein R1 is oxopyrrolidinyl substituted by amino or aminoC1-6alkyl; R2 is H; R3 is C1-6alkyl substituted three times by substituents independently selected from halogen, (hydroxyC1-6alkyl)C3-7cycloalkyl, 1,4-dioxanyl, 3-azabicyclo[3.1.0]hexanyl, 3- oxabicyclo[3.1.0]hexanyl, C1-6alkylhalopyrazinyl, C1-6alkylpyrazinyl, hydroxyC3- 7cycloalkyl, morpholinyl, oxetanyl, phenyl, pyridinyl and tetrahydropyranyl; R4 is halogen or C1-6alkyl; Q1 is CRa, wherein Ra is H or halogen; Q2 is CH; Q3 is N; Q4 is N or CH; Q5 is N or CH; Y is O; or a pharmaceutically acceptable salt thereof. 18. A compound according to claim 17, wherein R1 is oxopyrrolidinyl substituted by amino or aminomethyl; R2 is H; R3 is difluoro(1,4-dioxan-2-yl)methyl, difluoro(2-(hydroxymethyl)cyclopropyl)methyl, difluoro(2-pyridinyl)methyl, difluoro(3-hydroxycyclobutyl)methyl, difluoro(3- oxabicyclo[3.1.0]hexan-6-yl)methyl, difluoro(5-chloro-6-methyl-pyrazin-2-yl)methyl, difluoro(6-methylpyrazin-2-yl)methyl, difluoro(morpholin-2-yl)methyl, difluoro(oxetan-3- yl)methyl, difluoro(phenyl)methyl, difluoro(tetrahydropyran-4-yl)methyl or difluoro[3- azabicyclo[3.1.0]hexan-1-yl]methyl; R4 is chloro or methyl; Q1 is CRa, wherein Ra is H or fluoro; Q2 is CH; Q3 is N; Q4 is N or CH; Q5 is N or CH; Y is O; or a pharmaceutically acceptable salt thereof. 19. A compound selected from: 4-Amino-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl] pyrrolidin-2-one; 4-(Aminomethyl)-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonyl- phenyl]pyrrolidin-2-one; 5-(Aminomethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]oxazolidin-2-one; N-[[3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-2-oxo- oxazolidin-5-yl]methyl]acetamide; 4-Amino-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]- 3,3-dimethyl-pyrrolidin-2-one; 4-(Aminomethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]oxazolidin-2-one; (3S)-3-amino-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl] azetidin-2-one; 3-Amino-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl] pyrrolidin-2-one; 5-(2-Aminoethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]oxazolidin-2-one; 2-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-2,6- diazaspiro[4.5]decan-3-one; 2-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-2,8- diazaspiro[4.5]decan-3-one; 6-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-2,6- diazaspiro[3.4]octan-7-one; 7-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-5-oxa-2,7- diazaspiro[3.4]octan-6-one; 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-1-oxa-3,8- diazaspiro[4.5]decan-2-one; (3aR,6aS)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]- 4,5,6,6a-tetrahydro-3aH-pyrrolo[3,4-d]oxazol-2-one; (3aR,7aS)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]- 3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridin-2-one; (3aR,7aS)-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]- 3a,4,5,6,7,7a-hexahydrooxazolo[5,4-c]pyridin-2-one; (3aS,7aR)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]- 3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridin-2-one; (3aS,8aR)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]- 4,5,6,7,8,8a-hexahydro-3aH-oxazolo[4,5-c]azepin-2-one; 4-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]- 1,2,3,3a,6,6a-hexahydropyrrolo[3,2-b]pyrrol-5-one; 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-6-hydroxy- 5-(hydroxymethyl)-3a,5,6,6a-tetrahydrofuro[2,3-d]oxazol-2-one; 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-5- [(3S,4R,5R)-3,4-dihydroxy-5-methoxy-tetrahydrofuran-2-yl]-4-methyl-oxazolidin-2-one; (3aS,5S,6S,7S,7aR)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonyl phenyl]-5,7-dihydroxy-6-(hydroxymethyl)-5,6,7,7a-tetrahydro-3aH-pyrano[2,3-d]oxazol-2-one; 8-(2-Aminoethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-1-oxa-3,8-diazaspiro[4.5]decan-2-one; (3aS,8aR)-5-(2-aminoethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-3a,4,6,7,8,8a-hexahydrooxazolo[4,5-c]azepin-2-one; (3aS,8aR)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]- 5-[2-(methylamino)ethyl]-3a,4,6,7,8,8a-hexahydrooxazolo[4,5-c]azepin-2-one; 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-5- (methylaminomethyl)oxazolidin-2-one; 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-5- [(dimethylamino)methyl]oxazolidin-2-one; 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-5- (piperazin-1-ylmethyl)oxazolidin-2-one; 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-5-[[cis- 3,4-dihydroxypyrrolidin-1-yl]methyl]oxazolidin-2-one; 5-[(3-Aminoazetidin-1-yl)methyl]-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]oxazolidin-2-one; 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-5-(2,6- diazaspiro[3.3]heptan-2-ylmethyl)oxazolidin-2-one; 5-[[3-(Aminomethyl)azetidin-1-yl]methyl]-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]oxazolidin-2-one; 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-5- [(pyrrolidin-3-ylamino)methyl]oxazolidin-2-one; 4-[[3-(Aminomethyl)azetidin-1-yl]methyl]-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-4-[(4- methylpiperazin-1-yl)methyl]pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-4-[[3- [(dimethylamino)methyl]azetidin-1-yl]methyl]pyrrolidin-2-one; 4-[(Azetidin-3-ylmethylamino)methyl]-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; 4-[[3-(1-Aminocyclopropyl)azetidin-1-yl]methyl]-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; 4-[(3-Aminoazetidin-1-yl)methyl]-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-4- (piperazin-1-ylmethyl)pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-4-[[3- (dimethylamino)azetidin-1-yl]methyl]pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-4-[[3- (methylamino)azetidin-1-yl]methyl]pyrrolidin-2-one; 1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-4- (methylamino)pyrrolidin-2-one; 4-(2-Aminoethylamino)-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-4-(2- hydroxyethylamino)pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-4- piperazin-1-yl-pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-4- [(3S,4R)-3,4-dihydroxypyrrolidin-1-yl]pyrrolidin-2-one; 4-[3-(Aminomethyl)azetidin-1-yl]-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; 4-(3-Aminopyrrolidin-1-yl)-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-(3-Aminoazetidin-1-yl)-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-4-(2,6- diazaspiro[3.3]heptan-2-yl)pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-4-[(3S)-3- (hydroxymethyl)piperazin-1-yl]pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-4-[(3R)-3- (hydroxymethyl)piperazin-1-yl]pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-4-[(1S,4S)- 2,5-diazabicyclo[2.2.1]heptan-2-yl]pyrrolidin-2-one; 4-[(3R,4R)-3-amino-4-hydroxy-pyrrolidin-1-yl]-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; 4-((3R,4R)-3-amino-4-hydroxypiperidin-1-yl)-1-(4-((4-(6-chloro-4- (trifluoromethyl)pyridin-2-yl)piperazin-1-yl)sulfonyl)phenyl)pyrrolidin-2-one; 4-(Azetidin-3-ylamino)-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-4- (pyrrolidin-3-ylamino)pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-4-(4- piperidylamino)pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl]- 5-methyl-pyrrolidin-2-one; 4-Amino-1-[5-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonyl-2- pyridyl]pyrrolidin-2-one; 5-(Aminomethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonyl- 2-fluoro-phenyl]oxazolidin-2-one; 5-(Aminomethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonyl- 3-fluoro-phenyl]oxazolidin-2-one; 4-Amino-1-[4-[4-[4-methyl-6-(trifluoromethyl)pyrimidin-2-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[4-(trideuteriomethyl)-6-(trifluoromethyl)pyrimidin-2-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 1-[4-(4-Amino-2-oxo-pyrrolidin-1-yl)phenyl]sulfonyl-4-[6-chloro-4-(trifluoromethyl)-2- pyridyl]piperazine-2-carbonitrile; 4-Amino-1-[4-[4-[6-chloro-4-(difluoromethyl)-2-pyridyl]piperazin-1-yl]sulfonylphenyl] pyrrolidin-2-one; 4-Amino-1-[4-[4-(6-chloro-4-cyclopropylsulfonyl-2-pyridyl)piperazin-1-yl]sulfonylphenyl] pyrrolidin-2-one; 4-Amino-1-[4-[4-(6-chloro-4-tetrahydropyran-4-ylsulfonyl-2-pyridyl)piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 1-[2-[4-[4-(4-Amino-2-oxo-pyrrolidin-1-yl)phenyl]sulfonylpiperazin-1-yl]-6-chloro-4- pyridyl]cyclopropanecarbonitrile; 4-Amino-1-[4-[4-(6-chloro-4-phenoxy-2-pyridyl)piperazin-1-yl]sulfonylphenyl]pyrrolidin- 2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(phenyl)methyl]-2-pyridyl]piperazin-1- yl]sulfonylphenyl] pyrrolidin-2-one; 4-Amino-1-[4-[4-(4-benzyl-6-chloro-2-pyridyl)piperazin-1-yl]sulfonylphenyl]pyrrolidin-2- one; 4-Amino-1-[4-[4-[6-chloro-4-(1-phenylcyclopropyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl] pyrrolidin-2-one; 4-Amino-1-[4-[4-[4-[difluoro(phenyl)methyl]-6-methyl-pyrimidin-2-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[2-chloro-6-[difluoro(phenyl)methyl]pyrimidin-4-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[4-chloro-6-[difluoro(phenyl)methyl]pyrimidin-2-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[2-cyclopropyl-6-[difluoro(phenyl)methyl]pyrimidin-4-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[4-cyclopropyl-6-[difluoro(phenyl)methyl]pyrimidin-2-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-[difluoro(phenyl)methyl]-2-methoxy-pyrimidin-4-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[4-[difluoro(phenyl)methyl]-6-methoxy-pyrimidin-2-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-Amino-1-[4-[4-[6-[difluoro(phenyl)methyl]-2-methyl-pyrimidin-4-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-[difluoro(phenyl)methyl]-2-vinyl-pyrimidin-4-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 2-[4-[4-[6-Chloro-4-[difluoro(phenyl)methyl]-2-pyridyl]piperazin-1-yl]sulfonylphenyl]- 2,6,9-triazaspiro[4.5]decan-3-one; 5-(Aminomethyl)-1-[4-[4-[6-chloro-4-[difluoro(phenyl)methyl]-2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; (3aR,7aS)-3-[4-[4-[6-chloro-4-[difluoro(phenyl)methyl]-2-pyridyl]piperazin-1-yl]sulfonyl phenyl]-3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridin-2-one; 5-(Aminomethyl)-3-[4-[4-[6-chloro-4-[difluoro(phenyl)methyl]-2-pyridyl]piperazin-1- yl]sulfonylphenyl]oxazolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(2-pyridyl)methyl]-2-pyridyl]piperazin-1-yl] sulfonylphenyl] pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(3-pyridyl)methyl]-2-pyridyl]piperazin-1- yl]sulfonylphenyl] pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(4-pyridyl)methyl]-2-pyridyl]piperazin-1-yl] sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(pyrimidin-5-yl)methyl]-2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(pyrazin-2-yl)methyl]-2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(pyrimidin-2-yl)methyl]-2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(tetrahydropyran-4-yl)methyl]-2-pyridyl]piperazin- 1-yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(tetrahydrofuran-3-yl)methyl]-2-pyridyl]piperazin- 1-yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(oxetan-3-yl)methyl]-2-pyridyl]piperazin-1- yl]sulfonylphenyl] pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[3,6-dihydro-2H-pyran-4-yl(difluoro)methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(tetrahydropyran-3-yl)methyl]-2-pyridyl]piperazin- 1-yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(8-oxabicyclo[3.2.1]octan-3-yl)methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro(3-oxabicyclo[3.1.0]hexan-6-yl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[[(2S)-1,4-dioxan-2-yl]-difluoro-methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[[(2R)-1,4-dioxan-2-yl]-difluoro-methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-[(2S)-morpholin-2-yl]methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-[(2R)-morpholin-2-yl]methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro(1,2,3,6-tetrahydropyridin-4-yl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-[(3R)-3-piperidyl]methyl]-2-pyridyl]piperazin- 1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-[(3S)-3-piperidyl]methyl]-2-pyridyl]piperazin- 1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[4-[azetidin-3-yl(difluoro)methyl]-6-chloro-2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 3-[4-[4-[6-Chloro-4-[difluoro-[(2S)-morpholin-2-yl]methyl]-2-pyridyl]piperazin-1- yl]sulfonylphenyl] -5-(hydroxymethyl)oxazolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-(5-methylpyrazin-2-yl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-(6-methylpyrazin-2-yl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[(5-chloro-6-methyl-pyrazin-2-yl)-difluoro-methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-[cis-6-methylmorpholin-2-yl]methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; ((4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-(5-methylmorpholin-2-yl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[4-[[cis-3-azabicyclo[3.1.0]hexan-1-yl]-difluoro-methyl]-6-chloro-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[trans-difluoro-[2- (hydroxymethyl)cyclopropyl]methyl]-2-pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[4-[[trans-4-(aminomethyl)cyclohexyl]-difluoro-methyl]-6-chloro-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[trans-difluoro-(3-hydroxycyclobutyl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; and Trans-4-[[2-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-1-yl]phenyl]sulfonylpiperazin-1-yl]-6- chloro-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide; or a pharmaceutically acceptable salt thereof. 20. A process for the preparation of a compound according to any one of claims 1 to 19 comprising the following step: a) Coupling reaction between compound of formula (VII), with lactam or carbamate, R1H, in the presence of catalyst and base; wherein in step a), the catalyst is CuI, the base is Cs2CO3; R1, R2, R3, R4, Y, Q1, Q2, Q3, Q4 and Q5 are defined as in any one of claims 1 to 18. 21. A compound or pharmaceutically acceptable salt according to any one of claims 1 to 19 for use as therapeutically active substance. 22. A pharmaceutical composition comprising a compound in accordance with any one of claims 1 to 19 and a therapeutically inert carrier. 23. The use of a compound according to any one of claims 1 to 19 for the inhibition of LpxH. 24. The use of a compound according to any one of claims 1 to 19 for the treatment or prophylaxis of bacterial infection, particularly the bacteria is gram-negative bacteria. 25. The use of a compound according to any one of claims 1 to 19 for the preparation of a medicament for the treatment or prophylaxis of bacterial infection, particularly the bacteria is gram-negative bacteria.

26. The use of a compound according to claim 24 or 25, wherein the gram-negative bacteria is selected from Enterobacteriaceae, Neisseria gonorrhoeae, Haemophilus influenzae, Helicobacter pylorus, Acinetobacter baumannii and Pseudomonas aeruginosa. 27. The use of 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 19 for the treatment or prophylaxis of bacterial infection, particularly the bacteria is gram-negative bacteria. 29. A compound according to claim 28, wherein the gram-negative bacteria is selected from Enterobacteriaceae, Neisseria gonorrhoeae, Haemophilus influenzae, Helicobacter pylorus, Acinetobacter baumannii and Pseudomonas aeruginosa. 30. A compound according to claim 29, wherein the gram-negative bacteria is Enterobacteriaceae, wherein Enterobacteriaceae is Klebsiella pneumoniae or Escherichia coli. 31. A compound or pharmaceutically acceptable salt according to any one of claims 1 to 19, when manufactured according to a process of claim 20. 32. 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 19.

Description:
Piperazinylsulfonylaryl 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., Enterobacteriaceae 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 Enterobacteriaceae 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 Lipopolysaccha- ride (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 2-oxo-1-oxa-3,8-diazaspiro[4.5]decanyl unsubstituted or substituted by aminoC 1-6 alkyl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridinyl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[5,4-c]pyridinyl, 2-oxo-3a,4,6,7,8,8a-hexahydrooxazolo[4,5-c]azepinyl substituted by aminoC 1-6 alkyl or C1- 6alkylaminoC 1-6 alkyl, 2-oxo-3a,5,6,6a-tetrahydrofuro[2,3-d]oxazolyl substituted twice by substituents independently selected from hydroxy and hydroxyC 1-6 alkyl, 2-oxo-4,5,6,6a-tetrahydro-3aH-pyrrolo[3,4-d]oxazolyl, 2-oxo-4,5,6,7,8,8a-hexahydro-3aH-oxazolo[4,5-c]azepinyl, 2-oxo-5,6,7,7a-tetrahydro-3aH-pyrano[2,3-d]oxazolyl substituted three times by substituents independently selected from hydroxy and hydroxyC 1-6 alkyl, 3-oxo-2,6,9-triazaspiro[4.5]decanyl, 3-oxo-2,6-diazaspiro[4.5]decanyl, 3-oxo-2,8-diazaspiro[4.5]decanyl, 5-oxo-1,2,3,3a,6,6a-hexahydropyrrolo[3,2-b]pyrrolyl, 6-oxo-5-oxa-2,7-diazaspiro[3.4]octanyl, 7-oxo-2,6-diazaspiro[3.4]octanyl, oxoazetidinyl substituted by amino, oxooxazolidinyl substituted once or twice by substituents independently selected from ((aminoC 1-6 alkyl)azetidinyl)C 1-6 alkyl, (C 1-6 alkyl)2aminoC 1-6 alkyl, (dihydroxypyrrolidinyl)C 1-6 alkyl, (pyrrolidinylamino)C 1-6 alkyl, 2,6- diazaspiro[3.3]heptanylC 1-6 alkyl, dihydroxy(C 1-6 alkoxy)tetrahydrofuranyl, aminoazetidinylC 1-6 alkyl, aminoC 1-6 alkyl, C 1-6 alkyl, C 1-6 alkylaminoC 1-6 alkyl, C1- 6alkylcarbonylaminoC 1-6 alkyl, hydroxyC 1-6 alkyl and piperazinylC 1-6 alkyl, or oxopyrrolidinyl substituted once, twice or three times by substituents independently selected from (((C 1-6 alkyl)2aminoC 1-6 alkyl)azetidinyl)C 1-6 alkyl, ((aminoC1- 6alkyl)azetidinyl)C 1-6 alkyl, ((aminoC 3-7 cycloalkyl)azetidinyl)C 1-6 alkyl, ((C 1- 6 alkylamino)azetidinyl)C 1-6 alkyl, (aminoazetidinyl)C 1-6 alkyl, (aminoC 1- 6alkyl)azetidinyl, (azetidinylC 1-6 alkylamino)C 1-6 alkyl, (C 1-6 alkylpiperazinyl)C 1-6 alkyl, (hydroxyC 1-6 alkyl)piperazinyl, 2,5-diazabicyclo[2.2.1]heptanyl, 2,6- diazaspiro[3.3]heptanyl, amino, amino(hydroxy)piperidinyl, amino(hydroxy)pyrrolidinyl, aminoazetidinyl, aminoC 1-6 alkyl, (aminoC1- 6alkyl)amino, aminopyrrolidinyl, azetidinylamino, C 1-6 alkyl, C 1-6 alkylamino, dihydroxypyrrolidinyl, (hydroxyC 1-6 alkyl)amino, piperazinyl, piperazinylC 1-6 alkyl, piperidinylamino and pyrrolidinylamino; R 2 is H or cyano; R 3 is benzyl, C 3-7 cycloalkylsulfonyl, cyanoC3-7cycloalkyl, phenoxy, phenylC 3-7 cycloalkyl, tetrahydropyranylsulfonyl, or C 1-6 alkyl substituted twice or three times by substituents independently selected from halogen, (aminoC 1-6 alkyl)C 3-7 cycloalkyl, (hydroxyC 1-6 alkyl)C 3-7 cycloalkyl, 1,2,3,6- tetrahydropyridinyl, 1,4-dioxanyl, 3,6-dihydro-2H-pyranyl, 3- azabicyclo[3.1.0]hexanyl, 3-oxabicyclo[3.1.0]hexanyl, 8-oxabicyclo[3.2.1]octanyl, azetidinyl, C 1-6 alkylhalopyrazinyl, C 1-6 alkylmorpholinyl, C 1-6 alkylpyrazinyl, carbamoylC 3-7 cycloalkyl, hydroxyC 3-7 cycloalkyl, morpholinyl, oxetanyl, piperidinyl, pyrazinyl, pyridinyl, pyrimidinyl, tetrahydrofuranyl, tetrahydropyranyl, phenyl and pyridinyl; R 4 is halogen, C 1-6 alkyl, C 3-7 cycloalkyl, C 1-6 alkoxy or C 2-6 alkynyl; Q 1 is N or CR a , wherein R a is H or halogen; Q 2 is N or CR b , wherein R b is H or halogen; Q 3 is N or CH; Q 4 is N or CH; Q 5 is N or CH; Y is O or NH; 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, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl and the like. Particular “C 1-6 alkyl” groups are methyl, ethyl and n-propyl. The term “halogen” and “halo” are used interchangeably herein and denote fluoro, chloro, bromo, or iodo. The term “halopyrazinyl” denotes a pyrazinyl group wherein at least one of the hydrogen atoms of the pyrazinyl group has been replaced by same or different halogen atoms, particularly fluoro atoms. Examples of halopiperidinyl include fluoropyrazinyl and difluoropyrazinyl. The term “C2-6alkynyl” denotes a monovalent linear or branched saturated hydrocarbon group of 2 to 6 carbon atoms comprising one, two or three triple bonds. In particular embodiments alkynyl has from 2 to 4 carbon atoms comprising one or two triple bonds. Examples of C2-6alkynyl include ethynyl, propynyl, prop-2-ynyl, isopropynyl, and n-butynyl. 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 “C3-7cycloalkyl” denotes a monovalent saturated monocyclic or bicyclic hydrocarbon group of 3 to 7 ring carbon atoms. Bicyclic means consisting of two saturated carbocycles having one or more carbon atoms in common. Examples for monocyclic cycloalkyl are cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cycloheptyl. Examples for bicyclic cycloalkyl are bicyclo[1.1.0]butyl, bicyclo[2.2.1]heptanyl, or bicyclo[2.2.2]octanyl. The term “oxo” denotes a divalent oxygen atom =O. The term “cis-isomers” and “trans-isomers” denote the relative stereochemistry of the molecule or moiety. For example: tert-butyl cis-1-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]-3- azabicyclo[3.1.0]hexane-3-carboxylate (compound 116a) ( ) in Example 116 as the “cis-isomers” refers to a mixture of and ; similarly, trans-ethyl 2-[(2,6-dichloro-4-pyridyl)-difluoro- methyl]cyclopropanecarboxylate ( compound 117c) ( ) in Example 117 as the “trans-isomers” refers to a mixture of . The way of showing relative stereochemistry also applies to the final compound. 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, N-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 2-oxo-1-oxa-3,8-diazaspiro[4.5]decanyl unsubstituted or substituted by aminoC 1-6 alkyl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridinyl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[5,4-c]pyridinyl, 2-oxo-3a,4,6,7,8,8a-hexahydrooxazolo[4,5-c]azepinyl substituted by aminoC 1-6 alkyl or C 1- 6alkylaminoC 1-6 alkyl, 2-oxo-3a,5,6,6a-tetrahydrofuro[2,3-d]oxazolyl substituted twice by substituents independently selected from hydroxy and hydroxyC 1-6 alkyl, 2-oxo-4,5,6,6a-tetrahydro-3aH-pyrrolo[3,4-d]oxazolyl, 2-oxo-4,5,6,7,8,8a-hexahydro-3aH-oxazolo[4,5-c]azepinyl, 2-oxo-5,6,7,7a-tetrahydro-3aH-pyrano[2,3-d]oxazolyl substituted three times by substituents independently selected from hydroxy and hydroxyC 1-6 alkyl, 3-oxo-2,6,9-triazaspiro[4.5]decanyl, 3-oxo-2,6-diazaspiro[4.5]decanyl, 3-oxo-2,8-diazaspiro[4.5]decanyl, 5-oxo-1,2,3,3a,6,6a-hexahydropyrrolo[3,2-b]pyrrolyl, 6-oxo-5-oxa-2,7-diazaspiro[3.4]octanyl, 7-oxo-2,6-diazaspiro[3.4]octanyl, oxoazetidinyl substituted by amino, oxooxazolidinyl substituted once or twice by substituents independently selected from ((aminoC 1-6 alkyl)azetidinyl)C 1-6 alkyl, (C 1-6 alkyl) 2 aminoC 1-6 alkyl, (dihydroxypyrrolidinyl)C 1-6 alkyl, (pyrrolidinylamino)C 1-6 alkyl, 2,6- diazaspiro[3.3]heptanylC 1-6 alkyl, dihydroxy(C 1-6 alkoxy)tetrahydrofuranyl, aminoazetidinylC 1-6 alkyl, aminoC 1-6 alkyl, C 1-6 alkyl, C 1-6 alkylaminoC 1-6 alkyl, C 1- 6alkylcarbonylaminoC 1-6 alkyl, hydroxyC 1-6 alkyl and piperazinylC 1-6 alkyl, or oxopyrrolidinyl substituted once, twice or three times by substituents independently selected from (((C 1-6 alkyl) 2 aminoC 1-6 alkyl)azetidinyl)C 1-6 alkyl, ((aminoC 1- 6 alkyl)azetidinyl)C 1-6 alkyl, ((aminoC 3-7 cycloalkyl)azetidinyl)C 1-6 alkyl, ((C 1- 6alkylamino)azetidinyl)C 1-6 alkyl, (aminoazetidinyl)C 1-6 alkyl, (aminoC1- 6alkyl)azetidinyl, (azetidinylC 1-6 alkylamino)C 1-6 alkyl, (C 1-6 alkylpiperazinyl)C 1-6 alkyl, (hydroxyC 1-6 alkyl)piperazinyl, 2,5-diazabicyclo[2.2.1]heptanyl, 2,6- diazaspiro[3.3]heptanyl, amino, amino(hydroxy)piperidinyl, amino(hydroxy)pyrrolidinyl, aminoazetidinyl, aminoC 1-6 alkyl, (aminoC1- 6alkyl)amino, aminopyrrolidinyl, azetidinylamino, C 1-6 alkyl, C 1-6 alkylamino, dihydroxypyrrolidinyl, (hydroxyC 1-6 alkyl)amino, piperazinyl, piperazinylC 1-6 alkyl, piperidinylamino and pyrrolidinylamino; R 2 is H or cyano; R 3 is benzyl, C3-7cycloalkylsulfonyl, cyanoC3-7cycloalkyl, phenoxy, phenylC3-7cycloalkyl, tetrahydropyranylsulfonyl, or C 1-6 alkyl substituted twice or three times by substituents independently selected from halogen, (aminoC 1-6 alkyl)C3-7cycloalkyl, (hydroxyC 1-6 alkyl)C3-7cycloalkyl, 1,2,3,6- tetrahydropyridinyl, 1,4-dioxanyl, 3,6-dihydro-2H-pyranyl, 3- azabicyclo[3.1.0]hexanyl, 3-oxabicyclo[3.1.0]hexanyl, 8-oxabicyclo[3.2.1]octanyl, azetidinyl, C 1-6 alkylhalopyrazinyl, C 1-6 alkylmorpholinyl, C 1-6 alkylpyrazinyl, carbamoylC3-7cycloalkyl, hydroxyC3-7cycloalkyl, morpholinyl, oxetanyl, piperidinyl, pyrazinyl, pyridinyl, pyrimidinyl, tetrahydrofuranyl, tetrahydropyranyl, phenyl and pyridinyl; R 4 is halogen, C 1-6 alkyl, C3-7cycloalkyl, C 1-6 alkoxy or C2-6alkynyl; Q 1 is N or CR a , wherein R a is H or halogen; Q 2 is N or CR b , wherein R b is H or halogen; Q 3 is N or CH; Q 4 is N or CH; Q 5 is N or CH; Y is O or NH; or a pharmaceutically acceptable salt thereof. Another embodiment of present invention is (ii) a compound of formula (Ia), (Ia), wherein R 1 is 2-oxo-1-oxa-3,8-diazaspiro[4.5]decanyl unsubstituted or substituted by aminoC 1-6 alkyl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridinyl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[5,4-c]pyridinyl, 2-oxo-3a,4,6,7,8,8a-hexahydrooxazolo[4,5-c]azepinyl substituted by aminoC 1-6 alkyl or C1- 6alkylaminoC 1-6 alkyl, 2-oxo-3a,5,6,6a-tetrahydrofuro[2,3-d]oxazolyl substituted twice by substituents independently selected from hydroxy and hydroxyC 1-6 alkyl, 2-oxo-4,5,6,6a-tetrahydro-3aH-pyrrolo[3,4-d]oxazolyl, 2-oxo-4,5,6,7,8,8a-hexahydro-3aH-oxazolo[4,5-c]azepinyl, 2-oxo-5,6,7,7a-tetrahydro-3aH-pyrano[2,3-d]oxazolyl substituted three times by substituents independently selected from hydroxy and hydroxyC 1-6 alkyl, 3-oxo-2,6,9-triazaspiro[4.5]decanyl, 3-oxo-2,6-diazaspiro[4.5]decanyl, 3-oxo-2,8-diazaspiro[4.5]decanyl, 5-oxo-1,2,3,3a,6,6a-hexahydropyrrolo[3,2-b]pyrrolyl, 6-oxo-5-oxa-2,7-diazaspiro[3.4]octanyl, 7-oxo-2,6-diazaspiro[3.4]octanyl, oxoazetidinyl substituted by amino, oxooxazolidinyl substituted once or twice by substituents independently selected from ((aminoC 1-6 alkyl)azetidinyl)C 1-6 alkyl, (C 1-6 alkyl)2aminoC 1-6 alkyl, (dihydroxypyrrolidinyl)C 1-6 alkyl, (pyrrolidinylamino)C 1-6 alkyl, 2,6- diazaspiro[3.3]heptanylC 1-6 alkyl, dihydroxy(C 1-6 alkoxy)tetrahydrofuranyl, aminoazetidinylC 1-6 alkyl, aminoC 1-6 alkyl, C 1-6 alkyl, C 1-6 alkylaminoC 1-6 alkyl, C1- 6alkylcarbonylaminoC 1-6 alkyl, hydroxyC 1-6 alkyl and piperazinylC 1-6 alkyl, or oxopyrrolidinyl substituted once, twice or three times by substituents independently selected from (((C 1-6 alkyl)2aminoC 1-6 alkyl)azetidinyl)C 1-6 alkyl, ((aminoC1- 6alkyl)azetidinyl)C 1-6 alkyl, ((aminoC 3-7 cycloalkyl)azetidinyl)C 1-6 alkyl, ((C 1- 6 alkylamino)azetidinyl)C 1-6 alkyl, (aminoazetidinyl)C 1-6 alkyl, (aminoC 1- 6alkyl)azetidinyl, (azetidinylC 1-6 alkylamino)C 1-6 alkyl, (C 1-6 alkylpiperazinyl)C 1-6 alkyl, (hydroxyC 1-6 alkyl)piperazinyl, 2,5-diazabicyclo[2.2.1]heptanyl, 2,6- diazaspiro[3.3]heptanyl, amino, amino(hydroxy)piperidinyl, amino(hydroxy)pyrrolidinyl, aminoazetidinyl, aminoC 1-6 alkyl, (aminoC1- 6alkyl)amino, aminopyrrolidinyl, azetidinylamino, C 1-6 alkyl, C 1-6 alkylamino, dihydroxypyrrolidinyl, (hydroxyC 1-6 alkyl)amino, piperazinyl, piperazinylC 1-6 alkyl, piperidinylamino and pyrrolidinylamino; R 2 is H or cyano; R 3 is benzyl, C 3-7 cycloalkylsulfonyl, cyanoC3-7cycloalkyl, phenoxy, phenylC 3-7 cycloalkyl, tetrahydropyranylsulfonyl, or C 1-6 alkyl substituted twice or three times by substituents independently selected from halogen, (aminoC 1-6 alkyl)C 3-7 cycloalkyl, (hydroxyC 1-6 alkyl)C 3-7 cycloalkyl, 1,2,3,6- tetrahydropyridinyl, 1,4-dioxanyl, 3,6-dihydro-2H-pyranyl, 3- azabicyclo[3.1.0]hexanyl, 3-oxabicyclo[3.1.0]hexanyl, 8-oxabicyclo[3.2.1]octanyl, azetidinyl, C 1-6 alkylhalopyrazinyl, C 1-6 alkylmorpholinyl, C 1-6 alkylpyrazinyl, carbamoylC 3-7 cycloalkyl, hydroxyC 3-7 cycloalkyl, morpholinyl, oxetanyl, piperidinyl, pyrazinyl, pyridinyl, pyrimidinyl, tetrahydrofuranyl, tetrahydropyranyl, phenyl and pyridinyl; R 4 is halogen, C 1-6 alkyl, C 3-7 cycloalkyl, C 1-6 alkoxy or C 2-6 alkynyl; Q 1 is N or CR a , wherein R a is H or halogen; Q 2 is CR b , wherein R b is H or halogen; Q 3 is N; Q 4 is N or CH; Q 5 is N or CH; Y is O; or a pharmaceutically acceptable salt thereof. A further embodiment of present invention is (iii) a compound of formula (I) or (Ia) according to (i) or (ii), or a pharmaceutically acceptable salt thereof, wherein R 1 is 2-oxo-1-oxa-3,8-diazaspiro[4.5]decanyl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridinyl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[5,4-c]pyridinyl, 2-oxo-3a,5,6,6a-tetrahydrofuro[2,3-d]oxazolyl substituted twice by substituents independently selected from hydroxy and hydroxyC 1-6 alkyl, 2-oxo-4,5,6,6a-tetrahydro-3aH-pyrrolo[3,4-d]oxazolyl, 2-oxo-4,5,6,7,8,8a-hexahydro-3aH-oxazolo[4,5-c]azepinyl, 3-oxo-2,6-diazaspiro[4.5]decanyl, 5-oxo-1,2,3,3a,6,6a-hexahydropyrrolo[3,2-b]pyrrolyl, 6-oxo-5-oxa-2,7-diazaspiro[3.4]octanyl, oxooxazolidinyl substituted once or twice by substituents independently selected from ((aminoC 1-6 alkyl)azetidinyl)C 1-6 alkyl, (C 1-6 alkyl)2aminoC 1-6 alkyl, dihydroxy(C1- 6alkoxy)tetrahydrofuranyl, aminoC 1-6 alkyl, C 1-6 alkyl, C 1-6 alkylaminoC 1-6 alkyl, hydroxyC 1-6 alkyl and piperazinylC 1-6 alkyl, or oxopyrrolidinyl substituted by substituent selected from ((aminoC 1-6 alkyl)azetidinyl)C1- 6alkyl, ((aminoC3-7cycloalkyl)azetidinyl)C 1-6 alkyl, (aminoazetidinyl)C 1-6 alkyl, (C1- 6alkylpiperazinyl)C 1-6 alkyl, (hydroxyC 1-6 alkyl)piperazinyl, amino, amino(hydroxy)piperidinyl, amino(hydroxy)pyrrolidinyl, aminoC 1-6 alkyl, aminopyrrolidinyl, C 1-6 alkylamino, dihydroxypyrrolidinyl, (hydroxyC 1-6 alkyl)amino, piperazinyl, piperazinylC 1-6 alkyl and piperidinylamino. A further embodiment of present invention is (iv) a compound of formula (I) or (Ia), according to any one of (i) to (iii), or a pharmaceutically acceptable salt thereof, wherein R 1 is 2-oxo-1-oxa-3,8-diazaspiro[4.5]decanyl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridinyl, 2-oxo-3a,4,5,6,7,7a-hexahydrooxazolo[5,4-c]pyridinyl, 2-oxo-3a,5,6,6a-tetrahydrofuro[2,3-d]oxazolyl substituted twice by substituents independently selected from hydroxy and hydroxymethyl, 2-oxo-4,5,6,6a-tetrahydro-3aH-pyrrolo[3,4-d]oxazolyl, 2-oxo-4,5,6,7,8,8a-hexahydro-3aH-oxazolo[4,5-c]azepinyl, 3-oxo-2,6-diazaspiro[4.5]decanyl, 5-oxo-1,2,3,3a,6,6a-hexahydropyrrolo[3,2-b]pyrrolyl, 6-oxo-5-oxa-2,7-diazaspiro[3.4]octanyl, oxooxazolidinyl substituted once or twice by substituents independently selected from (3- (aminomethyl)azetidin-1-yl)methyl, (dimethylamino)methyl, 3,4-dihydroxy-5- methoxy-tetrahydrofuran-2-yl, aminomethyl, aminomethyl, hydroxymethyl, methyl, (methylamino)methyl and piperazin-1-ylmethyl, or oxopyrrolidinyl substituted by substituent selected from (3-(1-aminocyclopropyl)azetidin- 1-yl)methyl, (3-(aminomethyl)azetidin-1-yl)methyl, (3-aminoazetidin-1-yl)methyl, (4-methylpiperazin-1-yl)methyl, 3-(hydroxymethyl)piperazin-1-yl, 3,4- dihydroxypyrrolidin-1-yl, 3-amino-4-hydroxypiperidin-1-yl, 3-amino-4-hydroxy- pyrrolidin-1-yl, 3-aminopyrrolidin-1-yl, 4-piperidinylamino, amino, aminomethyl, (hydroxyethyl)amino, methylamino, piperazin-1-yl and piperazin-1-ylmethyl. A further embodiment of present invention is (v) a compound of formula (I) or (Ia) according to any one of (i) to (iv), wherein R 1 is 2-oxo-1-oxa-3,8-diazaspiro[4.5]decan-3-yl, 2- oxo-3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridin-3-yl, 2-oxo-3a,4,5,6,7,7a- hexahydrooxazolo[5,4-c]pyridin-1-yl, 2-oxo-4,5,6,6a-tetrahydro-3aH-pyrrolo[3,4-d]oxazol-3-yl, 2-oxo-4,5,6,7,8,8a-hexahydro-3aH-oxazolo[4,5-c]azepin-3-yl, 3-oxo-2,6-diazaspiro[4.5]decan-2- yl, 4-((3-(1-aminocyclopropyl)azetidin-1-yl)methyl)-2-oxo-pyrrol idin-1-yl, 4-((3-aminoazetidin- 1-yl)methyl)-2-oxo-pyrrolidin-1-yl, 4-((4-methylpiperazin-1-yl)methyl)-2-oxo-pyrrolidin-1-yl, 4-(2-hydroxyethylamino)-2-oxo-pyrrolidin-1-yl, 4-(3-(aminomethyl)azetidin-1-yl)methyl)-2- oxo-pyrrolidin-1-yl, 4-(3-(hydroxymethyl)piperazin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(3,4- dihydroxypyrrolidin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(3-amino-4-hydroxypiperidin-1-yl)-2-oxo- pyrrolidin-1-yl, 4-(3-amino-4-hydroxy-pyrrolidin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(3- aminopyrrolidin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(4-piperidinylamino)-2-oxo-pyrrolidin-1-yl, 4- (methylamino)-2-oxo-pyrrolidin-1-yl, 4-(piperazin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(piperazin-1- ylmethyl)-2-oxo-pyrrolidin-1-yl, 4-amino-2-oxo-pyrrolidin-1-yl, 4-aminomethyl-2-oxo- oxazolidin-3-yl, 4-aminomethyl-2-oxo-pyrrolidin-1-yl, 4-methyl-2-oxo-5-(3,4-dihydroxy-5- methoxy-tetrahydrofuran-2-yl)oxazolidin-3-yl, 5-((3-(aminomethyl)azetidin-1-yl)methyl)-2-oxo- oxazolidin-3-yl, 5-((dimethylamino)methyl)-2-oxo-oxazolidin-3-yl, 5-(2-aminoethyl)-2-oxo- oxazolidin-3-yl, 5-(methylaminomethyl)-2-oxo-oxazolidin-3-yl, 5-(piperazin-1-ylmethyl)-2-oxo- oxazolidin-3-yl, 5-aminomethyl-2-oxo-oxazolidin-3-yl, 5-hydroxymethyl-2-oxo-oxazolidin-3-yl, 5-oxo-1,2,3,3a,6,6a-hexahydropyrrolo[3,2-b]pyrrol-4-yl, 6-hydroxy-5-hydroxymethyl-2-oxo- 3a,5,6,6a-tetrahydrofuro[2,3-d]oxazol-3-yl or 6-oxo-5-oxa-2,7-diazaspiro[3.4]octan-7-yl. A further embodiment of present invention is (vi) a compound of formula (I) or (Ia), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (v), wherein R 1 is oxopyrrolidinyl substituted by substituent selected from ((aminoC 1-6 alkyl)azetidinyl)C 1-6 alkyl, ((aminoC 3-7 cycloalkyl)azetidinyl)C 1-6 alkyl, (aminoazetidinyl)C 1-6 alkyl, (C 1-6 alkylpiperazinyl)C 1- 6alkyl, (hydroxyC 1-6 alkyl)piperazinyl, amino, amino(hydroxy)piperidinyl, amino(hydroxy)pyrrolidinyl, aminoC 1-6 alkyl, aminopyrrolidinyl, C 1-6 alkylamino, dihydroxypyrrolidinyl, (hydroxyC 1-6 alkyl)amino, piperazinyl, piperazinylC 1-6 alkyl and piperidinylamino. A further embodiment of present invention is (vii) a compound of formula (I) or (Ia), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (vi), wherein R 1 is 4-((3- (1-aminocyclopropyl)azetidin-1-yl)methyl)-2-oxo-pyrrolidin-1 -yl, 4-((3-aminoazetidin-1- yl)methyl)-2-oxo-pyrrolidin-1-yl, 4-((4-methylpiperazin-1-yl)methyl)-2-oxo-pyrrolidin-1-yl, 4- (2-hydroxyethylamino)-2-oxo-pyrrolidin-1-yl, 4-(3-(aminomethyl)azetidin-1-yl)methyl)-2-oxo- pyrrolidin-1-yl, 4-(3-(hydroxymethyl)piperazin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(3,4- dihydroxypyrrolidin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(3-amino-4-hydroxypiperidin-1-yl)-2-oxo- pyrrolidin-1-yl, 4-(3-amino-4-hydroxy-pyrrolidin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(3- aminopyrrolidin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(4-piperidinylamino)-2-oxo-pyrrolidin-1-yl, 4- (methylamino)-2-oxo-pyrrolidin-1-yl, 4-(piperazin-1-yl)-2-oxo-pyrrolidin-1-yl, 4-(piperazin-1- ylmethyl)-2-oxo-pyrrolidin-1-yl, 4-amino-2-oxo-pyrrolidin-1-yl or 4-aminomethyl-2-oxo- pyrrolidin-1-yl. A further embodiment of present invention is (viii) a compound of formula (I) or (Ia), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (vii), wherein R 3 is benzyl, phenylC 3-7 cycloalkyl, or C 1-6 alkyl substituted three times by substituents independently selected from halogen, (hydroxyC 1-6 alkyl)C 3-7 cycloalkyl, 1,2,3,6-tetrahydropyridinyl, 1,4- dioxanyl, 3,6-dihydro-2H-pyranyl, 3-azabicyclo[3.1.0]hexanyl, 3-oxabicyclo[3.1.0]hexanyl, 8- oxabicyclo[3.2.1]octanyl, C 1-6 alkylhalopyrazinyl, C 1-6 alkylpyrazinyl, carbamoylC 3-7 cycloalkyl, hydroxyC 3-7 cycloalkyl, morpholinyl, oxetanyl, pyrazinyl, pyridinyl, tetrahydropyranyl and phenyl. A further embodiment of present invention is (ix) a compound of formula (I) or (Ia), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (viii), wherein R 3 is C 1- 6 alkyl substituted three times by substituents independently selected from halogen, (hydroxyC 1- 6alkyl)C3-7cycloalkyl, 1,4-dioxanyl, 3-azabicyclo[3.1.0]hexanyl, 3-oxabicyclo[3.1.0]hexanyl, C1- 6 alkylhalopyrazinyl, C 1-6 alkylpyrazinyl, hydroxyC 3-7 cycloalkyl, morpholinyl, oxetanyl, phenyl, pyridinyl and tetrahydropyranyl. A further embodiment of present invention is (x) a compound of formula (I) or (Ia), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (ix), wherein R 3 is difluoro(1,4-dioxan-2-yl)methyl, difluoro(2-(hydroxymethyl)cyclopropyl)methyl, difluoro(2- pyridinyl)methyl, difluoro(3-hydroxycyclobutyl)methyl, difluoro(3-oxabicyclo[3.1.0]hexan-6- yl)methyl, difluoro(5-chloro-6-methyl-pyrazin-2-yl)methyl, difluoro(6-methylpyrazin-2- yl)methyl, difluoro(morpholin-2-yl)methyl, difluoro(oxetan-3-yl)methyl, difluoro(phenyl)methyl, difluoro(tetrahydropyran-4-yl)methyl or difluoro[3-azabicyclo[3.1.0]hexan-1-yl]methyl. A further embodiment of present invention is (xi) a compound of formula (I) or (Ia), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (x), wherein R 2 is H. A further embodiment of present invention is (xii) a compound of formula (I) or (Ia), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (xi), wherein R 4 is halogen or C 1-6 alkyl. A further embodiment of present invention is (xiii) a compound of formula (I) or (Ia), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (xii), wherein R 4 is chloro or methyl. A further embodiment of present invention is (xiv) a compound of formula (I) or (Ia), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (xiii), wherein Q 1 is CR a , wherein R a is H or halogen. A further embodiment of present invention is (xv) a compound of formula (I) or (Ia), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (xiv), wherein Q 1 is CR a , wherein R a is H or fluoro. A further embodiment of present invention is (xvi) a compound of formula (I) or (Ia), or a pharmaceutically acceptable salt thereof, according to any one of (i) to (xv), wherein Q 2 is CH. A further embodiment of present invention is (xvii) a compound of formula (I) or (Ia) according to any one of (i) to (xvi), wherein R 1 is oxopyrrolidinyl substituted by amino or aminoC 1-6 alkyl; R 2 is H; R 3 is C 1-6 alkyl substituted three times by substituents independently selected from halogen, (hydroxyC 1-6 alkyl)C 3-7 cycloalkyl, 1,4-dioxanyl, 3-azabicyclo[3.1.0]hexanyl, 3- oxabicyclo[3.1.0]hexanyl, C 1-6 alkylhalopyrazinyl, C 1-6 alkylpyrazinyl, hydroxyC3- 7cycloalkyl, morpholinyl, oxetanyl, phenyl, pyridinyl and tetrahydropyranyl; R 4 is halogen or C 1-6 alkyl; Q 1 is CR a , wherein R a is H or halogen; Q 2 is CH; Q 3 is N; Q 4 is N or CH; Q 5 is N or CH; Y is O; or a pharmaceutically acceptable salt thereof. A further embodiment of present invention is (xviii) a compound of formula (I) or (Ia) according to any one of (i) to (xvii), wherein R 1 is oxopyrrolidinyl substituted by amino or aminomethyl; R 2 is H; R 3 is difluoro(1,4-dioxan-2-yl)methyl, difluoro(2-(hydroxymethyl)cyclopropyl)methyl, difluoro(2-pyridinyl)methyl, difluoro(3-hydroxycyclobutyl)methyl, difluoro(3- oxabicyclo[3.1.0]hexan-6-yl)methyl, difluoro(5-chloro-6-methyl-pyrazin-2-yl)methyl, difluoro(6-methylpyrazin-2-yl)methyl, difluoro(morpholin-2-yl)methyl, difluoro(oxetan-3- yl)methyl, difluoro(phenyl)methyl, difluoro(tetrahydropyran-4-yl)methyl or difluoro[3- azabicyclo[3.1.0]hexan-1-yl]methyl; R 4 is chloro or methyl; Q 1 is CR a , wherein R a is H or fluoro; Q 2 is CH; Q 3 is N; Q 4 is N or CH; Q 5 is N or CH; Y is O; or a pharmaceutically acceptable salt thereof. Another embodiment of present invention is a compound of formula (I) or (Ia) selected from the following: 4-Amino-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pipe razin-1-yl]sulfonylphenyl] pyrrolidin-2-one; 4-(Aminomethyl)-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyri dyl]piperazin-1-yl]sulfonyl- phenyl]pyrrolidin-2-one; 5-(Aminomethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyri dyl]piperazin-1- yl]sulfonylphenyl]oxazolidin-2-one; N-[[3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazi n-1-yl]sulfonylphenyl]-2-oxo- oxazolidin-5-yl]methyl]acetamide; 4-Amino-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pipe razin-1-yl]sulfonylphenyl]- 3,3-dimethyl-pyrrolidin-2-one; 4-(Aminomethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyri dyl]piperazin-1- yl]sulfonylphenyl]oxazolidin-2-one; (3S)-3-amino-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl ]piperazin-1- yl]sulfonylphenyl] azetidin-2-one; 3-Amino-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pipe razin-1-yl]sulfonylphenyl] pyrrolidin-2-one; 5-(2-Aminoethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyr idyl]piperazin-1- yl]sulfonylphenyl]oxazolidin-2-one; 2-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-2,6- diazaspiro[4.5]decan-3-one; 2-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-2,8- diazaspiro[4.5]decan-3-one; 6-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-2,6- diazaspiro[3.4]octan-7-one; 7-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-5-oxa-2,7- diazaspiro[3.4]octan-6-one; 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-1-oxa-3,8- diazaspiro[4.5]decan-2-one; (3aR,6aS)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pi perazin-1-yl]sulfonylphenyl]- 4,5,6,6a-tetrahydro-3aH-pyrrolo[3,4-d]oxazol-2-one; (3aR,7aS)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pi perazin-1-yl]sulfonylphenyl]- 3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridin-2-one; (3aR,7aS)-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pi perazin-1-yl]sulfonylphenyl]- 3a,4,5,6,7,7a-hexahydrooxazolo[5,4-c]pyridin-2-one; (3aS,7aR)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pi perazin-1-yl]sulfonylphenyl]- 3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridin-2-one; (3aS,8aR)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pi perazin-1-yl]sulfonylphenyl]- 4,5,6,7,8,8a-hexahydro-3aH-oxazolo[4,5-c]azepin-2-one; 4-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]- 1,2,3,3a,6,6a-hexahydropyrrolo[3,2-b]pyrrol-5-one; 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-6-hydroxy- 5-(hydroxymethyl)-3a,5,6,6a-tetrahydrofuro[2,3-d]oxazol-2-on e; 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-5- [(3S,4R,5R)-3,4-dihydroxy-5-methoxy-tetrahydrofuran-2-yl]-4- methyl-oxazolidin-2-one; (3aS,5S,6S,7S,7aR)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-p yridyl]piperazin-1-yl]sulfonyl phenyl]-5,7-dihydroxy-6-(hydroxymethyl)-5,6,7,7a-tetrahydro- 3aH-pyrano[2,3-d]oxazol-2-one; 8-(2-Aminoethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyr idyl]piperazin-1- yl]sulfonylphenyl]-1-oxa-3,8-diazaspiro[4.5]decan-2-one; (3aS,8aR)-5-(2-aminoethyl)-3-[4-[4-[6-chloro-4-(trifluoromet hyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-3a,4,6,7,8,8a-hexahydrooxazolo[4,5-c]azep in-2-one; (3aS,8aR)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pi perazin-1-yl]sulfonylphenyl]- 5-[2-(methylamino)ethyl]-3a,4,6,7,8,8a-hexahydrooxazolo[4,5- c]azepin-2-one; 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-5- (methylaminomethyl)oxazolidin-2-one; 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-5- [(dimethylamino)methyl]oxazolidin-2-one; 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-5- (piperazin-1-ylmethyl)oxazolidin-2-one; 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-5-[[cis- 3,4-dihydroxypyrrolidin-1-yl]methyl]oxazolidin-2-one; 5-[(3-Aminoazetidin-1-yl)methyl]-3-[4-[4-[6-chloro-4-(triflu oromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]oxazolidin-2-one; 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-5-(2,6- diazaspiro[3.3]heptan-2-ylmethyl)oxazolidin-2-one; 5-[[3-(Aminomethyl)azetidin-1-yl]methyl]-3-[4-[4-[6-chloro-4 -(trifluoromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]oxazolidin-2-one; 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-5- [(pyrrolidin-3-ylamino)methyl]oxazolidin-2-one; 4-[[3-(Aminomethyl)azetidin-1-yl]methyl]-1-[4-[4-[6-chloro-4 -(trifluoromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-[(4- methylpiperazin-1-yl)methyl]pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-[[3- [(dimethylamino)methyl]azetidin-1-yl]methyl]pyrrolidin-2-one ; 4-[(Azetidin-3-ylmethylamino)methyl]-1-[4-[4-[6-chloro-4-(tr ifluoromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; 4-[[3-(1-Aminocyclopropyl)azetidin-1-yl]methyl]-1-[4-[4-[6-c hloro-4-(trifluoromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; 4-[(3-Aminoazetidin-1-yl)methyl]-1-[4-[4-[6-chloro-4-(triflu oromethyl)-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4- (piperazin-1-ylmethyl)pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-[[3- (dimethylamino)azetidin-1-yl]methyl]pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-[[3- (methylamino)azetidin-1-yl]methyl]pyrrolidin-2-one; 1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4- (methylamino)pyrrolidin-2-one; 4-(2-Aminoethylamino)-1-[4-[4-[6-chloro-4-(trifluoromethyl)- 2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-(2- hydroxyethylamino)pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4- piperazin-1-yl-pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4- [(3S,4R)-3,4-dihydroxypyrrolidin-1-yl]pyrrolidin-2-one; 4-[3-(Aminomethyl)azetidin-1-yl]-1-[4-[4-[6-chloro-4-(triflu oromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; 4-(3-Aminopyrrolidin-1-yl)-1-[4-[4-[6-chloro-4-(trifluoromet hyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-(3-Aminoazetidin-1-yl)-1-[4-[4-[6-chloro-4-(trifluoromethy l)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-(2,6- diazaspiro[3.3]heptan-2-yl)pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-[(3S)-3- (hydroxymethyl)piperazin-1-yl]pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-[(3R)-3- (hydroxymethyl)piperazin-1-yl]pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-[(1S,4S)- 2,5-diazabicyclo[2.2.1]heptan-2-yl]pyrrolidin-2-one; 4-[(3R,4R)-3-amino-4-hydroxy-pyrrolidin-1-yl]-1-[4-[4-[6-chl oro-4-(trifluoromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; 4-((3R,4R)-3-amino-4-hydroxypiperidin-1-yl)-1-(4-((4-(6-chlo ro-4- (trifluoromethyl)pyridin-2-yl)piperazin-1-yl)sulfonyl)phenyl )pyrrolidin-2-one; 4-(Azetidin-3-ylamino)-1-[4-[4-[6-chloro-4-(trifluoromethyl) -2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4- (pyrrolidin-3-ylamino)pyrrolidin-2-one; 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-(4- piperidylamino)pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pipe razin-1-yl]sulfonylphenyl]- 5-methyl-pyrrolidin-2-one; 4-Amino-1-[5-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pipe razin-1-yl]sulfonyl-2- pyridyl]pyrrolidin-2-one; 5-(Aminomethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyri dyl]piperazin-1-yl]sulfonyl- 2-fluoro-phenyl]oxazolidin-2-one; 5-(Aminomethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyri dyl]piperazin-1-yl]sulfonyl- 3-fluoro-phenyl]oxazolidin-2-one; 4-Amino-1-[4-[4-[4-methyl-6-(trifluoromethyl)pyrimidin-2-yl] piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[4-(trideuteriomethyl)-6-(trifluoromethyl)py rimidin-2-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 1-[4-(4-Amino-2-oxo-pyrrolidin-1-yl)phenyl]sulfonyl-4-[6-chl oro-4-(trifluoromethyl)-2- pyridyl]piperazine-2-carbonitrile; 4-Amino-1-[4-[4-[6-chloro-4-(difluoromethyl)-2-pyridyl]piper azin-1-yl]sulfonylphenyl] pyrrolidin-2-one; 4-Amino-1-[4-[4-(6-chloro-4-cyclopropylsulfonyl-2-pyridyl)pi perazin-1-yl]sulfonylphenyl] pyrrolidin-2-one; 4-Amino-1-[4-[4-(6-chloro-4-tetrahydropyran-4-ylsulfonyl-2-p yridyl)piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 1-[2-[4-[4-(4-Amino-2-oxo-pyrrolidin-1-yl)phenyl]sulfonylpip erazin-1-yl]-6-chloro-4- pyridyl]cyclopropanecarbonitrile; 4-Amino-1-[4-[4-(6-chloro-4-phenoxy-2-pyridyl)piperazin-1-yl ]sulfonylphenyl]pyrrolidin- 2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(phenyl)methyl]-2-pyrid yl]piperazin-1- yl]sulfonylphenyl] pyrrolidin-2-one; 4-Amino-1-[4-[4-(4-benzyl-6-chloro-2-pyridyl)piperazin-1-yl] sulfonylphenyl]pyrrolidin-2- one; 4-Amino-1-[4-[4-[6-chloro-4-(1-phenylcyclopropyl)-2-pyridyl] piperazin-1- yl]sulfonylphenyl] pyrrolidin-2-one; 4-Amino-1-[4-[4-[4-[difluoro(phenyl)methyl]-6-methyl-pyrimid in-2-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[2-chloro-6-[difluoro(phenyl)methyl]pyrimidi n-4-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[4-chloro-6-[difluoro(phenyl)methyl]pyrimidi n-2-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[2-cyclopropyl-6-[difluoro(phenyl)methyl]pyr imidin-4-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[4-cyclopropyl-6-[difluoro(phenyl)methyl]pyr imidin-2-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-[difluoro(phenyl)methyl]-2-methoxy-pyrimi din-4-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[4-[difluoro(phenyl)methyl]-6-methoxy-pyrimi din-2-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-Amino-1-[4-[4-[6-[difluoro(phenyl)methyl]-2-methyl-py rimidin-4-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-[difluoro(phenyl)methyl]-2-vinyl-pyrimidi n-4-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 2-[4-[4-[6-Chloro-4-[difluoro(phenyl)methyl]-2-pyridyl]piper azin-1-yl]sulfonylphenyl]- 2,6,9-triazaspiro[4.5]decan-3-one; 5-(Aminomethyl)-1-[4-[4-[6-chloro-4-[difluoro(phenyl)methyl] -2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; (3aR,7aS)-3-[4-[4-[6-chloro-4-[difluoro(phenyl)methyl]-2-pyr idyl]piperazin-1-yl]sulfonyl phenyl]-3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridin-2-one; 5-(Aminomethyl)-3-[4-[4-[6-chloro-4-[difluoro(phenyl)methyl] -2-pyridyl]piperazin-1- yl]sulfonylphenyl]oxazolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(2-pyridyl)methyl]-2-py ridyl]piperazin-1-yl] sulfonylphenyl] pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(3-pyridyl)methyl]-2-py ridyl]piperazin-1- yl]sulfonylphenyl] pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(4-pyridyl)methyl]-2-py ridyl]piperazin-1-yl] sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(pyrimidin-5-yl)methyl] -2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(pyrazin-2-yl)methyl]-2 -pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(pyrimidin-2-yl)methyl] -2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(tetrahydropyran-4-yl)m ethyl]-2-pyridyl]piperazin- 1-yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(tetrahydrofuran-3-yl)m ethyl]-2-pyridyl]piperazin- 1-yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(oxetan-3-yl)methyl]-2- pyridyl]piperazin-1- yl]sulfonylphenyl] pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[3,6-dihydro-2H-pyran-4-yl(diflu oro)methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(tetrahydropyran-3-yl)m ethyl]-2-pyridyl]piperazin- 1-yl]sulfonylphenyl]pyrrolidin-2-one; 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(8-oxabicyclo[3.2.1]oct an-3-yl)methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro(3-oxabicyclo[3.1. 0]hexan-6-yl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[[(2S)-1,4-dioxan-2-yl]-dif luoro-methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[[(2R)-1,4-dioxan-2-yl]-dif luoro-methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-[(2S)-morpholin-2 -yl]methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-[(2R)-morpholin-2 -yl]methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro(1,2,3,6-tetrahydr opyridin-4-yl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-[(3R)-3-piperidyl ]methyl]-2-pyridyl]piperazin- 1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-[(3S)-3-piperidyl ]methyl]-2-pyridyl]piperazin- 1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[4-[azetidin-3-yl(difluoro)methyl]-6-ch loro-2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one; 3-[4-[4-[6-Chloro-4-[difluoro-[(2S)-morpholin-2-yl]methyl]-2 -pyridyl]piperazin-1- yl]sulfonylphenyl] -5-(hydroxymethyl)oxazolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-(5-methylpyrazin- 2-yl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-(6-methylpyrazin- 2-yl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[(5-chloro-6-methyl-pyrazin -2-yl)-difluoro-methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-[cis-6-methylmorp holin-2-yl]methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; ((4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-(5-methylmorphol in-2-yl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[4-[[cis-3-azabicyclo[3.1.0]hexan-1-yl] -difluoro-methyl]-6-chloro-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[trans-difluoro-[2- (hydroxymethyl)cyclopropyl]methyl]-2-pyridyl]piperazin-1-yl] sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[4-[[trans-4-(aminomethyl)cyclohexyl]-d ifluoro-methyl]-6-chloro-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; (4R)-4-amino-1-[4-[4-[6-chloro-4-[trans-difluoro-(3-hydroxyc yclobutyl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one; and Trans-4-[[2-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-1-yl]phenyl ]sulfonylpiperazin-1-yl]-6- chloro-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide; or a pharmaceutically acceptable salt thereof. 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 distasonis, 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 examples. All substituents, in particular, R 1 , R 2 , R 3 , R 4 , Y, Q 1 , Q 2 , Q 3 , Q 4 and Q 5 are defined above. 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 organic chemistry. General synthetic routes for preparing the compound of formula (I) are shown below. Scheme 1

Wherein X 1 and X 2 are halogen, such as Cl, Br, I; PG is a protecting group such as tert- Boc or Cbz group. Step (i) could be a sulfonated reaction between halogenated phenyl sulfonyl chlorides (II) and protected piperazine (III) under in basic conditions (for example, in the presence of a suitable base such as trimethylamine) to provide compound of formula (IV). Compounds of formula (V) may be obtained in step (ii) by de-protection of compound of formula (IV) with a suitable acid such as trifluoroacetic acid or by a reducing agent, such as H2 with palladium. Step (iii) may be a nucleophilic substitution by reacting Intermediate (V) with halogenated compound of formula (VI) using a suitable base to provide compound of formula (VII). The final compound of formula (I) could be obtained in step (iv) through the coupling reaction of compound of formula (VII)with an cyclic-amide such as lactam or carbamate, R 1 H, using catalysts such as CuI in the presence of bases such as Cs 2 CO 3 . 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) Coupling reaction between compound of formula (VII), with lactam or carbamate, R 1 H, in the presence of catalyst and base; wherein in step a), the catalyst can be CuI, the base can be Cs 2 CO 3 ; 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: ACN acetonitrile aq. aqueous BAST bis(2-methoxyethyl)aminosulfur Trifluoride DAST diethylaminosulfur trifluoride DBU 1,8-diazabicyclo[5.4.0]undec-7-ene DCM dichloromethane DIPEA N,N-diisopropylethylamine DMAP 4-dimethylaminopyridine DMEDA 1,2-dimethylethylenediamine DMF dimethylformamide DMP Dess-Martin periodinane DMSO dimethyl sulfoxide EA ethyl acetate FA formic acid h(s) or hr(s) hour HATU 1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyri dinium 3- oxid hexafluorophosphate HPLC high performance liquid chromatography IPA isopropyl alcohol m-CPBA meta-chloroperoxybenzoic acid MIC minimum inhibitory concentration NMP N-methyl-2-pyrrolidone obsd. observed PE petroleum ether prep-HPLC preparative high performance liquid chromatography rt room temperature SFC supercritical fluid chromatography TEA triethylamine TFA trifluoroacetic acid TLC thin layer 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 Å, particle size: 40-60 µm; 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 XBridge TM Prep-C18 (5 µm, OBDTM 30 × 100 mm) column, SunFire TM Prep-C18 (5 µm, OBD TM 30 × 100 mm) column, Phenomenex Synergi-C18 (10 µm, 25 × 150 mm) or Phenomenex Gemini-C18 (10 µm, 25 × 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% HCl 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 × 250 mm), AS (10 µm, 30 × 250 mm) or AD (10 µm, 30 × 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 IPA) or CO 2 and MeOH (0.1% NH 3 ∙H 2 O in MeOH), back pressure 100bar, detection UV@ 254 or 220 nm. LC/MS spectra of compounds were obtained using a LC/MS (Waters TM 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 H2O; 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% NH3·H2O in H2O; B: acetonitrile; Neutral condition: A: H2O; 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 A 1-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]-4-(4-iodophenyl)s ulfonyl-piperazine The title compound was prepared according to the following procedures: Step 1: 1-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazine(interme diate A1) To a solution of piperazine (47.86 g, 555.58 mmol) in 1,4-dioxane (150 mL) were added 2,6-dichloro-4-(trifluoromethyl)pyridine (15.0 g, 69.45 mmol) slowly. The mixture was stirred at 60 °C for 1 h, then diluted with water (300 mL) and extracted with EA (200 mL) three times. The combined organic layer was washed with brine (300 mL) four times, dried over anhydrous Na 2 SO 4 , filtered and concentrated to give intermediate A1 (18 g, 67.76 mmol, 97.6% yield) as a white solid. MS obsd.: 265.8 (MH + ). Step 2: 1-[6-chloro-4-(trifluoromethyl)-2-pyridyl]-4-(4-iodophenyl)s ulfonyl-piperazine (intermediate A) To a solution of intermediate A1 (7.9 g, 29.75 mmol) and TEA (12.44 mL, 89.25 mmol) of in THF (100 mL) was added 4-iodobenzenesulfonyl chloride (9.0 g, 29.75 mmol) and the mixture was stirred at 20 °C for 12 h. The mixture was diluted with EA (400 mL) and filtered. The filtrate was washed with brine (100 mL) three times, dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was triturated with PE (150 mL) to give intermediate A (14.5 g, 27.27 mmol, 91.7% yield) as a white solid. MS obsd.: 531.8 (MH + ). Intermediate B 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-2H-pyrrol- 5-one The title compound was prepared according to the following procedures: Step 1: 1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4- hydroxy-pyrrolidin-2-one (intermediate B1) To a solution of intermediate A (1.0 g, 1.88 mmol) in DMF (10 mL) were added 4- hydroxy-2-pyrrolidone (209.15 mg, 2.07 mmol), Cs 2 CO 3 (1.23 g, 3.76 mmol), DMEDA (33.16 mg, 0.380 mmol), CuI (0.01 mL, 0.380 mmol) in one portion. The reaction was stirred at 70 °C for 2 hrs under N 2 . The mixture was diluted with water (50 mL), extracted with EA (30 mL) three times. The combined organic layer was washed with brine (50 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by column chromatography on silica gel (PE: EA=10:1 to 0:1) and prep-HPLC to give intermediate B1 (320 mg, 0.630 mmol, 33.7% yield) as a white solid. Step 2: 1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-2H- pyrrol-5-one (intermediate B) To a solution of intermediate B1 (320.0 mg, 0.630 mmol) in DCM (4 mL) was added TEA (0.18 mL, 1.27 mmol) in one portion. After cooled to 0°C, methanesulfonyl chloride (0.07 mL, 0.950 mmol) was added and then the reaction was warmed to 20 °C for 1 h. The mixture was diluted with DCM (20 mL), washed with water (20 mL) and brine (20 mL). The organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated to give intermediate B (290 mg, 0.60 mmol, 93.98% yield) was obtained as yellow oil. Intermediate C Tert-butyl N-[5-oxo-1-(4-piperazin-1-ylsulfonylphenyl)pyrrolidin-3-yl]c arbamate The title compound was prepared according to the following procedures: Step 1: benzyl 4-(4-iodophenyl) sulfonylpiperazine-1-carboxylate (intermediate C1) 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 25 °C for 2 hrs.. The mixture was concentrated to give the residue which was triturated with petroleum ether and washed by water to give intermediate C1 (18.0 g, 37.0 mmol, 95.2% yield) as a white solid. MS obsd.: 487.0 (MH + ). 1 H NMR (400 MHz, CDCl3): δ ppm 7.94 (br d, J = 8.8 Hz, 2 H), 7.89 (m, 2 H), 7.43 (m, 1 H), 7.32 - 7.33 (m, 4 H), 5.01 (s, 2 H), 3.12 - 3.14 (m, 8 H). Step 2: benzyl 4-((4-(4-((tert-butoxycarbonyl)amino)-2-oxopyrrolidin-1-yl)p henyl)sulfonyl) piperazine-1-carboxylate (intermediate C2) To a mixture of intermediate C1 (18.0 g, 37.0 mmol), tert-butyl N-(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, 1.00 eq) and N1,N2-dimethylcyclohexane-1,2-diamine (5.26 g, 37.0 mmol). The mixture was stirred at 90 °C for 2 hrs under N2. The mixture was diluted with EA (200 mL) and filtered. The filtrate was washed with NH 3 • H 2 O (2% wt, 200 mL), aq CaCl 2 (150 mL, 2 N) two times and NaCl (150 mL) two times, dried over Na 2 SO 4 , filtered and concentrated to give yellow oil. The oil was purified by pre-HPLC to give intermediate C2 (13.5 g, 24.2 mmol, 65.3% yield) as a gray solid. MS obsd.: 559.3 (MH + ). Step 3: tert-butyl N-[5-oxo-1-(4-piperazin-1-ylsulfonylphenyl)pyrrolidin-3-yl]c arbamate (intermediate C) To a solution of intermediate C2 (500.0 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 25 °C for 2 hrs. The mixture was diluted with MeOH (100 mL), filtered through a pad of Celite and concentrated to give Intermediate C (300 mg, 0.710 mmol, 78.96% yield) as a grey solid. MS obsd.: 425.3 (MH + ). Intermediate D Tert-butyl N-[(3R)-5-oxo-1-(4-piperazin-1-ylsulfonylphenyl)pyrrolidin-3 -yl]carbamate The title compound was prepared in analogy to the preparation of intermediate C by using tert- butyl N-[(3R)-5-oxopyrrolidin-3-yl]carbamate instead of tert-butyl N-(5-oxopyrrolidin-3- yl)carbamate. Intermediate D (700 mg) was obtained as a white solid. MS obsd.: 425.2 (MH + ). Intermediate E and Intermediate F 2,6-Dichloro-4-[[(2S)-1,4-dioxan-2-yl]-difluoro-methyl]pyrid ine and 2,6-Dichloro-4-[[(2R)- 1,4-dioxan-2-yl]-difluoro-methyl]pyridine The title compounds were prepared according to the following procedures: Step 1: 1,4-dioxane-2-carbaldehyde (intermediate E1) To a mixture of DMSO (15.75 g, 315.1 mmol) in DCM (450 mL) was added (COCl)2 (19.84 g, 157.6 mmol) dropwise at -60 o C. After stirring for 1 h, (1,4-dioxan-2-yl)methanol (15.5 g, 131.3 mmol) was added and the mixture was stirred at -60 o C for 2 hrs. TEA (39.83 g, 393.9 mmol) was added at -60 o C and the mixture was warmed to 10 o C slowly. The suspension was filtered and the filtrate was concentrated to give the crude, which was distilled under vacuum pump and collected the 70 o C co-boiled to give intermediate E1 (21.5 g, yield: 70.7 %) as colorless liquid. 1 H NMR (400 MHz, CDCl 3 ): δ ppm 9.54 - 9.64 (m, 1 H), 3.97 - 4.09 (m, 1 H) 3.82 - 3.95 (m, 2 H) 3.52 - 3.79 (m, 4 H). Step 2: (2,6-dichloropyridin-4-yl)(1,4-dioxan-2-yl)methanol (intermediate E2) To a mixture of 2,6-dichloro-4-iodopyridine (21.46 g, 78.37 mmol) in THF (70 mL) was added CH 3 MgCl/LiCl (64.92 mL, 84.4 mmol) under N 2 at -50 °C. The mixture was stirred at 25 °C for 2 hrs and then 1,4-dioxane-2-carbaldehyde (14.0 g, 60.28 mmol) in THF (10 mL) was added at -50 °C. The mixture was stirred at 25 °C for 1h. The reaction was then poured into saturated aq. NH 4 Cl solution and extracted with EA (300 mL) three times. The organic layer were combined, washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by column chromatography on silica gel (PE: EA=3:2) to give intermediate E2 (8.3 g, 31.43 mmol, 52.13% yield) as yellow oil. MS obsd.: 264.1 (MH + ). Step 3: (2,6-dichloropyridin-4-yl)(1,4-dioxan-2-yl)methanone(interme diate E3) To a solution of intermediate E2 (6.4 g, 24.23 mmol) in DCM (60 mL) was added DMP (11.31 g, 26.66 mmol) at 0°C and the reaction was stirred at 20°C for 0.5 h. The mixture was poured into a mixture of aq. Na 2 CO 3 (400 mL) and EA: THF=5:1 (500 mL). After stirring for 0.5h, the reaction was filtered and the organic layer was extracted with aq. Na2CO3 (300 mL) two times and dried over anhydrous Na 2 SO 4 , filtered and concentrated to give intermediate E3 (6 g, 22.89 mmol, 94.47% yield) as a light yellow solid. Step 4: 2,6-dichloro-4-[1,4-dioxan-2-yl(difluoro)methyl]pyridine (intermediate E4) To a solution of intermediate E3 (7.0 g, 26.71 mmol) in DCM (150 mL) was added DAST (140.0 mL, 1060 mmol) and then the mixture was stirred at 20°C for 12 hrs. The reaction was extracted by EA (400 mL × 3) and the organic layer was washed with aq. NaHCO3 (200 × 3 mL), brine (200 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by pre-HPLC (TFA as addictive) to give intermediate E4 (4 g, 14.08 mmol, 52.72% yield) as a white solid. MS obsd.: 284.0 (MH + ). 1 H NMR (400 MHz, CDCl3): δ ppm 7.35 (s, 2H ), 3.87 - 4.00 (m, 2H ), 3.79-3.81 (m, 1H), 3.62-3.71 (m, 2H ), 3.53-3.60 (m, 2H). Step 5: 2,6-dichloro-4-[[(2S)-1,4-dioxan-2-yl]-difluoro-methyl]pyrid ine and 2,6-dichloro- 4-[[(2R)-1,4-dioxan-2-yl]-difluoro-methyl]pyridine(intermedi ates E and F) 4.0 g of intermediate E4 was purified by SFC using the following condition to give intermediate E (1.48 g, 36.5% yield, 99% ee, faster eluting) and intermediate F (1.5 g, 37.5% yield, 99% ee, slower eluting) as white solids. SFC condition: Column: OJ (250mm×50mm, 10 um); 0.1% NH3H2O EtOH; begin B 15%, end B 15%; Flow Rate (ml/min): 180. Intermediate G Tert-butyl (2R)-2-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]morpholine- 4-carboxylate The title compound was prepared according to the following procedures: Step 1: tert-butyl (2R)-2-formylmorpholine-4-carboxylate (intermediate G1) To a solution of (COCl) 2 (21.91 g, 172.6 mmol) in DCM (80 mL) was added DMSO (17.97 g, 230.14 mmol) at -60 °C and the mixture was stirred at -60 °C for 0.5 h under N2. Then a solution of (R)-N-Boc-2-hydroxymethylmorpholine (25.0 g, 115.07 mmol) in DCM (50 mL) was added and the reaction was stirred at -60 °C for additional 1 h. DIPEA (59.49 g, 460.28 mmol) was added and the mixture was stirred at - 60 ~ -20 °C for 1 h and then at 0 °C for 1 h. The mixture was diluted with DCM (500 mL), washed with aq. CaCl2 (100 mL) three times and brine (50 mL) two times, dried over anhydrous Na 2 SO 4 , filtered and concentrated to give the crude, which was purified by silica gel column (PE:EA=10:1 to 1:5) to give intermediate G1 (8 g, 37.17 mmol, 32.3% yield) as colorless oil. MS obsd.: 115.9 (MH + -100). Step 2: tert-butyl-(2R)-2-[(2,6-dichloro-4-pyridyl)-hydroxy-methyl]m orpholine-4- carboxylate (intermediate G2) To a solution of 2,6-dichloro-4-iodopyridine (9.16 g, 33.45 mmol) in THF (30.0 ml) was added CH3MgCl/LiCl in THF (28.3 mL, 36.79 mmol) at -40 °C under N2 and the mixture was stirred at 25 °C for 1 h. Then intermediate G1 (7.2 g, 33.45 mmol) in THF (130 mL) was added at -40 °C and the mixture was stirred at 25 °C for 1 h. The mixture was poured into water (100 mL) and extracted with EA (600 mL). The organic layer was washed with brine (100 mL) two times, dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by column chromatography on silica gel (PE: EA = 100:1~1:2) to give intermediate G2 (6 g, 16.52 mmol, 49.38% yield) as a white solid. MS obsd.: 363.3 (MH + ). Step 3: tert-butyl-(2R)-2-(2,6-dichloropyridine-4-carbonyl)morpholin e-4-carboxylate (intermediate G3) To a solution of intermediate G2 (5.5 g, 15.14 mmol) in THF (80 mL) was added DMP (7.06 g, 16.66 mmol) slowly at 0 °C under N 2 and then the mixture was stirred for 2 h. The mixture was poured into aq. K 2 CO 3 (100 mL) at 0 °C and then extracted with EA (150 mL) three times. The organic layer was washed with aq. K 2 CO 3 (50 mL) three times and brine (100 mL) two times, dried over anhydrous Na 2 SO 4 , filtered and concentrated to give intermediate G3 (5.3 g, 14.67 mmol, 96.9% yield) as a white solid. MS obsd.: 305.2 (MH + -56). Step 4: tert-butyl-(2R)-2-[(2,6-dichloro-4-pyridyl)-difluoro-methyl] morpholine-4- carboxylate (intermediate G) A solution of intermediate G3 (5.0 g, 13.84 mmol) in DCM (8 mL) was added DAST (3.0 mL, 207.63 mmol) and stirred at 40°C for 3 hrs. The mixture was poured into aq. NaHCO3 at 0 °C and then extracted with EA (200 mL) three times. The combined organic layer was washed with brine (100 mL) two times, dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by column chromatography on silica gel (PE: EA= 10:1~1:1) to give intermediate G (2.2 g, 5.74 mmol, 41.47% yield, 90% ee) as a white solid. MS obsd.: 327.3 (MH + -56). Intermediate H Tert-butyl-(2S)-2-[(2,6-dichloro-4-pyridyl)-difluoro-methyl] morpholine-4-carboxylate The title compound was prepared in analogy to the preparation of intermediate G by using (S)-N-Boc-2-hydroxymethylmorpholine instead of (R)-N-Boc-2-hydroxymethylmorpholine. Intermediate H (1.2 g, 95.5% ee) was obtained as a white solid. MS obsd.: 327.2 (MH + -56). Intermediate I Tert-butyl 4-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]-3,6-dihydro-2H- pyridine-1- carboxylate The title compound was prepared according to the following procedures: Step 1: tert-butyl 4-(methoxy(methyl)carbamoyl)-5,6-dihydropyridine-1(2H)-carbo xylate (intermediate I1) To a solution of 1-(tert-butoxycarbonyl)-1,2,3,6-tetrahydropyridine-4-carboxy lic acid (4.5 g, 19.80 mmol), HATU (11.29 g, 29.70 mmol) and DIPEA (8.96 g, 69.30 mmol) in DMF (50 mL) was added N,O-dimethylhydroxylamine hydrochloride (4.83 g, 49.50 mmol). The reaction was stirred at 15 °C for 2 hrs. The mixture was poured into water (30 mL) and extracted by EA (40 mL) two times. The organic layer was washed by brine (50 mL) two times, dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by column chromatography on silica gel (PE: EA=1:0 to 5:1) to give intermediate I1 (4.7 g, yield: 87.80%) as a white solid. MS obsd.: 215.4 (MH + -56). Step 2: tert-butyl 4-(2,6-dichloropyridine-4-carbonyl)-3,6-dihydro-2H-pyridine- 1- carboxylate (intermediate I2) To a solution of 2,6-dichloro-4-iodopyridine (5.07 g, 18.5 mmol) in THF (15.0 ml) was added CH 3 MgCl/LiCl in THF (14.23 mL, 18.5 mmol) at -40 °C under N 2 . After stirred at 25 °C for 1 h, intermediate I1 (2.5 g, 9.25 mmol) in THF (25 mL) was added into the solution at -40 °C. The mixture was poured into water (100 mL) and extracted with EA (600 mL). The organic layer was washed with brine (100 mL) two times, dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by column chromatography on silica gel (PE: EA=100:1~1:1) to give intermediate I2 (1.5 g, 4.2 mmol, 45.4% yield) as light yellow oil. MS obsd.: 301.3 (MH + -56). Step 3: tert-butyl 4-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]-3,6-dihydro-2H- pyridine-1- carboxylate(intermediate I) A solution of intermediate I2 (1.5 g, 4.2 mmol) in DCM (3 mL) was added DAST (6.77 g, 41.99 mmol) and stirred at 40°C for 12 hrs. The mixture was poured into saturated NaHCO3 (aq.) at 0 °C and then extracted with EA (100 mL) three times. The combined organic layer was washed with brine (500 mL) two times, dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by column chromatography on silica gel (PE: EA=10:1~1:1) to give intermediate I (1 g, 2.64 mmol, 62.8% yield) as a light yellow oil. MS obsd.: 323.2 (MH + -56). Intermediate J Tert-butyl 3-(2,6-dichloropyridine-4-carbonyl)piperidine-1-carboxylate The title compound was prepared in analogy to the preparation of intermediate G3 by using tert- butyl 3-formylpiperidine-1-carboxylate instead of intermediate G1. Intermediate J (3.1 g) was obtained as yellow oil. MS obsd.: 359.0 (MH + ). Intermediate K Tert-butyl 3-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]azetidine-1-carb oxylate The title compound was prepared in analogy to the preparation of intermediate G by using tert-butyl 3-formylazetidine-1-carboxylate instead of intermediate G1. Intermediate K (1.1 g) was obtained as a yellow solid. MS obsd.: 353.1 (MH + ). Example 1 4-Amino-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pipe razin-1-yl]sulfonylphenyl] pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: tert-butyl N-[1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin -1- yl]sulfonylphenyl]-5-oxo-pyrrolidin-3-yl]carbamate (compound 1a) A mixture of intermediate A (100 mg, 188 µmol), tert-butyl (5-oxopyrrolidin-3-yl) carbamate (75.3 mg, 376 µmol), DMEDA (3.32 mg, 37.6 µmol), CuI (7.17 mg, 37.6 µmol) and Cs 2 CO 3 (122 mg, 376 µmol) in DMF (5 mL) was heated at 75 °C for 1 h. The mixture was then poured into ice water, extracted with DCM (10 mL) three times, dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by column chromatography on silica gel (PE: EA= 5:1) to give compound 1a (97 mg, 85% yield) as a white solid. MS obsd.: 604.2 (MH + ). Step 2: 4-Amino-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pipe razin-1- yl]sulfonylphenyl] pyrrolidin-2-one(Example 1) A solution of compound 1a (100 mg, 166 µmol) in TFA (5 mL) was stirred at rt for 5 hrs. The mixture was concentrated and the crude was purified by prep-HPLC to give Example 1 (10 mg, 12 % yield) as a white solid. MS obsd.: 504.2 (MH + ). 1 H NMR (400 MHz, CD 3 OD): δ ppm 7.78 - 7.89 (m, 2H), 7.64 - 7.75 (m, 2H), 6.81 (s, 1H), 6.72 (s, 1H), 4.29 (dd, J = 7.2, 11.2 Hz, 1H), 3.97 – 4.16 (m, 1H), 3.85 (dd, J = 2.4, 11.2 Hz, 1H), 3.56 - 3.70 (m, 4H), 3.09 (dd, J = 8.4, 18.0 Hz, 1H), 2.95 - 3.00 (m, 4H), 2.46 - 2.69 (m, 1H). Example 2 4-(Aminomethyl)-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyri dyl]piperazin-1-yl]sulfonyl- phenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 1 by using tert-butyl N-[(5-oxopyrrolidin-3-yl)methyl]carbamate instead of tert-butyl (5-oxopyrrolidin-3- yl)carbamate. Example 2 (17.1 mg) was obtained as a white solid. MS obsd.: 518.0 (MH + ). 1 H NMR (400 MHz, CD 3 OD): δ ppm 7.75 - 7.790 (m, 2H), 7.68 - 7.73 (m, 2H), 6.81 (s, 1H), 6.73 (s, 1H), 4.02 (dd, J = 8.0, 9.6 Hz, 1H), 3.57 - 3.73 (m, 5H), 2.92 - 3.15 (m, 6H), 2.66 - 2.83 (m, 2H), 2.35 - 2.55 (m, 1H). Example 3 5-(Aminomethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyri dyl]piperazin-1- yl]sulfonylphenyl]oxazolidin-2-one The title compound was prepared in analogy to the preparation of Example 1 by using tert- butyl N-[(2-oxooxazolidin-5-yl)methyl]carbamate instead of tert-butyl (5-oxopyrrolidin-3- yl)carbamate. Example 3 (30 mg) was obtained as a white solid. MS obsd.: 520.0 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 8.15 (brs, 3H), 7.53 - 7.96 (m, 4H), 7.10 (s, 1H), 6.98 (s, 1H), 4.81 - 5.06 (m, 1H), 4.24 (t, J = 9.2 Hz, 1H), 3.87 (dd, J = 6.4, 9.2 Hz, 1H), 3.63 - 3.77 (m, 4H), 3.27 (s, 2H), 2.96 (t, J = 4.8 Hz, 4H). Example 4 N-[[3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazi n-1-yl]sulfonylphenyl]-2-oxo- oxazolidin-5-yl]methyl]acetamide To a solution of Example 3 (50 mg, 96.2 µmol) and DIPEA (62 mg, 481 µmol) in DCM (5 mL) was added acetyl chloride (15.1 mg, 192 µmol) dropwise at rt. The mixture was stirred at rt for 16 hrs. Solvent was evaporated and the crude was purified by prep-HPLC to give Example 4 as a white solid (15 mg, 25 % yield). MS obsd.: 561.9 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 8.23 (t, J = 6.0 Hz, 1H), 7.78 (s, 4H), 7.09 (s, 1H), 6.97 (s, 1H), 4.58 - 4.86 (m, 1H), 4.16 (t, J = 9.2 Hz, 1H), 3.78 (dd, J = 6.4, 9.12 Hz, 1H), 3.68 - 3.74 (m, 4H), 3.42 (t, J = 5.6 Hz, 2H), 2.96 (t, J = 4.8 Hz, 4H), 1.81 (s, 3H). Example 5 4-Amino-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pipe razin-1-yl]sulfonylphenyl]- 3,3-dimethyl-pyrrolidin-2-one A mixture of intermediate A (0.060 g, 113 µmol), 4-amino-3,3-dimethylpyrrolidin-2-one (14.5 mg, 113 µmol), DMEDA (1.99 mg, 22.6 µmol), CuI (4.3 mg, 22.6 µmol) and K 3 PO 4 (71.8 mg, 339 µmol) in DMF (1.5 mL) was heated at 80 °C for 1.5 hrs. The mixture was then poured into water, extracted with DCM. The organic layer was concentrated and the crude was purified by pre-HPLC to give Example 5 as a white solid (40 mg, 64.4% yield). MS obsd.: 521.1 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 8.31(br s, 2H), 7.83 - 7.89 (m, 2H), 7.71 - 7.76 (m, 2H), 7.02 (s, 1H), 6.90 (s, 1H), 4.22 (dd, J = 6.4, 11.2 Hz, 1H), 3.61 - 3.80 (m, 6H), 2.90 (br t, J = 4.8 Hz, 4H), 1.16 (s, 3H), 1.11 (s, 3H). Example 6 4-(Aminomethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyri dyl]piperazin-1- yl]sulfonylphenyl]oxazolidin-2-one The title compound was prepared in analogy to the preparation of Example 5 by using 4- (aminomethyl)oxazolidin-2-one instead of 4-amino-3,3-dimethylpyrrolidin-2-one. Example 6 (10 mg) was obtained as a white solid. MS obsd.: 520.1 (MH + ). 1 H NMR (400 MHz, CD3OD): δ ppm 7.95 (br s, 3H), 7.84 - 7.90 (m, 2H), 7.78 - 7.83 (m, 2H), 7.10 (s, 1H), 6.98 (s, 1H), 4.81 - 4.98 (m, 1H), 4.53 - 4.60 (m, 1H), 4.45 - 4.51 (m, 1H), 3.66 - 3.68 (m, 4H), 3.11 - 3.22 (m, 1H), 2.91 -3.09 (m, 5H). Example 7 (3S)-3-amino-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl ]piperazin-1-yl]sulfonylphenyl] azetidin-2-one The title compound was prepared in analogy to the preparation of Example 1 by using (3S)-3-aminoazetidin-2-one instead of tert-butyl (5-oxopyrrolidin-3-yl)carbamate. Example 7 (10 mg) was obtained as a white solid. MS obsd.: 490.0 (MH + ). 1 H NMR (400 MHz, CD3OD): δ ppm 7.67 - 7.85 (m, 2H), 7.38 - 7.61 (m, 2H), 6.82 (s, 1H), 6.74 (s, 1H), 4.62 (dd, J = 2.8, 5.6 Hz, 1H), 4.07 (dd, J = 5.6, 7.2 Hz, 1H), 3.53 - 3.76 (m, 5H), 2.92 - 3.03 (m, 4H). Example 8 3-Amino-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pipe razin-1-yl]sulfonylphenyl] pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 1 by using tert- butyl N-(2-oxopyrrolidin-3-yl)carbamate instead of tert-butyl (5-oxopyrrolidin-3-yl)carbamate. Example 8 (20 mg) was obtained as a white solid. MS obsd.: 504.1 (MH + ). 1 H NMR (400 MHz, CD 3 OD): δ ppm 7.82 - 8.02 (m, 2H), 7.65 - 7.82 (m, 2H), 6.82 (s, 1H), 6.73 (s, 1H), 4.19 (dd, J = 8.8, 11.2 Hz, 1H), 3.82 - 4.04 (m, 2H), 3.59 - 3.68 (m, 4H), 2.91 – 3.04 (m, 4H), 2.51 - 2.69 (m, 1H), 1.95 - 2.19 (m, 1H). Example 9 5-(2-Aminoethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyr idyl]piperazin-1- yl]sulfonylphenyl]oxazolidin-2-one The title compound was prepared in analogy to the preparation of Example 1 by using tert- butyl N-[2-(2-oxooxazolidin-5-yl)ethyl]carbamate instead of tert-butyl (5-oxopyrrolidin-3- yl)carbamate. Example 9 (66.4 mg) was obtained as a white solid. MS obsd.: 578.1 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 8.02 - 8.26 (m, 3 H), 7.74 - 7.84 (m, 4 H), 7.08 - 7.13 (m, 1 H), 6.95 - 7.00 (m, 1 H), 4.79 - 4.92 (m, 1 H), 4.18 - 4.29 (m, 1 H), 3.67 - 3.90 (m, 5 H), 2.86 - 3.07 (m, 6 H), 1.99 - 2.13 (m, 2 H). Example 10 2-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-2,6- diazaspiro[4.5]decan-3-one The title compound was prepared in analogy to the preparation of Example 1 by using tert- butyl 3-oxo-2,6-diazaspiro[4.5]decane-6-carboxylate instead of tert-butyl (5-oxopyrrolidin-3- yl)carbamate. Example 10 (10 mg) was obtained as a white solid. MS obsd.: 558.2 (MH + ). 1 H NMR (400 MHz, CD 3 OD): δ ppm 7.78 - 7.90 (m, 2H), 7.67 - 7.78 (m, 2H), 6.81 (s, 1H), 6.73 (s, 1H), 4.07 (s, 2H), 3.51 - 3.81 (m, 4H), 3.24 - 3.33 (m, 4H), 2.95 - 3.02 (m, 4H), 1.92 (s, 2H), 1.64 - 1.77 (m, 4H). Example 11 2-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-2,8- diazaspiro[4.5]decan-3-one The title compound was prepared in analogy to the preparation of Example 1 by using tert- butyl 3-oxo-2,8-diazaspiro[4.5]decane-8-carboxylate instead of tert-butyl (5-oxopyrrolidin-3- yl)carbamate. Example 11 (15 mg) was obtained as a white solid. MS obsd.: 558.1 (MH + ). 1 H NMR (400 MHz, CD3OD): δ ppm 8.50 (br s, 2H), 7.93 (d, J = 8.8 Hz, 2H), 7.77 (d, J = 8.8 Hz, 2H), 7.09 (s, 1H), 6.97 (s, 1H), 3.79 (s, 2H), 3.66 - 3.75 (m, 4H), 3.03 - 3.22 (m, 4H), 2.96 (t, J = 4.8 Hz, 4H), 2.61 (s, 2H), 1.65 - 1.88 (m, 4H). Example 12 6-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-2,6- diazaspiro[3.4]octan-7-one The title compound was prepared in analogy to the preparation of Example 1 by using tert- butyl 6-oxo-2,7-diazaspiro[3.4]octane-2-carboxylate instead of tert-butyl (5-oxopyrrolidin-3- yl)carbamate. Example 11 (10 mg) was obtained as a white solid. MS obsd.: 530.0 (MH + ). 1 H NMR (400 MHz, CD 3 OD): δ ppm 8.73 (br d, J = 1.6 Hz, 2H), 7.83 - 7.91 (m, 2H), 7.74 - 7.83 (m, 2H), 7.09 (s, 1H), 6.98 (s, 1H), 4.14 (s, 2H), 4.06 (td, J = 5.2, 10.8 Hz, 4H), 3.65 - 3.80 (m, 4H), 2.90 - 3.03 (m, 6H). Example 13 7-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-5-oxa-2,7- diazaspiro[3.4]octan-6-one The title compound was prepared in analogy to the preparation of Example 5 by using 5- oxa-2,7-diazaspiro[3.4]octan-6-one instead of 4-amino-3,3-dimethylpyrrolidin-2-one. Example 13 (8 mg) was obtained as a white solid. MS obsd.: 532.2 (MH + ). 1 H NMR (400 MHz, CD 3 OD): δ ppm 7.80 - 7.87 (m, 2H), 7.71 - 7.79 (m, 2H), 7.10 (s, 1H), 6.98 (s, 1H), 4.19 - 4.45 (m, 6H), 3.71 (t, J = 5.2 Hz, 4H), 2.96 (br t, J = 4.8 Hz, 4H). Example 14 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-1-oxa-3,8- diazaspiro[4.5]decan-2-one The title compound was prepared in analogy to the preparation of Example 5 by using 1- oxa-3,8-diazaspiro[4.5]decan-2-one hydrochloride instead of 4-amino-3,3-dimethylpyrrolidin-2- one. Example 14 (58 mg) was obtained as a white solid. MS obsd.: 560.1 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.66 - 7.80 (m, 4H), 6.97 - 7.04 (m, 1H), 6.84 - 6.93 (m, 1H), 3.81 - 3.87 (m, 2H), 3.62 - 3.66 (m, 4H), 2.86 - 2.91 (m, 4H), 2.72 - 2.81 (m, 2H), 2.59 - 2.67 (m, 2H), 1.59 - 1.80 (m, 4H). Example 15 (3aR,6aS)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pi perazin-1-yl]sulfonylphenyl]- 4,5,6,6a-tetrahydro-3aH-pyrrolo[3,4-d]oxazol-2-one The title compound was prepared in analogy to the preparation of Example 5 by using (3aR,6aS)-3,3a,4,5,6,6a-hexahydropyrrolo[3,4-d]oxazol-2-one instead of 4-amino-3,3- dimethylpyrrolidin-2-one. Example 15 (10 mg) was obtained as a white solid. MS obsd.: 532.1 (MH + ). 1 H NMR (400 MHz, CD3OD): δ ppm 7.73 - 8.02 (m, 4H), 6.94 (s, 1H), 6.85 (s, 1H), 5.48 (dd, J = 4.8, 7.6 Hz, 1H), 5.36 - 5.43 (m, 1H), 3.90 (d, J = 13.2 Hz, 1H), 3.73 - 3.81 (m, 4H), 3.747 - 3.72 (m, 3H), 3.05 - 3.19 (m, 4H). Example 16 (3aR,7aS)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pi perazin-1-yl]sulfonylphenyl]- 3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridin-2-one The title compound was prepared in analogy to the preparation of Example 1 by using tert- butyl (3aR,7aS)-2-oxo-3,3a,4,6,7,7a-hexahydrooxazolo[4,5-c]pyridin e-5-carboxylate instead of tert-butyl (5-oxopyrrolidin-3-yl)carbamate. Example 16 (10 mg) was obtained as a white solid. MS obsd.: 546.1 (MH + ). 1 H NMR (400 MHz, CD 3 OD): δ ppm 7.74 (d, J = 2.4 Hz, 4H), 6.82 (s, 1H), 6.73 (s, 1H), 4.82 - 4.99 (m, 2H), 3.60 - 3.70 (m, 4H), 3.55 (dd, J = 4.4, 14.0 Hz, 1H), 3.10 - 3.31 (m, 3H), 2.91 - 3.04 (m, 4H), 2.16 - 2.30 (m, 2H). Example 17 (3aR,7aS)-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pi perazin-1-yl]sulfonylphenyl]- 3a,4,5,6,7,7a-hexahydrooxazolo[5,4-c]pyridin-2-one The title compound was prepared in analogy to the preparation of Example 1 by using tert- butyl (3aR,7aS)-2-oxo-1,3a,4,6,7,7a-hexahydrooxazolo[5,4-c]pyridin e-5-carboxylate instead of tert-butyl (5-oxopyrrolidin-3-yl)carbamate. Example 17 (33 mg) was obtained as a white solid. MS obsd.: 546.1(MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 9.09 - 9.67 (m, 2H), 7.83 - 7.89 (m, 2H), 7.76 - 7.82 (m, 2H), 7.09 (s, 1H), 6.97 (s, 1H), 4.98 (br d, J = 7.2 Hz, 1H), 4.78 - 4.89 (m, 1H), 3.61 - 3.677 (m, 5H), 3.39 - 3.45 (m, 1H), 3.06 - 3.14 (m, 1H), 2.98 (br t, J = 4.4 Hz, 5H), 2.30 - 2.41 (m, 1H), 1.66 - 1.78 (m, 1H). Example 18 (3aS,7aR)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pi perazin-1-yl]sulfonylphenyl]- 3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridin-2-one The title compound was prepared in analogy to the preparation of Example 1 by using tert- butyl (3aS,7aR)-2-oxo-3,3a,4,6,7,7a-hexahydrooxazolo[4,5-c]pyridin e-5-carboxylate instead of tert-butyl (5-oxopyrrolidin-3-yl)carbamate. Example 18 (120 mg) was obtained as a white solid. MS obsd.: 546.0 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 9.19 - 9.31 (m, 2H), 7.80 (s, 4H), 7.10 (s, 1H), 6.98 (s, 1H), 4.09 - 5.02 (m, 2H), 3.68 - 3.78 (m, 4H), 3.49 - 3.56 (m, 1H), 3.11 - 3.22 (m, 2H), 3.02 - 3.09 (m, 1H), 2.99 (br s, 4H), 2.31 (br s, 1H), 2.09 - 2.020 (m, 1H). Example 19 (3aS,8aR)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pi perazin-1-yl]sulfonylphenyl]- 4,5,6,7,8,8a-hexahydro-3aH-oxazolo[4,5-c]azepin-2-one The title compound was prepared in analogy to the preparation of Example 1 by using tert- butyl (3aS,8aR)-2-oxo-3a,4,6,7,8,8a-hexahydro-3H-oxazolo[4,5-c]aze pine-5-carboxylate instead of tert-butyl (5-oxopyrrolidin-3-yl)carbamate. Example 19 (10 mg) was obtained as a white solid. MS obsd.: 560.1(MH + ). 1 H NMR (400 MHz, CD 3 OD): δ ppm 7.67 - 7.85 (m, 4H), 6.82 (s, 1H), 6.74 (s, 1H), 4.97 - 5.12 (m, 2H), 3.59 - 3.72 (m, 4H), 3.41 - 3.454 (m, 1H), 3.24 - 3.30 (m, 1H), 3.13 - 3.19 (m, 2H), 2.92 - 3.07 (m, 4H), 2.20 (d, J = 4.4 Hz, 2H), 1.95 - 2.11 (m, 1H), 1.69 - 1.86 (m, 1H). Example 20 4-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]- 1,2,3,3a,6,6a-hexahydropyrrolo[3,2-b]pyrrol-5-one The title compound was prepared in analogy to the preparation of Example 1 by using tert- butyl 5-oxo-2,3,3a,4,6,6a-hexahydropyrrolo[3,2-b]pyrrole-1-carboxy late instead of tert-butyl (5- oxopyrrolidin-3-yl)carbamate. Example 20 (100 mg) was obtained as a white solid. MS obsd.: 530.2(MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 9.56 (br s, 2H), 7.84 - 7.93 (m, 2H), 7.74 - 7.84 (m, 2H), 7.09 (s, 1H), 6.92 - 7.01 (m, 1H), 5.02 - 5.16 (m, 1H), 4.32 - 4.47 (m, 1H), 3.68 - 3.73 (m, 4H), 3.20 - 3.29 (m, 2H), 2.99 (br d, J = 5.2 Hz, 6H), 2.29 (br s, 1H), 1.85 (br d, J = 7.2 Hz, 1H). Example 21 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-6-hydroxy- 5-(hydroxymethyl)-3a,5,6,6a-tetrahydrofuro[2,3-d]oxazol-2-on e The title compound was prepared in analogy to the preparation of Example 5 by using 6- hydroxy-5-(hydroxymethyl)-3a,5,6,6a-tetrahydro-3H-furo[2,3-d ]oxazol-2-one instead of 4- amino-3,3-dimethylpyrrolidin-2-one. Example 21 (5 mg) was obtained as a white solid. MS obsd.: 579.1 (MH + ). 1 H NMR (400 MHz, CD 3 OD): δ ppm 7.85 (d, J = 8.8 Hz, 2H), 7.71 (d, J = 8.8 Hz, 2H), 6.82 (s, 1H), 6.73 (s, 1H), 6.12 (d, J = 6.0 Hz, 1H), 4.84 (d, J = 5.2 Hz, 1H), 4.31 (d, J = 2.4 Hz, 1H), 4.02 (dt, J = 2.8, 5.2 Hz, 1H), 3.57 - 3.69 (m, 4H), 3.46 (d, J = 5.2 Hz, 2H), 2.91 - 3.05 (m, 4H). Example 22 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-5- [(3S,4R,5R)-3,4-dihydroxy-5-methoxy-tetrahydrofuran-2-yl]-4- methyl-oxazolidin-2-one The title compound was prepared in analogy to the preparation of Example 5 by using 5 [(3S,4R,5R)-3,4-dihydroxy-5-methoxy-tetrahydrofuran-2-yl]-4- methyl-oxazolidin-2-one instead of 4-amino-3,3-dimethylpyrrolidin-2-one. Example 22 (20 mg) was obtained as a white solid. MS obsd.: 637.1(MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.92 (d, J = 8.8 Hz, 2H), 7.77 (d, J = 8.8 Hz, 2H), 7.09 (s, 1H), 6.97 (s, 1H), 4.94 - 5.38 (m, 1H), 4.76 - 4.89 (m, 2H), 4.74 (d, J = 1.2 Hz, 1H), 4.51 (dd, J = 7.32, 9.6 Hz, 1H), 4.17 (t, J = 5.2 Hz, 1H), 4.03 (dd, J = 5.2, 9.8 Hz, 1H), 3.83 (dd, J = 1.2, 4.8 Hz, 1H), 3.67 - 3.75 (m, 4H), 3.27 (s, 3H), 2.99 (t, J = 4.8 Hz, 4H), 1.26 (d, J = 6.4 Hz, 3H). Example 23 (3aS,5S,6S,7S,7aR)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-p yridyl]piperazin-1-yl]sulfonyl phenyl]-5,7-dihydroxy-6-(hydroxymethyl)-5,6,7,7a-tetrahydro- 3aH-pyrano[2,3-d]oxazol-2- one The title compound was prepared in analogy to the preparation of Example 5 by using (3aS,5S,6S,7S,7aR)-5,7-dihydroxy-6-(hydroxymethyl)-3,3a,5,6, 7,7a-hexahydropyrano[2,3-d] oxazol-2-one of 4-amino-3,3-dimethylpyrrolidin-2-one. Example 23 (10 mg) was obtained as a white solid. MS obsd.: 609.2(MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.92 (d, J = 8.8 Hz, 2H), 7.78 (d, J = 8.8 Hz, 2H), 7.08 (s, 1H), 6.97 (s, 1H), 6.07 (d, J = 6.0 Hz, 1H), 5.61 (d, J = 5.2 Hz, 1H), 5.38 (d, J = 5.2 Hz, 1H), 4.81 (t, J = 5.6 Hz, 1H), 4.60 (dd, J = 4.4, 6.0 Hz, 1H), 3.80 - 3.87 (m, 1H), 3.67 - 3.75 (m, 4H), 3.57 - 3.63 (m, 2H), 3.44 - 3.54 (m, 2H), 2.93 - 3.04 (m, 4H). Example 24 8-(2-Aminoethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyr idyl]piperazin-1- yl]sulfonylphenyl]-1-oxa-3,8-diazaspiro[4.5]decan-2-one The title compound was prepared according to the following scheme: Step 1: tert-butyl N-[2-[3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pipera zin-1-yl] fonylphenyl]-2-oxo-1-oxa-3,8-diazaspiro[4.5]decan-8-yl]ethyl ]carbamate (compound 24a) A mixture of Example 11 (0.050 g, 89.3 µmol), tert-butyl (2-oxoethyl)carbamate (28.4 mg, 179 µmol) and NaBH(OAc) 3 (75.7 mg, 357 µmol) in DCM (1 mL) and was heated at 25 °C overnight. Then, the mixture was poured into water, extracted with DCM. The organic layer was concentrated to give the crude, which was used directly in next step. MS obsd.: 703.2 (MH + ). Step 2: 8-(2-aminoethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyr idyl]piperazin-1- yl]sulfonylphenyl]-1-oxa-3,8-diazaspiro[4.5]decan-2-one (Example 24) A mixture of compound 24a (0.062 g, 88.2 µmol) and TFA (450 mg, 0.3mL, 3.95 mmol) in DCM (1 mL) was heated at 25 °C for 2 hrs. The mixture was concentrated and the crude was purified by HPLC to give Example 24 (15 mg, 26.8% yield) as a white solid. MS obsd.: 603.1 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.77 - 7.84 (m, 4H), 7.06 - 7.11 (m, 1H), 6.93 - 7.00 (m, 1H), 3.97 - 4.04 (m, 2H), 3.70 - 3.74 (m, 4H), 3.50 - 3.56 (m, 4H), 3.07 - 3.22 (m, 4H), 2.94 - 2.99 (m, 4H), 1.90 - 2.23 (m, 4H). Example 25 (3aS,8aR)-5-(2-aminoethyl)-3-[4-[4-[6-chloro-4-(trifluoromet hyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-3a,4,6,7,8,8a-hexahydrooxazolo[4,5-c]azep in-2-one The title compound was prepared according to the following procedures: Step 1: tert-butyl N-[2-[(3aS,8aR)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyri dyl] piperazin-1-yl]sulfonylphenyl]-2-oxo-3a,4,6,7,8,8a-hexahydro oxazolo[4,5-c]azepin-5-yl]ethyl] carbamate (compound 25a) A mixture of Example 19 (50mg, 74.2 µmol), tert-butyl (2-oxoethyl)carbamate (23.6 mg, 148 µmol) and NaBH(OAc)3 (62.9 mg, 297 µmol) in DCM (2 mL) was stirred at 25 °C overnight. Then the mixture was poured into water, extracted with DCM. The organic layer was concentrated to give the crude, which was used directly in next step (52 mg, 100% yield). MS obsd.: 703.2 (MH + ). Step 2: (3aS,8aR)-5-(2-aminoethyl)-3-[4-[4-[6-chloro-4-(trifluoromet hyl)-2-pyridyl] piperazin-1-yl]sulfonylphenyl]-3a,4,6,7,8,8a-hexahydrooxazol o[4,5-c]azepin-2-one (Example 25) A solution of compound 24a (80 mg, 114 µmol) in TFA (5 mL) was stirred at rt for 2 hrs. The mixture was concentrated and the crude was purified by prep-HPLC to give Example 25 (15 mg, 21.9 % yield). MS obsd.: 603.2 (MH + ). 1 H NMR (400 MHz, CD 3 OD): δ ppm 7.85 (s, 4H), 6.92 (s, 1H), 6.84 (s, 1H), 4.99 (td, J = 6.0, 8.8 Hz, 1H), 4.69 - 4.83 (m, 1H), 3.63 - 3.87 (m, 4H), 3.07 - 3.16 (m, 4H), 2.95 (dd, J = 6.8, 14.8 Hz, 1H), 2.55 - 2.84 (m, 7H), 2.08 - 2.20 (m, 2H), 1.90 - 2.04 (m, 1H), 1.54 - 1.72 (m, 1H). Example 26 (3aS,8aR)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pi perazin-1-yl]sulfonylphenyl]-5- [2-(methylamino)ethyl]-3a,4,6,7,8,8a-hexahydrooxazolo[4,5-c] azepin-2-one The title compound was prepared in analogy to the preparation of Example 25 by using tert-butyl N-methyl-N-(2-oxoethyl)carbamate instead of tert-butyl (2-oxoethyl)carbamate. Example 26 (19 mg) was obtained as a white solid. MS obsd.: 617.2 (MH + ). 1 H NMR (400 MHz, CD3OD): δ ppm 7.85 (s, 4H), 6.92 (s, 1H), 6.84 (s, 1H), 4.99 (td, J = 5.6, 8.8 Hz, 1H), 4.77 - 4.84 (m, 1H), 3.69 - 3.80 (m, 4H), 3.07 - 3.18 (m, 4H), 2.97 (dd, J = 6.6, 14.8 Hz, 1H), 2.73 - 2.82 (m, 3H), 2.65 - 2.72 (m, 4H), 2.60 (s, 3H), 2.08 - 2.19 (m, 2H), 1.90 - 2.04 (m, 1H), 1.58 - 1.70 (m, 1H). Example 27 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-5- (methylaminomethyl)oxazolidin-2-one The title compound was prepared according to the following procedures: Step 1: 3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-5- (hydroxymethyl)oxazolidin-2-one (compound 27a) To a solution of intermediate A (3.4 g, 6.39 mmol), 5-(hydroxymethyl)oxazolidin-2-one (1.12 g, 9.59 mmol) and K 2 CO 3 (2.65 g, 19.18 mmol) in NMP (42.5 mL) were added CuI (243.56 mg, 1.28 mmol) and trans-N,N′-dimethylcyclohexane-1,2-diamine (181.91 mg, 1.28 mmol) under N 2 . The mixture was stirred at 80 °C for 5 hrs under N 2 . The mixture was diluted with EA (400 mL) and filtered. The filtrate was washed with aq. CaCl2 (100 mL) three times and brine (100 mL) twice, dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by column chromatography on silica gel (PE: EA= 2:1~1:5) to give compound 27a(2.7 g, 5.18 mmol, 81.1% yield) as a white solid. MS obsd.: 520.1 (MH + ). Step 2: [3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1 -yl]sulfonylphenyl]-2- oxo-oxazolidin-5-yl]methyl 4-methylbenzenesulfonate (compound 27b) To a solution of compound 27a (1.4 g, 2.69 mmol), p-toluenesulfonic anhydride (1.75 g, 5.38 mmol) and DIPEA (1.04 g, 8.06 mmol) in THF (8 mL) were added DMAP (65.67 mg, 0.540 mmol) under nitrogen. The mixture was stirred at 45 °C for 12 hrs under N 2 and then diluted with EA (300 mL). The solution washed with aq. K 2 CO 3 (100 mL), 1N aq. HCl (100 mL) twice and brine (100 mL) twice. The organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by column chromatography on silica gel (PE: EA= 2:1~1:5) to give compound 27b (1.7 g, 2.52 mmol, 93.69% yield) as a white solid. MS obsd.: 675.2 (MH + ). Step 3: 3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-5- (methylaminomethyl)oxazolidin-2-one (Example 27) To a solution of compound 27b (120.0 mg, 0.18 mmol) and KI (88.5 mg, 0.53 mmol) in DMSO (1.5 mL) were added monomethylamine in THF (3.0 mL, 6 mmol) and 18-crown-6 (4.7 mg, 0.02 mmol) under N2. The mixture was stirred at 60 °C for 3 hrs under N2. The mixture was diluted with EA (200 mL) and filtered. The filtrate was washed with aq CaCl 2 (50 mL) three times and brine (100 mL) two times, dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by prep-HPLC to give Example 27 (23.4 mg, 0.040 mmol, 22.64% yield) as a white solid. MS obsd.: 534.3 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 8.99 - 9.90 (m, 2 H), 7.75 - 7.84 (m, 4 H), 7.06 - 7.13 (m, 1 H), 6.94 - 6.99 (m, 1 H), 5.04 - 5.20 (m, 1 H), 4.20 - 4.34 (m, 1 H), 3.91 - 4.03 (m, 1 H), 3.61 - 3.79 (m, 4 H), 3.30 - 3.36 (m, 2 H), 2.86 - 3.06 (m, 4 H), 2.56 - 2.63 (m, 3 H). Example 28 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-5- [(dimethylamino)methyl]oxazolidin-2-one The title compound was prepared in analogy to the preparation of Example 27 by using dimethylamine instead of monomethylamine. Example 28 (17.1 mg) was obtained as a white solid. MS obsd.: 548.3 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 10.77 - 11.01 (m, 1 H), 7.73 - 7.86 (m, 4 H), 7.08 - 7.12 (m, 1 H), 6.95 - 7.00 (m, 1 H), 5.20 - 5.35 (m, 1 H), 4.21 - 4.35 (m, 1 H), 3.84 - 3.95 (m, 1 H), 3.50 - 3.75 (m, 6 H), 2.93 - 3.06 (m, 4 H), 2.80 - 2.90 (m, 6 H). Example 29 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-5- (piperazin-1-ylmethyl)oxazolidin-2-one The title compound was prepared in analogy to the preparation of Example 27 by using piperazine instead of monomethylamine. Example 29 (49.9 mg) was obtained as a white solid. MS obsd.: 589.3 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 9.51 - 10.01 (m, 2 H), 7.72 - 7.90 (m, 4 H), 7.07 - 7.14 (m, 1 H), 6.93 - 7.00 (m, 1 H), 5.16 - 5.38 (m, 1 H), 4.24 - 4.34 (m, 1 H), 3.49 - 3.92 (m, 14 H), 2.90 - 3.02 (m, 4 H). Example 30 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-5-[[cis-3,4- dihydroxypyrrolidin-1-yl]methyl]oxazolidin-2-one The title compound was prepared in analogy to the preparation of Example 27 by using cis-pyrrolidine-3,4-diol hydrochloride instead of monomethylamine. Example 30 (25.5 mg) was obtained as a white solid. MS obsd.: 606.2 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 10.69 - 11.44 (m, 1 H), 7.75 - 7.83 (m, 4 H), 7.08 - 7.13 (m, 1 H), 6.95 - 7.01 (m, 1 H), 5.09 - 5.71 (m, 3 H), 4.03 - 4.37 (m, 3 H), 3.59 - 3.94 (m, 8 H), 3.37 - 3.48 (m, 2 H), 2.92 - 3.18 (m, 5 H). Example 31 5-[(3-Aminoazetidin-1-yl)methyl]-3-[4-[4-[6-chloro-4-(triflu oromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]oxazolidin-2-one The title compound was prepared according to the following scheme: Step 1: tert-butyl N-[1-[[3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piper azin-1- yl]sulfonylphenyl]-2-oxo-oxazolidin-5-yl]methyl]azetidin-3-y l]carbamate(compound 31a) To a solution of compound 27b (220.0 mg, 0.330 mmol), 3-N-Boc-amino-azetidine (168.37 mg, 0.980 mmol) and KI (0.05 mL, 0.980 mmol) in ACN (5 mL) was added K 2 CO 3 (135.11 mg, 0.980 mmol) under N2. The mixture was stirred at 70 °C for 8 hrs. The mixture was diluted with EA (200 mL) and filtered. The filtrate was washed with brine (100 mL) two times, dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by prep-HPLC to give compound 31a (90 mg, 0.130 mmol, 40.91% yield) as a white solid. MS obsd.: 675.0 (MH + ). Step 2: 5-[(3-aminoazetidin-1-yl)methyl]-3-[4-[4-[6-chloro-4-(triflu oromethyl)-2-pyridyl] piperazin-1-yl]sulfonylphenyl]oxazolidin-2-one (Example 31) To a solution of compound 31a (90.0 mg, 0.130 mmol) in DCM (2 mL) was added TFA (1.0 mL, 12.98 mmol) and the reaction was stirred at 25°C for 2 hrs. The mixture was concentrated and the crude was purified by prep-HPLC to give Example 31 (40 mg, 0.060 mmol, 43.06% yield) as a white solid. MS obsd.: 575.2 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 8.29 - 8.76 (m, 2 H), 7.74 - 7.86 (m, 4 H), 7.07 - 7.14 (m, 1 H), 6.94 - 7.02 (m, 1 H), 4.86 - 5.02 (m, 1 H), 3.83 - 4.31 (m, 9 H), 3.70 - 3.76 (m, 4 H), 2.89 - 3.02 (m, 4 H). Example 32 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-5-(2,6- diazaspiro[3.3]heptan-2-ylmethyl)oxazolidin-2-one The title compound was prepared in analogy to the preparation of Example 31 by using tert-butyl 2,6-diazaspiro[3.3]heptane-2-carboxylate instead of 3-N-Boc-amino-azetidine. Example 32 (95.7 mg) was obtained as a white solid. MS obsd.: 601.2 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 8.84 - 9.07 (m, 2 H), 7.73 - 7.87 (m, 4 H), 7.07 - 7.12 (m, 1 H), 6.93 - 6.99 (m, 1 H), 4.89 - 5.03 (m, 1 H), 4.31 - 4.45 (m, 4 H), 4.11 - 4.28 (m, 5 H), 3.82 - 3.88 (m, 1 H), 3.70 - 3.73 (m, 4 H), 3.53 - 3.62 (m, 2 H), 2.89 - 3.03 (m, 4 H). Example 33 5-[[3-(Aminomethyl)azetidin-1-yl]methyl]-3-[4-[4-[6-chloro-4 -(trifluoromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]oxazolidin-2-one The title compound was prepared in analogy to the preparation of Example 31 by using tert-butyl N-(azetidin-3-ylmethyl)carbamate instead of 3-N-Boc-amino-azetidine. Example 33 (95.7 mg) was obtained as a white solid. MS obsd.: 589.2 (MH + ). + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 10.55 - 11.53 (m, 1 H), 7.96 - 8.22 (m, 3 H), 7.72 - 7.86 (m, 4 H), 7.06 - 7.14 (m, 1 H), 6.93 - 6.99 (m, 1 H), 4.89 - 5.07 (m, 1 H), 3.98 - 4.34 (m, 5 H), 3.82 - 3.90 (m, 1 H), 3.72 (br d, J =4.4 Hz, 4 H), 3.59 - 3.67 (m, 2 H), 2.90 - 3.26 (m, 7 H). Example 34 3-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-5- [(pyrrolidin-3-ylamino)methyl]oxazolidin-2-one The title compound was prepared in analogy to the preparation of Example 31 by using N- Boc-3-aminopyrrolidine instead of 3-N-Boc-amino-azetidine. Example 34 (57.3 mg) was obtained as a white solid. MS obsd.: 589.2 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 8.96 - 9.61 (m, 2 H), 7.73 - 7.87 (m, 4 H), 7.06 - 7.14 (m, 1 H), 6.92 - 7.00 (m, 1 H), 4.93 - 5.11 (m, 1 H), 4.20 - 4.35 (m, 1 H), 3.85 - 3.96 (m, 2 H), 3.70 - 3.75 (m, 4 H), 3.56 (br dd, J =12.0, 7.6 Hz, 1 H), 3.22 - 3.48 (m, 5 H), 2.91 - 3.02 (m, 4 H). Example 35 4-[[3-(Aminomethyl)azetidin-1-yl]methyl]-1-[4-[4-[6-chloro-4 -(trifluoromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 31 by using 4- (hydroxymethyl)pyrrolidin-2-one instead of 5-(hydroxymethyl)oxazolidin-2-one and tert-butyl N-(azetidin-3-ylmethyl)carbamate instead of 3-N-Boc-amino-azetidine. Example 35 (15 mg) was obtained as a white solid. MS obsd.: 587.2 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 9.71 - 11.11 (m, 1H), 7.84 - 7.95 (m, 4H), 7.73 - 7.82 (m, 2H), 7.09 (s, 1H), 6.98 (s, 1H), 3.81 - 4.30 (m, 5H), 3.68 - 3.75 (m, 4H), 3.64 (dd, J = 6.4, 9.2 Hz, 1H), 3.16 - 3.23 (m, 1H), 3.01 - 3.15 (m, 2H), 2.96 (t, J = 4.8 Hz, 4H), 2.62 - 2.82 (m, 2H), 2.39 - 2.55 (m, 3H). Example 36 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-[(4- methylpiperazin-1-yl)methyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 27 by using 4- (hydroxymethyl)pyrrolidin-2-one instead of 5-(hydroxymethyl)oxazolidin-2-one and 1- methylpiperazine instead of monomethylamine. Example 36 (25 mg) was obtained as a white solid. MS obsd.: 600.9 (MH + ). 1 H NMR (400 MHz, CD3OD): δ ppm 7.88 - 7.93 (m, 2H), 7.75 - 7.83 (m, J = 9.2 Hz, 2H), 6.91 (s, 1H), 6.82 (s, 1H), 4.03 (dd, J = 7.6, 9.6 Hz, 1H), 3.67 - 3.77 (m, 5H), 3.01 - 3.0114 (m, 4H), 2.62 - 2.87 (m, 3H), 2.39 - 2.356 (m, 10H), 2.31 (s, 3H). Example 37 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-[[3- [(dimethylamino)methyl]azetidin-1-yl]methyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 27 by using 4- (hydroxymethyl)pyrrolidin-2-one instead of 5-(hydroxymethyl)oxazolidin-2-one and 1-(azetidin- 3-yl)-N,N-dimethyl-methanamine instead of monomethylamine. Example 37 (25 mg) was obtained as a white solid. MS obsd.: 614.9 (MH + ). 1 H NMR (400 MHz, CD3OD): δ ppm 7.86 - 7.93 (m, 2H), 7.75 - 7.85 (m, 2H), 6.91 (s, 1H), 6.82 (s, 1H), 4.25 - 4.41 (m, 2H), 3.95 - 4.14 (m, 3H), 3.69 - 3.80 (m, 5H), 3.48 (dd, J = 1.6, 3.2 Hz, 2H), 3.34 - 3.42 (m, 2H), 3.11 - 3.16 (m, 1H), 3.03 - 3.10 (m, 4H), 2.88 (s, 6H), 2.72 - 2.86 (m, 2H), 2.42 - 2.55 (m, 1H). Example 38 4-[(Azetidin-3-ylmethylamino)methyl]-1-[4-[4-[6-chloro-4-(tr ifluoromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 31 by using 4- (hydroxymethyl)pyrrolidin-2-one instead of 5-(hydroxymethyl)oxazolidin-2-one and tert-butyl 3-(aminomethyl)azetidine-1-carboxylate instead of 3-N-Boc-amino-azetidine. Example 38 (23 mg) was obtained as a white solid. MS obsd.: 587.0 (MH + ). 1 H NMR (400 MHz, CD3OD): δ ppm 7.86 - 7.92 (m, 2H), 7.77 - 7.84 (m, 2H), 6.91 (s, 1H), 6.82 (s, 1H), 4.10 - 4.24 (m, 3H),3.99 - 4.07 (m, 2H), 3.67 - 3.83 (m, 5H), 3.37 - 3.39 (m, 3H), 3.16 - 3.27 (m, 2H), 3.04 - 3.11 (m, 4H), 2.81 - 2.94 (m, 2H), 2.47 - 2.60 (m, 1H). Example 39 4-[[3-(1-Aminocyclopropyl)azetidin-1-yl]methyl]-1-[4-[4-[6-c hloro-4-(trifluoromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 31 by using 4- (hydroxymethyl)pyrrolidin-2-one instead of 5-(hydroxymethyl)oxazolidin-2-one and tert-butyl N-[1-(azetidin-3-yl)cyclopropyl]carbamate instead of 3-N-Boc-amino-azetidine. Example 39 (25 mg) was obtained as a white solid. MS obsd.: 612.9 (MH + ). 1 H NMR (400 MHz, CD3OD): δ ppm 7.87 - 7.94 (m, 2H), 7.76 - 7.82 (m, 2H), 6.91 (s, 1H), 6.82 (s, 1H), 4.00 (dd, J = 7.6, 9.8 Hz, 1H), 3.61 - 3.78 (m, 5H), 3.38 - 3.47 (m, 2H), 3.04 - 3.11 (m, 4H), 2.95 (t, J = 7.6 Hz, 2H), 2.72 (dd, J = 8.0, 16.8 Hz, 1H), 2.46 - 2.66 (m, 4H), 2.29 - 2.44 (m, 1H), 0.47 - 0.67 (m, 4H). Example 40 4-[(3-Aminoazetidin-1-yl)methyl]-1-[4-[4-[6-chloro-4-(triflu oromethyl)-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 31 by using 4- (hydroxymethyl)pyrrolidin-2-one instead of 5-(hydroxymethyl)oxazolidin-2-one and tert-butyl N-(azetidin-3-yl)carbamate instead of 3-N-Boc-amino-azetidine. Example 40 (25 mg) was obtained as a white solid. MS obsd.: 573.0 (MH + ). 1 H NMR (400 MHz, CD 3 OD): δ ppm 7.89 (d, J = 8.8 Hz, 2H), 7.74 - 7.83 (m, 2H), 6.91 (s, 1H), 6.82 (s, 1H), 4.00 (dd, J = 7.6, 9.6 Hz, 1H), 3.62 - 3.77 (m, 7H), 3.57 (quin, J = 6.8 Hz, 1H), 3.03 - 3.11 (m, 4H), 2.85 (dt, J = 4.0, 7.2 Hz, 2H), 2.50 - 2.77 (m, 4H), 2.33 - 2.45 (m, 1H). Example 41 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4- (piperazin-1-ylmethyl)pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 31 by using 4- (hydroxymethyl)pyrrolidin-2-one instead of 5-(hydroxymethyl)oxazolidin-2-one and tert-butyl piperazine-1-carboxylate instead of 3-N-Boc-amino-azetidine. Example 41 (25 mg) was obtained as a white solid. MS obsd.: 587.0 (MH + ). 1 H NMR (400 MHz, CD 3 OD): δ ppm 7.88 - 7.94 (m, 2H), 7.80 (d, J = 8.8 Hz, 2H), 6.91 (s, 1H), 6.82 (s, 1H), 4.05 (dd, J = 7.6, 9.8 Hz, 1H), 3.68 - 3.77 (m, 5H), 3.23 (t, J = 5.2 Hz, 4H), 3.04 - 3.11 (m, 4H), 2.65 - 2.85 (m, 6H), 2.57 (d, J = 3.2 Hz, 2H), 2.38 - 2.50 (m, 1H). Example 42 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-[[3- (dimethylamino)azetidin-1-yl]methyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 27 by using 4- (hydroxymethyl)pyrrolidin-2-one instead of 5-(hydroxymethyl)oxazolidin-2-one and N,N- dimethylazetidin-3-amine instead of monomethylamine. Example 42 (25 mg) was obtained as a white solid. MS obsd.: 600.1 (MH + ). 1 H NMR (400 MHz, CD3OD): δ ppm 7.86 - 7.892 (m, 2H), 7.77 - 7.84 (m, 2H), 6.91 (s, 1H), 6.82 (s, 1H), 4.06 (t, J = 8.8 Hz, 3H), 3.82 (d, J = 2.8 Hz, 3H), 3.67 - 3.76 (m, 5H), 3.11 - 3.18 (m, 1H), 3.00 - 3.11 (m, 5H), 2.63 - 2.85 (m, 8H), 2.41 - 2.55 (m, 1H). Example 43 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-[[3- (methylamino)azetidin-1-yl]methyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 31 by using 4- (hydroxymethyl)pyrrolidin-2-one instead of 5-(hydroxymethyl)oxazolidin-2-one and tert-butyl N-(azetidin-3-yl)-N-methyl-carbamate instead of 3-N-Boc-amino-azetidine. Example 43 (25 mg) was obtained as a white solid. MS obsd.: 587.0 (MH + ). 1 H NMR (400 MHz, CD 3 OD): δ ppm 7.86 - 7.93 (m, 2H), 7.75 - 7.84 (m, 2H), 6.91 (s, 1H), 6.82 (s, 1H), 3.98 - 4.15 (m, 4H), 3.80 - 3.97 (m, 2H), 3.68 - 3.78 (m, 5H), 2.98 - 3.12 (m, 6H), 2.74 - 2.85 (m, 1H), 2.70 (s, 4H), 2.41 - 2.53 (m, 1H). Example 44 1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4- (methylamino)pyrrolidin-2-one To a solution of intermediate B (100.0 mg, 0.210 mmol) in THF (3 mL) was added methylamine (1.03 mL, 2.05 mmol) and the mixture was heated to 90 °C for 1 h. The mixture was concentrated and the crude was purified by prep-HPLC to give Example 44 (25 mg, 0.050 mmol, 23.2% yield) as a white solid. MS obsd.: 518.3 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 9.42 - 9.61 (m, 2H), 7.91 (s, 2H), 7.79 (d, J = 9.2 Hz, 2H), 7.09 (s, 1H), 6.97 (s, 1H), 4.21 - 4.28 (m, 1H), 4.04 - 4.10 (m, 1H), 3.95 - 4.03 (m, 1H), 3.71 (br s, 4H), 3.01 - 3.09 (m, 1H), 2.97 (br s, 4H), 2.80 - 2.88 (m, 1H), 2.61 (br s, 3H). Example 45 4-(2-Aminoethylamino)-1-[4-[4-[6-chloro-4-(trifluoromethyl)- 2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 44 by using ethylenediamine instead of monomethylamine. Example 45 (18 mg) was obtained as a white solid. MS obsd.: 487.0 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 10.21 - 10.97 (m, 1 H), 7.58 - 8.52 (m, 4 H), 6.73 - 7.31 (m, 2 H), 3.59 - 4.13 (m, 4 H), 2.91 - 3.27 (m, 3 H), 1.97 - 2.24 (m, 3 H). Example 46 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-(2- hydroxyethylamino)pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 44 by using hydroxyethylamine instead of monomethylamine. Example 46 (35 mg) was obtained as a white solid. MS obsd.: 548.2 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 9.31 - 9.47 (m, 2H), 7.90 (s, 2H), 7.81 (s, 2H), 7.09 (s, 1H), 6.97 (s, 1H), 5.27 - 5.38 (m, 1H), 4.21 - 4.29 (m, 1H), 4.03 - 4.15 (m, 2H), 3.66 - 3.74 (m, 6H), 3.08 - 3.13 (m, 2H), 3.01 - 3.07 (m, 1H), 2.97 (br t, J = 4.8 Hz, 4H), 2.82 - 2.90 (m, 1H). Example 47 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-piperazin- 1-yl-pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 44 by using piperazine instead of monomethylamine. Example 47 (11 mg) was obtained as a white solid. MS obsd.: 573.1 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 9.33 - 9.50 (m, 1H), 7.90 - 7.96 (m, 2H), 7.80 (s, 2H), 7.08 (s, 1H), 6.97 (s, 1H), 4.16 - 4.25 (m, 1H), 4.00 - 4.10 (m, 1H), 3.80 - 3.94 (m, 2H), 3.71 (br s, 4H), 3.29 - 3.35 (m, 4H), 3.08 - 3.24 (m, 4H), 3.00 - 3.01 (m, 1H), 2.95 - 2.99 (m, 4H), 2.73 - 2.92 (m, 2H). Example 48 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-[(3S,4R)- 3,4-dihydroxypyrrolidin-1-yl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 44 by using - [(3S,4R)-pyrrolidine-3,4-diol instead of monomethylamine. Example 48 (45 mg) was obtained as a white solid. MS obsd.: 590.0 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.90 - 7.97 (m, 2H), 7.70 - 7.78 (m, 2H), 7.08 (s, 1H), 6.93 - 6.99 (m, 1H), 4.56 - 4.63 (m, 2H), 3.86 - 3.99 (m, 3H), 3.66 - 3.75 (m, 5H), 3.14 - 3.23 (m, 1H), 2.87 - 3.00 (m, 6H), 2.68 - 2.75 (m, 1H), 2.52 (br s, 1H), 2.33 - 2.40 (m, 2H). Example 49 4-[3-(Aminomethyl)azetidin-1-yl]-1-[4-[4-[6-chloro-4-(triflu oromethyl)-2-pyridyl]piperazin- 1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared according to the following scheme: Step 1: tert-butyl N-[1-[1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pipera zin-1- yl]sulfonylphenyl]-5-oxo-pyrrolidin-3-yl]azetidin-3-yl]carba mate (compound 49a) To a solution of intermediate B (80.0 mg, 0.16 mmol) in DMF (1 mL) was added tert-butyl N-(azetidin-3-ylmethyl)carbamate (84.89 mg, 0.49 mmol) and the mixture was heated to 90 °C for 1 h. The mixture was diluted with water (20 mL) and then extracted with EA (10 mL) three times. The combined organic layer was washed with brine (50 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated. The crude was purified by prep-HPLC to give compound 49a (85 mg, 0.130 mmol, 78.49% yield) as a white solid. MS obsd.: 673.2 (MH + ). Step 2: 4-[3-(aminomethyl)azetidin-1-yl]-1-[4-[4-[6-chloro-4-(triflu oromethyl)-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one (Example 49) To a solution of compound 49a (80.0 mg, 0.12 mmol) in DCM (1 mL) was added TFA (0.5 mL, 6.49 mmol) in one portion. The mixture was stirred at 20 °C for 1 h and then concentrated. The crude was purified by prep-HPLC to give Example 49 (45 mg, 0.070 mmol, 58.2% yield) as a white solid. MS obsd.: 573.1 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 8.11 - 8.16 (m, 1H), 7.89 - 7.94 (m, 2H), 7.73 - 7.77 (m, 2H), 7.08 (s, 1H), 6.95 - 6.98 (m, 1H), 3.90 - 4.24 (m, 2H), 3.69 - 3.73 (m, 4H), 3.58 - 3.66 (m, 1H), 3.37 - 3.42 (m, 4H), 3.01 - 3.07 (m, 2H), 2.94 - 2.97 (m, 4H), 2.51 - 2.53 (m, 2H). Example 50 4-(3-Aminopyrrolidin-1-yl)-1-[4-[4-[6-chloro-4-(trifluoromet hyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 49 by using tert-butyl N-[pyrrolidin-3-yl]carbamate instead of tert-butyl N-(azetidin-3-ylmethyl)carbamate. Example 50 (16 mg) was obtained as a white solid. MS obsd.: 573.3 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 8.49 - 8.73 (m, 3H), 8.48 - 8.49 (m, 1H), 7.91 - 7.97 (m, 2H), 7.77 - 7.82 (m, 2H), 7.05 - 7.12 (m, 1H), 6.95 - 6.99 (m, 1H), 4.25 - 4.45 (m, 2H), 4.12 - 4.24 (m, 1H), 3.79 - 4.10 (m, 2H), 3.71 (br s, 4H), 3.51 - 3.59 (m, 1H), 3.03 - 3.18 (m, 2H), 2.98 (br s, 4H), 2.33 (br d, J = 1.6 Hz, 2H). Example 51 4-(3-Aminoazetidin-1-yl)-1-[4-[4-[6-chloro-4-(trifluoromethy l)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 49 by using tert-butyl N-(azetidin-3-yl)carbamate instead of tert-butyl N-(azetidin-3-ylmethyl)carbamate. Example 51 (21 mg) was obtained as a white solid. MS obsd.: 559.1 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 8.24 - 8.30 (m, 1H), 7.90 (s, 2H), 7.73 (d, J = 8.8 Hz, 2H), 7.08 (s, 1H), 6.96 (s, 1H), 3.89 (s, 1H), 3.69 - 3.72 (m, 4H), 3.50 - 3.52 (m, 4H), 3.05 - 3.08 (m, 1H), 2.96 (br t, J = 4.8 Hz, 4H), 2.67 - 2.75 (m, 3H), 2.16 - 2.22 (m, 1H). Example 52 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-(2,6- diazaspiro[3.3]heptan-2-yl)pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 49 by using tert-butyl 2,6-diazaspiro[3.3]heptane-2-carboxylate instead of tert-butyl N-(azetidin-3-ylmethyl) carbamate. Example 52 (26 mg) was obtained as a white solid. MS obsd.: 585.3 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 8.68 - 8.81 (m, 2H), 7.87 (s, 2H), 7.78 (d, J = 8.8 Hz, 2H), 7.09 (s, 1H), 6.97 (s, 1H), 4.16 - 4.40 (m, 4H), 4.12 (br s, 4H), 3.78 - 3.84 (m, 1H), 3.69 - 3.74 (m, 4H), 2.98 - 3.05 (m, 1H), 2.96 (br s, 4H), 2.52 - 2.61 (m, 2H). Example 53 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-[(3S)-3- (hydroxymethyl)piperazin-1-yl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 49 by using tert-butyl (2S)-2-(hydroxymethyl)piperazine-1-carboxylate instead of tert-butyl N-(azetidin-3- ylmethyl)carbamate. Example 53 (35 mg) was obtained as a white solid. MS obsd.: 603.3 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 9.34 - 9.58 (m, 1H), 8.93 - 9.26 (m, 1H), 7.93 (d, J = 8.8 Hz, 2H), 7.78 (d, J = 8.8 Hz, 2H), 7.04 - 7.10 (m, 1H), 6.97 (s, 1H), 4.11 - 4.21 (m, 1H), 3.92 - 4.02 (m, 1H), 3.76 - 3.64 (m, 7H), 3.55 - 3.63 (m, 4H), 3.06 - 3.21 (m, 2H), 2.97 (br t, J = 4.4 Hz, 4H), 2.85 - 2.92 (m, 1H), 2.70 - 2.84 (m, 2H), 2.54 - 2.69 (m, 1H). Example 54 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-[(3R)-3- (hydroxymethyl)piperazin-1-yl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 49 by using tert-butyl (2R)-2-(hydroxymethyl)piperazine-1-carboxylate instead of tert-butyl N-(azetidin-3- ylmethyl)carbamate. Example 54 (65 mg) was obtained as a white solid. MS obsd.: 603.1 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 9.57 - 9.69 (m, 1H), 9.18 - 9.38 (m, 1H), 7.93 (s, 2H), 7.79 (d, J = 8.8 Hz, 2H), 7.09 (s, 1H), 6.97 (s, 1H), 4.18 (br s, 1H), 3.99 - 4.06 (m, 1H), 3.67 - 3.77 (m, 7H), 3.58 - 3.66 (m, 4H), 3.14 - 3.25 (m, 2H), 2.98 (br t, J = 4.4 Hz, 4H), 2.70 - 2.94 (m, 4H). Example 55 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-[(1S,4S)- 2,5-diazabicyclo[2.2.1]heptan-2-yl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 49 by using (1S, 4S)-2-Boc-2,5-diazabicyclo[2.2.1]heptane instead of tert-butyl N-(azetidin-3- ylmethyl)carbamate. Example 55 (22 mg) was obtained as a white solid. MS obsd.: 589.0 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 9.31 - 10.01 (m, 2H), 7.93 (d, J = 8.8 Hz, 2H), 7.79 (d, J = 8.8 Hz, 2H), 7.09 (s, 1H), 6.97 (s, 1H), 4.34 - 4.61 (m, 2H), 3.96 - 4.29 (m, 3H), 3.57 - 3.75 (m, 6H), 2.86 - 3.07 (m, 6H), 1.92 - 2.37 (m, 2H). Example 56 4-[(3R,4R)-3-amino-4-hydroxy-pyrrolidin-1-yl]-1-[4-[4-[6-chl oro-4-(trifluoromethyl)-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 49 by using tert-butyl N-[(3R, 4R)-4-hydroxypyrrolidin-3-yl]carbamate instead of tert-butyl N-(azetidin-3- ylmethyl) carbamate. Example 56 (31 mg) was obtained as a white solid. MS obsd.: 589.1 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 8.54 - 9.03 (m, 3H), 7.91 - 7.97 (m, 2H), 7.80 (s, 2H), 7.06 - 7.10 (m, 1H), 6.94 - 6.98 (m, 1H), 4.43 - 4.53 (m, 1H), 4.23 - 4.29 (m, 1H), 4.08 - 4.21 (m, 1H), 3.64 - 3.86 (m, 7H), 3.00 - 3.13 (m, 2H), 2.98 (br d, J = 4.4 Hz, 4H). Example 57 4-((3R,4R)-3-amino-4-hydroxypiperidin-1-yl)-1-(4-((4-(6-chlo ro-4-(trifluoromethyl)pyridin- 2-yl)piperazin-1-yl)sulfonyl)phenyl)pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 49 by using tert-butyl N-[(3R, 4R)-4-hydroxy-3-piperidyl]carbamate instead of tert-butyl N-(azetidin-3- ylmethyl) carbamate. Example 57 (20.1 mg) was obtained as a white solid. MS obsd.: 603.2 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 8.28 - 8.65 (m, 3H), 7.93 (s, 2H), 7.80 (br d, J = 1.2 Hz, 2H), 7.08 (s, 1H), 6.96 (s, 1H), 3.89 - 4.42 (m, 4H), 3.58 - 3.87 (m, 7H), 3.18 - 3.25 (m, 1H), 2.94 - 3.01 (m, 1H), 2.98 (br d, J = 4.4 Hz, 7H), 1.97 - 2.12 (m, 1H), 1.70 - 1.93 (m, 1H). Example 58 4-(Azetidin-3-ylamino)-1-[4-[4-[6-chloro-4-(trifluoromethyl) -2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: tert-butyl 3-[[1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazi n-1- yl]sulfonyl phenyl]-5-oxo-pyrrolidin-3-yl]amino]azetidine-1-carboxylate (compound 58a) A solution of Example 1 (120.0 mg, 0.24 mmol), tert-butyl 3-oxoazetidine-1-carboxylate (203.8 mg, 1.2 mmol) in methanol (0.42 mL) and THF (0.42 mL) was added anhydrous Na 2 SO 4 (85.01 mg, 0.710 mmol). The mixture was stirred at 25 °C for 2 hrs and then NaBH₃CN (104.75 mg, 1.67 mmol) was added and the mixture was stirred at 45 °C for 6 hrs. The mixture was diluted with water (100 mL) and extracted with EA (100 mL) three times. The combined organic layer was washed with brine (100 mL) two times, dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by prep-HPLC (FA as additive) to give compound 58a (90 mg, 0.140 mmol, 57.34% yield) as a white solid. MS obsd.: 659.0 (MH + ). Step 2: 4-(azetidin-3-ylamino)-1-[4-[4-[6-chloro-4-(trifluoromethyl) -2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one(Example 58) To a solution of compound 58a (80.0 mg, 0.12 mmol) in DCM (2 mL) was added TFA (1.0 mL, 12.98 mmol). The mixture was stirred at 25°C for 2 hrs and then concentrated. The crude was dissolved in methanol (3.0 mL) and basified with ammonium hydroxide to pH 7.0. The solution was purified by prep-HPLC to give Example 58 (29.6 mg, 0.05 mmol, 39.7% yield) as a white solid. MS obsd.: 559.2 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 8.87 - 9.87 (m, 2 H), 7.71 - 7.99 (m, 4 H), 7.06 - 7.14 (m, 1 H), 6.93 - 7.00 (m, 1 H), 3.66 - 4.42 (m, 12 H), 2.88 - 3.08 (m, 5 H), 2.56 - 2.75 (m, 1 H). Example 59 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4- (pyrrolidin-3-ylamino)pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 58 by using tert-butyl 3-oxopyrrolidine-1-carboxylate instead of tert-butyl 3-oxoazetidine-1-carboxylate. Example 59 (33.5 mg) was obtained as a white solid. MS obsd.: 573.0 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 10.17 - 10.81 (m, 2 H), 9.29 - 9.92 (m, 2 H), 7.88 - 7.96 (m, 2 H), 7.77 - 7.86 (m, 2 H), 7.08 - 7.14 (m, 1 H), 6.95 - 7.01 (m, 1 H), 3.98 - 4.38 (m, 4 H), 3.68 - 3.81 (m, 4 H), 3.44 - 3.64 (m, 3 H), 2.91 - 3.28 (m, 7 H), 2.13 - 2.41 (m, 2 H). Example 60 1-[4-[4-[6-Chloro-4-(trifluoromethyl)-2-pyridyl]piperazin-1- yl]sulfonylphenyl]-4-(4- piperidylamino)pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 58 by using tert-butyl 4-oxopiperidine-1-carboxylate instead of tert-butyl 3-oxoazetidine-1-carboxylate. Example 60 (58 mg) was obtained as a white solid. MS obsd.: 587.3 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 8.77 - 10.68 (m, 4 H), 7.75 - 7.96 (m, 4 H), 7.05 - 7.16 (m, 1 H), 6.93 - 7.01 (m, 1 H), 4.07 - 4.38 (m, 3 H), 3.65 - 3.81 (m, 4 H), 3.39 - 3.61 (m, 3 H), 2.82 - 3.10 (m, 8 H), 2.14 - 2.29 (m, 2 H), 1.82 - 2.02 (m, 2 H). Example 61 4-Amino-1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pipe razin-1-yl]sulfonylphenyl]-5- methyl-pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 1 by using tert- butyl N-(2-methyl-5-oxo-pyrrolidin-3-yl)carbamate instead of tert-butyl (5-oxopyrrolidin-3- yl)carbamate. Example 61 (20 mg) was obtained as a white solid. MS obsd.: 518.2 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 7.82 - 7.93 (m, 2H), 7.70 - 7.79 (m, 2H), 7.08 (s, 1H), 6.97 (s, 1H), 3.91 - 4.17 (m, 3H), 3.65 - 378 (m, 6H), 2.92 - 3.04 (m, 5H), 2.22 (dd, J = 2.8, 17.2 Hz, 1H), 1.08 - 1.24 (m, 3H). Example 62 4-Amino-1-[5-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pipe razin-1-yl]sulfonyl-2- pyridyl]pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: 1-[(6-bromo-3-pyridyl)sulfonyl]-4-[6-chloro-4-(trifluorometh yl)-2-pyridyl] piperazine (compound 62a) A mixture of intermediate 1a (207.14 mg, 0.780 mmol), 6-bromo-pyridine-3-sulfonyl chloride (200.0 mg, 0.780 mmol) and DIPEA (0.27 mL, 1.56 mmol) in DCM (6.67 mL) was stirred at 25 °C for 2 hrs. After the reaction was completed, the solvent was removed under vacuum and the crude was purified by column chromatography on silica gel (PE: EA = 5:2) to give compound 62a (300 mg, 75.25% yield) as a white solid. MS obsd.: 485.0 (MH + ). Step 2 : tert-butyl N-[1-[5-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazin -1- yl]sulfonyl-2-pyridyl]-5-oxo-pyrrolidin-3-yl]carbamate (compound 62b) A mixture of tert-butyl N-(5-oxopyrrolidin-3-yl)carbamate (41.2 mg, 0.21 mmol), CuI (0.02 mmol), Cs 2 CO 3 (134.2 mg, 0.41 mmol), compound 62a (100.0 mg, 0.21 mmol) in DMF (2 mL) was heated at 85 °C in a microwave reactor for 1 h. The mixture was poured into ice water, extracted with DCM (15 mL) twice, dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by column chromatography on silica gel to give compound 62b (80 mg, 0.130 mmol, 61.01% yield) as a white solid. MS obsd.: 605.2 (MH + ). Step 3: 4-amino-1-[5-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]pipe razin-1-yl]sulfonyl-2- pyridyl]pyrrolidin-2-one (Example 62) A mixture of compound 62b (60.0 mg, 0.10 mmol) and HCl in EA (0.05 mL, 0.10 mmol) was stirred at 25 °C for 2 h. After the reaction was completed, the solvent was removed under vacuum and the crude was purified by prep-HPLC to give Example 62 (6 mg, 0.01 mmol, 11.58% yield) as a white solid. MS obsd.: 505.1 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 8.73 (d, J = 2.4 Hz, 1H), 8.50 (d, J = 9.2 Hz, 1H), 8.18 - 8.25 (m, 3H), 7.09 (s, 1H), 6.96 (s, 1H), 4.23 (dd, J = 12.6, 7.2 Hz, 1H), 4.11 (dd, J = 12.6, 2.0 Hz, 1H), 4.03 (s, 1H), 3.66 - 3.76 (m, 4H), 3.15 (dd, J = 18.0, 8.0 Hz, 1H), 2.97 - 3.05 (m, 4H), 2.60 - 2.67 (m, 1H). Example 63 5-(Aminomethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyri dyl]piperazin-1-yl]sulfonyl-2- fluoro-phenyl]oxazolidin-2-one The title compound was prepared in analogy to the preparation of Example 1 by using tert- butyl N-[(2-oxooxazolidin-5-yl)methyl]carbamate instead of tert-butyl (5-oxopyrrolidin-3- yl)carbamate and 4-bromo-3-fluorobenzenesulfonyl chloride instead of 4-iodobenzenesulfonyl chloride. Example 63 (35 mg) was obtained as a white solid. MS obsd.: 538.1 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 8.17-8.31 (m, 2H), 7.77-7.83 (m, 1H), 7.72 (br dd, J = 13.2, 2.0 Hz, 1H), 7.55 (br dd, J = 8.8, 2.0 Hz, 1H), 7.10 (s, 1H), 6.98 (s, 1H), 4.62 - 4.72 (m, 2H), 4.11 (m, 1H), 3.90 (br dd, J = 9.2, 6.0 Hz, 1H), 3.67 - 3.77 (m, 4H), 3.06 - 3.16 (m, 4H). Example 64 5-(Aminomethyl)-3-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyri dyl]piperazin-1-yl]sulfonyl-3- fluoro-phenyl]oxazolidin-2-one The title compound was prepared in analogy to the preparation of Example 1 by using tert- butyl N-[(2-oxooxazolidin-5-yl)methyl]carbamate instead of tert-butyl (5-oxopyrrolidin-3- yl)carbamate and 4-bromo-2-fluorobenzenesulfonyl chloride instead of 4-iodobenzenesulfonyl chloride. Example 64 (45 mg) was obtained as a white solid. MS obsd.: 538.1 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.82-7.92 (m, 1H), 7.74 (dd, 1H, J = 2.0, 10.8 Hz), 7.67 (dd, 1H, J = 2.0, 8.4 Hz), 7.11 (s, 1H), 6.98 (s, 1H), 4.83 - 4.93 (m, 1H), 4.11 - 4.34 (m, 1H), 3.93 - 4.02 (m, 1H), 3.61 - 3.85 (m, 4H), 3.11 (br d, 2H, J = 6.0 Hz), 3.03 - 3.14 (m, 4H). Example 65 4-Amino-1-[4-[4-[4-methyl-6-(trifluoromethyl)pyrimidin-2-yl] piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 1 by using 2- chloro-4-methyl-6-(trifluoromethyl)pyrimidine instead of 2,6-dichloro-4-(trifluoromethyl) pyridine. Example 65 (14 mg) was obtained as a white solid. MS obsd.: 484.8 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 8.23(br s, 2H), 7.86 - 7.99 (m, 2H), 7.72 - 7.83 (m, 2H), 6.85 - 7.04 (m, 1H), 4.21 - 4.30 (m, 1H), 4.03 - 4.10 (m, 1H), 3.74 - 3.90 (m, 5H), 3.02 - 3.13 (m, 1H), 2.97 (br t, J = 4.8 Hz, 4H), 2.53 - 2.61 (m, 1H), 2.33 - 2.40 (m, 3H). Example 66 4-Amino-1-[4-[4-[4-(trideuteriomethyl)-6-(trifluoromethyl)py rimidin-2-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: tert-butyl N-[1-[4-[4-[4-chloro-6-(trifluoromethyl)pyrimidin-2-yl]piper azin-1- yl]sulfonylphenyl]-5-oxo-pyrrolidin-3-yl]carbamate (compound 66a) A mixture of intermediate C (0.320 g, 754 µmol), 2,4-dichloro-6-(trifluoromethyl) pyrimidine (164 mg, 754 µmol) and DIPEA (292 mg, 2.26 mmol) in THF (2 mL) was stirred at 90 °C overnight. The mixture was concentrated and the residue was diluted with EA, washed with 1 N HCl (10 mL). The organic layer was dried, concentrated and used directly in next step. MS obsd.: 605.4 (MH + ). Step 2: tert-butyl N-[5-oxo-1-[4-[4-[4-(trideuteriomethyl)-6-(trifluoromethyl)p yrimidin-2- yl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-3-yl]carbamate (compound 66b) A mixture of compound 66a (0.15 g, 248 µmol), (methyl-d3)boronic acid (31.2 mg, 496 µmol) and K 2 CO 3 (103 mg, 744 µmol), Ag 2 O(144 mg) and Pd(dppf)Cl 2 (18.1 mg) in THF (0.5 mL) was stirred at 80 °C overnight. The organic layer was concentrated and used directly in next step. MS obsd.: 588.3 (MH + ). Step 3: 4-amino-1-[4-[4-[4-(trideuteriomethyl)-6-(trifluoromethyl)py rimidin-2-yl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one (Example 66) A mixture of compound 66b (0.120 g, 204 µmol) and TFA (460 mg) in DCM (0.5 mL) was stirred at 25 °C 1h. The mixture was concentrated and the crude was purified by pre-HPLC to give 4.5 mg of Example 66. MS obsd.: 488.3 (MH + ). 1 H NMR (400 MHz, CD 3 OD): δ ppm 7.92 - 8.01 (m, 2H), 7.80 - 7.84 (m, 2H), 6.93 (s, 1H), 4.24 (dd, 1H, J = 6.4, 10.4 Hz), 3.94 (br d, 1H, J=3.2 Hz), 3.87 (br s, 4H), 3.74 (dd, 1H, J = 2.8, 10.4 Hz), 3.11 - 3.15 (m, 4H), 3.02 (dd, 1H, J = 7.6, 17.6 Hz), 2.51 (dd, 1H, J=3.6, 17.6 Hz). Example 67 1-[4-(4-Amino-2-oxo-pyrrolidin-1-yl)phenyl]sulfonyl-4-[6-chl oro-4-(trifluoromethyl)-2- pyridyl]piperazine-2-carbonitrile The title compound was prepared according to the following procedures: Step 1: tert-butyl N-[1-(4-benzylsulfanylphenyl)-5-oxo-pyrrolidin-3-yl]carbamat e (compound 67a) A mixture of tert-butyl N-(5-oxopyrrolidin-3-yl)carbamate (215.15 mg, 1.07 mmol), CuI (0.100 eq), Cs 2 CO 3 (700.19 mg, 2.15 mmol), 1-benzylsulfanyl-4-bromo-benzene (300.0 mg, 1.07 mmol) in DMF (10.44 mL) was heated at 85 °C in a microwave reactor for 1h. The mixture was poured into ice water, extracted with DCM (15mL) two times, dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by chromatography on silica gel to give compound 67a (300 mg, 0.750 mmol, 65.16% yield) as a white solid. MS obsd.: 399.2 (MH + ). Step 2: tert-butyl N-[1-(4-chlorosulfonylphenyl)-5-oxo-pyrrolidin-3-yl]carbamat e (compound 67b) To a mixture of compound 67a (200.0 mg, 0.500 mmol) in ACN (7.53 mL), AcOH (0.753 mL) and water (0.753 mL) was added 1,3-dichloro-5,5-dimethylhydantoin (197.75 mg, 1 mmol) at 25 °C for 1 h. After completion, the mixture was poured into brine (25 mL) and extracted by EA (15mL) twice, washed with brine (15 mL) twice. The organic layer was concentrated and the crude was purified by chromatography on silica gel (PE: EA =13:3) to give compound 67b (42 mg, 0.110 mmol, 20.09% yield) as colorless oil. MS obsd.: 397 (MNa + ). Step 3: tert-butyl 4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]-2-cyano-piperazin e-1- carboxylate (compound 67c) A mixture of 2,6-dichloro-4-(trifluoromethyl)pyridine (239.91 mg, 1.11 mmol) and tert- butyl 2-cyanopiperazine-1-carboxylate (200.0 mg, 1.11 mmol) was stirred at 100 °C for 12 h. Then water (30 mL) was added and the solution was extracted with EA, dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by prep-HPLC to give compound 67c (150 mg, 0.380 mmol, 31.1% yield). MS obsd.: 391.1 (MH + ). Step 4: 4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]piperazine-2-carbo nitrile (compound 67d) Compound 67c (150.0 mg, 0.380 mmol) was dissolved in 2M HCl/EA (10 mL) and the mixture was stirred at rt for 4 hrs. The mixture was concentrated and the crude was purified by prep-HPLC to give compound 67d (75 mg, 0.26 mmol, 60.5% yield). MS obsd.: 291.1 (MH + ). Step 5: tert-butyl N-[1-[4-[4-[6-chloro-4-(trifluoromethyl)-2-pyridyl]-2-cyano- piperazin-1- yl]sulfonylphenyl]-5-oxo-pyrrolidin-3-yl]carbamate (compound 67e) To a mixture of compound 67b (75.0 mg, 0.200 mmol) and pyridine (0.5 mL) in DCM (4.52 mL) was added compound 67d (42.0 mg, 0.140 mmol) and the reaction was stirred at rt for 8 hrs. Water (10 mL) was added and the mixture was extracted with EA, dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by prep-HPLC to give compound 67e (50 mg, 0.080 mmol, 49.51% yield). MS obsd.:651.1 (MNa + ). Step 6: 1-[4-(4-amino-2-oxo-pyrrolidin-1-yl)phenyl]sulfonyl-4-[6-chl oro-4-(trifluoro- methyl)-2-pyridyl]piperazine-2-carbonitrile (Example 67) Compound 67e (49.76 mg, 0.080 mmol) was dissolved in 4M HCl/1,4-dioxane(10 mL) and the mixture was stirred at rt for 4 hrs. The mixture was concentrated and the crude was purified by prep-HPLC to give Example 67 (25 mg, 0.050 mmol, 58.74% yield) as a white solid. MS obsd.: 529.1 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.95 (d, J = 8.8 Hz, 2H), 7.86 (d, J = 8.8 Hz, 2H), 7.25 (s, 1H), 7.10 (s, 1H), 5.25 (s, 1H), 4.78 (d, J = 13.6 Hz, 1H), 4.45 (d, J = 12.8 Hz, 1H), 4.06 (d, J = 9.6 Hz, 1H), 3.82 (d, J = 12.0 Hz, 1H), 3.75 (s, 1H), 3.57 (d, J = 9.6 Hz, 1H), 3.28 (dd, J = 13.6, 3.2 Hz, 1H), 3.09 (t, J = 11.2 Hz, 1H), 2.71 - 2.90 (m, 2H), 2.33 (d, J = 16.4 Hz, 1H). Example 68 4-Amino-1-[4-[4-[6-chloro-4-(difluoromethyl)-2-pyridyl]piper azin-1-yl]sulfonylphenyl] pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 1 by using 2,6- dichloro-4-(difluoromethyl)pyridine instead of 2,6-dichloro-4-(trifluoromethyl)pyridine. Example 68 (42.4 mg) was obtained as a white solid. MS obsd.: 486.1 (MH + ). 1 H NMR (400 MHz, CD3OD): δ ppm 7.90 - 7.94 (m, 2 H), 7.81 - 7.86 (m, 2 H), 6.79 (s, 1 H), 6.73 (s, 1 H), 6.56 (s, 1 H), 4.39 (dd, J = 11.2, 6.8 Hz, 1 H), 4.18 (m, 1 H), 3.93 (dd, J = 11.2, 2.4 Hz, 1 H), 3.65 - 3.75 (m, 4 H), 3.19 (dd, J =18.0, 8.4 Hz, 1 H), 3.03 - 3.11 (m, 4 H), 2.67 (dd, J = 18.0, 2.8 Hz, 1 H). Example 69 4-Amino-1-[4-[4-(6-chloro-4-cyclopropylsulfonyl-2-pyridyl)pi perazin-1-yl]sulfonylphenyl] pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: 2,6-dichloro-4-cyclopropylsulfanyl-pyridine (compound 69a) To a solution of 4-bromo-2,6-dichloro-pyridine (100.0 mg, 0.440 mmol), cyclopropanethiol (32.68 mg, 0.440 mmol) in DMF (2 mL) were added K 2 CO 3 (0.25 mL, 1.32 mmol). The mixture was heated to 80 °C for 4 hrs, then diluted with water (40 mL) and extracted with EA (50 mL) three times. The combined organic layer was washed with brine (50 mL), dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by prep-HPLC to give compound 69a (100 mg, 0.450 mmol, 82.46% yield) as yellow oil. MS obsd.: 220.0 (MH + ). Step 2: 2,6-dichloro-4-(cyclopropylsulfonyl)pyridine (compound 69b) To a solution of compound 69a (80.0 mg, 0.360 mmol) in DCM (4 mL) were added m- CPBA (0.15 mL, 1.09 mmol) in one portion. The mixture was stirred at 20 °C for 2 hrs and then concentrated. The crude was purified by prep-HPLC to give compound 69b (60 mg, 0.240 mmol, 65.48% yield) as a yellow solid. MS obsd.: 252.9 (MH + ). Step 3: tert-butyl (1-(4-((4-(6-chloro-4-(cyclopropylsulfonyl)pyridin-2-yl)pipe razin-1- yl)sulfonyl)phenyl)-5-oxopyrrolidin-3-yl)carbamate (compound 69c) To a mixture of Intermediate C (80.0 mg, 0.190 mmol), Cs 2 CO 3 (184.19 mg, 0.570 mmol) in DMSO (4 mL) was added compound 69b (61.77 mg, 0.240 mmol) and the mixture was stirred at 80 °C for 2 hrs. Solvent was evaporated and the residue was dissolved in water, extracted with EA (50 mL) three times. The combined organic layer was washed with brine, dried over Na 2 SO 4 , filtered and concentrated to give compound 69c (100 mg, 0.160 mmol, 66.31% yield) as a yellow solid MS obsd.: 640.1 (MH + ). Step 4: 4-amino-1-(4-((4-(6-chloro-4-(cyclopropylsulfonyl)pyridin-2- yl)piperazin-1- yl)sulfonyl) phenyl)pyrrolidin-2-one (Example 69) To a solution of compound 69b (80.0 mg, 0.120 mmol) in DCM (2 mL) was added TFA (1.0 mL, 0.370 mmol). The mixture was stirred at 20 °C for 1 h and then concentrated. The crude was purified by prep-HPLC to give Example 69 (30.4 mg, 0.060 mmol, 43.67% yield) as a yellow solid. MS obsd.: 540.1 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 8.38 - 8.51 (m, 3H), 7.87 - 7.97 (m, 2H), 7.73 - 7.83 (m, 2H), 7.11 (d, J = 0.8 Hz, 1H), 7.03 (d, J = 0.8 Hz, 1H), 4.20 - 4.32 (m, 1H), 3.99 - 4.13 (m, 1H), 3.81 - 3.894 (m, 1H), 3.64 - 3.78 (m, 4H), 2.91 - 3.15 (m, 6H), 2.57 - 2.67 (m, 1H), 1.03 - 1.21 (m, 4H). Example 70 4-Amino-1-[4-[4-(6-chloro-4-tetrahydropyran-4-ylsulfonyl-2-p yridyl)piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: 2,6-dichloro-4-tetrahydropyran-4-ylsulfanyl-pyridine(compoun d 70a) To a solution of 2,6-dichloropyridine-4-thiol (90.0 mg, 0.500 mmol), K 2 CO 3 (0.08 mL, 1 mmol) in NMP (2 mL) were added 4-bromo-tetrahydropyran (82.49 mg, 0.500 mmol). The mixture was stirred at 70 °C for 15 hrs under N2. The mixture was diluted with water (50 mL) and extracted with EA (50 mL) three times. The organic layer was washed with brine (50 mL) two times, dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by prep-HPLC to give compound 70a (120 mg, 0.450 mmol, 90.88% yield) as a yellow solid. MS obsd.: 490.9 (MH + ). Step 2 : 4-Amino-1-[4-[4-(6-chloro-4-tetrahydropyran-4-ylsulfonyl-2-p yridyl)piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one (Example 70) The title compound was prepared in analogy to the preparation of Example 69 by using compound 70a instead of compound 69a. Example 70 (33.2 mg) was obtained as a white solid. MS obsd.: 584.2 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 8.48 - 8.64 (m, 3H), 7.87 - 8.01 (m, 2H), 7.70 - 7.84 (m, 2H), 7.00 - 7.12 (m, 1H), 6.89 - 6.98 (m, 1H), 4.21 - 4.33 (m, 1H), 4.03 - 4.10 (m, 1H), 3.86 - 3.94 (m, 3H), 3.69 - 3.78 (m, 5H), 3.22 - 3.31 (m, 2H), 2.97 - 3.11(m, 5H), 2.67 (s, 1H), 1.65 - 1.74 (m, 2H), 1.51 (br s, 2H). Example 71 1-[2-[4-[4-(4-Amino-2-oxo-pyrrolidin-1-yl)phenyl]sulfonylpip erazin-1-yl]-6-chloro-4- pyridyl]cyclopropanecarbonitrile The title compound was prepared according to the following procedures: Step 1: tert-butyl 2-cyano-2-(2,6-dichloropyridin-4-yl)acetate (compound 71a) A mixture of 2,4,6-trichloropyridine (1.82 g, 10 mmol), tert-butyl 2-cyanoacetate (1.69 g, 12 mmol) and Cs 2 CO 3 (4.9 g, 15 mmol) in DMF (20 mL) was stirred for 2 hrs at 90 o C. Then the mixture was poured into water (10 mL), extracted with EA. The organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by column chromatography on silica gel (PE: EA= 20:1~10:1) to give compound 71a (2.01 g, 8.4 mmol, 70% yield) as a yellow oil. MS obsd.: 287.0 (MH + ). Step 2: 2-(2,6-dichloropyridin-4-yl)acetonitrile (compound 71b) A mixture of intermediate 71a (2 g, 6.97 mmol) and TFA (7.94 g, 69.7 mmol) in DCM (3 mL) was stirred for 2 hrs at rt and then concentrated. The crude was poured into saturated NaHCO 3 solution (10 mL) and extracted with EA. The organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by column chromatography on silica gel (PE: EA= 10:1~10:5) to give compound 71b (1.17 g, 6.27 mmol, 90% yield) as a yellow solid. MS obsd.: 187.0 (MH + ). Step 3: 1-(2,6-dichloro-4-pyridyl)cyclopropanecarbonitrile (Intermediate 71c) A solution of intermediate 71b (0.8 g, 4.28 mmol) and diphenyl(vinyl)sulfonium trifluoromethanesulfonate (1.86 g, 5.13 mmol) in DMF (10 mL) was added DBU (1.95 g, 12.8 mmol). The mixture was stirred for 2 hrs at rt and then poured into water, extracted with EA. The organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated. The residue was purified by column chromatography on silica gel (PE: EA= 2:1~1:5) to give compound 71c (0.6 g, 2.81 mmol, 65.8% yield) as a white solid. MS obsd.: 214.1 (MH + ). Step 4: 1-[2-[4-[4-(4-amino-2-oxo-pyrrolidin-1-yl)phenyl]sulfonylpip erazin-1-yl]-6- chloro-4-pyridyl]cyclopropanecarbonitrile (Example 71) The title compound was prepared in analogy to the preparation of Example 69 by using compound 71b instead of compound 69b. Example 71 (6.0 mg) was obtained as a white solid. MS obsd.: 501.4 (MH + ). 1 H NMR (400 MHz, CD3OD): δ ppm 7.89 - 7.93 (m, 2 H), 7.77 - 7.82 (m, 2 H), 6.53 (d, J = 1.2 Hz, 1 H), 6.50 (d, J =1.2 Hz, 1 H), 4.58 (br s, 2 H), 4.15 (dd, J =10.0, 6.4 Hz, 1 H), 3.78 (s, 1 H), 3.61 - 3.69 (m, 5 H), 3.03 - 3.08 (m, 4 H), 2.91 (dd, J = 17.2, 7.2 Hz, 1 H), 2.38 - 2.43 (m, 1 H), 1.74 - 1.78 (m, 2 H), 1.55 - 1.59 (m, 2 H). Example 72 4-Amino-1-[4-[4-(6-chloro-4-phenoxy-2-pyridyl)piperazin-1-yl ]sulfonylphenyl]pyrrolidin- 2-one The title compound was prepared according to the following procedures: Step 1: 2,6-dichloro-4-phenoxy-pyridine (compound 72a) To a solution of 4-bromo-2,6-dichloro-pyridine (100.0 mg, 0.44 mmol) in DMF (1.0 mL) was added phenol (41.48 mg, 0.44 mmol), potassium (51.7 mg, 1.32 mmol) in one portion. The reaction was stirred at 100 °C for 1 h and diluted with water (20 mL), extracted with EA(10 mL) three times. The combined organic layer was washed with brine (50 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated to give compound 72a (100 mg, 0.42 mmol, 94.5% yield) as colorless oil. Step 2: 4-Amino-1-[4-[4-(6-chloro-4-phenoxy-2-pyridyl)piperazin-1-yl ]sulfonylphenyl] pyrrolidin-2-one(Example 72) The title compound was prepared in analogy to the preparation of Example 69 by using compound 72a instead of compound 69a. Example 72 (45 mg) was obtained as a white solid. MS obsd.: 528.2 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 8.47 - 8.70 (m, 3H), 8.45 - 8.72 (m, 4H), 7.88 - 7.91 (m, 2H), 7.78 (s, 2H), 7.42 - 7.48 (m, 2H), 7.24 - 7.30 (m, 1H), 7.09 - 7.13 (m, 2H), 6.25 - 6.28 (m, 1H), 6.27 (s, 1H), 6.11 - 6.14 (m, 1H), 4.24 - 4.29 (m, 1H), 4.05 - 4.11 (m, 1H), 3.95 - 3.99 (m, 1H), 3.53 - 3.58 (m, 1H), 3.01 - 3.06 (m, 5H), 2.69 - 2.75 (m, 1H). Example 73 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(phenyl)methyl]-2-pyrid yl]piperazin-1- yl]sulfonylphenyl] pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: 2,6-dichloro-N-methoxy-N-methyl-pyridine-4-carboxamide (compound 73a) A mixture of 2,6-dichloroisonicotinic acid (30 g, 156.25 mmol) in SOCl2 was stirred at 90 o C for 16 hrs and then concentrated. The crude was dissolved in DCM (300 mL) and a solution of O, N-dimethylhydroxylamine (18288.75 mg, 187.5 mmol) and DIPEA (20 mL) was added. The mixture was stirred at rt for 12 hrs and washed with brine (150 mL) three times. The organic layer was dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by flash column on silica gel (PE: EA=100:0~0:100) to give compound 73a (30000 mg, 127.62 mmol, 73.51% yield) as a white solid. MS obsd.: 235.1 (MH + ). Step 2: (2,6-dichloro-4-pyridyl)-phenyl-methanone (compound 73b) To a solution of compound 73a (500.0 mg, 2.13 mmol) in THF (15 mL) was added phenylmagnesium bromide (0.24 mL, 2.13 mmol) at -70 °C under N 2 and the mixture was stirred for 2 hrs. The mixture was quenched with saturated NH4Cl (100 mL) and extracted with EA (50 mL) three times. The organic layers were dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by flash chromatography on silica gel (EA in PE=0-30%) to give compound 73b (150 mg, 0.600 mmol, 25.2% yield) as a white solid. MS obsd.: 252 (MH + ). Step 3: 2,6-dichloro-4-[difluoro(phenyl)methyl]pyridine (compound 73c) A solution of compound 73b (1300.0 mg, 5.16 mmol) and BAST (10.0 mL) was stirred at 25 °C for 12 hrs. The reaction was quenched with ice water (50 mL) and extracted with EA (30 mL) three times. The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by flash column (PE: EA=1:0 to 1:1) to give compound 73c (800 mg, 2.92 mmol, 50.94% yield) as a light yellow solid. MS obsd.: 274(MH + ). Step 4: tert-butyl N-[1-[4-[4-[6-chloro-4-[difluoro(phenyl)methyl]-2-pyridyl]pi perazin-1-yl] sulfonylphenyl]-5-oxo-pyrrolidin-3-yl]carbamate (compound 73d) To a solution of intermediate C (309.76 mg, 0.730 mmol) and compound 73c (200.0 mg, 0.730 mmol) in DMSO (0.5 mL) was added DIPEA (2.0 mL) and the reaction was stirred at 115 °C for 12 hrs. The reaction was quenched with brine (50 mL) and extracted with DCM (30 mL) two times. The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by flash column (EA in PE=0-70%) to give compound 73d (400 mg, 0.600 mmol, 82.79% yield) as a white solid. MS obsd.: 662(MH + ). Step 5: 4-amino-1-[4-[4-[6-chloro-4-[difluoro(phenyl)methyl]-2-pyrid yl]piperazin-1- yl]sulfonyl phenyl]pyrrolidin-2-one(Example 73) Compound 73d (400.0 mg, 0.60 mmol) was dissolved in 2 M HCl/EA (10 mL) and the reaction was stirred at rt for 4 hrs. The mixture was concentrated and the crude was purified by prep-HPLC to give Example 73 (300 mg, 0.530 mmol, 86.47% yield) as a white solid. MS obsd.: 562(MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.92 (d, J = 8.8 Hz, 2H), 7.77 (d, J = 8.8 Hz, 2H), 7.55 - 7.62 (m, 2H), 7.44 - 7.54 (m, 3H), 6.95 (s, 1H), 6.76 (s, 1H), 4.14 (dd, J = 10.4, 6.4 Hz, 1H), 3.88 (s, 1H), 3.64 -3.72 (m, 5H), 2.86 - 3.01 (m, 5H), 2.46 (dd, J = 17.4, 3.2 Hz, 1H). Example 74 4-Amino-1-[4-[4-(4-benzyl-6-chloro-2-pyridyl)piperazin-1-yl] sulfonylphenyl]pyrrolidin-2- one The title compound was prepared according to the following procedures: Step 1: 4-benzyl-2,6-dichloro-pyridine (compound 74a) A mixture of methyltriphenylphosphonium bromide (1.7 g, 4.76 mmol), potassium t- Butoxide (0.45 g, 4.76 mmol) in THF (20 mL) was stirred for 0.5 h under N 2 , then compound 73b (1 g, 3.97 mmol) was added. The resulted mixture was stirred for 12 hrs at rt and then poured into water, extracted with EA. The organic layer was dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by column chromatography on silica gel (PE: EA= 20:1~10:1) to give compound 74a (0.23 g, 0.99 mmol, 25% yield) as yellow gum. MS obsd.: 236.0(MH + ). Step 2: 4-amino-1-[4-[4-(4-benzyl-6-chloro-2-pyridyl)piperazin-1-yl] sulfonylphenyl] pyrrolidin-2-one (Example 74) The title compound was prepared in analogy to the preparation of Example 73 by using compound 74a instead of compound 73c. Example 74 (3.6 mg) was obtained as a white solid. MS obsd.: 526 (MH + ). 1 H NMR (400 MHz, CD 3 OD): δ ppm 7.92 (d, J = 8.8 Hz, 2 H), 7.78 - 7.82 (m, 2 H), 7.24 - 7.31 (m, 2 H), 7.15 - 7.21 (m, 3 H), 6.52 (s, 1 H), 6.45 (s, 1 H), 4.26 (m, 1 H), 3.94 - 4.00 (m, 1 H), 3.83 (s, 2 H), 3.77 (dd, J = 10.8, 2.8 Hz, 1 H), 3.56 - 3.63 (m, 5 H), 2.93 - 3.15 (m, 8 H), 2.51 (m, 1 H). Example 75 4-Amino-1-[4-[4-[6-chloro-4-(1-phenylcyclopropyl)-2-pyridyl] piperazin-1-yl]sulfonylphenyl] pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: 2,6-dichloro-4-(1-phenylvinyl)pyridine (compound 75a) A mixture of methyltriphenylphosphonium bromide (1.7 g, 4.76 mmol), potassium t- Butoxide (0.45 g, 4.76 mmol) in THF (20 mL) was stirred for 0.5h under N2, then compound 73b (1 g, 3.97 mmol) was added. The resulted mixture was stirred for 12 hrs at rt and then poured into water, extracted with EA. The organic layer was dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by column chromatography on silica gel (PE: EA= 20:1~10:1) to give compound 75a (0.50 g, 2.38 mmol, 50% yield) as yellow gum. MS obsd.: 250.2(MH + ). Step 2: 2,6-dichloro-4-(1-phenylcyclopropyl)pyridine (compound 75b) A mixture of trimethylsulfoxonium iodide (2.2 g, 10 mmol), sodium hydride (0.4 g, 10 mmol) in THF (5 mL) was stirred for 0.5h 30 at rt, then compound 75a (500 mg, 2 mmol) in THF (5 mL) was added dropwise. The mixture was stirred for 2 hrs at rt and then poured into water, extracted with EA. The organic layer was dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by column chromatography on silica gel (PE: EA= 20:1~10:1) to compound 75b (0.32 g, 1.2 mmol, 60% yield) as a yellow gum. MS obsd.: 264.1(MH + ). Step 3: 4-amino-1-[4-[4-[6-chloro-4-(1-phenylcyclopropyl)-2-pyridyl] piperazin-1- yl]sulfonyl phenyl]pyrrolidin-2-one(Example 75) The title compound was prepared in analogy to the preparation of Example 73 by using compound 75b instead of compound 73c. Example 75 (7.4 mg) was obtained as a white solid. MS obsd.: 552 (MH + ). 1 H NMR (400 MHz, CD 3 OD): δ ppm 7.92 (m, 2 H), 7.78 (m, 2 H), 7.23 - 7.31 (m, 5 H), 6.24 - 6.39 (m, 2 H), 4.18 - 4.33 (m, 1 H), 3.86 - 4.00 (m, 1 H), 3.69 - 3.80 (m, 1 H), 3.48 - 3.50 (m, 4 H), 2.99 - 3.03 (m, 5 H), 2.45 - 2.58 (m, 1 H), 1.31 (s, 4H). Example 76 4-Amino-1-[4-[4-[4-[difluoro(phenyl)methyl]-6-methyl-pyrimid in-2-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 73 by using 2- chloro-6-methylpyrimidine-4-carboxylate instead of 2,6-dichloro-N-methoxy-N-methyl- pyridine-4-carboxamide. Example 76 (28 mg) was obtained as a white solid. MS obsd.: 543 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 8.18 (br s, 2H), 7.90 (d, J = 9.2 Hz, 2H), 7.77 (d, J = 9.2 Hz, 2H), 7.45 - 7.56 (m, 5H), 6.89 (s, 1H), 4.25 (dd, J = 6.8, 11.2 Hz, 1H), 4.07 (br s, 1H), 3.79 (br s, 5H), 3.43 - 3.63 (m, 1H), 3.21 - 3.31 (m, 1H), 3.17 (s, 1H), 3.07 (dd, J = 8.4, 17.6 Hz, 1H), 2.92 (br t, J = 4.8 Hz, 4H), 2.67 (s, 1H), 2.53 - 2.60 (m, 1H), 2.33 (s, 3H), 2.07 (s, 1H). Examples 77 and 78 4-Amino-1-[4-[4-[2-chloro-6-[difluoro(phenyl)methyl]pyrimidi n-4-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one 4-Amino-1-[4-[4-[4-chloro-6-[difluoro(phenyl)methyl]pyrimidi n-2-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compounds were prepared according to the following procedures: Step 1: (2,6-dichloropyrimidin-4-yl)-phenyl-methanone (compound 77a) A solution of methyl 2,6-dichloropyrimidine-4-carboxylate (1.1 g, 5.31 mmol) in THF (1.5 mL) was cooled to -78 °C under N2 and treated with PhMgBr (5.31 mL, 5.31 mmol) dropwise over a period of 0.5 h. The reaction was stirred at -78 °C for 30 min and then quenched with water (5 mL) and brine (5 mL). The mixture was extracted with EA (10 mL) twice and the organic phase was dried and concentrated to give compound 77a (500 mg, 37.2%). MS obsd.: 253.2 (MH + ). Step 2: 2,4-dichloro-6-[difluoro(phenyl)methyl]pyrimidine (compound 77b) Compound 77a (0.500 g, 1.98 mmol) and DAST (15.9 g, 13 mL, 98.4 mmol) was stirred at rt for 4 hrs. The mixture was cooled at 0 °C and quenched with water (10 mL). The mixture was extracted with DCM (20 mL). The organic phase was dried and concentrated to give the crude product (545 mg, 100% yield). Step 3: tert-butyl N-[1-[4-[4-[2-chloro-6-[difluoro(phenyl)methyl]pyrimidin-4-y l] piperazin-1-yl]sulfonylphenyl]-5-oxo-pyrrolidin-3-yl]carbama te (compound 77c) tert-butyl N-[1-[4-[4-[4-chloro-6-[difluoro(phenyl)methyl]pyrimidin-2-y l]piperazin-1-yl] sulfonylphenyl]-5-oxo-pyrrolidin-3-yl]carbamate (compound 78c) A mixture of intermediate C (23.1 mg, 54.5 µmol), compound 77a (0.015 g, 54.5 µmol) and DIPEA (21.1 mg, 29.2 µL, 164 µmol) in THF (200 µL) was stirred at 70 °C overnight. The mixture was concentrated to give the mixture of compounds 77c and 78c, which was used directly in next step. MS obsd.: 663.7 (MH + ). Step 4: 4-amino-1-[4-[4-[2-chloro-6-[difluoro(phenyl)methyl]pyrimidi n-4-yl]piperazin-1- yl] sulfonylphenyl]pyrrolidin-2-one (Example 77) and 4-amino-1-[4-[4-[4-chloro-6-[difluoro (phenyl)methyl]pyrimidin-2-yl]piperazin-1-yl]sulfonylphenyl] pyrrolidin-2-one (Example 78) A mixture of compounds 77c and 78c (0.035 g, 52.8 µmol) and TFA (181 mg) in DCM (1 mL) was stirred at 25 °C for 1h. The mixture was concentrated and purified by pre-HPLC to give Example 77 (24 mg, 80% yield) and Example 78 (5 mg, 16.8% yield) as white solids. Example 77: MS obsd.: 563.3 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.88 - 7.95 (m, J = 8.8 Hz, 2H), 7.69 - 7.79 (m, J = 8.8 Hz, 2H), 7.45 - 7.58 (m, 5H), 7.19 (s, 1H), 3.88 - 4.08 (m, 1H), 3.82 (br s, 4H), 3.56 - 3.74 (m, 1H), 3.45 - 3.50 (m, 1H), 3.18 (br s, 1H), 3.00 (br t, J = 4.8 Hz, 4H), 2.76 (dd, J = 6.8, 16.8 Hz, 1H), 2.25 (dd, J = 4.0, 16.8 Hz, 1H). Example 78: MS obsd.: 563.6 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.85 - 7.93 (m, J = 8.8 Hz, 2H), 7.67 - 7.77 (m, J = 8.8 Hz, 2H), 7.43 - 7.62 (m, 5H), 7.08 (s, 1H), 3.97 (br dd, J = 6.0, 9.6 Hz, 1H), 3.72 - 3.84 (m, 4H), 3.57 - 3.71 (m, 1H), 3.44 - 3.49 (m, 1H), 2.94 (br t, J = 4.8 Hz, 4H), 2.54 - 2.581 (m, 1H), 2.24 (dd, J = 4.0, 16.8 Hz, 1H). Examples 79 and 80 4-Amino-1-[4-[4-[2-cyclopropyl-6-[difluoro(phenyl)methyl]pyr imidin-4-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one 4-Amino-1-[4-[4-[4-cyclopropyl-6-[difluoro(phenyl)methyl]pyr imidin-2-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compounds were prepared according to the following procedures: Step 1: tert-butyl 4-[2-chloro-6-[difluoro(phenyl)methyl]pyrimidin-4-yl]piperaz ine-1- carboxylate (compound 79a) tert-butyl 4-[4-chloro-6-[difluoro(phenyl)methyl]pyrimidin-2-yl]piperaz ine-1-carboxylate (compound 80a) A mixture of tert-butyl piperazine-1-carboxylate (339 mg, 1.82 mmol), compound 77b (0.50 g, 1.82 mmol) and K 2 CO 3 (502 mg, 3.64 mmol) in DMSO (10 mL) was stirred at 85 °C overnight. The mixture was diluted with EA (20 mL) and washed with water (20 mL). The organic was dried, concentrated to give the mixture of compound 79a and 80a (772 mg, 100% yield) which was used in next step without further purification. MS obsd.: 425.3 (MH + ). Step 2: tert-butyl 4-[2-cyclopropyl-6-[difluoro(phenyl)methyl]pyrimidin-4-yl]pi perazine-1- carboxylate (compound 79b) tert-butyl 4-[4-cyclopropyl-6-[difluoro(phenyl)methyl] pyrimidin-2-yl]piperazine-1- carboxylate(compound 80b) A mixture of compound 79a and 80a (0.10 g, 235 µmol), potassium cyclopropyltrifluoro- borate (104 mg, 706 µmol), Pd(dppf)Cl 2 (17 mg, 23.5 µmol) and K 2 CO 3 (97.4 mg, 706 µmol) in 1, 4-dioxane (1mL) and water (0.2 mL) was stirred at 70 °C overnight. The mixture was diluted with EA and washed with water. The organic was concentrated to give the crude product (100 mg, 100% yield), which was used directly in next step. MS obsd.: 431.3 (MH + ). Step 3: 2-cyclopropyl-4-[difluoro(phenyl)methyl]-6-piperazin-1-yl-py rimidine (compound 79c) 4-cyclopropyl-6-[difluoro(phenyl)methyl]-2-piperazin-1-yl-py rimidine (compound 80c) A mixture of compounds 79b and 80b (0.100 g, 232 µmol) and TFA (750 mg, 0.5 mL, 6.58 mmol) in DCM (1mL) was stirred at 25 °C for 1 h. The mixture was concentrated to give the crude product (76 mg, 100% yield), which was used directly in next step. MS obsd.: 331.2 (MH + ). Step 4: 2-cyclopropyl-4-[difluoro(phenyl)methyl]-6-[4-(4-iodophenyl) sulfonylpiperazin-1- yl]pyrimidine (compound 79d) 4-cyclopropyl-6-[difluoro(phenyl)methyl]-2-[4-(4-iodophenyl) sulfonylpiperazin-1- yl]pyrimidine (compound 80d) A solution of 4-iodobenzenesulfonyl chloride (104 mg, 345 µmol) and a mixture of compounds 79c and 80c (0.076 g, 230 µmol), TEA (140 mg, 192 µl, 1.38 mmol) was stirred at rt for 1h. The mixture was diluted with EA (20 mL) and washed with water (10 mL) two times. The organic was dried and concentrated to give the crude (137 mg, 100% yield). MS obsd.: 597.3 (MH + ). Step 5: tert-butyl N-[1-[4-[4-[2-cyclopropyl-6-[difluoro(phenyl)methyl]pyrimidi n-4- yl]piperazin -1-yl]sulfonylphenyl]-5-oxo-pyrrolidin-3-yl]carbamate (compound 79e) tert-butyl N-[1-[4-[4-[4-cyclopropyl-6-[difluoro(phenyl)methyl]pyrimidi n-2-yl]piperazin- 1-yl]sulfonylphenyl]-5-oxo-pyrrolidin-3-yl]carbamate (compound 80e) A mixture of compounds 79d and 80d (0.137 g, 230 µmol), tert-butyl (5-oxopyrrolidin-3- yl)carbamate (69 mg, 345 µmol), DMEDA (4.05 mg, 45.9 µmol), CuI (8.75 mg, 45.9 µmol) and K 3 PO 4 (122 mg, 574 µmol) in DMF (1.5 mL) was heated at 90 °C overnight. This reaction was diluted with EA (20 mL), washed with water (10 mL) two times. The organic was dried, concentrated to give the crude products (150 mg, 98% yield) which was used directly in next step. MS obsd.: 669.4 (MH + ). Step 6: 4-amino-1-[4-[4-[2-cyclopropyl-6-[difluoro(phenyl)methyl]pyr imidin-4-yl] piperazin-1-yl] sulfonylphenyl]pyrrolidin-2-one(Example 79) and 4-amino-1-[4-[4-[4- cyclopropyl-6-[difluoro(phenyl)methyl]pyrimidin-2-yl]piperaz in-1-yl]sulfonylphenyl]pyrrolidin- 2 one(Example 80) A mixture of compounds 79e and 80e (0.150 g, 224 µmol) and TFA (750 mg, 0.5 mL, 6.58 mmol) in DCM (2 mL) was stirred at 25 °C for 2 hrs. The mixture was concentrated and purified by pre-HPLC to give Example 79 (15 mg, 11.8%) and Example 80 (3.5 mg, 2.7%) as white solids. Example 79: MS obsd.: 569.3 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.83 (d, J = 8.8 Hz, 2H), 7.67 (d, J = 8.8 Hz, 2H), 7.36 - 7.49 (m, 5H), 6.83 (s, 1H), 3.93 (br dd, J = 6.4, 9.6 Hz, 2H), 3.69 (br s, 4H), 2.87 (br t, J = 4.4 Hz, 4H), 2.71 (br dd, J = 7.2, 16.8 Hz, 1H), 2.57 - 2.64 (m, 1H), 2.212 - 2.29 (m, 1H), 1.80 - 1.86 (m, 1H), 0.71 - 0.84 (m, 4H). Example 80: MS obsd.: 569.3 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.82 (d, J = 8.8 Hz, 2H), 7.65 (d, J = 8.8 Hz, 2H), 7.45 - 7.49 (m, 2H), 7.37 - 7.45 (m, 3H), 6.87 (s, 1H), 3.92 (br dd, J = 6.4, 9.6 Hz, 2H), 3.67 (br s, 4H), 2.78 - 2.83 (m, 4H), 2.71 (br dd, J = 7.2, 16.8 Hz, 2H), 2.14 - 2.26 (m, 1H), 1.91 - 2.05 (m, 1H), 0.81 - 1.03 (m, 4H). Examples 81 and 82 4-Amino-1-[4-[4-[6-[difluoro(phenyl)methyl]-2-methoxy-pyrimi din-4-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one 4-Amino-1-[4-[4-[4-[difluoro(phenyl)methyl]-6-methoxy-pyrimi din-2-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compounds were prepared according to the following procedures: Step 1: tert-butyl 4-[4-[difluoro(phenyl)methyl]-2-methoxy-pyrimidin-2-yl]piper azine-1- carboxylate(compound 81a) tert-butyl 4-[6-[difluoro(phenyl)methyl]-6-methoxy-pyrimidin-4-yl]piper azine-1- carboxylate(Intermediate 82a) A mixture of compounds 79a and 80a (0.10 g, 235 µmol) and sodium methanolate (94.1 µL, 471 µmol) in MeOH (1 mL) was stirred at 80 °C overnight. This mixture was concentrated and diluted with EA (5 mL) and washed with water (10 mL). The organic layer was dried, concentrated to give the crude (99 mg, 100% yield) which was used directly in next step. MS obsd.: 421.5 (MH + ). Step 2: 4-amino-1-[4-[4-[6-[difluoro(phenyl)methyl]-2-methoxy-pyrimi din-4-yl]piperazin- 1-yl]sulfonylphenyl]pyrrolidin-2-one (Example 81) and 4-amino-1-[4-[4-[4 [difluoro(phenyl) methyl]-6-methoxy-pyrimidin-2-yl]piperazin-1-yl]sulfonylphen yl]pyrrolidin-2-one (Example 82) The title compounds were prepared in analogy to the preparation of Example 79 and 80 by using the mixture of compounds 81a and 82a instead of the mixture of compounds 79e and 80e. Examples 81 (28 mg) and 82 (4.2 mg) were obtained as white solids. Example 81: MS obsd.: 559.3 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.91 (d, J = 8.8 Hz, 2H), 7.74 (d, J = 8.8 Hz, 2H), 7.43 - 7.57 (m, 5H), 6.82 (s, 1H), 4.00 (br dd, J = 6.0, 9.6 Hz, 2H), 3.79 (br s, 4H), 3.73 (s, 3H), 3.67 - 3.68 (m, 1H), 2.96 (br t, J = 4.4 Hz, 4H), 2.79 (dd, J = 7.2, 16.8 Hz, 1H), 2.19 - 2.36 (m, 1H). Example 82: MS obsd.: 559.4 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.82 (br d, J = 8.8 Hz, 2H), 7.65 (d, J = 8.8 Hz, 2H), 7.41 - 7.49 (m, 5H), 6.30 (s, 1H), 3.92 (br dd, J = 6.0, 9.6 Hz, 2H), 3.77 (s, 3H), 3.72 (br s, 4H), 2.83 (br s, 4H), 2.64 - 2.76 (m, 1H), 2.18 (br dd, J = 4.0, 16.8 Hz, 1H), 2.00 (s, 1H). Example 83 (4R)-4-Amino-1-[4-[4-[6-[difluoro(phenyl)methyl]-2-methyl-py rimidin-4-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: tert-butyl 4-[6-[difluoro(phenyl)methyl]-2-methyl-pyrimidin-4-yl]pipera zine-1- carboxylate (compound 83a) A mixture of compound 79a (0.100 g, 235 µmol), 2,4,6-trimethyl-1,3,5,2,4,6- trioxatriborinane (202 µL, 706 µmol), Na2CO3 (62.4 mg, 588 µmol) and Pd(PPh3)4 (27 mg) in 1,4-dioxane (0.5 mL) and water (0.1 mL) was stirred at 130 °C overnight. The mixture was concentrated and the residue was used directly in next step. MS obsd.: 405.3 (MH + ). Step 2: (4R)-4-amino-1-[4-[4-[6-[difluoro(phenyl)methyl]-2-methyl-py rimidin-4-yl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one(Example 83) The title compound was prepared in analogy to the preparation of Example 79 by using compound 83a instead of compound 79e and intermediate D instead of intermediate C. Example 83 (12 mg) was obtained as a white solid. MS obsd.: 543.2 (MH + ). 1 H NMR (400 MHz, DMSO- d 6 ): δ ppm 7.80 - 7.87 (m, J = 8.8 Hz, 2H), 7.62 - 7.72 (m, J = 8.8 Hz, 2H), 7.36 - 7.49 (m, 5H), 6.91 (s, 1H), 3.91 (br dd, J = 6.0, 9.6 Hz, 2H), 3.73 (br s, 4H), 3.57 (br s, 1H), 2.88 (br t, J = 4.4 Hz, 4H), 2.69 (dd, J = 6.8, 16.8 Hz, 1H), 2.24 (s, 3H), 2.17 (dd, J = 4.0, 16.8 Hz, 1H). Example 84 4-Amino-1-[4-[4-[6-[difluoro(phenyl)methyl]-2-vinyl-pyrimidi n-4-yl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 79 by using 4,4,5,5-tetramethyl-2-vinyl-1,3,2-dioxaborolane instead of potassium cyclopropyltrifluoro-borate. Example 84 (2 mg) was obtained as a white solid. MS obsd.: 555.3 (MH + ). 1 H NMR (400 MHz, CD3OD): δ ppm 7.86 - 7.97 (m, 2H), 7.80 - 7.84 (m, 2H), 7.54 - 7.61 (m, 2H), 7.44 - 7.53 (m, 3H), 7.00 (s, 1H), 6.58 - 6.66 (m, 1H), 6.53 (d, J = 2.0 Hz, 1H), 5.66 (br dd, J = 2.0, 10.2 Hz, 1H), 4.40 (br dd, J = 7.2, 11.2 Hz, 1H), 4.21 (br t, J = 7.2 Hz, 1H), 3.97 (br dd, J = 2.0, 11.2 Hz, 1H), 3.90 (br s, 4H), 3.20 (br dd, J = 8.4, 18.0 Hz, 1H), 3.06 - 3.14 (m, 4H), 2.72 (br dd, J = 2.8, 18.0 Hz, 1H). Example 85 2-[4-[4-[6-Chloro-4-[difluoro(phenyl)methyl]-2-pyridyl]piper azin-1-yl]sulfonylphenyl]- 2,6,9-triazaspiro[4.5]decan-3-one The title compound was prepared according to the following procedures: Step 1: tert-butyl 4-[6-chloro-4-[difluoro(phenyl)methyl]-2-pyridyl]piperazine- 1- carboxylate(compound 85a) Compound 73c (150 mg, 547 µmol) and tert-butyl piperazine-1-carboxylate (204 mg, 1.09 mmol) were dissolved in DMSO (5 mL), followed by adding K 2 CO 3 (227 mg, 1.64 mmol). The reaction was stirred at 90 °C overnight. The mixture was partitioned between EA (300 mL) and water (300 mL). The organic layer was combined, wash with water (100 mL) three times and brine (300 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated to give the crude (230 mg, 0.542 mmol, 99.1%) as yellow oil. MS obsd.424.6: (MH + ). Step 2: 1-[6-chloro-4-[difluoro(phenyl)methyl]-2-pyridyl]piperazine( compound 85b) Compound 85a (0.23 g, 543 µmol) was dissolved in DCM (7.5 mL), followed by adding TFA (2.5 mL, 543 µmol). The mixture was stirred at rt for 2 hrs and then concentrated. The residue was re-dissolved in the EA (200 mL) and washed with 1M aq. K 2 CO 3 solution (200 mL), brine (300 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated to give the crude (0.25 g, 543 µmol, 100 % yield) as a light yellow solid. MS obsd.: 324.5 (MH + ). Step 3: 1-[6-chloro-4-[difluoro(phenyl)methyl]-2-pyridyl]-4-(4-iodop henyl)sulfonyl- piperazine (compound 85c) Compound 85b (50 mg, 154 µmol) was dissolved in DCM (5 mL), followed by adding TEA (64.6 µL, 463 µmol) and 4-iodobenzenesulfonyl chloride (93.4 mg, 309 µmol) and the reaction was stirred at rt for 2 hrs. The mixture was partitioned between EA (300 mL) and water (300 mL). The organic layer was combined, washed with brine (200 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated to give the crude (90 mg, 0.152 mmol, 99% yield) as a light yellow solid. MS obsd.: 590.1 (MH + ). Step 4: di-tert-butyl 2-[4-[4-[6-chloro-4-[difluoro(phenyl)methyl]-2-pyridyl]piper azin-1- yl]sulfonylphenyl]-3-oxo-2,6,9-triazaspiro[4.5]decane-6,9-di carboxylate (compound 85d) A mixture of compound 85c (0.07 g, 119 µmol), di-tert-butyl 3-oxo-2,6,9-triazaspiro[4.5] decane-6,9-dicarboxylate (65 mg, 183 µmol, WuXi AppTec), DMEDA (10.5 mg, 12.8 µL), CuI (6.78 mg, 35.6 µmol) and K3PO4 (78.7 mg, 356 µmol) in DMF (5 mL) was heated at 70 °C for 2 hrs. Then, the mixture was poured into water (250 mL), extracted with DCM (200 mL) three times. The combined organic layer was washed with brine (200 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by flash chromatography on silica gel (EA in PE=0-30%) to give compound 85d (90 mg, 110.2 µmol, 92.8% yield) as brown oil. MS obsd.: 817.5 (MH + ). Step 5: 2-[4-[4-[6-Chloro-4-[difluoro(phenyl)methyl]-2-pyridyl]piper azin-1-yl] sulfonylphenyl]-2,6,9-triazaspiro[4.5]decan-3-one(Example 85) Compound 85d (90 mg, 110 µmol) was dissolved in DCM (5 mL), followed by adding TFA (100 µl, 1.3 mmol). The reaction was stirred at rt for 1 h and concentrated. The crude was purified by prep-HPLC to give Example 85 (4.15 mg, 6.72 µmol, 5% yield) as a light brown solid. MS obsd.: 617.5 (MH + ). 1 H NMR (400 MHz, CD 3 OD): δ ppm 7.90 (d, J = 8.8 Hz, 2 H), 7.81 (d, J = 8.8 Hz, 2 H), 7.42 - 7.52 (m, 5 H), 6.78 (s, 1 H), 6.66 (s, 1 H), 4.61 - 4.67 (m, 1 H), 4.04 (d, J = 10.4 Hz, 1 H), 3.92 (d, J = 10.4 Hz, 1 H), 3.63 - 3.72 (m, 4 H), 3.11 - 3.19 (m, 4 H), 3.03 - 3.10 (m, 4 H), 2.89 (d, J = 17.6 Hz, 1 H), 2.75 (d, J = 17.6 Hz, 1 H), 2.19 (t, J= 7.6 Hz, 1 H), 2.03 (br d, J = 5.6 Hz, 1 H), 1.60 (br s, 1 H). Example 86 5-(Aminomethyl)-1-[4-[4-[6-chloro-4-[difluoro(phenyl)methyl] -2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one A mixture of compound 85c (91 mg, 154 µmol), 5-(aminomethyl)pyrrolidin-2-one (35.2 mg, 309 µmol), DMEDA (16.6 µL, 154 µmol), CuI (12 mg, 63 µmol) and K 3 PO 4 (102 mg, 463 µmol) in DMF (5 mL) was heated at 70 °C for 2 hrs. Then, the mixture was poured into water (200 mL), extracted with DCM (200 mL) three times. The organic layer was combined, washed with brine (200 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by prep-HPLC to give Example 86 (6 mg, 8.7 µmol, 5.35% yield) as a white solid. MS obsd.: 576.2 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.88 (br d, J = 9.2 Hz, 4 H), 7.75 - 7.83 (m, 2 H), 7.46 - 7.65 (m, 5 H), 6.93 - 6.97 (m, 1 H), 6.76 (s, 1 H), 4.61 - 4.67 (m, 1 H), 3.63 - 3.71 (m, 4 H), 2.93 - 3.06 (m, 4 H), 2.60 - 2.80 (m, 2 H), 2.52 - 2.52 (m, 1 H), 2.39 - 2.47 (m, 1 H), 2.26 - 2.36 (m, 1 H), 2.04 - 2.13 (m, 1 H). Example 87 (3aR,7aS)-3-[4-[4-[6-chloro-4-[difluoro(phenyl)methyl]-2-pyr idyl]piperazin-1-yl]sulfonyl phenyl]-3a,4,5,6,7,7a-hexahydrooxazolo[4,5-c]pyridin-2-one The title compound was prepared in analogy to the preparation of Example 85 by using tert-butyl (3aR,7aS)-2-oxohexahydrooxazolo[4,5-c]pyridine-5(4H)-carboxy late instead of di- tert-butyl 3-oxo-2,6,9-triazaspiro[4.5]decane-6,9-dicarboxylate. Example 87 (23 mg) was obtained as a white solid. MS obsd.: 585.3 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 7.78 (s, 4H), 7.52 - 7.56 (m, 2H), 7.45 - 7.52 (m, 2H), 7.37 - 7.60 (m, 1H), 6.90 (s, 1H), 4.99 (td, J = 4.0, 7.6 Hz, 1H), 4.84 - 4.94 (m, 1H), 3.81 (br s, 4H), 3.50 (br dd, J = 8.8, 13.2 Hz, 1H), 3.15 - 3.27 (m, 2H), 3.08 (br d, J = 9.6 Hz, 1H), 2.88 - 3.01 (m, 4H), 2.17 - 2.18 (m, 1H), 2.07 (s, 1H). Example 88 5-(Aminomethyl)-3-[4-[4-[6-chloro-4-[difluoro(phenyl)methyl] -2-pyridyl]piperazin-1- yl]sulfonylphenyl]oxazolidin-2-one The title compound was prepared in analogy to the preparation of Example 85 by using tert-butyl ((2-oxooxazolidin-5-yl)methyl)carbamate instead of di-tert-butyl 3-oxo-2,6,9- triazaspiro[4.5] decane-6,9-dicarboxylate. Example 88 (21 mg) was obtained as a white solid. MS obsd.: 559.3 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.71 - 7.85 (m, 4H), 7.45 - 7.58 (m, 5H), 6.89 (s, 1H), 4.84 - 4.97 (m, 1H), 4.22 (t, J = 9.2 Hz, 1H), 3.86 - 3.88 (m, 1H), 3.78 - 3.89 (m, 4H), 3.13 - 3.29 (m, 2H), 2.91 (br t, J = 4.8 Hz, 4H), 2.33 (s, 3H). Example 89 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(2-pyridyl)methyl]-2-py ridyl]piperazin-1-yl] sulfonylphenyl] pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: (2,6-dichloro-4-pyridyl)-(2-pyridyl)methanone (compound 89a) To a solution of 2-bromopyridine (0.41 mL, 4.25 mmol) and 2,6-dichloro-N-methoxy-N- methyl-pyridine-4-carboxamide (1000.0 mg, 4.25 mmol) in THF (20 mL) was added n-BuLi (1.77 mL, 4.25 mmol) at -70 °C for 40 min under N2. The mixture was stirred at -70 °C for 4 hrs and then quenched with saturated NH 4 Cl (50 mL), extracted with EA (30 mL × 3). The organic layer was dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by flash chromatography on silica gel (EA in PE= 0~30%) to give compound 89a (450 mg, 1.78 mmol, 37.62% yield) as a white solid. MS obsd.: 253 (MH + ). Step 2: 2,6-dichloro-4-[difluoro(2-pyridyl)methyl]pyridine (compound 89b) Compound 89a (150.0 mg, 0.590 mmol) was dissolved in BAST (2.0 mL) and the reaction was stirred at 25 °C for 12 hrs. The mixture was quenched with ice water (20 mL) and extracted with EA (20 mL) two times. The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by flash chromatography on silica gel (EA in PE= 0-50%) to give compound 89b (85 mg, 0.310 mmol, 46.9% yield) as a yellow solid. MS obsd.: 275.1 (MH + ). Step 3: tert-butyl N-[1-[4-[4-[6-chloro-4-[difluoro(2-pyridyl)methyl]-2-pyridyl ]piperazin- 1-yl]sulfonylphenyl]-5-oxo-pyrrolidin-3-yl]carbamate (compound 89c) Compound 89b (85.0 mg, 0.310 mmol) and intermediate D (131.17 mg, 0.310 mmol) was dissolved in a mixed solution of DMSO (0.5 mL) and DIPEA (2.0 mL). The reaction was stirred at 115 °C for 12 h. The reaction was quenched with brine (20 mL) and extracted with DCM (20 mL) twice. The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by flash chromatography on silica gel (EA in PE=0-70%) to give compound 89c (120 mg, 0.180 mmol, 52.71% yield) as a yellow solid. MS obsd.: 663.2 (MH + ). Step 4: 4-amino-1-[4-[4-[6-chloro-4-[difluoro(2-pyridyl)methyl]-2-py ridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one (Example 89) Compound 89c (100.0 mg, 0.150 mmol) was dissolved in 2 M HCl/EA (5 mL). The reaction mixture was stirred at rt for 12 hrs. The solvent was removed and the crude was purified by prep-HPLC to give Example 89 (83 mg, 0.150 mmol, 96.8% yield) as a white solid. MS obsd.: 563.2 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 8.63 (d, J = 4.4 Hz, 1H), 8.02 (td, J = 7.6, 1.6 Hz, 1H), 7.92 (d, J = 8.8 Hz, 2H), 7.86 (d, J = 7.8 Hz, 1H), 7.76 (d, J = 8.8 Hz, 2H), 7.55 (dd, J = 7.2, 5.2 Hz, 1H), 6.92 (s, 1H), 6.76 (s, 1H), 4.11 (dd, J = 10.4, 6.4 Hz, 1H), 3.84 (dd, J = 8.4, 5.2 Hz, 1H), 3.66 (d, J = 5.2 Hz, 5H), 2.86 - 3.01 (m, 5H), 2.42 (dd, J = 17.2, 3.2 Hz, 1H). Example 90 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(3-pyridyl)methyl]-2-py ridyl]piperazin-1- yl]sulfonylphenyl] pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 89 by using 3- bromopyridine instead of 2-bromopyridine. Example 90 (60 mg) was obtained as a white solid. MS obsd.: 563.1 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 8.85 (s, 1H), 8.73 (d, J = 4.4 Hz, 1H), 8.30 (s, 2H), 8.06 (d, J = 8.0 Hz, 1H), 7.92 (d, J = 8.8 Hz, 2H), 7.80 (d, J = 8.8 Hz, 2H), 7.54 (dd, J = 8.0, 4.8 Hz, 1H), 7.01 (s, 1H), 6.87 (s, 1H), 4.27 (dd, J = 11.2, 6.9 Hz, 1H), 4.09 (s, 1H), 3.83 (d, J = 10.0 Hz, 1H), 3.68 (s, 4H), 3.08 (dd, J = 18.0, 8.0 Hz, 1H), 2.96 (s, 4H), 2.59 (dd, J = 18.0, 2.4 Hz, 1H). Example 91 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(4-pyridyl)methyl]-2-py ridyl]piperazin-1-yl] sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 89 by using 4- bromopyridine instead of 2-bromopyridine. Example 91 (22 mg) was obtained as a white solid. MS obsd. 563.2 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 8.78 (s, 2H), 8.37 (s, 2H), 7.91 (d, J = 8.8 Hz, 2H), 7.79 (d, J = 8.8 Hz, 2H), 7.67 (s, 2H), 6.99 (s, 1H), 6.85 (s, 1H), 4.26 (dd, J = 11.2, 6.8 Hz, 1H), 4.09 (s, 1H), 3.82 (s, 1H), 3.68 (d, J = 4.0 Hz, 4H), 3.08 (dd, J = 18.0, 8.2 Hz, 1H), 2.96 (s, 4H), 2.60 (dd, J = 18.0, 2.4 Hz, 1H). Example 92 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(pyrimidin-5-yl)methyl] -2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 89 by using 4- bromopyridine instead of 5-bromopyrimidine. Example 92 (19.5 mg) was obtained as a white solid. MS obsd. MS obsd.: 563.2 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 1 H NMR (400 MHz, DMSO) 9.36 (s, 1H), 9.12 (s, 2H), 7.92 (d, J = 8.8 Hz, 2H), 7.79 (d, J = 8.8 Hz, 2H), 7.03 (s, 1H), 6.95 (s, 1H), 4.25 (dd, J = 11.2, 6.8 Hz, 1H), 4.07 (br s, 1H), 3.82 (br s, 1H), 3.68 (br s, 4H), 3.31 (br s, 1H), 3.11 - 3.03 (m, 1H), 2.97 (d, J = 4.4 Hz, 4H). Example 93 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(pyrazin-2-yl)methyl]-2 -pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 89 by using 2- bromopyrazine instead of 2-bromopyridine. Example 92 (50 mg) was obtained as a white solid. MS obsd.: 564 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 9.17 (t, J = 10.0 Hz, 1H), 8.86 (d, J = 2.4 Hz, 1H), 8.74 (s, 1H), 7.91 (d, J = 8.8 Hz, 2H), 7.75 (d, J = 8.8 Hz, 2H), 6.97 (d, J = 14.0 Hz, 1H), 6.84 (s, 1H), 4.03 (dd, J = 10.0, 6.0 Hz, 1H), 3.62 - 3.68 (m, 5H), 3.52 - 3.56 (m, 1H), 2.88 - 3.03 (m, 4H), 2.81 (dd, J = 17.2, 7.2 Hz, 1H), 2.30 (dd, J = 17.2, 3.6 Hz, 1H). Example 94 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(pyrimidin-2-yl)methyl] -2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: 2,6-dichloro-4-pyridyl)-pyrimidin-2-yl-methanone (compound 93a) To a solution of n-BuLi (2.82 mL, 6.76 mmol) in THF (30 mL) was added 2,6- dichloropyridine (1.0 g, 6.76 mmol) at -78 °C and then stirred for 30 min under N 2 . A solution of methyl pyrimidine-2-carboxylate (0.26 mL, 6.76 mmol) in THF (15 mL) was added dropwise. The reaction was stirred for further 5 hrs at -78 °C under N2. The reaction mixture was quenched with aq. NH 4 Cl (100 mL) and extracted with EA (40 mL) three times. The combined organic layer was washed with brine (80 mL), dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by silica gel (EA in PE=0~15%) to give compound 93a (450 mg, 1.77 mmol, 23.59% yield) as a colorless oil. MS obsd.: 254 (MH + ). Step 2: 4-amino-1-[4-[4-[6-chloro-4-[difluoro(pyrimidin-2-yl)methyl] -2-pyridyl]piperazin- 1-yl]sulfonylphenyl]pyrrolidin-2-one (Example 93) The title compound was prepared in analogy to the preparation of Example 89 by using compound 93a instead of compound 89b. Example 93 (7 mg) was obtained as a white solid. MS obsd.: 564.1 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 8.96 (d, J = 4.8 Hz, 2H), 7.91 (d, J = 8.8 Hz, 2H), 7.63 - 7.78 (m, 3H), 6.90 (s, 1H), 6.76 (s, 1H), 3.95 - 4.05 (m, 1H), 3.64 (s, 5H), 3.49 (d, J = 7.2 Hz, 1H), 2.94 (s, 4H), 2.77 (dd, J = 16.8, 6.8 Hz, 1H), 2.22 - 2.33 (m, 1H). Example 95 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(tetrahydropyran-4-yl)m ethyl]-2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: N-methoxy-N-methyl-tetrahydropyran-4-carboxamide (compound 95a) A mixture solution of tetrahydro-2H-pyran-4-carboxylic acid (2000.0 mg, 15.37 mmol) was added HATU (11688.3 mg, 30.74 mmol) and DIPEA (20 mL) in DCM (60 mL) was stirred at rt for 30 min. Then O, N-dimethylhydroxylamine (1798.8 mg, 18.44 mmol) was added and the reaction was stirred at rt for 12 hrs. The mixture was washed with brine (30 mL) three times, dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by flash chromatography on silica gel (EA in PE=0~100%) to give compound 95a (2000 mg, 11.55 mmol, 67.62% yield) as a light yellow oil. MS obsd.: 174 (MH + ). Step 2: (2,6-dichloro-4-pyridyl)-tetrahydropyran-4-yl-methanone (compound 95b) To a solution of n-BuLi (1.2 mL, 2.89 mmol) in THF (20 mL) was added 2,6- dichloropyridine (428 mg, 2.89 mmol) at -70 °C for 40 min under nitrogen and compound 95a (500.0 mg, 2.89 mmol) was added. The mixture was stirred at -70 °C for 4 hrs, quenched with saturated NH4Cl (50 mL) and extracted with EA (30 mL × 3). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by flash chromatography on silica gel (EA in PE = 0-30%) to give compound 95b (200 mg, 0.770 mmol, 23.97% yield) as a white solid. MS obsd.: 260, 262 (MH + ). Step 2: 2,6-dichloro-4-[difluoro(tetrahydropyran-4-yl)methyl]pyridin e (compound 95c) Compound 95b (200.0 mg, 0.770 mmol) was dissolved in BAST (3.0 mL) and the reaction was stirred at 25 °C for 12 hrs. The mixture was quenched with ice water (20 mL) and extracted with EA (20 mL) two times. The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by flash chromatography on silica gel (EA in PE=0~50%) to give compound 95c (150 mg, 0.530 mmol, 62.24% yield) as a white solid. MS obsd.: 282, 284 (MH + ). Step 3: tert-butyl N-[1-[4-[4-[6-chloro-4-[difluoro(tetrahydropyran-4-yl)methyl ]-2- pyridyl]piperazin -1-yl]sulfonylphenyl]-5-oxo-pyrrolidin-3-yl]carbamate (compound 95d) Compound 95c (150.0 mg, 0.530 mmol) and intermediate C (92.93 mg, 0.220 mmol) was dissolved in a mixed solution of DMSO (0.5 mL) and DIPEA (2.0 mL). The reaction was stirred at 115 °C for 12 h. The mixture was quenched with brine (50 mL) and extracted with DCM (30 mL) two times. The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by flash chromatography on silica gel (EA in PE=10~70%) to give compound 95d (100 mg, 0.150 mmol, 25.26% yield) as a yellow solid. MS obsd.: 670 (MH + ). Step 4: 4-amino-1-[4-[4-[6-chloro-4-[difluoro(tetrahydropyran-4-yl)m ethyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one (Example 95) Compound 95d (100.0 mg, 0.150 mmol) was dissolved in 2 M HCl/EA (10 mL) and the reaction was stirred at rt for 4 hrs. The mixture was then concentrated and the crude was purified by prep-HPLC to give Example 95 (28 mg, 0.05 mmol, 32.6% yield) as a white solid. MS obsd.: 570 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.92 (d, J = 8.8 Hz, 2H), 7.75 (d, J = 8.8 Hz, 2H), 6.79 (s, 1H), 6.71 (s, 1H), 3.98 (dd, J = 9.6, 6.0 Hz, 1H), 3.85 (dd, J = 11.2, 3.2 Hz, 2H), 3.65 (dd, J = 11.6, 7.2 Hz, 5H), 3.48 (dd, J = 9.6, 3.2 Hz, 1H), 3.24 (t, J = 10.8 Hz, 2H), 2.92 - 3.01 (m, 4H), 2.76 (dd, J = 16.8, 6.8 Hz, 1H), 2.45 (s, 1H), 2.24 (dd, J = 16.8, 4.0 Hz, 1H), 1.97 (s, 2H), 1.39 (dt, J = 12.4, 8.8 Hz, 4H). Example 96 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(tetrahydrofuran-3-yl)m ethyl]-2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 95 by using tetrahydrofuran-3-carboxylic acid instead of tetrahydro-2H-pyran-4-carboxylic acid. Example 96 (160 mg) was obtained as a white solid. MS obsd.: 556 (MH + ). 1 H NMR (400 MHz, CD3OD): δ ppm 7.92 (d, J = 8.8 Hz, 2H), 7.81 (d, J = 8.8 Hz, 2H), 6.74 (d, J = 8.8 Hz, 2H), 4.35 (dd, J = 11.2, 6.8 Hz, 1H), 4.11 (s, 1H), 3.89 (dd, J = 11.2, 2.8 Hz, 1H), 3.73 - 3.84 (m, 3H), 3.60 - 3.73 (m, 5H), 2.99 - 3.18 (m, 6H), 2.63 (dd, J = 18.0, 3.2 Hz, 1H), 1.88 - 2.00 (m, 2H). Example 97 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(oxetan-3-yl)methyl]-2- pyridyl]piperazin-1- yl]sulfonylphenyl] pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 95 by using oxetane-3-carboxylic acid instead of tetrahydro-2H-pyran-4-carboxylic acid. Example 97 (22 mg) was obtained as a white solid. MS obsd.: 542 (MH + ). 1 H NMR (400 MHz, CD3OD): δ ppm 7.91 (d, J = 8.8 Hz, 2H), 7.79 (d, J = 8.8 Hz, 2H), 6.73 (s, 1H), 6.67 (s, 1H), 5.60 (s, 1H), 5.50 (s, 1H), 4.15 (dd, J = 10.0, 6.4 Hz, 1H), 4.07 (s, 2H), 3.78 (td, J = 7.2, 3.6 Hz, 1H), 3.61 - 3.72 (m, 5H), 3.26 - 3.28 (m, 1H), 3.01 - 3.10 (m, 4H), 2.92 (dd, J = 17.2, 7.6 Hz, 1H), 2.41 (dd, J = 17.2, 4.0 Hz, 1H). Example 98 4-Amino-1-[4-[4-[6-chloro-4-[3,6-dihydro-2H-pyran-4-yl(diflu oro)methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 95 by using 3,6-dihydro-2H-pyran-4-carboxylic acid instead of tetrahydro-2H-pyran-4-carboxylic acid. Example 98 (60 mg) was obtained as a white solid. MS obsd.: 568 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.92 (d, J = 8.8 Hz, 2H), 7.76 (d, J = 8.8 Hz, 2H), 6.78 (s, 1H), 6.67 (s, 1H), 6.09 (s, 1H), 4.09 (d, J = 16.0 Hz, 3H), 3.80 (s, 1H), 3.58 - 3.70 (m, 7H), 2.96 (s, 4H), 2.87 (dd, J = 17.2, 7.2 Hz, 1H), 2.38 (dd, J = 17.2, 2.8 Hz, 1H), 2.04 (s, 2H). Example 99 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(tetrahydropyran-3-yl)m ethyl]-2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 95 by using tetrahydro-2H-pyran-3-carboxylic acid instead of tetrahydro-2H-pyran-4-carboxylic acid. Example 99 (200 mg) was obtained as a white solid. MS obsd.: 570.2 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.92 (d, J = 8.8 Hz, 2H), 7.77 (d, J = 8.8 Hz, 2H), 6.80 (s, 1H), 6.72 (s, 1H), 4.13 (dd, J = 10.4, 6.4 Hz, 1H), 3.73 - 3.88 (m, 3H), 3.59 - 3.73 (m, 5H), 3.21 (ddd, J = 11.2, 9.2, 4.4 Hz, 2H), 2.83 - 3.03 (m, 5H), 2.45 (dd, J = 17.4, 3.2 Hz, 2H), 1.61 (dd, J = 31.4, 12.4 Hz, 2H), 1.29 - 1.52 (m, 2H). Example 100 4-Amino-1-[4-[4-[6-chloro-4-[difluoro(8-oxabicyclo[3.2.1]oct an-3-yl)methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 95 by using 8- oxabicyclo[3.2.1]octane-3-carboxylic acid instead of tetrahydro-2H-pyran-4-carboxylic acid. Example 100 (70.4 mg) was obtained as a white solid. MS obsd.: 596.3 (MH + ). 1 H NMR (400 MHz, CD 3 OD): δ ppm 7.95 (d, J = 8.8 Hz, 2H), 7.85 (d, J = 8.8 Hz, 2H), 6.70 (d, J = 5.2 Hz, 2H), 4.42 (dd, J = 11.4, 6.8 Hz, 3H), 4.21 (t, J = 7.6 Hz, 1H), 3.97 (dd, J = 11.4, 2.0 Hz, 1H), 3.66 - 3.78 (m, 4H), 3.21 (dd, J = 18.0, 8.4 Hz, 1H), 3.14 - 3.03 (m, 4H), 2.71 (dd, J = 18.0, 2.8 Hz, 1H), 2.46 - 2.64 (m, 1H), 1.86 - 1.99 (m, 2H), 1.60 - 1.81 (m, 4H), 1.40 (dd, J = 13.2, 4.4 Hz, 2H). Example 101 (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro(3-oxabicyclo[3.1. 0]hexan-6-yl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 95 by using 8- oxabicyclo[3.2.1]octane-3-carboxylic acid instead of tetrahydro-2H-pyran-4-carboxylic acid and using intermediate D instead of intermediate C. Example 101 (300 mg) was obtained as a white solid. MS obsd.: 568.3 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.92 (d, J = 8.8 Hz, 2 H), 7.75 (m, J = 8.8 Hz, 2 H), 6.85 (s, 1 H), 6.79 (s, 1 H), 3.99 (br dd, J = 9.6, 6.0 Hz, 1 H), 3.75 (d, J = 8.8 Hz, 2 H), 3.56 (br d, J = 8.4 Hz, 2 H), 3.48 (br dd, J = 9.6, 3.2 Hz, 2 H), 3.33 (br s, 4 H), 2.96 (br s, 4 H), 2.76 (dd, J = 16.8, 6.8 Hz, 1 H), 2.24 (br dd, J = 16.8, 4.0 Hz, 2 H), 2.03 (br s, 2 H), 1.43 - 1.56 (m, 1 H). Example 102 and Example 103 (4R)-4-amino-1-[4-[4-[6-chloro-4-[[(2S)-1,4-dioxan-2-yl]-dif luoro-methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one (4R)-4-amino-1-[4-[4-[6-chloro-4-[[(2R)-1,4-dioxan-2-yl]-dif luoro-methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compounds were prepared in analogy to the preparation of Example 95 by using intermediate E or intermediate F instead of compound 95b, and using intermediate D instead of intermediate C. Example 102 (40.4 mg) was obtained as a white solid. MS obsd.: 572 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.92 (d, J = 9.2 Hz, 2 H), 7.72 - 7.77 (m, 2 H), 6.84 (s, 1 H), 6.76 (s, 1 H), 4.11 - 4.27 (m, 1 H), 3.99 (dd, J = 9.6, 6.0 Hz, 1 H), 3.71 - 3.85 (m, 2 H), 3.41 - 3.68 (m, 8 H), 2.96 (br t, J = 4.8 Hz, 4 H), 2.76 (dd, J = 16.8, 6.8 Hz, 1 H), 2.11 - 2.38 (m, 2 H), 1.97 - 2.05 (m, 1 H), 1.23 (br s, 1 H). Example 103 (35.8 mg) was obtained as a white solid. MS obsd.: 572 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 7.92 (d, J = 9.2 Hz, 2 H), 7.72 - 7.77 (m, 2 H), 6.84 (s, 1 H), 6.76 (s, 1 H), 4.11 - 4.27 (m, 1 H), 3.99 (dd, J = 9.6, 6.0 Hz, 1 H), 3.71 - 3.85 (m, 2 H), 3.41 - 3.68 (m, 8 H), 2.96 (br t, J = 4.8 Hz, 4 H), 2.76 (dd, J = 16.8, 6.8 Hz, 1 H), 2.11 - 2.38 (m, 2 H), 1.97 - 2.05 (m, 1 H), 1.23 (br s, 1 H). Example 104 (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-[(2S)-morpholin-2 -yl]methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: tert-butyl (2S)-2-[[2-[4-[4-[(4R)-4-(tert-butoxycarbonylamino)-2-oxo-py rrolidin-1- yl]phenyl]sulfonylpiperazin-1-yl]-6-chloro-4-pyridyl]-difluo ro-methyl]morpholine-4- carboxylate (compound 104a) To a solution of intermediate G (59.58 mg, 0.160 mmol) and intermediate D (60.0 mg, 0.140 mmol) in DMSO (2 mL) was added DIPEA (36.47 mg, 0.280 mmol). The mixture was stirred at 100 °C for 12 hrs. The mixture was added water (50 mL) and EA (50 mL). The organic phase was washed with aq.CaCl2 (30 mL), brine (30 mL), dried over Na 2 SO 4 and concentrated. The crude was purified by prep-TLC to give compound 104a (90 mg, 0.120 mmol, 82.56% yield) as a white solid. MS obsd.: 771.1 (MH + ). Step 2: (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-[(2S)-morpholin-2 -yl]methyl]-2- pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one (Example 104) A mixture of compound 104a (80.0 mg, 0.10 mmol) in TFA (1.33 mL, 18.01 mmol) was stirred at 15 °C for 2 hrs. The mixture was concentrated and the crude was purified by prep- HPLC to give Example 104 (56 mg, 0.090 mmol, 83.54% yield) as a white solid. MS obsd.: 571.1 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 7.89 - 7.95 (m, 2 H), 7.80 (d, J = 8.8 Hz, 3 H), 6.85 (s, 1 H), 6.76 (s, 1 H), 4.33 - 4.44 (m, 1 H), 4.26 (dd, J = 11.2, 7.2 Hz, 1 H), 3.99 - 4.11 (m, 2 H), 3.87 (d, J =12.8 Hz, 1 H), 3.77 (t, J = 12.8 Hz, 1 H), 3.64 - 3.71 (m, 4 H), 3.18 (d, J = 12.4 Hz, 3 H), 3.02 - 3.12 (m, 2 H), 2.94 - 3.02 (m, 5 H). Example 105 (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-[(2R)-morpholin-2 -yl]methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 104 by using intermediate H instead of intermediate G. Example 105 (35.5 mg) was obtained as a white solid. MS obsd.: 571.1 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 8.38 (br s, 3 H) 7.92 (d, J = 8.8 Hz, 2 H), 7.80 (d, J = 9.2 Hz, 2 H), 6.86 (s, 1 H), 6.76 (s, 1 H), 4.31 - 4.46 (m, 1 H), 4.27 (dd, J = 11.2, 6.8 Hz, 1 H), 4.03 (dd, J = 12.4, 2.4 Hz, 2 H), 3.86 (d, J = 10.4 Hz, 1 H), 3.64 - 3.82 (m, 5 H), 3.03 - 3.26 (m, 3 H), 2.95 - 3.03 (m, 5 H), 2.58 - 2.70 (m, 2 H). Example 106 (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro(1,2,3,6-tetrahydr opyridin-4-yl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 104 by using intermediate I instead of intermediate G. Example 106 (11.8 mg) was obtained as a white solid. MS obsd.: 567.1 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 9.53 (br s, 2 H), 8.66 (s, 3 H), 7.91 (d, J = 8.8 Hz, 2 H), 7.79 (d, J = 8.8 Hz, 2 H), 6.80 (s, 1 H), 6.72 (s, 1 H), 6.17 ( s, 1 H), 4.27 (dd, J = 11.2, 6.8 Hz, 1 H), 4.06 ( s, 1 H), 3.92 (d, J = 9.6 Hz, 1 H), 3.68 (br s, 6 H), 3.16 (br s, 2 H), 2.97 (br s, 4 H), 2.67 (d, J = 18.0 Hz, 1 H), 2.26 (br s, 2 H). Example 107 and Example 108 (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-[(3R)-3-piperidyl ]methyl]-2-pyridyl]piperazin- 1-yl]sulfonylphenyl]pyrrolidin-2-one (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-[(3S)-3-piperidyl ]methyl]-2-pyridyl]piperazin- 1-yl]sulfonylphenyl]pyrrolidin-2-one The title compounds were prepared according to the following procedures: Step 1: tert-butyl 3-[2-chloro-6-[4-[4-[(4R)-4-(benzyloxycarbonylamino)-2-oxo-p yrrolidin- 1-yl]phenyl]sulfonylpiperazin-1-yl]pyridine-4-carbonyl]piper idine-1-carboxylate (compound 107a) To a solution of benzyl N-[(3R)-5-oxo-1-(4-piperazin-1-ylsulfonylphenyl)pyrrolidin-3 - yl]carbamate (280 mg, 557 µmol) and intermediate J (200 mg, 557 µmol) in DMSO (10 mL) was added DIPEA (216 mg, 292 µL, 1.67 mmol). The reaction was stirred at 100 °C for 2 days. After cooling to rt, the mixture was diluted with water (30 mL) and extracted with EA (30 mL) three times. The combined organic layer was washed with brine (100 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by flash column on silica gel (EA in PE=70~90%) to give Intermediate 107a (320 mg, 69.6% yield) as yellow oil. MS obsd.: 725.2 (MH + ). Step 2: tert-butyl (3R)-3-[[2-[4-[4-[(4R)-4-(benzyloxycarbonylamino)-2-oxo-pyrr olidin-1- yl] phenyl]sulfonylpiperazin-1-yl]-6-chloro-4-pyridyl]-difluoro- methyl]piperidine-1-carboxylate and tert-butyl (3S)-3-[[2-[4-[4-[(4R)-4-(benzyloxycarbonylamino)-2-oxo-pyrr olidin-1- yl]phenyl]sulfonylpiperazin-1-yl]-6-chloro-4-pyridyl]-difluo ro-methyl]piperidine-1- carboxylate(compounds 107b and 108b) Compound 107a (280 mg, 358 µmol) was dissolved in the DAST (10 mL) and the mixture was stirred at rt for 2 days. The reaction was quenched by adding water dropwise in the ice/water bath and then extracted with DCM (50 mL) three times. The combined organic layer was washed with brine (100 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated. The crude was purified by flash chromatography on silica gel (70-100% EA in PE) and then chiral separation by SFC to give compound 107b (60 mg, 74.7 µmol, 20.8% yield, faster eluting) and compound 108b (56 mg, 69.7 µmol, 19.5% yield, slower eluting) as white solids. MS obsd.: 803.0 (MH + ). SFC conditions: Column: AD, 250×30 mm I.D., 5µm; Mobile phase: A for CO 2 and B for Ethanol (0.1% NH3H2O); Gradient: B 50%; Flow rate: 50 mL/min; Backpressure: 100 bar; Column temperature: 35 °C. Step 3: (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-[(3R)-3-piperidyl ]methyl]-2-pyridyl] piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one and (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro- [(3S)-3-piperidyl]methyl]-2-pyridyl]piperazin-1-yl]sulfonylp henyl]pyrrolidin-2-one (Example 107 and Example 108) A solution of compound 107b (50 mg, 62.2 µmol) and TFA was stirred at 65 °C for 2 hrs. The mixture was the concentrated and the crude was purified by prep-HPLC to give Example 107 (20.8 mg, 36.4 µmol, 58.6% yield) as a white solid. MS obsd.: 569.2 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 8.29 (br s, 2 H), 7.92 (m, J = 8.8 Hz, 2 H), 7.80 (m, J = 8.8 Hz, 2 H), 6.81 (s, 1 H), 6.72 (s, 1 H), 4.26 (dd, J = 11.2, 6.8 Hz, 1 H), 4.08 (br s, 1 H), 3.74 - 3.92 (m, 1 H), 3.62 - 3.72 (m, 4 H), 3.18 - 3.29 (m, 2 H), 3.08 (dd, J = 18.0, 8.0 Hz, 2 H), 2.96 - 3.02 (m, 4 H), 2.77 (br d, J= 12.0 Hz, 2 H), 1.90 - 2.12 (m, 1 H), 1.69 - 1.87 (m, 2 H), 1.57 (q, J = 13.2 Hz, 1 H), 1.28 - 1.47 (m, 2 H), 1.24 (br s, 1 H). Example 108 (23.7 mg) was prepared in analogy to the preparation of Example 107 by using compound 108b instead of compound 107b. MS obsd.: 569.2 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ): δ ppm 7.92 (m, J = 8.98 Hz, 2 H), 7.80 (m, J = 8.8 Hz, 2 H), 6.80 (s, 1 H), 6.73 (s, 1 H), 4.26 (dd, J = 11.2, 6.98 Hz, 1 H), 4.08 (br s, 1 H), 3.81 (m, 1 H), 3.62 - 3.72 (m, 4 H), 3.15 - 3.28 (m, 2 H), 3.08 (dd, J = 18.0, 8.4 Hz, 2 H), 2.98 (m, 4 H), 2.77 (m, 2 H), 1.90 - 2.12 (m, 1 H), 1.69 - 1.87 (m, 2 H), 1.57 (q, J = 13.2 Hz, 1 H), 1.28 - 1.47 (m, 2 H)1.24 (br s, 1 H). Example 109 (4R)-4-amino-1-[4-[4-[4-[azetidin-3-yl(difluoro)methyl]-6-ch loro-2-pyridyl]piperazin-1- yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 104 by using intermediate I instead of intermediate K. Example 109 (30 mg) was obtained as a white solid. MS obsd.: 540.9 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.92 (d, J = 9.2 Hz, 2H), 7.77 (d, J = 9.2 Hz, 2H), 6.83 (s, 1H), 6.78 (s, 1H), 4.08 (dd, J = 6.4, 9.6 Hz, 1H), 3.80 (t, J = 9.2 Hz, 5H), 3.55 - 3.72 (m, 6H), 2.96 (t, J = 4.8 Hz, 4H), 2.87 (dd, J = 7.2, 16.8 Hz, 1H), 2.30 - 2.40 (m, 1H). Example 110 3-[4-[4-[6-Chloro-4-[difluoro-[(2S)-morpholin-2-yl]methyl]-2 -pyridyl]piperazin-1- yl]sulfonylphenyl] -5-(hydroxymethyl)oxazolidin-2-one The title compound was prepared according to the following procedures: Step 1: tert-butyl 4-[4-[5-(hydroxymethyl)-2-oxo-oxazolidin-3-yl]phenyl]sulfony l piperazine-1-carboxylate (compound 110a) A mixture of tert-butyl 4-((4-iodophenyl)sulfonyl)piperazine-1-carboxylate (0.430 g, 951 µmol), 5-(hydroxymethyl)oxazolidin-2-one (111 mg, 951 µmol), DMEDA (29.3 mg, 333 µmol), CuI (63.4 mg, 333 µmol) and K 3 PO 4 (404 mg, 1.9 mmol) in DMF (4 mL) was heated at 75 o C in a microwave for 1h. The mixture was poured into ice water, extracted with DCM, dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by flash chromatography on silica gel to give compound 110a (310 mg, 702 µmol, 73.9 % yield) as a white solid. MS obsd.: 442 (MH + ). Step 2: 5-(hydroxymethyl)-3-(4-piperazin-1-ylsulfonylphenyl)oxazolid in-2-one (compound 110b) A mixture of compound 110a (0.060 g, 136 µmol) and TFA (877 mg) in CH 2 Cl 2 (1 mL) was stirred at rt for 1h. The mixture was concentrated and used directly in next step. MS obsd.: 342 (MH + ). Step 3: tert-butyl (2S)-2-[[2-chloro-6-[4-[4-[5-(hydroxymethyl)-2-oxo-oxazolidi n-3- yl]phenyl]sulfonylpiperazin-1-yl]-4-pyridyl]-difluoro-methyl ]morpholine-4-carboxylate (compound 110c) A mixture of compound 110b (0.045 g, 132 µmol), intermediate H (50.5 mg, 132 µmol) and DIPEA (85.1 mg, 115 µl, 659 µmol) in NMP (1.5 mL) was stirred at 100 °C overnight. The mixture was diluted with EA (10 mL), washed with water (10 mL) twice. The organic layer was dried, concentrated and used directly in next step. MS obsd.: 688 (MH + ). Step 4: 3-[4-[4-[6-chloro-4-[difluoro-[(2S)-morpholin-2-yl]methyl]-2 -pyridyl]piperazin-1- yl]sulfonylphenyl]-5-(hydroxymethyl)oxazolidin-2-one (Example 110) A mixture of compound 110c (0.090 g, 131 µmol,) and TFA (844 mg) in CH2Cl2 (1.5 mL) was stirred at rt for 1h. The mixture was concentrated and isolated by pre-HPLC to give Example 110 (24 mg, 29.6% yield) as a white solid. MS obsd.: 588 (MH + ). 1 H NMR (400 MHz, DMSO-d6): δ ppm 7.73 - 7.78 (m, 2H), 7.69 - 7.71 (d, J = 8.4 Hz, 2H), 6.75 (s, 1H), 6.67 (s, 1H), 5.16 (br s, 1H), 4.61 - 4.73 (m, 1H), 3.94 - 4.16 (m, 2H), 3.81 (br dd, J = 6.0, 9.2 Hz, 2H), 3.55 - 3.59 (m, 4H), 3.41- 3.53 (m, 2H), 2.99 (br d, J = 12.4 Hz, 1H), 2.90 - 2.91 (m, 4H), 2.79 - 2.84 (m, 1H), 2.62 - 2.77 (m, 2H). Example 111 (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-(5-methylpyrazin- 2-yl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 89 by using 2- bromo-5-methyl-pyrazine instead of 2-bromopyridine. Example 111 (89 mg) was obtained as a white solid. MS obsd.: 578.0 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ) δ ppm 8.87 - 9.07 (m, 1 H), 8.50 - 8.68 (m, 1 H), 7.66 - 8.06 (m, 3 H), 6.64 - 7.01 (m, 2 H), 3.92 - 4.15 (m, 2 H), 3.51 - 3.79 (m, 7 H), 2.89 - 3.08 (m, 4 H), 2.71 - 2.91 (m, 1 H), 2.51 (br s, 3 H), 2.21 - 2.39 (m, 1 H). 19 F NMR (376 MHz, DMSO-d 6 ) δ ppm -97.90 - -96.24 (m, 2 F). Example 112 (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-(6-methylpyrazin- 2-yl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 89 by using 2- bromo-6-methyl-pyrazine instead of 2-bromopyridine. Example 112 (59 mg) was obtained as a white solid. MS obsd.: 578.2 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ) δ ppm 8.82 - 8.94 (m, 1 H), 8.73 (s, 1 H), 7.91 (d, J =9.05 Hz, 2 H), 7.74 (d, J =9.05 Hz, 2 H), 6.93 (s, 1 H), 6.81 (s, 1 H), 4.00 (dd, J =10.03, 6.11 Hz, 1 H), 3.60 - 3.71 (m, 5 H), 3.49 (br dd, J =9.66, 3.30 Hz, 1 H), 3.33 - 3.34 (m, 3 H), 2.89 - 3.01 (m, 4 H), 2.78 (dd, J =16.87, 7.09 Hz, 1 H), 2.26 (dd, J =16.63, 3.91 Hz, 1 H). 19 F NMR (376 MHz, DMSO-d 6 ) δ ppm -97.18 (s, 2 F). Example 113 (4R)-4-amino-1-[4-[4-[6-chloro-4-[(5-chloro-6-methyl-pyrazin -2-yl)-difluoro-methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 94 by using methyl 5-chloro-6-methylpyrazine-2-carboxylate instead of methyl pyrimidine-2-carboxylate. Example 113 (20 mg) was obtained as a white solid. MS obsd.: 612.3 (MH + ). 1 H NMR (400 MHz, DMSO-d6) δ ppm 8.79 (s, 1 H), 7.91 (d, J = 8.9 Hz, 2 H), 7.74 (d, J = 8.9 Hz, 2 H), 6.92 (s, 1 H), 6.82 (s, 1 H), 4.0-4.0 (m, 1 H), 3.6-3.7 (m, 5 H), 3.50 (br dd, J = 3.1, 9.8 Hz, 1 H), 2.95 (t, J = 4.7 Hz, 4 H), 2.7-2.8 (m, 1 H), 2.56 (s, 3 H), 2.26 (dd, J = 3.8, 16.9 Hz, 1 H). 19 F NMR (376 MHz, DMSO-d6) δ ppm -96.54 (s, 2 F). Example 114 (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-[cis-6-methylmorp holin-2-yl]methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: Tert-butyl cis-2-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]-6-methyl-mo rpholine-4- carboxylate (compound 114a) Compound 114a was prepared in analogy to the preparation of intermediate G by using (cis-6-methylmorpholin-2-yl)methanol instead of (R)-N-Boc-2-hydroxymethylmorpholine. Compound 114a (1.2 g) was obtained as a white solid. MS obsd.: 341.0 (MH + -56). Step 2: (4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-[cis-6-methylmorp holin-2-yl]methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one (Example 114) The title compound was prepared in analogy to the preparation of Example 104 by using compound 114a instead of Intermediate G. Example 114 (23.6 mg) was obtained as a white solid. MS obsd.: 585.4 (MH + ). 1 H NMR (400 MHz, DMSO-d6) δ ppm 8.59 (br d, J = 2.57 Hz, 3 H), 7.90 (d, J = 8.93 Hz, 2 H), 7.79 (d, J = 8.93 Hz, 2 H), 6.85 (s, 1 H), 6.74 (s, 1 H), 4.49 (q, J = 10.43 Hz, 1 H), 4.26 (dd, J = 11.07, 6.91 Hz, 1 H), 3.84 - 4.12 (m, 3 H), 3.67 (br s, 4 H), 3.17 - 3.28 (m, 2 H), 3.06 (dd, J = 17.85, 8.19 Hz, 1 H), 2.97 (br d, J = 4.16 Hz, 4 H), 2.72 - 2.85 (m, 1 H), 2.65 (br dd, J = 17.79, 2.75 Hz, 2 H), 1.09 (d, J = 6.24 Hz, 3 H). 19 F NMR (376 MHz, DMSO-d6) δ ppm -109.50 - -105.5 (m, 2 F). Example 115 ((4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-(5-methylmorphol in-2-yl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: Tert-butyl 2-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]-5-methyl-morpho line-4- carboxylate (compound 115a) Compound 115a was prepared in analogy to the preparation of intermediate G by using (5- methylmorpholin-2-yl)methanol instead of (R)-N-Boc-2-hydroxymethylmorpholine. Compound 115a (580 mg) was obtained as a white solid. MS obsd.: 341.2 (MH + -56). Step 2: ((4R)-4-amino-1-[4-[4-[6-chloro-4-[difluoro-(5-methylmorphol in-2-yl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one (Example 115) The title compound was prepared in analogy to the preparation of Example 104 by using compound 115a instead of Intermediate G. Example 115 (25.4 mg) was obtained as a white solid. MS obsd.: 585.2 (MH + ). 1 H NMR (400 MHz, DMSO-d 6 ) δ ppm 8.66 ( s, 3 H), 7.91 (d, J = 8.93 Hz, 2 H), 7.79 (d, J = 8.93 Hz, 2 H), 6.89 (s, 1 H), 6.77 (s, 1 H), 4.37 - 4.54 (m, 1 H), 4.27 (dd, J = 11.07, 6.91 Hz, 1 H), 4.06 ( s, 1 H), 3.92 ( d, J = 11.25 Hz, 2 H), 3.76 ( d, J = 12.10 Hz, 1 H), 3.68 ( s, 4 H), 3.52 ( d, J = 7.09 Hz, 1 H), 3.02 - 3.15 (m, 3 H), 2.97 ( s, 4 H), 2.67 (dd, J = 17.85, 2.81 Hz, 1 H), 1.29 (d, J = 6.85 Hz, 3 H). Example 116 (4R)-4-amino-1-[4-[4-[4-[[cis-3-azabicyclo[3.1.0]hexan-1-yl] -difluoro-methyl]-6-chloro-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: Tert-butyl cis-1-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]-3-azabicycl o[3.1.0]hexane-3- carboxylate (compound 116a) Compound 116a was prepared in analogy to the preparation of intermediate G by using tert-butyl cis-1-(hydroxymethyl)-3-azabicyclo[3.1.0]hexane-3-carboxylat e instead of (R)-N-Boc- 2-hydroxymethylmorpholine. Compound 116a (2.6 g) was obtained as a white solid. MS obsd.: 323.1 (MH + -56). Step 2: (4R)-4-amino-1-[4-[4-[4-[[cis-3-azabicyclo[3.1.0]hexan-1-yl] -difluoro-methyl]-6-chloro- 2-pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared in analogy to the preparation of Example 104 by using compound 116a instead of Intermediate G. Example 115 (80.2 mg) was obtained as a white solid. MS obsd.: 567.2 (MH + ). 1 H NMR (400 MHz, DMSO-d6) δ ppm 8.55 - 8.35 (m, 3 H), 7.95 - 7.88 (m, 2 H), 7.82 - 7.74 (m, 2 H), 6.84 - 6.80 (m, 1 H), 6.80 - 6.77 (m, 1 H), 4.30 - 4.22 (m, 1 H), 4.11 - 4.02 (m, 1 H), 3.91 - 3.84 (m, 1 H), 3.71 - 3.62 (m, 4 H), 3.28 - 3.21 (m, 2 H), 3.12 - 3.01 (m, 1 H), 3.01 - 2.93 (m, 4 H), 2.66 - 2.58 (m, 1 H), 2.24 - 2.16 (m, 1 H), 1.27 - 1.15 (m, 2 H). 19 F NMR (376 MHz, DMSO-d6) δ -97.642 (s, 2 F). Example 117 (4R)-4-amino-1-[4-[4-[6-chloro-4-[trans-difluoro-[2-(hydroxy methyl)cyclopropyl]methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: 2-(2,6-dichloro-4-pyridyl)-2,2-difluoro-acetaldehyde (117a) To a solution of 2-(2,6-dichloro-4-pyridyl)-2,2-difluoro-ethanol (2.5 g, 10.96 mmol) in DCM (20 mL) was added Dess-Martin periodinane (5.12 g, 12.06 mmol) at 0 °C under N 2 . The mixture was stirred at 25 °C for 2 hours. After the starting material was consumed, the mixture was poured into water (100 mL) at 0 °C, and extracted with EA (150 mL × 3). The organic layer was washed with aq. NH 4 Cl solution (100 mL × 3) and brine (100 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated to dryness to give crude product. The residue was purified by column chromatography on silica gel (EA in PE = 1:100 - 2:1) to give compound 117a (1.8 g, 72.64% yield) as colorless oil. MS obsd.: 243.7 (MH+18 + ). Step 2: Ethyl (E)-4-(2,6-dichloro-4-pyridyl)-4,4-difluoro-but-2-enoate (117b) To a solution of compound 117a (1.8 g, 7.96 mmol) in THF (15 mL) was added (carbethoxymethylene)triphenylphosphorane (5.55 g, 15.93 mmol) under N 2, and the reaction mixture was stirred at 70 °C for 5 hours. After the starting material was consumed, the mixture was poured into water (100 mL). Then, the mixture was extracted with EA (150 mL × 3). The combined organic layer was washed with aq. NH 4 Cl (100 mL × 3) and brine (100 mL × 2), dried over anhydrous Na 2 SO 4 , filtered and concentrated to dryness to give crude product. The crude product was purified by column chromatography on silica gel (EA in PE = 1:100 - 1:5 to give compound 117b (1.5 g, 60.34% yield) as colorless oil. MS obsd.: 295.8 (MH + ). Step 3: Trans-ethyl 2-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]cyclopropanecarb oxylate ( compound 117c) To a solution of sodium hydride (303.95 mg, 7.6 mmol) in THF (15 mL) and DMSO (15 mL) were added trimethylsulfoxonium iodide (2.23 g, 10.13 mmol) at 0 °C under N 2 and compound 117b (1.5 g, 5.07 mmol). The reaction was stirred at 15 °C for 1 hour. After the starting material was consumed, the mixture was poured into water (100 mL) at 0 °C. The resulting mixture was extracted with EA (150 mL × 3). The combined organic layer was washed with aq. NH 4 Cl solution (100 mL × 3) and brine (100 mL × 2), dried over anhydrous Na 2 SO 4 , filtered and concentrated to dryness to give crude product. The crude product was purified by column chromatography on silica gel (EA:PE=1:10 - 1:1) to give 117c (900 mg, 57.29% yield) as colorless oil. MS obsd.: 310.2 (MH + ). Step 4: [Tran-2-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]cyclopropy l]methanol (117d) To a solution of compound 117d (600.0 mg, 1.93 mmol) in ethanol (4 mL) was added sodium borohydride (146.39 mg, 3.87 mmol) slowly at 0 °C. The mixture was stirred at 45 °C for 2 hours. After the starting material was consumed, the mixture was poured into water (100 mL) at 0 °C, and extracted with EA (100 mL × 3). The combined organic layer was washed with aq NH4Cl solution (100 mL × 3) and brine (100 mL × 2), dried over anhydrous Na 2 SO 4 , filtered and concentrated to dryness to give the crude product, which was purified by prep-HPLC to give 117d (400 mg, 77.12% yield) as a colorless oil. MS obsd.: 267.8 (MH + ). Step 4: Tert-butyl N-[(3R)-1-[4-[4-[6-chloro-4-[trans-difluoro-[2- (hydroxymethyl)cyclopropyl]methyl]-2-pyridyl]piperazin-1-yl] sulfonylphenyl]-5-oxo- pyrrolidin-3-yl]carbamate (117e) To a solution of compound 117d (100.0 mg, 0.370 mmol) in DMSO (0.5 mL) were added Intermediate D (158.35 mg, 0.370 mmol) and N,N-diisopropylethylamine (0.13 mL, 0.750 mmol) in one portion. The reaction was stirred at 100 °C for 16 hours. After completion, the mixture was diluted with water (20 mL) and extracted with EA (20 mL × 3). The combined organic layer was washed with brine (50 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give a crude product. The crude was purified by prep-HPLC to give 117e (205 mg, 83.76% yield) as a yellow oil. MS obsd.: 656.14 (MH + ). Step 5: (4R)-4-amino-1-[4-[4-[6-chloro-4-[trans-difluoro-[2- (hydroxymethyl)cyclopropyl]methyl]-2-pyridyl]piperazin-1-yl] sulfonylphenyl]pyrrolidin-2-one To a solution of 117e (190.0 mg, 0.290 mmol) in DCM (2 mL) was added hydrochloric acid methanol solution (3.62 mL, 14.48 mmol) in one portion. The reaction 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 117 (98 mg, 59.67% yield) as a white solid. MS obsd.: 556.1 (MH + ). 1 H NMR (400 MHz, DMSO-d6) δ ppm 8.56 - 8.46 (m, 3 H), 7.94 - 7.89 (m, 2 H), 7.80 (s, 2 H), 6.98 - 6.94 (m, 1 H), 6.80 (s, 1 H), 4.78 - 4.68 (m, 1 H), 4.29 - 4.21 (m, 1 H), 4.09 - 4.02 (m, 1 H), 3.92 - 3.86 (m, 1 H), 3.71 - 3.62 (m, 4 H), 3.47 - 3.40 (m, 1 H), 3.24 - 3.17 (m, 1 H), 3.10 - 3.01 (m, 1 H), 3.00 - 2.91 (m, 4 H), 2.67 - 2.60 (m, 1 H), 1.63 - 1.49 (m, 1 H), 1.36 - 1.26 (m, 1 H), 0.86 - 0.78 (m, 1 H). 19 F NMR (376 MHz, DMSO-d 6 ) δ -103.881 (s, 2 F). Example 118 (4R)-4-amino-1-[4-[4-[4-[[trans-4-(aminomethyl)cyclohexyl]-d ifluoro-methyl]-6-chloro-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: Trans-methyl 4-(hydroxymethyl)cyclohexanecarboxylate (118a) To a solution of trans-4-methoxycarbonylcyclohexanecarboxylic acid (50.0 g, 268.51 mmol) in THF (150 mL) was added BH3•Me2S (53.7 mL, 537.03 mmol) dropwise at 0-10°C for 1 hour. 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 (EA:PE = 0-50%) to give compound 118a (28 g, 60.55% yield) as colorless oil. Step 2: Trans-methyl 4-formylcyclohexanecarboxylate (118b) 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 N2. The solution of compound 118b (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 EA (600 mL) and washed with aq. CaCl2 solution (100 mL × 3) and brine (50 mL × 2). The combined organic layer was dried over anhydrous Na 2 SO 4 , filtered, and concentrated in vacuo to give the crude residue, which was purified by flash chromatography on silica gel (EA:PE = 3-50%) to give compound 118b (24 g, 86.73% yield) as light yellow oil. Step 3: Trans-methyl 4-[(2,6-dichloro-4-pyridyl)-hydroxy-methyl]cyclohexanecarbox ylate (118c) To a solution of 2,6-dichloro-4-iodo-pyridine (42.48 g, 155 mmol) in THF (50 mL) was added isopropylmagnesium chloride lithium chloride complex in THF (130.16 mL, 169.2 mmol) at -40°C under N 2 . The mixture was stirred at 25 °C for 1 hour. The reaction mixture was then added into a solution of compound 118b (24.0 g, 141 mmol) in THF (100 mL) dropwise at -40°C. Upon addition, the mixture was stirred at 25 °C for 3 hours. After completion, the reaction mixture was poured into aq. NH 4 Cl solution (200 mL) and extracted with EA (300 mL × 3). The combined organic layer was washed with brine (100 mL × 2), dried over anhydrous Na 2 SO 4 , filtered, and concentrated in vacuo to give the crude residue, which was purified by flash chromatography on silica gel (EA:PE = 0-30%) to give compound 118c (33 g, 73.55% yield) as a white solid. MS obsd. (ESI + ) [(M+Cl) + ]: 317.8. Step 4: Trans-methyl 4-(2,6-dichloropyridine-4-carbonyl)cyclohexanecarboxylate (118d) To a solution of compound 118c (33.0 g, 103.71 mmol) in THF (150 mL) was added Dess- Martin periodinane (48.39 g, 114 mmol) slowly at 0 °C under N 2 . The reaction was stirred at 25 °C for 3 hours. After completion, the mixture was diluted with EA (500 mL), and poured into aq. K 2 CO 3 solution (200 mL). The mixture was kept stirring at room temperature for 0.5 hour. The combined organic layer was washed with brine (100 mL × 2), dried over anhydrous Na 2 SO 4 , filtered, and concentrated in vacuo to give crude compound 118d (30 g, 91.5% 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 (118e) To a solution of compound 118d (29.5 g, 93.3 mmol) in DCM (20 mL) was added DAST (135.35 g, 839.71 mmol), and the reaction was stirred at 45°C for 12 hours. After completion, the reaction mixture was poured into the ice-water and extracted with EA (200 mL × 3). The combined organic layer was washed with aq. K 2 CO 3 solution (100 mL × 2) and brine (100 mL × 2), dried over anhydrous Na 2 SO 4 , filtered, and concentrated in vacuo to give a crude residue. The residue was recrystallized with methanol (150 mL) to give compound 118e (25.6 g, 81.13% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 337.8. Step 6: Trans-[4-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]cyclohexy l]methanol (118f) To a solution of 118e (250.0 mg, 0.740 mmol) in THF (5 mL) was added LiAlH4 (42.14 mg, 1.11 mmol) at -10 °C and the reaction was stirred at -10 °C for 2 hours. After completion, the mixture was allowed to warm to 0 ℃, and quenched by addition of 0.1 mL H 2 O, followed by 0.15 mL of 15% aq. NaOH solution. After stirring for 0.5 hour, the mixture was filtered through a Celite pad, and the filtrate was concentrated in vacuo to give crude product. The residue was purified by prep-HPLC to give 118f (210 mg, 91.59% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 310.1. Step 7: tert-butyl N-[(3R)-1-[4-[4-[4-[[trans-4-(aminomethyl)cyclohexyl]-difluo ro-methyl]-6- chloro-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-5-oxo-pyrrol idin-3-yl]carbamate (compound 118g) To a solution of compound 118f (380.0 mg, 0.540 mmol) in DCM (5 mL) was added trifluoromethanesulfonic anhydride (0.18 mL, 1.09 mmol) at -20 °C, and the reaction was stirred at -20 °C for 0.5 hour. To the mixture was added ammonia gas in THF (92.68 mg, 5.44 mmol) and stirred for 1 hour at 45 °C. After completion, the mixture was quenched by slow addition of saturated aqueous ammonium chloride (20 mL). The resulting mixture was extracted with EA (20 mL × 3). The combined organic layer was washed with brine (50 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated under reduced pressure to afford the crude product, which was purified by prep-HPLC to give 118g (120 mg, 0.170 mmol, 31.62% yield) as white solid. MS obsd. (ESI + ) [(M+H) + ]: 697.2. Step 8: (4R)-4-amino-1-[4-[4-[4-[[trans-4-(aminomethyl)cyclohexyl]-d ifluoro-methyl]-6-chloro- 2-pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one To a solution of compound 118 (40.0 mg, 0.060 mmol) in DCM (1 mL) was added TFA (65.4 mg, 0.570 mmol) at 25 °C. The reaction was stirred at 25 °C for 15 minutes. After completion, the mixture was concentrated under reduced pressure to afford the crude product, which was purified by prep-HPLC to give Example 118 (24.7 mg, 59.1% yield) as a white solid. MS obsd. (ESI + ) [(M+H) + ]: 597.2. 1 H NMR (400 MHz, DMSO-d6) δ ppm 8.74 - 8.26 (m, 3 H), 7.95 - 7.91 (m, 2 H), 7.85 - 7.75 (m, 2 H), 6.86 - 6.76 (m, 1 H), 6.76 - 6.68 (m, 1 H), 4.35 - 4.21 (m, 1 H), 4.13 - 4.00 (m, 1 H), 3.97 - 3.82 (m, 1 H), 3.75 - 3.60 (m, 4 H), 3.15 - 2.88 (m, 5 H), 2.71 - 2.60 (m, 3 H), 2.22 - 2.04 (m, 1 H), 1.90 - 1.74 (m, 2 H), 1.73 - 1.61 (m, 2 H), 1.56 - 1.42 (m, 1 H), 1.18 - 1.01 (m, 2 H), 0.99 - 0.83 (m, 2 H). 19 F NMR (376 MHz, DMSO-d6) δ -103.881 (s, 2 F). Example 119 (4R)-4-amino-1-[4-[4-[6-chloro-4-[trans-difluoro-(3-hydroxyc yclobutyl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one The title compound was prepared according to the following procedures: Step 1: Trans-(3-benzyloxycyclobutyl)methanol (compound 119a) To a solution of 3-benzyloxycyclobutanecarboxylic acid (1.2 g, 5.82 mmol) in THF (5 mL) was added borane-methyl sulfide complex (1.16 mL, 11.64 mmol) slowly. The reaction was stirred at 20 °C for 2 hours. After completion, the reaction was quenched with methanol (5 mL), and diluted with aq. NH 4 Cl solution (20 mL). The aqueous phase was extracted with EA (50 mL × 2). The combined organic layer was washed with brine (100 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give compound 119a (1.05 g, 93.87% yield) as colorless oil. Step 2: Trans-3-benzyloxycyclobutanecarbaldehyde (compound 119b) To a solution of DMSO (0.85 mL, 11.96 mmol) in DCM (20 mL) was added 119a (1000 mg, 5.2 mmol) dropwise at -78 °C. After being stirred for 10 minutes, oxalyl chloride (1056 mg, 8.32 mmol) in DCM (1 mL) was added and the reaction was stirred at -78 °C for 0.5 hour. Then triethylamine (2.9 mL, 20.81 mmol) was added and the mixture was stirred at -78 °C for 3 hours. After completion, the reaction was diluted with water (30 mL) and then extracted with DCM (20 mL × 2). The combined organic layer was washed with brine (50 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated in vacuo to give a crude residue, which was purified by column chromatography on silica gel (EA:PE = 1:100 - 1:10) to afford 119b (980 mg, 99.04% yield) as light yellow oil. Step 3: Trans-(3-Benzyloxycyclobutyl)-(2,6-dichloro-4-pyridyl)methan ol (compound 119c) To a solution of 2,6-dichloro-4-iodopyridine (1.55 g, 5.67 mmol) in THF (10 mL) was added isopropylmagnesium chloride lithium chloride complex in THF (4.76 mL, 6.18 mmol) dropwise at -78 °C. After addition, the reaction was warmed up to 25 °C and stirred for 1 hour. Then, compound 119b (980.0 mg, 5.15 mmol) in THF (5 mL) was added at 0 °C. The reaction was stirred at 25 °C for 1 hour. The reaction was quenched with aq. NH4Cl solution (50 mL), extracted with EA (100 mL × 2). The combined organic layer was washed with brine (100 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give the crude product, which was purified by prep-HPLC to afford 119c (760 mg, 43.62% yield) as a yellow oil. MS obsd.: 338.1 (MH + ). Step 4: Trans-(3-Benzyloxycyclobutyl)-(2,6-dichloro-4-pyridyl)methan one (compound 119d) To a solution of compound 119c (660 mg, 1.95 mmol) in DCM (5 mL) was added Dess- Martin periodinane (993 mg, 2.34 mmol) in one portion, and the reaction was stirred at 25 °C for 2 hours. After completion, the reaction was poured into aq. K 2 CO 3 solution (100 mL) and stirred for 30 minutes. The aqueous phase was extracted with EA (50 mL × 3). The combined organic layer was washed with brine (100 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated in vacuo to give 119d (650 mg, 99.08% yield) as colorless oil. MS obsd.: 336.1 (MH + ). Step 5: trans-4-[(3-Benzyloxycyclobutyl)-difluoro-methyl]-2,6-dichlo ro-pyridine (compound 119e) To a solution of 119d (600 mg, 1.78 mmol) in DCM (5 mL) was added DAST (2.88 g, 17.85 mmol) in one portion, and the reaction was stirred at 25 °C for 16 hours. After completion, the reaction mixture was poured into ice water and the pH was adjusted to 7~8 with aq. NaHCO3 solution. The aqueous phase was extracted with EA (30 mL × 3). The combined organic layer was washed with brine (100 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give the crude product, which was purified by flash column on silica gel (EA:PE = 1:100-1:10) to afford 119e (590 mg, 92.29% yield) as a yellow solid. MS obsd.: 358.1 (MH + ). Step 6: Trans-3-[(2,6-dichloro-4-pyridyl)-difluoro-methyl]cyclobutan ol (compound 119f) A solution of compound 119e (550 mg, 1.54 mmol) in DCM (5 mL) was added boron trichloride (4.61 mL, 4.61 mmol) and the reaction was stirred at -78 °C for 1 hour. After completion, the mixture was quenched with water (20 mL) and extracted with EA (30 mL × 2). The combined organic layer was washed with brine (50 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated in vacuo to give the crude product, which was purified by chromatography on silica gel (EA:PE = 1:100-1:10) to afford 119f (340 mg, 82.6% yield) as yellow oil. MS obsd.: 267.9 (MH + ). Step 7: tert-butyl N-[(3R)-1-[4-[4-[6-chloro-4-[trans-difluoro-(3-hydroxycyclob utyl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]-5-oxo-pyrrolidin-3-yl ]carbamate (compound 119g) To a solution of compound 119f (40 mg, 0.150 mmol) in DMSO (1 mL) were added Intermediate D (63.34 mg, 0.150 mmol), N,N-diisopropylethylamine (0.05 mL, 0.300 mmol) in one portion. The reaction was stirred at 100 °C for 16 hours. After completion, the reaction was poured into water (10 mL) and the aqueous phase was extracted with EA (10 mL × 3). The combined organic layer was washed with brine (50 mL), dried over anhydrous Na 2 SO 4, filtered, and concentrated in vacuo to give crude product, which was purified by prep-HPLC to give 119g (60 mg, 61.29% yield) as a white solid. MS obsd.: 656.1 (MH + ). Step 7: (4R)-4-amino-1-[4-[4-[6-chloro-4-[trans-difluoro-(3-hydroxyc yclobutyl)methyl]-2- pyridyl]piperazin-1-yl]sulfonylphenyl]pyrrolidin-2-one To solution of compound 119g (60 mg, 0.090 mmol) in DCM (0.5 mL) was added TFA (0.5 mL, 6.49 mmol) in one portion. The reaction was stirred at 25 °C for 1 hour. After completion, the reaction was concentrated in vacuo to give crude product, which was purified by prep-HPLC to afford Example 119 (32 mg, 57.63% yield) as a white solid. MS obsd.: 556.1 (MH + ). 1 H NMR (400 MHz, DMSO-d6) δ ppm 8.48 - 8.33 (m, 3 H), 7.91 (br d, J = 8.8 Hz, 2 H), 7.79 (br d, J = 8.6 Hz, 2 H), 6.84 - 6.78 (m, 1 H), 6.76 - 6.68 (m, 1 H), 5.25 - 5.15 (m, 1 H), 4.32 - 4.21 (m, 1 H), 4.19 - 4.10 (m, 1 H), 4.09 - 4.00 (m, 1 H), 3.88 (br s, 1 H), 3.65 (br s, 4 H), 3.11 - 3.03 (m, 1 H), 2.96 (br s, 4 H), 2.64 (br s, 1 H), 2.23 (br d, J = 4.9 Hz, 2 H), 2.02 - 1.88 (m, 2 H), 1.32 - 1.14 (m, 1 H). 19 F NMR (376 MHz, DMSO-d6) δ -103.606 (s, 2 F). Example 120 Trans-4-[[2-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-1-yl]phenyl ]sulfonylpiperazin-1-yl]-6- chloro-4-pyridyl]-difluoro-methyl]cyclohexanecarboxamide The title compound was prepared according to the following procedures: Step 1: Trans-methyl 4-[[2-chloro-6-[4-[4-[(4R)-4-(tert-butoxycarbonylamino)-2-ox o-pyrrolidin- 1-yl]phenyl]sulfonylpiperazin-1-yl]-4-pyridyl]-difluoro-meth yl]cyclohexanecarboxylate (compound 120a) To a solution of compound Intermediate D (50 mg, 0.118 mmol) and compound 118d (60 mg, 0.177 mmol) in DMSO (1 mL) was added K 2 CO 3 (48 mg, 0.353 mmol). The mixture was stirred at 90 °C for 12 hours. After completion, the mixture was diluted with EA (50 mL) and water (30 mL). The organic phase was washed with brine (50 mL), dried over anhydrous Na 2 SO 4 , concentrated in vacuo to give the crude compound 120a (60 mg, 70.73% yield) as a yellow solid, which was used directly in the next step. MS obsd. (ESI + ) [(M+H) + ]: 726.4. Step 2: Trans-4-[[2-chloro-6-[4-[4-[(4R)-4-(tert-butoxycarbonylamino )-2-oxo-pyrrolidin-1- yl]phenyl]sulfonylpiperazin-1-yl]-4-pyridyl]-difluoro-methyl ]cyclohexanecarboxylic acid (compound 120b) To a solution of compound 120a (2.4 g, 3.3 mmol) in MeCN (30 mL) and water (10 mL) were added LiBr (2.87 g, 33.06 mmol) and TEA (2.3 mL, 16.52 mmol). The mixture was stirred at 50 °C for 5 hours. After completion, the mixture was diluted with EA (200 ml) and adjusted to pH=5 with 1M HCl solution. The organic layer was washed with brine (200 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated in vacuo to give the crude product, which was purified by flash column on silica gel (EA:PE = 1:5-3:1) to afford compound 120b (640 mg, 27.19% yield) as a yellow solid. MS obsd. (ESI + ) [(M+H) + ]: 712.2. Step 3: Trans-tert-butyl N-[(3R)-1-[4-[4-[4-[(4-carbamoylcyclohexyl)-difluoro-methyl] -6- chloro-2-pyridyl]piperazin-1-yl]sulfonylphenyl]-5-oxo-pyrrol idin-3-yl]carbamate (compound 120c) A mixture of ammonium chloride (20 mg, 0.374 mmol), compound 120b (100 mg, 0.140 mmol), HATU (106.71 mg, 0.281 mmol) and DIPEA (54.38 mg, 73.49 µL, 0.421 mmol) in DMF (5 mL) was stirred at room temperature for 2 hours. After completion, the mixture was diluted with EA, washed with water (100 mL) and brine (100 mL). The combined organic layer was dried over anhydrous Na 2 SO 4 , filtered, concentrated in vacuo to give crude product 120c (50 mg, 50.07%) as a white solid, which was used directly in the next step. MS obsd. (ESI + ) [(M+H) + ]: 710.9. Step 4: Trans-4-[[2-chloro-6-[4-[4-[(4R)-4-amino-2-oxo-pyrrolidin-1- yl]phenyl]sulfonylpiperazin-1-yl]-4-pyridyl]-difluoro-methyl ]cyclohexanecarboxamide Compound 120c (90 mg, 0.127 mmol) and TFA (1 mL) were dissolved in the DCM (5 mL). The reaction was stirred at room temperature for 2 hours. After completion, the reaction was concentrated in vacuo to give crude product, which was purified by prep-HPLC to give Example 120 (50.8 mg, 62.41% yield) as a white solid. MS obsd.: 611.1 (MH + ). 1 H NMR (400 MHz, METHANOL-d 6 ) δ ppm 7.88 - 7.98 (m, 2 H), 7.70 - 7.79 (m, 2 H), 6.75 - 6.80 (m, 1 H), 6.69 - 6.72 (m, 1 H), 3.90 - 4.07 (m, 1 H), 3.59 - 3.78 (m, 5 H), 3.46 - 3.59 (m, 1 H), 2.88 - 3.03 (m, 4 H), 2.74 - 2.89 (m, 1 H), 2.26 - 2.40 (m, 1 H), 2.10 - 2.25 (m, 1 H), 1.92 - 2.07 (m, 2 H), 1.70 - 1.81 (m, 2 H), 1.58 - 1.70 (m, 2 H), 1.18 - 1.37 (m, 4 H), 0.96 - 1.18 (m, 3 H). 19 F NMR (376 MHz, METHANOL-d 4 ) δ ppm -107.25 (br d, J = 14.99 Hz, 2 F). BIOLOGICAL EXAMPLES Example 121: 50% Inhibitory Concentration (IC50) to UDP-2,3-diacylglucosamine hydrolase (LpxH) The inhibitory potency of molecules to UDP-2,3-diacylglucosamine hydrolase (LpxH) was determined using the UMP/CMP-Glo™ Glycosyltransferase Assay kit. LpxH hydrolyzes the pyrophosphate bond of UDP-2,3-diacylglucosamine (UDP-DAG) to yield 2,3-diacylglucosamine 1-phosphate (lipid X) and UMP. The assay kit could quantify the UMP production by convert the UMP to ATP and generate light in a luciferase reaction. The compound’s effect on the LpxH activity is detected by measuring the light using a luminometer (Envision). Table 1: Enzymatic IC50 values of the compounds of this invention against Escherichia coli LpxH Example 122: Minimal Inhibitory Concentration Protocol (MIC) Assay: The in vitro potency of compounds to inhibit E. coli (ATCC 25922) and K. pneumonia (ATCC 43816) growth was assessed by MIC (Minimal Inhibitory Concentration) assay. Samples were prepared from 10 mM DMSO stock solutions. After serial 2-fold dilution in DMSO in a master plate (Greiner, Cat No: 651201), 180 µL sterile distilled water was added to 20 µL each aliquot of the samples. Then 10µL diluted compounds were transferred into a new assay plate (Costar, 3599). The growth medium Cation-Adjusted Mueller Hinton Broth (CAMHB) was prepared by adding the 20 mg per liter CaCl 2 and 20 mg per liter MgCl 2 into the MHB medium (Jianglai Company, sterilized). Vials of each of the test microorganisms were maintained frozen in the vapor phase of a liquid nitrogen freezer. The bacteria E.coli ATCC 25922 (KWIKSTIK, 0335K) and K. pneumonia ATCC 43816 were taken out from liquid nitrogen freezer, then thawed in room temperature, and diluted in the CAMHB medium to achieve a final inoculum of 5 × 105 CFU/ mL.90 µL CAMHB with bacteria was dispensed to the assay plate and pipetted 5 times. Then the assay plates were incubated for 20 hours at 35°C in ambient air. Following incubation, the MIC (µg/mL), the lowest concentration of drug that inhibits visible growth of the microorganism was read and recorded throw the microscope. Table 2: MIC values of the compounds of this invention against E. coli and K. pneumonia