Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
BICYCLIC TETRAHYDROTHIAZEPINE DERIVATIVES
Document Type and Number:
WIPO Patent Application WO/2024/033388
Kind Code:
A1
Abstract:
The present invention provides new bicyclic tetrahydrothiazepine derivatives having the general formula (I) wherein Y, R1, R2 and R4 are as defined herein, compositions including the compounds, processes of manufacturing the componds and methods of using the compounds.

Inventors:
BRANDSTAETTER MARCO (CH)
DI FRANCESCO MARIA EMILIA (CH)
HUTTER ROMAN (CH)
KUEHNE HOLGER (CH)
LUEBBERS THOMAS (CH)
MARTIN LAETITIA JANINE (CH)
MUELLER BARBARA JOHANNA (CH)
Application Number:
PCT/EP2023/071988
Publication Date:
February 15, 2024
Filing Date:
August 09, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
HOFFMANN LA ROCHE (US)
HOFFMANN LA ROCHE (US)
International Classes:
C07D417/04; A61K31/554; A61P35/00; C07D417/14
Domestic Patent References:
WO2016139181A12016-09-09
WO2014023708A12014-02-13
WO2022171745A12022-08-18
WO1996040210A11996-12-19
WO1998050433A21998-11-12
WO1998014451A11998-04-09
WO1998050038A11998-11-12
WO1999009016A11999-02-25
WO1999024037A11999-05-20
WO1999009016A11999-02-25
WO1998043960A11998-10-08
WO1997038983A11997-10-23
WO1999006378A11999-02-11
WO1999006396A11999-02-11
WO1996030347A11996-10-03
WO1996033978A11996-10-31
WO1996003397A11996-02-08
WO1996033980A11996-10-31
WO2012145493A12012-10-26
WO2010077634A12010-07-08
WO2007005874A22007-01-11
WO2013079174A12013-06-06
WO2010019570A22010-02-18
WO2014008218A12014-01-09
WO2008132601A12008-11-06
WO2009044273A22009-04-09
WO2012032433A12012-03-15
WO2006105021A22006-10-05
WO2009009116A22009-01-15
WO2011028683A12011-03-10
WO2006122150A12006-11-16
WO2007075598A22007-07-05
WO2008036653A22008-03-27
WO2008036642A22008-03-27
WO2009073620A22009-06-11
WO2009115652A22009-09-24
WO2011056652A12011-05-12
WO2012142237A12012-10-18
WO2006029879A22006-03-23
WO2011109400A22011-09-09
WO2016139181A12016-09-09
Foreign References:
US4943533A1990-07-24
US5212290A1993-05-18
US5891996A1999-04-06
US6235883B12001-05-22
EP0659439A21995-06-28
US5616582A1997-04-01
US5457105A1995-10-10
US5475001A1995-12-12
US5654307A1997-08-05
US5679683A1997-10-21
US6084095A2000-07-04
US6265410B12001-07-24
US6455534B22002-09-24
US6521620B12003-02-18
US6596726B12003-07-22
US6713484B22004-03-30
US5770599A1998-06-23
US6140332A2000-10-31
US5866572A1999-02-02
US6399602B12002-06-04
US6344459B12002-02-05
US6602863B12003-08-05
US6391874B12002-05-21
US6344455B12002-02-05
US5760041A1998-06-02
US6002008A1999-12-14
US5747498A1998-05-05
US5804396A1998-09-08
Other References:
MELLMAN, I. ET AL., CANCER IMMUNOTHERAPY COMES OF AGE, NATURE, vol. 480, no. 7378, 2011, pages 480 - 489
MERIDA, I.AVILA-FLORES, A.MERINO, E.: "Diacylglycerol kinases: at the hub of cell signalling", BIOCHEM. J., vol. 409, no. 1, 2008, pages 1 - 18
ARRANZ-NICOLAS, J.MERIDA, I.: "Biological regulation of diacylglycerol kinases in normal and neoplastic tissues: New opportunities for cancer immunotherapy", ADVANCES IN BIOLOGICAL REGULATION, vol. 75, 2020
MA, Q.GABELLI, S.B.RABEN, D.M.: "Diacylglycerol kinases: relationship to other lipid kinases", ADV BIOL REGUL, vol. 71, 2019, pages 104 - 110, XP085582566, DOI: 10.1016/j.jbior.2018.09.014
RIESE, M.J.MOON, E.K.JOHNSON, B.D.ALBELDA, S.M.: "Diacylglycerol kinases (DGKs): novel targets for improving T cell activity in cancer", FRONT CELL DEV BIOL, vol. 4, 2016, pages 108
NOESSNER, E.: "DGK-alpha: a checkpoint in cancer-mediated immuno-inhibition and target for immunotherapy", FRONT CELL DEV BIOL, vol. 5, 2017, pages 16
SAKANE, F.MIZUNO, S.KOMENOI, S.: "Diacylglycerol kinases as emerging potential drug targets for a variety of diseases: an update", FRONT CELL DEV BIOL, vol. 4, 2016, pages 82
MERIDA, I.ANDRADA, EGHARBI, S.I.AVILA-FLORES, A: "Redundant and specialized roles for diacylglycerol kinases alpha and zeta in the control of T cell functions", SCI. SIGNAL., vol. 8, no. 374, 2015
SHULGA, Y.V.TOPHAM, M.K.EPAND, R.M.: "Regulation and functions of diacylglycerol kinases", CHEM. REV., vol. 111, no. 10, 2011, pages 6186 - 6208
SIM, J.A.KIM, J.YANG, D: "Beyond Lipid Signaling: Pleiotropic Effects of Diacylglycerol Kinases in Cellular Signaling", INT. J. MOL. SCI., vol. 21, 2020, pages 6861
RIESE, M.J.GREWAL, J.DAS, J.ZOU, T.PATIL, V.CHAKRABORTY, A.K.KORETZKY, G.A.: "Decreased diacylglycerol metabolism enhances ERK activation and augments CD8+ T cell functional responses", J. BIOL. CHEM., vol. 286, no. 7, 2011, pages 5254 - 5265
ZHA, Y.MARKS, R.HO, A.W.PETERSON, A.C.JANARDHAN, S.BROWN, I.PRAVEEN, K.STANG, S.STONE, J.C.GAJEWSKI, T.F.: "T cell anergy is reversed by active Ras and is regulated by diacylglycerol kinase-alpha", NAT. IMMUNOL., vol. 7, no. 11, 2006, pages 1166 - 1173, XP055916654, DOI: 10.1038/ni1394
OLENCHOCK, B.A., GUO, R., CARPENTER, J.H., JORDAN, M., TOPHAM, M.K., KORETZKY, G.A., ZHONG, X.P.: " Disruption of diacylglycerol metabolism impairs the induction of T cell anergy.", NAT. IMMUNOL., vol. 7, no. 11, 2006, pages 1174 - 1181
"IUPAC - Compendium of Chemical Terminology", 1997, BLACKWELL SCIENTIFIC PUBLICATIONS
BARANYR. B. MERRIFIELD, J. AM. CHEM. SOC., vol. 99, 1977, pages 7363
H. WALDMANN ET AL., ANGEW. CHEM. INT. ED. ENGL., vol. 35, 1996, pages 2056
RICHARD C. LAROCK.: "Transformations: A Guide to Functional Group Preparations", 1999, JOHN WILEY & SONS
PETER G. M. WUTS: "Greene's protective groups in organic synthesis", 15 February 2001, LIPPINCOTT WILLIAMS & WILKINS PUBLISHERS
ANGEW CHEM. INTI. ED. ENGL., vol. 33, 1994, pages 183 - 186
STRAGLIOTTO ET AL., EUR. J. CANCER, vol. 32A, 1996, pages 636 - 640
JOHNS ET AL., J. BIOL. CHEM., vol. 279, no. 29, 2004, pages 30375 - 30384
CAS , no. 1250528-75-6
CAS, no. 2089150-62-7
TAKAHASHI, D.; SAKANE, F.: "Expression and purification of human diacylglycerol kinase alpha from baculovirus-infected insect cells for structural studies. ", PEERJ, vol. 6, no. e5449, 2018
Attorney, Agent or Firm:
BELKACEMI, Doria (CH)
Download PDF:
Claims:
Claims

1. A compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein:

Y is S(O), S(O)2 and S(O)N(Ry);

R1 is 5-membered heteroaryl, wherein R1 is optionally substituted with one or more R10 which can be the same or different;

R2 is selected from hydrogen and halogen;

R4 is selected from Cs-w-aryl and 5-14 membered heteroaryl, wherein R4 is optionally substituted with one or more R11 which can be the same or different;

R10 is -C(R10aR10b)2-S(O)2(R10c);

R10a and R10b are each independently selected from hydrogen and Ci-6-alkyl, or R10a and R10b, taken together with the carbon atom to which they are attached, form a Cs-e-cycloalkyl;

R10c is selected from hydrogen, Ci-6-alkyl, Cs-e-cycloalkyl, -N(Ci-6-alkyl), halo-Ci-6-alkyl, and phenyl, wherein phenyl is optionally substituted with one or more halogen, or Ci-6-alkyl; or R10a and R10c, taken together with the carbon atom and the sulphur atom to which they are attached, form a 3-10 membered heterocyclyl;

R11 is selected from: i) halogen; ii) 3-10 membered heterocyclyl, optionally substituted with one or more halo-Ci-6-alkyl, Cs-io-cycloalkyl; iii) 5-6 membered heteroaryl, optionally substituted with one or more halogen, Ci-6-alkyl, halo-Ci-6-alkyl, Cs-io-cycloalkyl, Ci-6-haloalkoxy; iv) phenyl, optionally substituted with one or more halogen, cyano, Ci-6-alkoxy, Ci-6- haloalkyl, Ci-6-alkyl; v) -O(Rl la); vi) -N(Rl lgRl lh); and vii) -S(Rl lk)

Rl la is selected from Ci-12-alkyl, halo-Ci-6-alkyl, C3-7-cycloalkyl, 3-10 membered -(C1-6- alkyl)heterocyclyl, phenyl and -Ci-6-alkyl-phenyl, wherein said Ci-12-alkyl, C3-7-cycloalkyl, phenyl and -Ci-6-alkyl-phenyl are optionally substituted with one or more halogen, Ci-6-alkyl, halo-Ci-6-alkyl, Ci-6-alkoxy, Ci-6-haloalkoxy;

Rl lg and Rllh are each independently selected from hydrogen, Ci-6-alkyl, -(Ci-6-alkyl)phenyl;

Rl lk is selected from hydrogen and halo-Ci-6-alkyl;

Ry is selected from hydrogen and Ci-6-alkyl.

2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R1 is oxadiazole or tetrazole, wherein R1 is optionally substituted with one or more R10 which can be the same or different.

3. The compound of claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein R1 is oxadiazole, wherein R1 is optionally substituted with one or more R10 which can be the same or different.

4. The compound of any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, wherein R2 is selected from hydrogen and fluorine.

5. The compound of any one of claims 1 to 4, or a pharmaceutically acceptable salt thereof, wherein R4 is phenyl.

6. The compound of any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, wherein R10a and R10b are each independently selected from hydrogen and Ci-6-alkyl, or R10a and R10b, taken together with the carbon atom to which they are attached, form a C3-6- cycloalkyl.

7. The compound of any one of claims 1 to 6, or a pharmaceutically acceptable salt thereof, wherein R10 is C3-6-cycloalkyl substituted with one -S(O)2(Ci-6-alkyl).

8. The compound of any one of claims 1 to 7, or a pharmaceutically acceptable salt thereof, wherein R10 is 3-6 membered heterocyclyl, wherein the heteroatom is sulphur, substituted with one or more Ci-10-alkyl, oxo, wherein the oxo substitution is on the sulphur atom.

9. The compound of any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, wherein R10 is selected from 1-methyl-methylsulfonyl-ethyl.

10. The compound of any one of claims 1 to 9, or a pharmaceutically acceptable salt thereof, wherein R11 is i) halogen; ii) 3-10 membered heterocyclyl; iii) 5-6 membered heteroaryl, optionally substituted with one or more halogen, halo-Ci-6- alkyl, Cs-io-cycloalkyl, Ci-6-haloalkoxy; iv) phenyl, optionally substituted with one or more cyano, Ci-6-alkoxy, Ci-6-haloalkyl, C1-6- alkyl; v) -O(Rl la); Rlla is selected from Ci-12-alkyl, halo-Ci-6-alkyl, C3-7-cycloalkyl, 5-6 membered -(Ci-6-alkyl)heteroaryl, phenyl and -Ci-6-alkyl-phenyl, wherein said Ci-12-alkyl, phenyl, 5-6 membered -(Ci-6-alkyl)heteroaryl and -Ci-6-alkyl-phenyl are optionally substituted with one or more halogen, Ci-6-alkyl, halo-Ci-6-alkyl, Ci-6-alkoxy, C1-6- haloalkoxy; vi) -N(Rl lgRl lh); Rllg and Rl lh are each independently selected from Ci-6-alkyl, -(C1-6- alkyl)phenyl; vii) -S(halo-Ci-6-alkyl); viii) CH2-O-phenyl ;

11. The compound of any one of claims 1 to 10, or a pharmaceutically acceptable salt thereof, wherein R11 is selected from: i) -O(Rl la), wherein Rl lais selected from halo-Ci-6-alkyl and phenyl, wherein said phenyl is optionally substituted with one or more halogen; or ii) phenyl, optionally substituted with one or more Ci-6-alkoxy, Ci-6-haloalkyl; iii) halogen iv) 5-6 membered heteroaryl, optionally substituted with one or more halo-Ci-6-alkyl, C3-10- cycloalkyl, Ci-6-haloalkoxy.

12. The compound of any one of claims 1 to 11, or a pharmaceutically acceptable salt thereof, wherein R11 is selected from (trifluoromethyl)phenyl, (trifluoromethyl)-oxadiazolyl, cyclopropyl-oxadiazolyl, phenoxyl, methoxyphenyl, tetrafluoroethoxyl, dichlorophenoxyl, chloro, trifluoromethoxy-2-pyridyl.

13. The compound of any one of claims 1 to 12, or a pharmaceutically acceptable salt thereof, wherein Y is S(O)2.

14. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein:

Y is S(O), S(O)2 and S(O)N(Ry);

R1 is 5-membered heteroaryl, wherein R1 is optionally substituted with one or more R10 which can be the same or different;

R2 is selected from hydrogen and fluorine;

R4 is selected from Ce-aryl and 6-membered heteroaryl, wherein R4 is optionally substituted with one or more R11 which can be the same or different;

R10 is C3-6-cycloalkyl substituted with one -S(O)2(Ci-6-alkyl); R11 is i) halogen; ii) 3-10 membered heterocyclyl; iii) 5-6 membered heteroaryl, optionally substituted with one or more halogen, halo-Ci-6- alkyl, Cs-io-cycloalkyl, Ci-6-haloalkoxy; iv) phenyl, optionally substituted with one or more cyano, Ci-6-alkoxy, Ci-6-haloalkyl, Ci-6- alkyl; v) -O(Rl la); Rlla is selected from Ci-12-alkyl, halo-Ci-6-alkyl, C3-7-cycloalkyl, 5-6 membered -(Ci-6-alkyl)heteroaryl, phenyl and -Ci-6-alkyl-phenyl, wherein said Ci-12-alkyl, phenyl, 5-6 membered -(Ci-6-alkyl)heteroaryl and -Ci-6-alkyl-phenyl are optionally substituted with one or more halogen, Ci-6-alkyl, halo-Ci-6-alkyl, Ci-6-alkoxy, C1-6- haloalkoxy; vi) -N(Rl lgRl lh); Rllg and Rl lh are each independently selected from Ci-6-alkyl, -(C1-6- alkyl)phenyl; vii) -S(halo-Ci-6-alkyl); viii) CH2-O-phenyl ;

15. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein:

Y is S(O), S(O)2 and S(O)N(Ry);

R1 is oxadiazole, wherein R1 is optionally substituted with one or more R10 which can be the same or different;

R2 is selected from hydrogen and fluorine;

R4 is phenyl;

R10 is selected from methyl-methylsulfonyl-ethyl, cyclopropylsulfonyl-methyl-ethyl, methyldi oxo-thiolanyl, methylsulfonylmethyl, methylsulfonyl cyclobutyl;

R11 is selected from: i) -O(Rl la), wherein Rl lais selected from halo-Ci-6-alkyl, phenyl, wherein said phenyl is optionally substituted with one or more halogen; ii) phenyl, optionally substituted with one or more Ci-6-alkoxy, Ci-6-haloalkyl; iii) halogen; and iv) 5-6 membered heteroaryl, optionally substituted with one or more halo-Ci-6-alkyl, C3-10- cycloalkyl, Ci-6-haloalkoxy.

16. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein:

Y is S(O), S(O)2 and S(O)N(Ry);

R1 is oxadiazole, wherein R1 is optionally substituted with one or more R10 which can be the same or different;

R2 is selected from hydrogen and fluorine;

R4 is phenyl;

R10 is methyl-methylsulfonyl-ethyl;

R11 is selected from (trifluoromethyl )phenyl, (trifluoromethyl)-oxadiazolyl, cyclopropyl - oxadiazolyl, phenoxyl, methoxyphenyl, tetrafluoroethoxyl, dichlorophenoxyl, chloro and trifluoromethoxy)-2-pyridyl.

17. The compound of any one of claims 1 to 16, or a pharmaceutically acceptable salt thereof, selected from:

(3R)-3-amino-5-[[4-(l-ethylpropoxy)phenyl]methyl]-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4- oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[[4-(2,2,2- tri fluoroethoxy )phenyl]methyl]-2,3-dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-(cyclopentoxy)phenyl]methyl]-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4- oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-5-[(4-benzyloxyphenyl)methyl]-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4- oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-[(4-fluorophenyl)methoxy]phenyl]methyl]-7-[5-(l -methyl- 1 -methylsulfonyl- ethyl)-l, 3, 4-oxadiazol-2-yl]- 1,1 -di oxo-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one; (3 R)-3-amino-5-[[4-[(2-fluorophenyl)methoxy]phenyl]methyl]-7-[5-(l -methyl- 1-methylsulfonyl- ethyl)-!, 3, 4-oxadiazol-2-yl]- 1,1 -di oxo-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-[(3-fluorophenyl)methoxy]phenyl]methyl]-7-[5-(l -methyl- 1 -methylsulfonyl- ethyl)-l, 3, 4-oxadiazol-2-yl]- 1,1 -di oxo-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[[4-(3,3,3- trifluoropropoxy)phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[[4- (trifluoromethylsulfanyl)phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[[4-

(phenoxymethyl)phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-[3-(difluoromethoxy)phenoxy]phenyl]methyl]-7-[5-(l-methyl-l- methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-5-[[4-(3- methylphenoxy)phenyl]methyl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3 R)-3-amino-5-[[4-(2,3-dichlorophenoxy)phenyl]methyl]-7-[5-(l -methyl- 1-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-(3-fluorophenoxy)phenyl]methyl]-7-[5-(l -methyl- 1-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-(3-chlorophenoxy)phenyl]methyl]-7-[5-(l -methyl- 1-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3 R)-3-amino-5-[[4-(3-chloro-4-fluoro-phenoxy)phenyl]methyl]-7-[5-(l -methyl- 1-methylsulfonyl- ethyl)-!, 3, 4-oxadiazol-2-yl]- 1,1 -di oxo-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[[4-[3-

(trifluoromethyl)phenoxy]phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-[3-fhroro-5-(trifluoromethyl)phenoxy]phenyl]methyl]-7-[5-(l-methyl-l- methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[[4-[3-

(trifluoromethoxy)phenoxy]phenyl]methyl]-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-(3-chloro-5-methoxy-phenoxy)phenyl]methyl]-7-[5-(l-methyl-l- methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one; (3R)-3-amino-5-[[4-(3-chloro-5-methoxy-phenoxy)phenyl]methyl]-7-[5-(l-methyl-l- methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

4-[4-[[(3R)-3-amino-7-[5-(l -methyl- 1 -methyl sulfonyl-ethyl)-l, 3, 4-oxadiazol-2-yl]-l,l,4-tri oxo-2, 3- dihydro-lX6,5-benzothiazepin-5-yl]methyl]phenyl]benzonitrile;

(3R)-3-amino-5-[[4-(4-methoxyphenyl)phenyl]methyl]-7-[5-(l-methyl-l-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-(3-methoxyphenyl)phenyl]methyl]-7-[5-(l-methyl-l-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-(4-methoxy-2-methyl-phenyl)phenyl]methyl]-7-[5-(l -methyl- 1 -methylsulfonyl- ethyl)-l, 3, 4-oxadiazol-2-yl]- 1,1 -di oxo-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[[4-[4-

(trifluoromethyl)phenyl]phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[[4-

(l,l,2,2-tetrafluoroethoxy)phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-(3-bromophenoxy)phenyl]methyl]-7-[5-(l-methyl-l-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[[4-

(trifluoromethoxy)phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-l,l-diketo-7-[5-(l-mesyl-l-methyl-ethyl)-l,3,4-oxadiazol-2-yl]-5-[4-[5-

(trifluoromethoxy)-2-pyridyl]benzyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3 R)-3-amino-5-[[4-[benzyl(methyl)amino]phenyl]methyl]-7-[5-(l -methyl- 1-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-[isopropyl(methyl)amino]phenyl]methyl]-7-[5-(l -methyl- 1 -methylsulfonyl- ethyl)-l, 3, 4-oxadiazol-2-yl]- 1,1 -di oxo-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[[4-[5-

(trifluoromethyl)-l,2,4-oxadiazol-3-yl]phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[[4-[5-

(trifluoromethyl)-2-pyridyl]phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-(5-fluoro-2-pyridyl)phenyl]methyl]-7-[5-(l -methyl- 1-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[[6-[4-

(trifluoromethyl)phenyl]-3-pyridyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one; (3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-5- [(6-phenoxy-3-pyridyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fluoro-5-[[4-(4-methoxyphenyl)phenyl]methyl]-7-[5-(l -methyl- 1 -methylsulfonyl- ethyl)-l, 2, 4-oxadiazol-3-yl]- 1,1 -di oxo-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-5-[(4-chlorophenyl)methyl]-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2,4- oxadiazol-3-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-5-

[[4-[5-(trifluoromethyl)-l,2,4-oxadiazol-3-yl]phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4- one;

(3R)-3-amino-5-[[4-(5-cyclopropyl-l,2,4-oxadiazol-3-yl)phenyl]methyl]-8-fluoro-7-[5-(l-methyl-l- methylsulfonyl-ethyl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-5- [(4-phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-5-

[[4-(trifluoromethoxy)phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-5-[(4-chlorophenyl)methyl]-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4- oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3 R)-3-amino-5-[(4-chlorophenyl)methyl]-8-fluoro-7-[5-(l -methylsulfonyl cyclopropyl)-l, 3, 4- oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fhioro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-5-

[(4-tetrahydropyran-4-yl phenyl )methyl]-2,3-dihydro- IX6,5-benzothiazepin-4-one;

(3 R)-3-amino-8-fhroro-5-[[4-(2-methoxy-l,l -dimethyl-ethoxy )phenyl]methyl]-7-[5-(l -methyl- 1- methylsulfonyl-ethyl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fhioro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-

[[4-[4-(tri fluoromethyl)phenyl]phenyl]methyl]-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-8-fhroro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-

[[4-(2,2,2-trifluoroethoxy)phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fhroro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-

[[4-[5-(trifluoromethyl)-2-pyridyl]phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fhroro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-

[[4-(l,l,2,2-tetrafluoroethoxy)phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one; (3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5- [(4-phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5- [[4-[5-(trifluoromethyl)-l,2,4-oxadiazol-3-yl]phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4- one;

(3R)-3-amino-8-fluoro-5-[[4-(4-methoxyphenyl)phenyl]methyl]-7-[5-(l -methyl- 1 -methylsulfonyl- ethyl)-l, 3, 4-oxadiazol-2-yl]- 1,1 -di oxo-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5- [[4-[6-(tri fl uorom ethyl )-3-pyridyl]phenyl]methyl]-2,3-dihydro-IX6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-5-

[[4-[[3-(tri fluoromethyl)- 1,2, 4-oxadiazol-5-yl]methoxy]phenyl]methyl]-2, 3 -dihydro- 1X6, 5- benzothiazepin-4-one;

(3R)-3-amino-8-fhroro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2,4-oxadiazol-3-yl]-l-oxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-lX4,5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-(4-methoxyphenyl)phenyl]methyl]-7-[5-(l-methyl-l-methylsulfonyl-ethyl)- l,2,4-oxadiazol-3-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methylsulfonylcyclobutyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methylsulfonylcyclopropyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-cyclopropylsulfonyl-l-methyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(2-methyl-l,l-dioxo-thiolan-2-yl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

2-[5-[(3R)-3-amino-l,l,4-trioxo-5-[[4-[4-(trifluoromethyl)phenyl]phenyl]methyl]-2, 3 -dihydro- lX6,5-benzothiazepin-7-yl]-l,3,4-oxadiazol-2-yl]-N,N-dimethyl-propane-2-sulfonamide;

(3R)-3-amino-7-[5-(l-methyl-l-pyrrolidin-l-ylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5- [[4-[4-(tri fluoromethyl)phenyl]phenyl]methyl]-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-8-fhroro-7-[2-(methylsulfonylmethyl)tetrazol-5-yl]-l,l-dioxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one; and (3R)-3-amino-8-fluoro-7-[l-(methylsulfonylmethyl)tetrazol-5-yl]-l,l-dioxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one. 18. The compound of any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, selected from (3R)-3-amino-8-fhroro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5- [[4-(l,l,2,2-tetrafluoroethoxy)phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fhroro-5-[[4-(4-methoxyphenyl)phenyl]methyl]-7-[5-(l -methyl- 1 -methylsulfonyl- ethyl)-l, 3, 4-oxadiazol-2-yl]- 1,1 -di oxo-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-8-fhroro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-5- [(4-phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3 R)-3-amino-5-[[4-(2,3-dichlorophenoxy)phenyl]methyl]-7-[5-(l -methyl- 1-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-(3-chlorophenoxy)phenyl]methyl]-7-[5-(l -methyl- 1-methylsulfonyl-ethyl)-

1.3.4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5- [[4-[4-(tri fluoromethyl)phenyl]phenyl]methyl]-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-5-[(4-chlorophenyl)methyl]-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2,4- oxadiazol-3-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-5- [[4-[5-(trifluoromethyl)-l,2,4-oxadiazol-3-yl]phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4- one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[[4-[4- (trifluoromethyl)phenyl]phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-(4-methoxyphenyl)phenyl]methyl]-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-

1.2.4-oxadiazol-3-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4-one; and (3R)-3-amino-l,l-diketo-7-[5-(l-mesyl-l-methyl-ethyl)-l,3,4-oxadiazol-2-yl]-5-[4-[5- (trifluoromethoxy)-2-pyridyl]benzyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one.

19. A process for the preparation of a compound according to any one of claims 1 to 18, or a pharmaceutically acceptable salt thereof, comprising reacting of compound of formula (IX) wherein Y, R1, R2 and R4 are as defined in any one of claims 1 to 18 and PG is an amino protecting group, with a suitable deprotection agent to form said compound of formula (I).

20. The compound of any one of claims 1 to 18, or a pharmaceutically acceptable salt thereof, when manufactured according to the process of claim 19.

21. The compound of any one of claims 1 to 18, or a pharmaceutically acceptable salt thereof, for use as therapeutically active substance.

22. A pharmaceutical composition comprising a compound according to any one of claims 1 to 18, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

23. The pharmaceutical composition according to claim 22, further comprising an additional therapeutic agent.

24. The compound according to any one of claims 1 to 18, or a pharmaceutically acceptable salt thereof, for use in the treatment, prevention and/or delay of progression of cancer.

25. The compound for use according to claim 24, wherein the cancer is associated with aberrant di acyl glycerol kinase signaling, wherein the diacylglycerol kinase is selected from DGKa and/or DGK^.

26. The compound for use according to claim 24 or 25, wherein the cancer is selected from the group consisting of B-cell acute lymphoid leukemia, T-cell acute lymphoid leukemia, acute lymphoid leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia B-cell prolymphocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitt's lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma, Marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin's lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, Waldenstrom’s macroglobulinemia, preleukemia, sarcoma, carcinoma, melanoma, neuroblastoma, renal cell carcinoma, colon cancer, colorectal cancer, breast cancer, epithelial squamous cell cancer, melanoma, stomach cancer, brain cancer, lung cancer (e.g., NSCLC), pancreatic cancer, cervical cancer, ovarian cancer, liver cancer, bladder cancer, prostate cancer, testicular cancer, thyroid cancer, uterine cancer, adrenal cancer and head and neck cancer.

27. The use of a compound according to any one of claims 1 to 18, or a pharmaceutically acceptable salt thereof, for the treatment, prevention and/or delay of progression of cancer.

28. The use of a compound according to any one of claims 1 to 18, or a pharmaceutically acceptable salt thereof, for the preparation of a medicament for the treatment, prevention and/or delay of progression of cancer.

29. A method for the treatment, prevention and/or delay of progression of cancer, which method comprises administering a therapeutically effective amount of a compound according to any one of claims 1 to 18, or a pharmaceutically acceptable salt thereof.

30. Use of a compound according to any one of claims 1 to 18, or a pharmaceutically acceptable salt thereof, for inhibiting activity of at least one of diacylglycerol kinases selected from DGKa and DGK(^.

31. A method of inhibiting activity of at least one of di acylglycerol kinases selected from DGKa and DGK^ comprising administering to a subject in need thereof a therapeutically effective amount of at least one compound according to any one of claims 1 to 18, or a pharmaceutically acceptable salt thereof.

32. The invention as hereinbefore described.

Description:
Case P37530

BICYCLIC TETRAHYDROTHIAZEPINE DERIVATIVES

Field of the invention

The present invention relates to bicyclic tetrahydrothiazepine compounds which inhibit Diacylglycerol kinases (DGK) a and C, and are useful as T-Cell activators, their manufacture and pharmaceutical compositions comprising said compounds.

The present compounds may be useful as immunotherapeutic agents for the treatment of human diseases. More specifically, the present compounds can be used alone or in combination with other immunotherapeutic agents in order to boost anti-cancer immunity.

Background of the invention

Cancer immunity is a multistep process that is regulated by a series of negative immune checkpoint and positive co-stimulatory receptors and related intracellular signaling cascades that when effectively triggered can achieve antitumor response (Mellman, I., et al. (2011) Cancer Immunotherapy Comes of Age, Nature 480(7378), 480-489). Indeed, PD1/PDL1 targeting and other immune-checkpoint inhibitors have revolutionized cancer immunotherapy, but still more than 70% of patients do not benefit from immune-checkpoint inhibition. Similarly, for T-cell bispecific antibodies, even in the most promising indication (Non-Hodgkin lymphoma), these T-cell binders (TCBs) achieve complete remissions in less than 50% of patients. T-cell exhaustion seems to play an important role in many of these examples of primary or secondary resistance to cancer immunotherapy. A possible reason for this lack of efficacy is that T-cell activation occurs via targeting and crosslinking of CD3 (signal 1), but costimulation e.g. via CD28 or 4-1BB (signal 2) is missing. This hypothesis was verified clinically for CAR T-cell therapy where it was shown that only after the incorporation of costimulatory domains, clinically relevant efficacy was observed.

Diacylglycerol kinases (DGKs) are lipid kinases that catalyze the conversion of Diacylglycerol (DAG) to phosphatidic acid (PA), thus limiting DAG-regulated and promoting PA-dependent functions (Merida, I., Avila-Flores, A., and Merino, E. 2008: Diacylglycerol kinases: at the hub of cell signalling. Biochem. J. 409 (1), 1-18). The DGK family consist of ten isoforms that can be grouped into five subtypes based on the presence of different regulatory domains within their structure. Beyond that, the lack of structural data as of now still hinders a more thorough understanding of the DGKs mode of action. Also information on certain prokaryotic DGK and other lipid kinases like sphingosine kinase and phosphatidylinositol-3-kinase (PI3K) has provided only limited insight into the DGK catalytic mechanisms which seems to be distinct from classical kinases (Arranz-Nicolas, J. and Merida,

1., 2020. Biological regulation of diacylglycerol kinases in normal and neoplastic tissues: New opportunities for cancer immunotherapy, Advances in Biological Regulation, Volume 75; Ma, Q., Gabelli, S.B., Raben, D.M., 2019: Diacylglycerol kinases: relationship to other lipid kinases. Adv Biol Regul 71, 104-110).

Although several isoforms within the DGK family have been described to play a role in cancer, the a and C, isoforms are the ones that have been most deeply studied in this regard. As PA producers, both enzymes have been implicated in various processes promoting tumor growth and metastasis. On the other hand, as DAG consumers, DGKa and C, have been extensively characterized as negative regulators of T cell responses (Riese, M.J., Moon, E.K., Johnson, B.D., Albelda, S.M., 2016. Diacylglycerol kinases (DGKs): novel targets for improving T cell activity in cancer. Front Cell Dev Biol 4,108; Noessner, E., 2017. DGK- alpha: a checkpoint in cancer -mediated immuno-inhibition and target for immunotherapy. Front Cell Dev Biol 5, 16; Sakane, F., Mizuno, S., Komenoi, S., 2016. Diacylglycerol kinases as emerging potential drug targets for a variety of diseases: an update. Front Cell Dev Biol 4, 82; Arranz-Nicolas, J. and Merida, I., 2020. Biological regulation of diacylglycerol kinases in normal and neoplastic tissues: New opportunities for cancer immunotherapy, Advances in Biological Regulation, Volume 75)

These two isozymes DGKa and DGK^ are active downstream of CD28 and other costimulatory receptors as well as the T cell receptor (TCR), and their function is to limit the amount of DAG generated - and ultimately T-cell activation (Merida, I., Andrada, E., Gharbi,

5.1., Avila-Flores, A., 2015. Redundant and specialized roles for diacylglycerol kinases alpha and zeta in the control ofT cell functions. Sci. Signal. 8 (374); Shulga, Y.V., Topham, M.K., Epand, R.M., 2011. Regulation and functions of diacylglycerol kinases. Chem. Rev. I l l (10), 6186-6208.) A summary of representative DGK-regulated signaling pathways is shown in Figure 1 (Sim, J. A.; Kim, J.; Yang, D. Beyond Lipid Signaling: Pleiotropic Effects of Diacylglycerol Kinases in Cellular Signaling. Int. J. Mol. Sci. 2020, 21, 6861): Activated PLCI cleaves PIP2 in the plasma membrane to generate two secondary messengers, DAG and IP3. DAG activates PKC, Ras/MEK/ERK/AP-1 and NF-kB, while IP3 is involved in the activation of intracellular Ca2+ flux. The upregulated Ca2+ signaling in turn activates the transcription factor NF AT. In short, DAG production and levels determine the duration and intensity of the Ras/MEK/ERK and PKC-dependent signaling pathways, and they are central to T-cell activation. Thus, DGKs serve as intracellular checkpoints and inhibition of DGKs is expected to enhance T cell signaling pathways and T cell activation.

Experimental evidence suggests that enhanced DGK function and / or expression in tumor infiltrating T-cells (TILs) limits tumor destruction. Experiments with CAR T cells directed against human mesothelioma engrafted into nude mice demonstrated that tumorinfiltrating CAR T cells express elevated concentrations of surface inhibitory receptors, as well as the inhibitory enzymes SHIP-1, DGKa and DGK^ (Moon et al., 2014). Further, high DGKa expression was also observed in TIL isolated from human renal tumors (Prinz et al., 2012). In mouse mesoCAR T cells, dual deletion of DGKa and DGK^ results in enhanced cytokine expression and cytotoxicity against tumor cells (Riese et al., 2013). Similar results have been reported for human CAR T cells in which both DGKa and DGK^ expression were silenced using CRISPR/

Cas9 (Jung et al., 2018). All these studies support a rationale for targeting DGKa/(^ in the development of anti-cancer therapies (Arranz-Nicolas, J. and Merida, I., 2020. Biological regulation of diacylglycerol kinases in normal and neoplastic tissues: New opportunities for cancer immunotherapy, Advances in Biological Regulation, Volume 75; Riese, M.J., Moon, E.K., Johnson, B.D., Albelda, S.M., 2016. Diacylglycerol kinases (DGKs): novel targets for improving T cell activity in cancer. Front Cell Dev Biol 4, 108.). Knock out mouse models provide further evidence: Mice lacking either DGKa or DGK(^ showed a hyper-responsive T cell phenotype and improved anti-tumor immune activity (Riese, M. J., Grewal, J., Das, J., Zou, T., Patil, V., Chakraborty, A.K., Koretzky, G.A., 2011. Decreased diacylglycerol metabolism enhances ERK activation and augments CD8+ T cell functional responses. J. Biol. Chem. 286 (7), 5254-5265; Zha, Y., Marks, R., Ho, A.W., Peterson, A.C., Janardhan, S., Brown, I., Praveen, K., Stang, S., Stone, J.C., Gajewski, T.F., 2006. T cell anergy is reversed by active Ras and is regulated by diacylglycerol kinase-alpha. Nat. Immunol. 7 (11), 1166-1173; Olenchock, B.A., Guo, R., Carpenter, J.H., Jordan, M., Topham, M.K., Koretzky, G.A., Zhong, X.P., 2006a. Disruption of diacylglycerol metabolism impairs the induction of T cell anergy. Nat. Immunol. 7 (11), 1174-1181.)

Taken together, there is substantial evidence that DGKa and DGK(^ are high value targets for cancer immunotherapy. At the same time, there is a lack of compounds with the ability to potently inhibit both DGKa and DGK(^ with good selectivity over other di acylglycerol kinases, protein kinases, and/or other lipid kinases.

This invention describes such dual DGK a/ inhibitors with excellent selectivity over other protein kinases, across safety / off-target panels and vs. other lipid kinases. These compounds potently activate suboptimally stimulated T-cells and thereby act as intracellular enhancers of co- stimulatory signaling cascades. These DGK a/ inhibitors have the potential to increase proliferation, cytotoxicity and the life span of targeted T-cells which may result in improved anticancer activity of CPIs, CD3 engaging T-cell bispecifics and CAR T-cells. Further, by engaging a signaling node central to both TCR and co-stimulatory receptors, it is plausible that these molecules enhance both signals 1 and 2 and thus single agent activity can be achieved, e.g. in inflamed tumors.

There is an ongoing need for new compounds capable of activating and proliferating T- cells, thus enabling the treatment, prevention and/or delay of progression of cancer.

It is, therefore, an object of this invention to provide compounds useful as DGKa/^ inhibitors for the treatment or prevention or amelioration of such diseases with improved therapeutic properties, in particular improved pharmacokinetic properties.

Summary of the invention

A first object of the present invention is a compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein:

Y is S(O), S(O) 2 and S(O)N(R y );

R 1 is 5-membered heteroaryl, wherein R 1 is optionally substituted with one or more R 10 which can be the same or different;

R 2 is selected from hydrogen and halogen;

R 4 is selected from Cs-w-aryl and 5-14 membered heteroaryl, wherein R 4 is optionally substituted with one or more R 11 which can be the same or different;

R 10 is -C(R 10a R 10b )2-S(O)2(R 10c );

R 10a and R 10b are each independently selected from hydrogen or Ci-6-alkyl, or R 10a and R 10b , taken together with the carbon atom to which they are attached, form a Cs-e-cycloalkyl;

R 10c is selected from hydrogen, Ci-6-alkyl, Cs-e-cycloalkyl, -N(Ci-6-alkyl), halo-Ci-6-alkyl, and phenyl, wherein phenyl is optionally substituted with one or more halogen, or Ci-6-alkyl; or R 10a and R 10c , taken together with the carbon atom and the sulphur atom to which they are attached, form a 3-10 membered heterocyclyl;

R 11 is selected from: i) halogen; ii) 3-10 membered heterocyclyl, optionally substituted with one or more halo-Ci-6-alkyl, Cs-io-cycloalkyl; iii) 5-6 membered heteroaryl, optionally substituted with one or more halogen, Ci-6-alkyl, halo-Ci-6-alkyl, Cs-io-cycloalkyl, Ci-6-haloalkoxy; iv) phenyl, optionally substituted with one or more halogen, cyano, Ci-6-alkoxy, Ci-6- haloalkyl, Ci-6-alkyl; v) -O(R l la ); vi) -N(R l lg R l lh ); and vii) -S(R l lk );

R l la is selected from Ci-12-alkyl, halo-Ci-6-alkyl, C3-7-cycloalkyl, 3-10 membered -(C1-6- alkyl)heterocyclyl, phenyl, and -Ci-6-alkyl-phenyl, wherein said Ci-12-alkyl, C3-7-cycloalkyl, phenyl and -Ci-6-alkyl-phenyl are optionally substituted with one or more halogen, Ci-6-alkyl, halo-Ci-6-alkyl, Ci-6-alkoxy, Ci-6-haloalkoxy;

R l lg and R llh are each independently selected from hydrogen, Ci-6-alkyl, -(Ci-6-alkyl)phenyl;

R l lk is selected from hydrogen and halo-Ci-6-alkyl;

R y is selected from hydrogen and Ci-6-alkyl.

A second object of the present invention is a process for the preparation of a compound of formula (I) as described above, or a pharmaceutically acceptable salt thereof, comprising reacting a compound of formula (IX) wherein Y, R 1 , R 2 and R 4 are as defined above and PG is an amino protecting group, with a suitable deprotection agent to form said compound of formula (I).

A third object of the present invention is a pharmaceutical composition comprising a compound of formula (I) as described above, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. A forth object of the present invention is a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment, prevention and/or delay of progression of cancer.

Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.

Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the invention, suitable methods and materials are described below.

All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety.

The nomenclature used in this application is based on IUPAC systematic nomenclature, unless indicated otherwise.

Detailed description of the invention

Definitions

"Alkoxy" refers to an alkyl group, as previously defined, attached to the parent molecular moiety via an oxygen atom. Unless otherwise specified, the alkoxy group contains 1 to 12 carbon atoms (“Ci-12-alkoxy”), preferably 1 to 10 carbon atoms (“Ci-io-alkoxy”), more preferably 1 to 6 carbon atoms (“Ci-6-alkoxy”). In some preferred embodiments, the alkoxy group contains 1 to 4 carbon atoms. In still other embodiments, the alkoxy group contains 1 to 3 carbon atoms. Some non-limiting examples of alkoxy groups include methoxy, ethoxy, n- propoxy, isopropoxy, n-butoxy, isobutoxy and tert-butoxy.

"Alkoxyalkyl" refers toan alkyl group, wherein at least one of the hydrogen atoms of the alkyl group has been replaced by an alkoxy group. Preferably, “alkoxyalkyl” refers to an alkyl group wherein 1, 2 or 3 hydrogen atoms, most preferably one hydrogen atom of the alkyl group have been replaced by an alkoxy group. Particularly preferred, yet non-limiting examples of alkoxyalkyl is methoxymethyl and 2-meth oxy ethyl. "Alkyl" refers to a saturated linear (i.e. unbranched) or branched univalent hydrocarbon chain or combination thereof, having the number of carbon atoms designated (i.e., Ci-io means one to ten carbon atoms). Particular alkyl groups are those having 1 to 20 carbon atoms (a “Ci- 20 alkyl”), having 1 to 12 carbon atoms (a “C1-12 alkyl”), having 1 to 10 carbon atoms (a “C1-10 alkyl”), having 1 to 8 carbon atoms (a “C1-8 alkyl”), having 1 to 6 carbon atoms (a “C1-6 alkyl”), having 2 to 6 carbon atoms (a “C2-6 alkyl”), or having 1 to 4 carbon atoms (a “C1-4 alkyl”). Examples of alkyl group include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like.

"Alkynyl" refers to an unsaturated linear (i.e. unbranched) or branched univalent hydrocarbon chain or combination thereof, having at least one site of acetylenic unsaturation i.e., having at least one moiety of the formula C=C) having the number of carbon atoms designated (i.e. C2-10 means two to ten carbon atoms). Particular alkynyl groups are those having 2 to 20 carbon atoms (a “C2-20 alkynyl”), having 2 to 8 carbon atoms (a “C2-8 alkynyl”), having 2 to 6 carbon atoms (a “C2-6 alkynyl”), having 2 to 4 carbon atoms (a “C2-4 alkynyl”). Examples of alkynyl group include, but are not limited to, groups such as ethynyl (or acetylenyl), prop-l-ynyl, prop-2 -ynyl (or propargyl), but-l-ynyl, but-2-ynyl, but-3-ynyl, homologs and isomers thereof, and the like.

"Amino", alone or in combination with other groups, refers to NH2.

"Aminoalkyl" refers to an alkyl group wherein one or more of the hydrogen atoms of the alkyl group have been replaced by an amino moiety.

"Aromatic" denotes the conventional idea of aromaticity as defined in the literature, in particular in IUPAC - Compendium of Chemical Terminology, 2 nd Edition, A. D. McNaught & A. Wilkinson (Eds). Blackwell Scientific Publications, Oxford (1997).

"Aryl" refers to a cyclic aromatic hydrocarbon moiety having a mono-, bi- or tricyclic aromatic ring of 5 to 14 carbon ring atoms (“Cs-w-aryl”). Bicyclic aryl ring systems include fused bicyclics having two fused five-membered aryl rings (denoted as 5-5), having a fivemembered aryl ring and a fused six-membered aryl ring (denoted as 5-6 and as 6-5), and having two fused six-membered aryl rings (denoted as 6-6). The aryl group can be optionally substituted as defined herein. Examples of aryl moieties include, but are not limited to, phenyl, naphthyl, phenanthryl, fluorenyl, indenyl, pentalenyl, azulenyl, and the like. The term “aryl” also includes partially hydrogenated derivatives of the cyclic aromatic hydrocarbon moiety provided that at least one ring of the cyclic aromatic hydrocarbon moiety is aromatic, each being optionally substituted.

"Cancer" refers to a disease characterized by the presence of a neoplasm or tumor resulting from abnormal uncontrolled growth of cells (such cells being "cancer cells"). As used herein, the term cancer explicitly includes, but is not limited to, hepatocellular cancer, malignancies and hyperproliferative disorders of the colon (colon cancer), lung cancer, breast cancer, prostate cancer, melanoma, and ovarian cancer.

"Cyano", alone or in combination with other groups, refers to CN (i.e. nitrile).

"Cyanoalkyl" refers to an alkyl group wherein one or more of the hydrogen atoms of the alkyl group have been replaced by a cyano moiety.

"Cycloalkyl" refers to a saturated or partially unsaturated carbocyclic moiety having mono-, bi- (including bridged bicyclic and cycloalkyl spiro moieties) or tricyclic rings and 3 to 10 carbon atoms i.e., (C3-Cio)cycloalkyl) in the ring. The cycloalkyl moiety can optionally be substituted with one or more substituents. In particular aspects cycloalkyl contains from 3 to 8 carbon atoms (i.e., (C3-C8)cycloalkyl). In other particular aspects cycloalkyl contains from 3 to 6 carbon atoms (i.e., (C3-C6)cycloalkyl). Examples of cycloalkyl moieties include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and partially unsaturated (cycloalkenyl) derivatives thereof (e.g. cyclopentenyl, cyclohexenyl, and cycloheptenyl), bicyclo[3.1.0]hexanyl, bicyclo[3.1.0]hexenyl, bicyclo[3.1.1]heptanyl, bicyclo[3.1.1]heptenyl and bicyclo[l. l.l]pentane. The cycloalkyl moiety can be attached in a “spiro-cycloalkyl” or “cycloalkyl spiro” fashion such as “spirocyclopropyl”. "EC X " refers to the effective concentration e.g. in medium or in the plasma of a particular compound required for obtaining x% of the maximum of a particular effect in vitro or in vivo. Examples of “EC X ” are EC20, EC50 and EC100 denoting the concentration of a particular compound in medium or plasma required for obtaining 20%, 50% and 100%, respectively, of the maximum of a particular effect in vitro or in vivo. "Halo" or “Halogen” refers to fluoro, chloro, bromo and/or iodo. Where a residue is substituted with more than one halogen, it can be referred to by using a prefix corresponding to the number of halogen moieties attached, e.g., dihaloaryl, dihaloalkyl, trihaloaryl etc. refer to aryl and alkyl substituted with two (“di”) or three (“tri”) Halo groups, which can be but are not necessarily the same Halo; thus 4-chl oro-3 -fluorophenyl is within the scope of dihaloaryl. An alkyl group in which one or more hydrogen is replaced with a Halo group is referred to as a “haloalkyl”, for example, “C1-6 haloalkyl.” A preferred haloalkyl group is trifluoroalkyl (-CF3).

"Haloalkoxy" refers to an alkoxy group in which at least one halogen takes the place of each H in the hydrocarbon making up the alkyl moiety of the alkoxy group. An example of a haloalkoxy group is difluoromethoxy (-OCHF2), trifluoromethoxy (-OCF3).

"Haloalkyl" refers to alkyl group, as defined above, containing at least 1 carbon atom substituted with at least one Halo group, Halo being as defined herein. Examples of "haloalkyl" groups useful in the present invention include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, isobutyl and n-butyl substituted independently with one or more Halo groups, e.g., fluoro, chloro, bromo and iodo.

"Haloaryl" refers to refers to an aryl wherein at least one hydrogen has been substituted with a halogen.

"Heteroatom" refers to an atom that is other than carbon or hydrogen.

"Heteroaryl" refers to an aromatic heterocyclic mono-, bi- or tricyclic ring system of 5 to 14 ring atoms, preferably from 5 to 10 ring atoms, more preferably from 5 to 6 ring atoms, comprising 1, 2, 3 or 4 heteroatoms selected from N, O and S, the remaining ring atoms being carbon. In some aspects, monocyclic heteroaryl rings may be 5-6 membered. Bicyclic heteroaryl ring systems include fused bicyclics having two fused five-membered heteroaryl rings (denoted as 5-5), having a five-membered heteroaryl ring and a fused six-membered heteroaryl ring (denoted as 5-6 and 6-5), and having two fused six-membered heteroaryl rings (denoted as 6-6). The heteroaryl group can be optionally substituted as defined herein.

Examples of heteroaryl moieties include pyrrolyl, furanyl, thienyl, imidazolyl, oxazolyl, thiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, tetrazolyl, pyridinyl, pyrazinyl, pyrazolyl, pyridazinyl, pyrimidinyl, triazinyl, isoxazolyl, benzofuranyl, isothiazolyl, benzothienyl, benzothiophenyl, indolyl, aza-indolyl, isoindolyl, isobenzofuranyl, benzimidazolyl, benzoxazolyl, benzoisoxazolyl, benzothiazolyl, benzoisothiazolyl, benzooxadiazolyl, benzothiadiazolyl, benzotriazolyl, purinyl, quinolinyl, isoquinolinyl, quinazolinyl, quinoxalinyl, pyrrolopyridinyl, furopyridinyl, thienopyridinyl, pyrrol opyridazinyl, pyrrolopyrimidinyl, pyrrolopyrazinyl, thienopyridazinyl, thienopyrimidinyl, thienopyrazinyl, furopyridazinyl, furopyrimidinyl, and furopyrazinyl. Most preferably, “5-membered heteroaryl” refers to the following groups:

"Heterocycle" or “heterocyclyl” refers to a 3, 4, 5, 6, 7, 8, 9, 10-membered monocyclic, 7, 8, 9 and 10-membered bicyclic (including bridged bicyclic and cycloalkyl spiro moieties) or 10, 11, 12, 13, 14 and 15-membered bicyclic heterocyclic moiety that is saturated or partially unsaturated, and has one or more (e.g., 1, 2, 3 or 4 heteroatoms selected from oxygen, nitrogen and sulfur in the ring with the remaining ring atoms being carbon. In some aspects, the heterocycle is a heterocycloalkyl. In particular aspects heterocycle or heterocyclyl refers to a 4, 5, 6 or 7-membered heterocycle. When used in reference to a ring atom of a heterocycle, a nitrogen or sulfur may also be in an oxidized form, and a nitrogen may be substituted with one or more (Ci-Ce)alkyl or groups. The heterocycle can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure. Any of the heterocycle ring atoms can be optionally substituted with one or more substituents described herein. Examples of such saturated or partially unsaturated heterocycles include, without limitation, tetrahydrofuranyl, tetrahydrothienyl, pyrrolidinyl, pyrrolidonyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, pyrrolidine 1 -oxide, N-hydroxypiperidine, 1- methylpyrrolidine N-oxide, diazirinyl and quinuclidinyl. The term heterocycle also includes groups in which a heterocycle is fused to one or more aryl, heteroaryl, or cycloalkyl rings, such as indolinyl, 3H-indolyl, chromanyl, azabicyclo[2.2. l]heptanyl, azabicyclo[3.1.0]hexanyl, azabicyclo[3.1.1]heptanyl, octahydroindolyl, or tetrahydroquinolinyl.

"Hydroxy", alone or in combination with other groups, refers to OH.

"Hydroxyalkyl" refers to an alkyl group wherein one or more of the hydrogen atoms of the alkyl group have been replaced by a hydroxy moiety. Examples include alcohols and diols.

"Moiety" and “substituent” refer to an atom or group of chemically bonded atoms that is attached to another atom or molecule by one or more chemical bonds thereby forming part of a molecule.

When indicating the number of substituents, the term “one or more” refers to the range from one substituent to the highest possible number of substitution, i.e. replacement of one hydrogen up to replacement of all hydrogens by substituents, in particular wherein “one or more” refers to one, two or three, most particularly “one or more” refers to one or two.

"Optional" or "optionally" means that the subsequently described event or circumstance may but need not occur, and that the description includes instances where the event or circumstance occurs and instances in which it does not. For example, "aryl group optionally substituted with an alkyl group" means that the alkyl may but need not be present, and the description includes situations where the aryl group is substituted with an alkyl group and situations where the aryl group is not substituted with the alkyl group.

"Optionally substituted" refers to means unsubstituted or substituted. Generally these substituents can be the same or different.

"Oxo", alone or in combination with other groups, refers to =0.

"Pharmaceutically acceptable salt" refers to those salts which retain the biological effectiveness and properties of the free bases or free acids, which are not biologically or otherwise undesirable. The salts are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, particularly hydrochloric acid, and organic acids such as acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, N-acetylcystein.

Particularly preferred pharmaceutically acceptable salts of compounds of formula (I) are the salts of hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid and methanesulfonic acid.

"Protecting group" (PG) denotes the group which selectively blocks a reactive site in a multifunctional compound such that a chemical reaction can be carried out selectively at another unprotected reactive site in the meaning conventionally associated with it in synthetic chemistry. Protective groups can be removed at the appropriate point. Exemplary protective groups are amino-protective groups, carb oxy-protective groups or hydroxy-protective groups. Particular protective groups are the tert-butoxy carbonyl (Boc), benzyl oxy carbonyl (Cbz), fluorenylmethoxycarbonyl (Fmoc) and benzyl (Bn). Further particular protective groups are the tert-butoxy carbonyl (Boc) and the fluorenylmethoxycarbonyl (Fmoc). More particular protective group is the tert-butoxy carbonyl (Boc). Exemplary protective groups and their application in organic synthesis are described, for example, in “Protective Groups in Organic Chemistry” by T. W. Greene and P. G. M. Wutts, 5th Ed., 2014, John Wiley & Sons, N.Y.

"Prophylaxis" as used herein includes: preventing or delaying the appearance of clinical symptoms of the state, disorder or condition developing in a mammal and especially a human that may be afflicted with or predisposed to the state, disorder or condition but does not yet experience or display clinical or subclinical symptoms of the state, disorder or condition.

"Substituted" refers to the replacement of at least one of hydrogen atoms of a compound or moiety with another substituent or moiety. Examples of such substituents include, without limitation, halogen, -OH, -CN, oxo, alkoxy, alkyl, alkylene, aryl, heteroaryl, haloalkyl, haloalkoxy, cycloalkyl and heterocycle. For example, the term “haloalkyl” refers to the fact that one or more hydrogen atoms of an alkyl (as defined below) is replaced by one or more halogen atoms (e.g., trifluoromethyl, difluoromethyl, fluoromethyl, chloromethyl, etc.). In one aspect, substituted as used herein can refer to replacement of at least one hydrogen atom of a compound or moiety described herein with halogen or alkyl.

"Therapeutically effective amount" refers to 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.

"Therapeutically inert carrier" refers to any ingredient having no therapeutic activity and being non-toxic such as disintegrators, binders, fillers, solvents, buffers, tonicity agents, stabilizers, antioxidants, surfactants or lubricants used in formulating pharmaceutical products.

In particular, the chemical groups whose definitions are given above are those specifically exemplified in the examples.

The following abbreviations are used in the present text:

BOP = benzotriazol-l-yloxytris(dimethylamino)phosphonium hexafluorophosphate, Brine = saturated aqueous NaCl solution, CAS = chemical abstracts registration number, CDI = l,l'-Carbonyldiimidazole, DBU = l,8-diazabicyclo[5,4,0]undec-7-ene, DCM = dichloromethane, DDQ = 2,3-dichloro-5,6-dicyano-l,4-benzoquinone, DMF = N,N- dimethylformamide, DIPEA = N,N-diisopropylethylamine, EDC = l-ethyl-3-(3- dimethylaminopropyljcarbodiimide, ESI = electrospray ionization, EtOAc = ethyl acetate, EtOH = ethanol, h = hour(s), HATU = l-[bis(dimethylamino)methylene]-lH-l,2,3-triazolo[4,5- b]pyridinium-3 -oxide hexafluorophosphate, HBTU = O-benzotriazole-N,N,N’,N’-tetram ethyl - uronium-hexafluoro-phosphate, HFIP = hexafluoroisopropanol, HOBt = hydroxybenzotriazole, HPLC = high performance liquid chromatography, m-CPBA = meta-chloroperoxybenzoic acid, MeCN = acetonitrile, Mel = methyliodide, MeOH = methanol, min = minute(s), MS = mass spectrum, NBS = N-bromosuccinimide, PE = petroleum ether, PyBroP = bromo-tris- pyrrolidino-phosphonium hexafluorophosphate, RT = room temperature, T3P = propylphosphonic anhydride, TBAF = tetrabutylammonium fluoride, TBAOH = tetrabutyl ammonium hydroxide, TBDMS = tert-butyldimethylsilyl, TEA = triethylamine, TFA = trifluoroacetic acid, THF = tetrahydrofuran, TMSOTF = trifluoromethanesulfonic acid trimethyl silyl ester, TLC = thin layer chromatography.

In the description herein, if there is a discrepancy between a depicted structure and a name given to that structure, then the depicted structure controls. Additionally, if the stereochemistry of a structure or a portion of a structure is not indicated with, for example, bold wedged, or dashed lines, the structure or portion of the structure is to be interpreted as encompassing all stereoisomers of it. In some cases, however, where more than one chiral center exists, the structures and names may be represented as single enantiomers to help describe the relative stereochemistry.

Unless otherwise indicated, the terms “a compound of the formula” or “a compound of formula” or “compounds of the formula” or “compounds of formula” refer to any compound selected from the genus of compounds as defined by the formula (including any pharmaceutically acceptable salt of any such compound if not otherwise noted).

Certain compounds may exhibit tautomerism. Tautomeric compounds can exist as two or more interconvertable species. Prototropic tautomers result from the migration of a covalently bonded hydrogen atom between two atoms. Tautomers generally exist in equilibrium and attempts to isolate an individual tautomers usually produce a mixture whose chemical and physical properties are consistent with a mixture of compounds. The position of the equilibrium is dependent on chemical features within the molecule. For example, in many aliphatic aldehydes and ketones, such as acetaldehyde, the keto form predominates while; in phenols, the enol form predominates. Common prototropic tautomers include keto/enol (- C(=O)-CH- -C(-OH)=CH-), amide/imidic acid (-C(=O)-NH- -C(-OH)=N-) and amidine (-

C(=NR)-NH- -C(-NHR)=N-) tautomers. The latter two are particularly common in heteroaryl and heterocyclic rings and the present invention encompasses all tautomeric forms of the compounds. Furthermore, the invention includes all optical isomers, i.e. diastereoisomers, diastereomeric mixtures, racemic mixtures, all their corresponding enantiomers and/or tautomers as well as their solvates of the compounds of formula (I).

The compounds of formula (I) may contain one or more asymmetric centers and can therefore occur as racemates, racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. Additional asymmetric centers may be present depending upon the nature of the various substituents on the molecule. Each such asymmetric center will independently produce two optical isomers and it is intended that all of the possible optical isomers and diastereomers in mixtures and as pure or partially purified compounds are included within this invention. The present invention is meant to encompass all such isomeric forms of these compounds. The independent syntheses of these diastereomers or their chromatographic separations may be achieved as known in the art by appropriate modification of the methodology disclosed herein. Their absolute stereochemistry may be determined by the x-ray crystallography of crystalline products or crystalline intermediates which are derivatized, if necessary, with a reagent containing an asymmetric center of known absolute configuration. If desired, racemic mixtures of the compounds may be separated so that the individual enantiomers are isolated. The separation can be carried out by methods well known in the art, such as the coupling of a racemic mixture of compounds to an enantiomerically pure compound to form a diastereomeric mixture, followed by separation of the individual diastereomers by standard methods, such as fractional crystallization or chromatography.

In the embodiments, where optically pure enantiomers are provided, optically pure enantiomer means that the compound contains > 90% of the desired isomer by weight, particularly > 95% of the desired isomer by weight, or more particularly > 99% of the desired isomer by weight, said weight percent based upon the total weight of the isomer(s) of the compound. Chirally pure or chirally enriched compounds may be prepared by chirally selective synthesis or by separation of enantiomers. The separation of enantiomers may be carried out on the final product or alternatively on a suitable intermediate.

In some embodiments, the compounds of formula (I) are isotopically-labeled by having one or more atoms therein replaced by an atom having a different atomic mass or mass number. Such isotopically-labeled (i.e., radiolabeled) compounds of formula (I) are considered to be within the scope of this disclosure. Examples of isotopes that can be incorporated into the compounds of formula (I) include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine, chlorine, and iodine, such as, but not limited to, 2 H, 3 H, n C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 31 P, 32 P, 35 S, 18 F, 36 C1, 123 I, and 125 I, respectively. Certain isotopically-labeled compounds of formula (I), for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3 H, and carbon-14, i.e., 14 C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection. For example, a compound of formula (I) can be enriched with 1, 2, 5, 10, 25, 50, 75, 90, 95, or 99 percent of a given isotope.

Substitution with heavier isotopes such as deuterium, i.e. 2 H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements.

Substitution with positron emitting isotopes, such as n C, 18 F, 15 O and 13 N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Examples as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.

Compounds of the invention

In one embodiment, there is provided a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein:

Y is S(O), S(O) 2 and S(O)N(R y ); R 1 is 5-membered heteroaryl, wherein R 1 is optionally substituted with one or more R 10 which can be the same or different;

R 2 is selected from hydrogen and halogen;

R 4 is selected from Cs-w-aryl and 5-14 membered heteroaryl, wherein R 4 is optionally substituted with one or more R 11 which can be the same or different;

R 10 is -C(R 10a R 10b )2-S(O)2(R 10c );

R 10a and R 10b are each independently selected from hydrogen and Ci-6-alkyl, or R 10a and R 10b , taken together with the carbon atom to which they are attached, form a Cs-e-cycloalkyl;

R 10c is selected from hydrogen, Ci-6-alkyl, Cs-e-cycloalkyl, -N(Ci-6-alkyl), halo-Ci-6-alkyl, and phenyl, wherein phenyl is optionally substituted with one or more halogen, or Ci-6-alkyl; or R 10a and R 10c , taken together with the carbon atom and the sulphur atom to which they are attached, form a 3-10 membered heterocyclyl;

R 11 is selected from: i) halogen; ii) 3-10 membered heterocyclyl, optionally substituted with one or more halo-Ci-6-alkyl, Cs-io-cycloalkyl; iii) 5-6 membered heteroaryl, optionally substituted with one or more halogen, Ci-6-alkyl, halo-Ci-6-alkyl, Cs-io-cycloalkyl, Ci-6-haloalkoxy; iv) phenyl, optionally substituted with one or more halogen, cyano, Ci-6-alkoxy, Ci-6- haloalkyl, Ci-6-alkyl; v) -O(R l la ); vi) -N(R l lg R l lh ); and vii) -S(R l lk ); R l la is selected from Ci-12-alkyl, halo-Ci-6-alkyl, C3-7-cycloalkyl, 3-10 membered -(C1-6- alkyl)heterocyclyl, phenyl, and -Ci-6-alkyl-phenyl, wherein said Ci-12-alkyl, C3-7-cycloalkyl, phenyl and -Ci-6-alkyl-phenyl are optionally substituted with one or more halogen, Ci-6-alkyl, halo-Ci-6-alkyl, Ci-6-alkoxy, Ci-6-haloalkoxy;

R l lg and R llh are each independently selected from hydrogen, Ci-6-alkyl, -(Ci-6-alkyl)phenyl;

R l lk is selected from hydrogen and halo-Ci-6-alkyl;

R y is selected from hydrogen and Ci-6-alkyl.

In another embodiment, there is provided a compound of formula (I) as described herein, wherein R 1 is optionally substituted with one or more R 10 which can be the same or different.

In another embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 2 is selected from hydrogen and fluorine.

In another embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 4 is phenyl.

In another embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 10a and R 10b are each independently selected from hydrogen or Ci-6-alkyl, or R 10a and R 10b , taken together with the carbon atom to which they are attached, form a C3-6-cycloalkyl.

In another embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 10 is selected from C3-6-cycloalkyl, substituted with one -S(O)2(Ci-6-alkyl).

In another embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 10 is 3-6 membered heterocyclyl, wherein the heteroatom is sulphur, substituted with one or more Ci-10-alkyl, oxo, wherein the oxo substitution is on the sulphur atom. In another embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 10 is selected from 1 -methylmethyl sulfonyl -ethyl .

In another embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 11 is selected from -O(R l la ), wherein R l la is selected from Ci-12-alkyl, halo-Ci-6-alkyl, C3-7-cycloalkyl, 3-10 membered-(Ci-6- alkyl)heterocyclyl, phenyl, -Ci-6-alkyl-phenyl, wherein said Ci-12-alkyl, C3-7-cycloalkyl, phenyl and -Ci-6-alkyl-phenyl are optionally substituted with one or more halogen, Ci-6-alkyl, halo-Ci- 6-alkyl, Ci-6-alkoxyl, Ci-6-haloalkoxyl.

In one embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 11 is selected from viii) halogen; ix) 3-10 membered heterocyclyl; x) 5-6 membered heteroaryl, optionally substituted with one or more halogen, halo-Ci-6- alkyl, Cs-io-cycloalkyl, Ci-6-haloalkoxy; xi) phenyl, optionally substituted with one or more cyano, Ci-6-alkoxy, Ci-6-haloalkyl, C1-6- alkyl; xii) -O(R l la ); R lla is selected from Ci-12-alkyl, halo-Ci-6-alkyl, C3-7-cycloalkyl, 5-6 membered -(Ci-6-alkyl)heteroaryl, phenyl and -Ci-6-alkyl-phenyl, wherein said Ci-12-alkyl, phenyl, 5-6 membered -(Ci-6-alkyl)heteroaryl and -Ci-6-alkyl-phenyl are optionally substituted with one or more halogen, Ci-6-alkyl, halo-Ci-6-alkyl, Ci-6-alkoxy, C1-6- haloalkoxy; xiii)-N(R l lg R l lh ); R llg and R l lh are each independently selected from Ci-6-alkyl, -(C1-6- alkyl)phenyl; xiv)-S(halo-Ci-6-alkyl); xv) CH2-O-phenyl.

In another embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 11 is selected from i) -O(R l la ), wherein R l la is selected from halo-Ci-6-alkyl and phenyl, wherein said phenyl is optionally substituted with one or more halogen; or ii) phenyl, optionally substituted with one or more Ci-6-alkoxy, Ci-6-haloalkyl; iii) halogen iv) 5-6 membered heteroaryl, optionally substituted with one or more halo-Ci-6-alkyl, C3-10- cycloalkyl, Ci-6-haloalkoxy, chloro, trifluoromethoxy-2-pyridyl.

In another embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein R 11 is selected from (trifluoromethyl)phenyl, (trifluoromethyl)-oxadiazolyl, cyclopropyl-oxadiazolyl, phenoxyl, methoxyphenyl, tetrafluoroethoxyl , di chi orophenoxyl .

In another embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein Y is S(O)2.

In another embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:

Y is S(O), S(O) 2 and S(O)N(R y );

R 1 is 5-membered heteroaryl, wherein R 1 is optionally substituted with one or more R 10 which can be the same or different;

R 2 is selected from hydrogen and fluorine;

R 4 is selected from Ce-aryl and 6-membered heteroaryl, wherein R 4 is optionally substituted with one or more R 11 which can be the same or different;

R 10 is C3-6-cycloalkyl substituted with one -S(O)2(Ci-6-alkyl);

R 11 is i) halogen; ii) 3-10 membered heterocyclyl; iii) 5-6 membered heteroaryl, optionally substituted with one or more halogen, halo-Ci-6- alkyl, Cs-io-cycloalkyl, Ci-6-haloalkoxy; iv) phenyl, optionally substituted with one or more cyano, Ci-6-alkoxy, Ci-6-haloalkyl, C1-6- alkyl; v) -0(R l la ); R lla is selected from Ci-12-alkyl, halo-Ci-6-alkyl, C3-7-cycloalkyl, 5-6 membered -(Ci-6-alkyl)heteroaryl, phenyl and -Ci-6-alkyl-phenyl, wherein said C1-12- alkyl, phenyl, 5-6 membered -(Ci-6-alkyl)heteroaryl and -Ci-6-alkyl-phenyl are optionally substituted with one or more halogen, Ci-6-alkyl, halo-Ci-6-alkyl, C1-6- alkoxy, Ci-6-haloalkoxy; vi) -N(R l lg R l lh ); R llg and R l lh are each independently selected from Ci-6-alkyl, -(C1-6- alkyl)phenyl; vii) -S(halo-Ci-6-alkyl); viii) CH2-O-phenyl.

In another embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:

Y is S(O), S(O) 2 and S(O)N(R y );

R 1 is oxadiazole, wherein R 1 is optionally substituted with one or more R 10 which can be the same or different;

R 2 is selected from hydrogen and fluorine;

R 4 is phenyl;

R 10 is selected from methyl-methylsulfonyl-ethyl, cyclopropylsulfonyl-methyl-ethyl, methyldi oxo-thiolanyl, methylsulfonylmethyl, methylsulfonyl cyclobutyl;

R 11 is selected from: i) -O(R l la ), wherein R l la is selected from halo-Ci-6-alkyl, phenyl, wherein said phenyl is optionally substituted with one or more halogen; ii) phenyl, optionally substituted with one or more Ci-6-alkoxy, Ci-6-haloalkyl; iii) halogen; and iv) 5-6 membered heteroaryl, optionally substituted with one or more halo-Ci-6-alkyl, C3-10- cycloalkyl, Ci-6-haloalkoxy. In another embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:

Y is S(O), S(O) 2 and S(O)N(R y );

R 1 is oxadiazole, wherein R 1 is optionally substituted with one or more R 10 which can be the same or different;

R 2 is selected from hydrogen and fluorine;

R 4 is phenyl;

R 10 is methyl-methylsulfonyl-ethyl;

R 11 is selected from (trifluoromethyl )phenyl, (trifluoromethyl)-oxadiazolyl, cyclopropyl - oxadiazolyl, phenoxyl, methoxyphenyl, tetrafluoroethoxyl, dichlorophenoxyl, chloro and trifluoromethoxy)-2-pyridyl.

In another embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:

Y is S(O), S(O) 2 and S(O)N(R y );

R 1 is 5-membered heteroaryl, wherein R 1 is optionally substituted with one or more R 10 which can be the same or different;

R 2 is selected from hydrogen and fluorine;

R 4 is selected from Ce-aryl and 6 membered heteroaryl, wherein R 4 is optionally substituted with one or more R 11 which can be the same or different;

R 10 is selected from C3-6-cycloalkyl, substituted with one -S(O) 2 (Ci-6-alkyl);

R 11 is selected from -O(R l la ), wherein R l la is selected from Ci-i 2 -alkyl, halo-Ci-6-alkyl, C3-7- cycloalkyl, 3-10 membered-(Ci-6-alkyl)heterocyclyl, phenyl, -Ci-6-alkyl-phenyl, wherein said Ci-12-alkyl, C3-7-cycloalkyl, phenyl and -Ci-6-alkyl-phenyl are optionally substituted with one or more halogen, Ci-6-alkyl, halo-Ci-6-alkyl, Ci-6-alkoxyl, Ci-6-haloalkoxyl.

In another embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:

Y is S(O), S(O) 2 and S(O)N(R y );

R 1 is oxadiazole, wherein R 1 is optionally substituted with one or more R 10 which can be the same or different;

R 2 is selected from hydrogen and fluorine;

R 4 is phenyl;

R 10 is selected from methyl-methylsulfonyl-ethyl, cyclopropylsulfonyl-methyl-ethyl, methyldi oxo-thiolanyl, methylsulfonylmethyl, methylsulfonyl cyclobutyl;

R 11 is selected from 2-methoxy- 1,1 -dimethyl-ethoxy, phenoxyl, bromophenoxyl, (trifluoromethyl)-oxadiazolyl]methoxyl, (trifluoromethyl)phenyl, tetrahydropyranyl, methoxyphenyl, methoxy-methyl-phenyl, (difluoromethoxy)phenoxyl, (trifluoromethyl)- oxadiazolyl, cyclopropyl-oxadiazolyl, trifluoromethoxyl, chloro, methoxyphenoxyl, chloro- methoxy-phenoxyl, trifluoromethoxy)phenoxyl, fluoro-(trifluoromethyl)phenoxyl, chloro- fluoro-phenoxyl, trifluoromethyl-pyridyl, chlorophenoxyl, fluorophenoxyl, dichlorophenoxyl, methylphenoxyl, tetrafluoroethoxyl, trifluoroethoxyl, trifluoropropoxyl, benzonitrileyl, ethylpropoxyl, isopropyl(methyl)aminoyl, cyclopentoxyl, benzyl(methyl)aminoyl, (fluorophenyl)methoxyl, benzyloxyl, fluoro-pyridyl, trifluoromethylsulfanyl.

In another embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, wherein:

Y is S(O), S(O) 2 and S(O)N(R y );

R 1 is oxadiazole, wherein R 1 is optionally substituted with one or more R 10 which can be the same or different; R 2 is selected from hydrogen and fluorine;

R 4 is phenyl;

R 10 is selected from methyl-methylsulfonyl-ethyl;

R 11 is selected from (trifluoromethyl )phenyl, (trifluoromethyl)-oxadiazolyl, cyclopropyl - oxadiazolyl, phenoxyl, methoxyphenyl, tetrafluoroethoxyl, dichlorophenoxyl.

In another embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, selected from:

(3R)-3-amino-5-[[4-(l-ethylpropoxy)phenyl]methyl]-7-[5-(l -methyl-l-methylsulfonyl-ethyl)-l,3,4- oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4 -one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4 -oxadiazol-2-yl]-l,l-dioxo-5-[[4-(2,2,2- tri fluoroethoxy )phenyl]methyl]-2,3-dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-(cyclopentoxy)phenyl]methyl]-7-[5-(l-m ethyl-l-methylsulfonyl-ethyl)-l,3,4- oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4 -one;

(3R)-3-amino-5-[(4-benzyloxyphenyl)methyl]-7-[5-(l-methyl -l-methylsulfonyl-ethyl)-l,3,4- oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4 -one;

(3R)-3-amino-5-[[4-[(4-fluorophenyl)methoxy]phenyl]methyl ]-7-[5-(l -methyl- 1 -methylsulfonyl- ethyl)-l, 3, 4-oxadiazol-2-yl]- 1,1 -di oxo-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-[(2-fluorophenyl)methoxy]phenyl]methyl ]-7-[5-(l -methyl- 1 -methylsulfonyl- ethyl)-l, 3, 4-oxadiazol-2-yl]- 1,1 -di oxo-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-[(3-fluorophenyl)methoxy]phenyl]methyl ]-7-[5-(l -methyl- 1 -methylsulfonyl- ethyl)-l, 3, 4-oxadiazol-2-yl]- 1,1 -di oxo-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4 -oxadiazol-2-yl]-l,l-dioxo-5-[[4-(3,3,3- trifluoropropoxy)phenyl]methyl]-2,3-dihydro-lX6,5-benzothiaz epin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4 -oxadiazol-2-yl]-l,l-dioxo-5-[[4- (trifluoromethylsulfanyl)phenyl]methyl]-2,3-dihydro-lX6,5-be nzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4 -oxadiazol-2-yl]-l,l-dioxo-5-[[4- (phenoxymethyl)phenyl]methyl]-2,3-dihydro-lX6,5-benzothiazep in-4-one; (3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-ox adiazol-2-yl]-l,l-dioxo-5-[(4- phenoxyphenyl )methyl]-2,3-dihydro- IX6,5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-[3-(difluoromethoxy)phenoxy]phenyl]met hyl]-7-[5-(l-methyl-l- methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-di hydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4 -oxadiazol-2-yl]-5-[[4-(3- methylphenoxy)phenyl]methyl]-l,l-dioxo-2,3-dihydro-lX6,5-ben zothiazepin-4-one;

(3 R)-3-amino-5-[[4-(2,3-dichlorophenoxy)phenyl]methyl]-7-[5-(l -methyl- 1-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiaz epin-4-one;

(3R)-3-amino-5-[[4-(3-fluorophenoxy)phenyl]methyl]-7-[5-( l -methyl- 1-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiaz epin-4-one;

(3R)-3-amino-5-[[4-(3-chlorophenoxy)phenyl]methyl]-7-[5-( l -methyl- 1-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiaz epin-4-one;

(3 R)-3-amino-5-[[4-(3-chloro-4-fluoro-phenoxy)phenyl]methyl]-7 -[5-(l -methyl- 1-methylsulfonyl- ethyl)-!, 3, 4-oxadiazol-2-yl]- 1,1 -di oxo-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4 -oxadiazol-2-yl]-l,l-dioxo-5-[[4-[3-

(trifluoromethyl)phenoxy]phenyl]methyl]-2,3-dihydro-lX6,5 -benzothiazepin-4-one;

(3R)-3-amino-5-[[4-[3-fhroro-5-(trifluoromethyl)phenoxy]p henyl]methyl]-7-[5-(l-methyl-l- methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-di hydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4 -oxadiazol-2-yl]-l,l-dioxo-5-[[4-[3-

(trifluoromethoxy)phenoxy]phenyl]methyl]-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-(3-chloro-5-methoxy-phenoxy)phenyl]met hyl]-7-[5-(l-methyl-l- methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-di hydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-(3-chloro-5-methoxy-phenoxy)phenyl]met hyl]-7-[5-(l-methyl-l- methylsulfonyl-ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-di hydro-lX6,5-benzothiazepin-4-one;

4-[4-[[(3R)-3-amino-7-[5-(l -methyl- l-methylsulfonyl-ethyl)-l, 3, 4-oxadiazol-2-yl]-l,l,4-tri oxo-2, 3- dihydro- 1X6, 5-benzothiazepin-5-yl]methyl]phenyl]benzonitrile;

(3R)-3-amino-5-[[4-(4-methoxyphenyl)phenyl]methyl]-7-[5-( l-methyl-l-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiaz epin-4-one;

(3R)-3-amino-5-[[4-(3-methoxyphenyl)phenyl]methyl]-7-[5-( l-methyl-l-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiaz epin-4-one;

(3 R)-3-amino-5-[[4-(4-methoxy-2-methyl-phenyl)phenyl]methyl]-7 -[5-(l -methyl- 1-methylsulfonyl- ethyl)-!, 3, 4-oxadiazol-2-yl]- 1,1 -di oxo-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one; (3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-ox adiazol-2-yl]-l,l-dioxo-5-[[4-[4-

(trifluoromethyl)phenyl]phenyl]methyl]-2,3-dihydro-lX6,5- benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4 -oxadiazol-2-yl]-l,l-dioxo-5-[[4-

(l,l,2,2-tetrafluoroethoxy)phenyl]methyl]-2,3-dihydro-lX6 ,5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-(3-bromophenoxy)phenyl]methyl]-7-[5-(l -methyl-l-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiaz epin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4 -oxadiazol-2-yl]-l,l-dioxo-5-[[4-

(trifluoromethoxy)phenyl]methyl]-2,3-dihydro-lX6,5-benzot hiazepin-4-one;

(3R)-3-amino-l,l-diketo-7-[5-(l-mesyl-l-methyl-ethyl)-l,3 ,4-oxadiazol-2-yl]-5-[4-[5-

(trifluoromethoxy)-2-pyridyl]benzyl]-2,3-dihydro-lX6,5-be nzothiazepin-4-one;

(3 R)-3-amino-5-[[4-[benzyl(methyl)amino]phenyl]methyl]-7-[5-(l -methyl- 1-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiaz epin-4-one;

(3R)-3-amino-5-[[4-[isopropyl(methyl)amino]phenyl]methyl] -7-[5-(l -methyl- 1 -methylsulfonyl- ethyl)-l, 3, 4-oxadiazol-2-yl]- 1,1 -di oxo-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4 -oxadiazol-2-yl]-l,l-dioxo-5-[[4-[5-

(trifluoromethyl)-l,2,4-oxadiazol-3-yl]phenyl]methyl]-2,3 -dihydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4 -oxadiazol-2-yl]-l,l-dioxo-5-[[4-[5-

(trifluoromethyl)-2-pyridyl]phenyl]methyl]-2,3-dihydro-lX 6,5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-(5-fluoro-2-pyridyl)phenyl]methyl]-7-[ 5-(l -methyl- 1-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiaz epin-4-one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4 -oxadiazol-2-yl]-l,l-dioxo-5-[[6-[4-

(trifluoromethyl)phenyl]-3-pyridyl]methyl]-2,3-dihydro-lX 6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fhioro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-5-

[(6-phenoxy-3-pyridyl)methyl]-2,3-dihydro-lX6,5-benzothia zepin-4-one;

(3 R)-3-amino-8-fluoro-5-[[4-(4-methoxyphenyl)phenyl]methyl]-7- [5-(l -methyl- 1-methylsulfonyl- ethyl)-!, 2, 4-oxadiazol-3-yl]- 1,1 -di oxo-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-5-[(4-chlorophenyl)methyl]-8-fluoro-7-[5-(l- methyl-l-methylsulfonyl-ethyl)-l,2,4- oxadiazol-3-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4 -one;

(3R)-3-amino-8-fhroro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-5-

[[4-[5-(tri fluoromethyl)-!, 2, 4-oxadiazol-3-yl]phenyl]methyl]-2, 3-dihydro- 1X6, 5-benzothiazepin-4- one; (3R)-3-amino-5-[[4-(5-cyclopropyl-l,2,4-oxadiazol-3-yl)pheny l]methyl]-8-fluoro-7-[5-(l-methyl-l- methylsulfonyl-ethyl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-2,3-di hydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-5-

[(4-phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepi n-4-one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-5-

[[4-(trifluoromethoxy)phenyl]methyl]-2,3-dihydro-lX6,5-be nzothiazepin-4-one;

(3R)-3-amino-5-[(4-chlorophenyl)methyl]-8-fluoro-7-[5-(l- methyl-l-methylsulfonyl-ethyl)-l,3,4- oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4 -one;

(3 R)-3-amino-5-[(4-chlorophenyl)methyl]-8-fluoro-7-[5-(l -methylsulfonyl cyclopropyl)-l, 3, 4- oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4 -one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-5-

[(4-tetrahydropyran-4-yl phenyl )methyl]-2,3-dihydro- IX6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fluoro-5-[[4-(2-methoxy-l J -dimethyl-ethoxy )phenyl]methyl]-7-[5-(l -methyl- 1- methylsulfonyl-ethyl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-2,3-di hydro-lX6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-

[[4-[4-(tri fluoromethyl)phenyl]phenyl]methyl]-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-

[[4-(2,2,2-trifluoroethoxy)phenyl]methyl]-2,3-dihydro-lX6 ,5-benzothiazepin-4-one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-

[[4-[5-(tri fl uorom ethyl )-2-pyridyl]phenyl]methyl]-2,3-dihydro-IX6,5-benzothiazepin- 4-one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-

[[4-(l,l,2,2-tetrafluoroethoxy)phenyl]methyl]-2,3-dihydro -lX6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-

[(4-phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepi n-4-one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-

[[4-[5-(trifluoromethyl)-l,2,4-oxadiazol-3-yl]phenyl]meth yl]-2,3-dihydro-lX6,5-benzothiazepin-4- one;

(3R)-3-amino-8-fluoro-5-[[4-(4-methoxyphenyl)phenyl]methy l]-7-[5-(l -methyl- 1 -methylsulfonyl- ethyl)-l, 3, 4-oxadiazol-2-yl]- 1,1 -di oxo-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-8-fhroro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-

[[4-[6-(trifluoromethyl)-3-pyridyl]phenyl]methyl]-2,3-dih ydro-lX6,5-benzothiazepin-4-one; (3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl) -l,2,4-oxadiazol-3-yl]-l,l-dioxo-5- [[4-[[3-(tri fluoromethyl)- 1, 2, 4-oxadiazol-5-yl]methoxy]phenyl]methyl]-2,3-dihydro-lX6, 5- benzothiazepin-4-one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,2,4-oxadiazol-3-yl]-l-oxo-5-[(4- phenoxyphenyl )methyl]-2,3-dihydro- IX4,5-benzothiazepin-4-one;

(3R)-3-amino-5-[[4-(4-methoxyphenyl)phenyl]methyl]-7-[5-( l-methyl-l-methylsulfonyl-ethyl)- l,2,4-oxadiazol-3-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiaz epin-4-one;

(3R)-3-amino-7-[5-(l-methylsulfonylcyclobutyl)-l,3,4-oxad iazol-2-yl]-l,l-dioxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one ;

(3R)-3-amino-7-[5-(l-methylsulfonylcyclopropyl)-l,3,4-oxa diazol-2-yl]-l,l-dioxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one ;

(3R)-3-amino-7-[5-(l-cyclopropylsulfonyl-l-methyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-5-[(4- phenoxyphenyl )methyl]-2,3-dihydro- IX6,5-benzothiazepin-4-one;

(3R)-3-amino-7-[5-(2-methyl-l,l-dioxo-thiolan-2-yl)-l,3,4 -oxadiazol-2-yl]-l,l-dioxo-5-[(4- phenoxyphenyl )methyl]-2,3-dihydro- IX6,5-benzothiazepin-4-one;

2-[5-[(3R)-3-amino-l,l,4-trioxo-5-[[4-[4-(trifluoromethyl )phenyl]phenyl]methyl]-2,3-dihydro- lX6,5-benzothiazepin-7-yl]-l,3,4-oxadiazol-2-yl]-N,N-dimethy l-propane-2-sulfonamide;

(3R)-3-amino-7-[5-(l-methyl-l-pyrrolidin-l-ylsulfonyl-eth yl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5- [[4-[4-(trifluorom ethyl)phenyl]phenyl]methyl]-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-8-fluoro-7-[2-(methylsulfonylmethyl)tetrazol -5-yl]-l,l-dioxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one ;

(3R)-3-amino-8-fluoro-7-[l-(methylsulfonylmethyl)tetrazol -5-yl]-l,l-dioxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro- IX6,5-benzothiazepin-4-one.

In a preferred embodiment, there is provided a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, selected from (3R)-3-amino-8-fhroro-7-[5-(l-methyl-l-methylsulfonyl-ethyl) -l,3,4-oxadiazol-2-yl]-l,l-dioxo-5- [[4-(l,l,2,2-tetrafluoroethoxy)phenyl]methyl]-2,3-dihydro-lX 6,5-benzothiazepin-4-one;

(3R)-3-amino-8-fhroro-5-[[4-(4-methoxyphenyl)phenyl]methy l]-7-[5-(l -methyl- 1 -methylsulfonyl- ethyl)-l, 3, 4-oxadiazol-2-yl]- 1,1 -di oxo-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one; (3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl) -l,2,4-oxadiazol-3-yl]-l,l-dioxo-5- [(4-phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-4 -one;

(3 R)-3-amino-5-[[4-(2,3-dichlorophenoxy)phenyl]methyl]-7-[5-(l -methyl- 1-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiaz epin-4-one;

(3R)-3-amino-5-[[4-(3-chlorophenoxy)phenyl]methyl]-7-[5-( l -methyl- 1-methylsulfonyl-ethyl)-

1.3.4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzoth iazepin-4-one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-5- [[4-[4-(tri fluoromethyl)phenyl]phenyl]methyl]-2, 3 -dihydro- 1X6, 5-benzothiazepin-4-one;

(3R)-3-amino-5-[(4-chlorophenyl)methyl]-8-fluoro-7-[5-(l- methyl-l-methylsulfonyl-ethyl)-l,2,4- oxadiazol-3-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzothiazepin-4 -one;

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-5- [[4-[5-(trifluoromethyl)-l,2,4-oxadiazol-3-yl]phenyl]methyl] -2,3-dihydro-lX6,5-benzothiazepin-4- one;

(3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4 -oxadiazol-2-yl]-l,l-dioxo-5-[[4-[4- (trifluoromethyl)phenyl]phenyl]methyl]-2,3-dihydro-lX6,5-ben zothiazepin-4-one;

(3R)-3-amino-5-[[4-(4-methoxyphenyl)phenyl]methyl]-7-[5-( l-methyl-l-methylsulfonyl-ethyl)-

1.2.4-oxadiazol-3-yl]-l,l-dioxo-2,3-dihydro-lX6,5-benzoth iazepin-4-one; and (3R)-3-amino-l,l-diketo-7-[5-(l-mesyl-l-methyl-ethyl)-l,3,4- oxadiazol-2-yl]-5-[4-[5- (trifluoromethoxy)-2-pyridyl]benzyl]-2,3-dihydro-lX6,5-benzo thiazepin-4-one.

Processes of manufacturing

Processes for the manufacture of compounds of formula (I), or pharmaceutically acceptable salts thereof, as described herein are also an object of the invention.

The present invention provides a process for the preparation of a compound as described herein, or a pharmaceutically acceptable salt thereof, comprising reacting a compound of formula (IX) wherein Y, R 1 , R 2 and R 4 are as defined herein and PG is an amino protecting group, with a suitable deprotection agent to form said compound of formula (I).

The preparation of compounds of formula (I) of the present invention may be carried out in sequential or convergent synthetic routes. Syntheses of the invention are shown in the following general schemes. The skills required for carrying out the reaction and purification of the resulting products are known to those persons skilled in the art. The substituents and indices used in the following description of the processes have the significance given herein, unless indicated to the contrary.

If one of the starting materials, intermediates or compounds of formula (I) contain one or more functional groups which are not stable or are reactive under the reaction conditions of one or more reaction steps, appropriate protective groups (as described e.g., in “Protective Groups in Organic Chemistry” by T. W. Greene and P. G. M. Wutts, 5th Ed., 2014, John Wiley & Sons, N.Y.) can be introduced before the critical step applying methods well known in the art. Such protective groups can be removed at a later stage of the synthesis using standard methods described in the literature.

If starting materials or intermediates contain stereogenic centers, compounds of formula (I) can be obtained as mixtures of diastereomers or enantiomers, which can be separated by methods well known in the art e.g., chiral HPLC, chiral SFC or chiral crystallization. Racemic compounds can e.g., be separated into their antipodes via diastereomeric salts by crystallization with optically pure acids or by separation of the antipodes by specific chromatographic methods using either a chiral adsorbent or a chiral eluent. It is equally possible to separate starting materials and intermediates containing stereogenic centers to afford diastereomerically/enantiomerically enriched starting materials and intermediates. Using such diastereomerically/enantiomerically enriched starting materials and intermediates in the synthesis of compounds of formula (I) will typically lead to the respective diastereomerically/enantiomerically enriched compounds of formula (I). A person skilled in the art will acknowledge that in the synthesis of compounds of formula (I) - insofar not desired otherwise - an “orthogonal protection group strategy” will be applied, allowing the cleavage of several protective groups one at a time each without affecting other protective groups in the molecule. The principle of orthogonal protection is well known in the art and has also been described in literature (e.g. Barany and R. B. Merrifield, J. Am. Chem. Soc. 1977, 99, 7363; H. Waldmann et al., Angew. Chem. Int. Ed. Engl. 1996, 35, 2056).

A person skilled in the art will acknowledge that the sequence of reactions may be varied depending on reactivity and nature of the intermediates.

In more detail, the compounds of formula (I) can be manufactured by the methods given below, by the methods given in the examples or by analogous methods. Appropriate reaction conditions for the individual reaction steps are known to a person skilled in the art. Also, for reaction conditions described in literature affecting the described reactions see for example: Comprehensive Organic Transformations: A Guide to Functional Group Preparations, 2nd Edition, Richard C. Larock. John Wiley & Sons, New York, NY. 1999). It was found convenient to carry out the reactions in the presence or absence of a solvent. There is no particular restriction on the nature of the solvent to be employed, provided that it has no adverse effect on the reaction or the reagents involved and that it can dissolve the reagents, at least to some extent. The described reactions can take place over a wide range of temperatures, and the precise reaction temperature is not critical to the invention. It is convenient to carry out the described reactions in a temperature range between -78 °C to reflux. The time required for the reaction may also vary widely, depending on many factors, notably the reaction temperature and the nature of the reagents. However, a period of from 0.5 hours to several days will usually suffice to yield the described intermediates and compounds. The reaction sequence is not limited to the one displayed in the schemes, however, depending on the starting materials and their respective reactivity, the sequence of reaction steps can be freely altered.

If starting materials or intermediates are not commercially available or their synthesis not described in literature, they can be prepared in analogy to existing procedures for close analogues or as outlined in the experimental section. The present compounds of formula (I), or their pharmaceutically acceptable salts, may be prepared by a process described below (Scheme 1), together with synthetic methods known in the art of organic chemistry, or modifications and derivatizations that are familiar to those of ordinary skilled in the art. Suitable starting materials for the preparation of compounds of formula (I) are nitro compounds of formula (II) wherein X 2 is F or Cl and X 1 is either already R 1 or a group such as Br, CN or -CChAlkyl which can later be elaborated into R 1 . Compounds of formula (II) can be reacted with suitably protected cysteine derivatives (III) in the presence of a base such as DIPEA at elevated temperatures in a solvent such as 1,2-di chloroethane to obtain compounds of formula (IV). The preferred protecting group (PG) of the cysteine derivative (III) is Boc. The nitro group in formula (IV) compounds can be reduced using iron in the presence of either hydrogen chloride or ammonium chloride at elevated temperatures in a solvent mixture of water and ethanol to obtain compounds of formula (V). Alternatively, this conversion can be achieved by catalytic hydrogenation. Compounds of formula (V) can be cyclized to compounds of formula (VI) using standard amide coupling conditions. Preferably, this cyclization is conducted using 2,4,6-tripropyl-l,3,5,2,4,6-trioxatriphosphinane 2,4,6-trioxide (50% solution in EtOAc) and employing a base such as DIPEA in a solvent such as DMF at room temperature. Reaction of formula (VI) compounds with compounds of formula (VII) wherein Y 1 is Cl, Br, I or a sulfonate group in the presence of a base such as potassium carbonate and if necessary with an additive such as potassium iodide in a solvent such as DMSO or DMF at room temperature affords compounds of formula (VIII). Alternatively, compounds of cormula (VI) can be reacted with compounds of formula (VII) wherein Y 1 is OH in the presence of PPhs with an additive such as DIAD in a solvent such as toluene at elevated temperature to afford compounds of formula (VIII). For compounds of formula (VIII) wherein X 1 is Br or -CO2Alkyl, these groups can be elaborated into substituents R 1 at this stage as described in the schemes below. Compounds of formula (VIII) can then be converted into compounds of formula (IX) wherein Y is S(O) or S(O)2 by reaction with an appropriate amount of an oxidant such as m-CPBA in a solvent such as DCM at room temperature. Final deprotection provides compounds of formula (I). If the N-protecting group (PG) is Boc, typical conditions for this deprotection step include TFA in a solvent such as DCM at room temperature, hydrogen chloride in solvents such as dioxane, diethyl ether or ethyl acetate at room temperature or hexafluoroisopropanol at reflux temperature. Additionally, substituents R 1 and R 4 might contain functional groups that could be either modified prior to the removal of the N-protecting group (PG) or that might require the use of suitable protecting groups during the synthesis. These protecting groups might be removed prior to the removal of the N-protecting group (PG) or they might be removed simultaneously using suitable methods [Peter G. M. Wuts, Greene's protective groups in organic synthesis, 5th edition, Hoboken, N.J.: Wiley-Interscience],

Alternatively, compounds of formula (I) wherein Y is S(O)2 may be prepared as illustrated in scheme 2.

(VI) (X) (XI)

Scheme 2

Compounds of formula (VI) can be converted into compounds of formula (X) upon reaction with an oxidant such as m-CPBA in a solvent such as DCM at room temperature. The reaction of formula (X) compounds with compounds of formula (VII) to afford compounds of formula (XI) and the subsequent conversion into compounds of formula (I) wherein Y is S(O)2 can be achieved using reaction conditions as described for the similar steps in scheme 1. If X 1 is Br,-CO2Alkyl, or CN, these groups can be elaborated into substituents R 1 at any stage of the synthesis (for compounds of formula (VI), (X) or (XI)) using methods as described for the schemes below).

Compounds of formula (I) wherein the 5-membered heteroaryl R 1 is a 1,3,4-oxadiazolyl group may be prepared as illustrated in scheme 3.

(XVI) (I) wherein R 1 is 1 ,3,4-oxadiazolyl Scheme 3

Compounds of formula (VIII) wherein X 1 is CCEMe can be converted into compounds of formula (XII) by reaction with alkali hydroxides such as LiOH, NaOH or KOH in a mixture of solvents such as MeOH, THF and water at room temperature. Compounds of formula (XII) can be reacted with hydrazine hydrate after activation with suitable reagents such as CDI in a solvent such as THF at room temperature to obtain compounds of formula (XIII). Compounds of formula (XIII) can be reacted with carboxylic acids R 10 CO2H using standard amide coupling conditions such as HATU in the presence of a base such as DIPEA in a solvent such as THF at room temperature. The coupling products of formula (XIV) can be cyclized to compounds of formula (XV) using a dehydrating reagent such as Burgess reagent or can be reacted with tosyl chloride in the presence of a base such as DIPEA at room temperature. The conversion of compounds of formula (XV) into compounds of formula (XVI) and the subsequent conversion into compounds of formula (I) can be achieved using reaction conditions as described for the similar steps in scheme 1.

Compounds of formula (I) wherein the 5-membered heteroaryl R 1 is a 1,2,4-oxadiazolyl group may be prepared as illustrated in scheme 4.

(XIX) (I) wherein R 1 is 1 ,2,4-oxadiazolyl

Scheme 4

Compounds of formula (VIII) wherein X 1 is CN can be reacted with hydroxylamine hydrochloride in presence of a base such as potassium carbonate in a solvent such as ethanol at elevated temperatures to obtain amidoxime compounds of formula (XVII wherein R is H). Reaction of compounds of formula (XVII wherein R is H) with carboxylic acids R 10 CO2H using standard amide coupling conditions such as CDI, HATU or EDCI and HOBt in the presence of a base such as DIPEA in a solvent such as acetonitrile, DMF or THF provides coupling intermediates (XVII wherein R is - C(O)R 10 ) which upon heating cyclize to the corresponding compounds of formula (XVIII). Alternatively, coupling intermediates (XVII wherein R is -C(O)R 10 ) can be isolated and the cyclization step can be conducted either by heating in a solvent such as toluene or reaction with TBAOH in a solvent such as THF. The conversion of compounds of formula (XVIII) into compounds of formula (XIX) and the subsequent conversion into compounds of formula (I) can be achieved using reaction conditions as described for the similar steps in scheme 1.

Compounds of formula (I) wherein the 5-membered heteroaryl R 1 is a tetrazolyl group may be prepared as illustrated in scheme 5.

Scheme 5

Compounds of formula (VIII) wherein X 1 is CN can be converted into compounds of formula (XX) by reaction with azide reagents such as azidotrimethylsilane in the presence of tetra-N- butylammonium fluoride trihydrate in toluene at elevated temperature. To obtain compounds of formula (XXI) from compounds of formula (XX), substituents R 10 can be introduced by a variety of methods such as reaction with a reagent R 10 Y (wherein Y is Cl, Br or I) in the presence of a base such as potassium carbonate in a solvent such as DMF at room temperature. Alternatively, compounds of cormula (XX) can be reacted with a reagent R 10 Y (wherein Y is OH) in the presence of PPF13 with an additive such as DIAD in a solvent such as toluene at elevated temperature to afford compounds of formula (XXI). The conversion of compounds of formula (XXI) into compounds of formula (XXII) and the subsequent conversion into compounds of formula (I) can be achieved using reaction conditions as described for the similar steps in scheme 1

Pharmaceutical compositions and administration

Another object of the present invention is a pharmaceutical composition comprising a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

The compounds of formula (I) and their pharmaceutically acceptable salts can be used as medicaments, in the form of pharmaceutical preparations. The pharmaceutical preparations can be administered internally, such as orally (e.g. in the form of tablets, coated tablets, dragees, hard and soft gelatine capsules, solutions, emulsions or suspensions), nasally (e.g. in the form of nasal sprays) or rectally (e.g. in the form of suppositories). However, the administration can also be effected parenterally, such as intramuscularly or intravenously (e.g. in the form of injection solutions). The administration can also be effected topically, e.g. transdermal administration, or in form of eye drops or ear drops.

The compounds of formula (I) and their pharmaceutically acceptable salts can be processed with pharmaceutically inert, inorganic or organic carriers for the production of pharmaceutical preparations, such as tablets, coated tablets, dragees, hard gelatin capsules, injection solutions or topical formulations. Lactose, corn starch or derivatives thereof, talc, stearic acids or salts thereof, and the like can be used, for example, as such carriers for tablets, coated tablets, dragees and hard gelatin capsules.

Suitable carriers for soft gelatin capsules are, for example, vegetable oils, waxes, fats, semi-solid substances and liquid polyols and the like. Depending on the nature of the active substance no carriers are, however, usually required in the case of soft gelatin capsules.

Suitable carriers for the production of solutions and syrups are, for example, water, alcohols, polyols, saccharose, glucose, invert sugar, vegetable oil, etc. Suitable carriers for injection solutions are, for example, water, alcohols, polyols, glycerol, vegetable oils, etc.

Suitable carriers for suppositories are, for example, natural or hardened oils, waxes, fats, semi-liquid or liquid polyols, etc.

Suitable carriers for topical ocular formulations are, for example, cyclodextrins, mannitol or many other carriers and excipients known in the art.

Moreover, the pharmaceutical preparations can contain preservatives, solubilizers, viscosity increasing substances, stabilizers, wetting agents, emulsifiers, sweeteners, colorants, flavorants, salts for varying the osmotic pressure, buffers, masking agents or antioxidants. They can also contain other therapeutically valuable substances.

Medicaments containing a compound of formula (I) or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient are also an object of the present invention, as is a process for their production, which comprises bringing one or more compounds of formula (I) and/or pharmaceutically acceptable salts thereof and, if desired, one or more other therapeutically valuable substances into a galenical administration form together with one or more pharmaceutically acceptable excipients.

The dosage can vary within wide limits and will, of course, have to be adjusted to the individual requirements in each particular case. In general, in the case of oral administration a daily dosage of about 0.1 mg to 20 mg per kg body weight, preferably 0.5 mg to 4 mg per kg body weight (e.g. about 300 mg per person), divided into preferably 1-3 individual doses, which can consist, for example, of the same amounts, should be appropriate. In the case of topical administration, the formulation can contain 0.001% to 15% by weight of medicament and the required dose, which can be between 0.1 and 25 mg, and can be administered either by single dose per day or per week, or by multiple doses (2 to 4) per day, or by multiple doses per week. It will, however, be clear that the upper or lower limit given herein can be exceeded when this is shown to be indicated.

The pharmaceutical composition according to the invention may be prepared as follows.

Preparation of pharmaceutical compositions comprising compounds of the invention Tablet Formulation (Wet Granulation)

Ingredient mg/tablet

5 25 100 500

1) Compound of formula (I) 5 25 100 500

2) Lactose Anhydrous DTG 125 105 30 150

3) Sta-Rx 1500 6 6 6 30

4) Microcrystalline Cellulose 30 30 30 150

5) Magnesium Stearate 1 1 1 1

Total 167 167 167 831

Manufacturing Procedure:

1. Mix ingredients 1, 2, 3 and 4 and granulate with purified water.

2. Dry the granules at 50°C. 3. Pass the granules through suitable milling equipment.

4. Add ingredient 5 and mix for three minutes; compress on a suitable press.

Capsule Formulation

Ingredient mg/capsule

5 25 100 500

1) Compound of formula (I) 5 25 100 500

2) Hydrous Lactose 159 123 148

3) Corn Starch 25 35 40 70

4) Talc 10 15 10 25 5) Magnesium Stearate 1 2 2 5

Total 200 200 300 600

Manufacturing Procedure:

1. Mix ingredients 1, 2 and 3 in a suitable mixer for 30 minutes.

2. Add ingredients 4 and 5 and mix for 3 minutes.

3. Fill into a suitable capsule. Injection Solutions

Ingredient mg/injection solution

Compound of formula I 3

Polyethylene Glycol 400 150 acetic acid q.s. ad pH 5.0 water for injection solutions ad 1.0 ml

Manufacturing Procedure:

A compound of formula (I) is dissolved in a mixture of Polyethylene Glycol 400 and water for injection (part). The pH is adjusted to 5.0 by acetic acid. The volume is adjusted to 1.0 ml by addition of the residual amount of water. The solution is filtered, filled into vials using an appropriate overage and sterilized.

Indications

The compounds of formula (I) can be used in an effective amount to treat a subject, in particular a human, affected by cancer.

In one aspect, the present invention provides a compound of formula (I) described herein, or a pharmaceutically acceptable thereof, for use as a therapeutically active substance. In a further aspect, the present invention provides a compound of formula (I) as described herein, or a pharmaceutically acceptable thereof, for use in the treatment, prevention and/or delay of progression of cancer.

In a further aspect, the present invention provides the use of a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, for the treatment, prevention and/or delay of progression of cancer.

In a further aspect, the present invention provides the use of a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof, for the preparation of a medicament for the treatment, prevention and/or delay of progression of cancer.

In a further aspect, the present invention provides a method for the treatment, prevention and/or delay of progression of cancer, which method comprises administering a therapeutically effective amount of a compound of formula (I) as described herein, or a pharmaceutically acceptable salt thereof.

By the term “treatment” or "treating" and grammatical variations thereof as used herein, is meant therapeutic therapy. In reference to a particular condition, treating means: (1) to ameliorate the condition or one or more of the biological manifestations of the condition, (2) to interfere with (a) one or more points in the biological cascade that leads to or is responsible for the condition or (b) one or more of the biological manifestations of the condition, (3) to alleviate one or more of the symptoms, effects or side effects associated with the condition or treatment thereof, or (4) to slow the progression of the condition or one or more of the biological manifestations of the condition. Prophylactic therapy using the methods and/or compositions of the invention is also contemplated. The skilled artisan will appreciate that "prevention" is not an absolute term. In medicine, "prevention" is understood to refer to the prophylactic administration of a drug to substantially diminish the likelihood or severity of a condition or biological manifestation thereof, or to delay the onset of such condition or biological manifestation thereof. Prophylactic therapy is appropriate, for example, when a subject is considered at high risk for developing cancer, such as when a subject has a strong family history of cancer or when a subject has been exposed to a carcinogen. As immunotherapeutic agents acting on immune cells rather than directly acting on the cancer cells, the present disclosure could also be foreseen for the use as anti-cancer vaccines. This also comprises approaches in which immune cells are cultured and manipulated ex vivo and the herein disclosed molecules are used as a way of conferring co-stimulation of the ex vivo manipulated cells.

In one embodiment, the cancer is a hematologic cancer such as lymphoma, a leukemia or a myeloma. A hematologic cancer contemplated herein includes, but is not limited to, one or more leukemias such as B-cell acute lymphoid leukemia ("BALL"), T-cell acute lymphoid leukemia ("TALL"), acute lymphoid leukemia (ALL); one or more chronic leukemias including but not limited to chronic myelogenous leukemia (CML) and chronic lymphocytic leukemia (CLL); additional hematologic cancers or hematologic conditions including, but not limited to B cell prolymphocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitt's lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, mucosa-associated lymphoid tissue (MALT) lymphoma, mantle cell lymphoma, Marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin's lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, Waldenstrom macroglobulinemia, and "preleukemia," which are a diverse collection of hematological conditions united by ineffective production (or dysplasia) of myeloid blood cells.

In a further embodiment, the cancer is a non-hematologic cancer such as a sarcoma, a carcinoma, or a melanoma. A non-hematologic cancer contemplated herein includes, but is not limited to, a neuroblastoma, renal cell carcinoma, colon cancer, colorectal cancer, breast cancer, epithelial squamous cell cancer, melanoma, stomach cancer, brain cancer, lung cancer (e.g. nonsmall cell lung cancer - NSCLC), pancreatic cancer, cervical cancer, ovarian cancer, liver cancer, bladder cancer, prostate cancer, testicular cancer, thyroid cancer, uterine cancer, adrenal cancer and head and neck cancer.

Co-administration of compounds of formula (I) and other agents The compounds of formula (I) or salts thereof or a compound disclosed herein or a pharmaceutically acceptable salt thereof may be employed alone or in combination with other agents for treatment. For example, the second agent of the pharmaceutical combination formulation or dosing regimen may have complementary activities to the compound of formula (I) such that they do not adversely affect each other. The compounds may be administered together in a unitary pharmaceutical composition or separately. In one embodiment a compound or a pharmaceutically acceptable salt can be co-administered with a cytotoxic agent to treat proliferative diseases and cancer.

The term "co-administering" refers to either simultaneous administration, or any manner of separate sequential administration, of a compound of formula (I) or a salt thereof or a compound disclosed herein or a pharmaceutically acceptable salt thereof and a further active pharmaceutical ingredient or ingredients, including cytotoxic agents and radiation treatment. If the administration is not simultaneous, the compounds are administered in a close time proximity to each other. Furthermore, it does not matter if the compounds are administered in the same dosage form, e.g. one compound may be administered topically and another compound may be administered orally.

Typically, any agent that has anti-cancer activity may be co-administered. Examples of such agents can be found in Cancer Principles and Practice of Oncology by V.T. Devita and S. Heilman (editors), 6th edition (February 15, 2001), Lippincott Williams & Wilkins Publishers. A person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the disease involved.

In one aspect, the present invention provides a pharmaceutical composition described herein, further comprising an additional therapeutic agent.

In one embodiment, said additional therapeutic agent is a chemotherapeutic agent.

In one embodiment, said additional therapeutic agent is a cytotoxic agent.

In one embodiment, said additional therapeutic agent is an immuno-oncology agent.

The term "cytotoxic agent” as used herein refers to a substance that inhibits or prevents a cellular function and/or causes cell death or destruction. Cytotoxic agents include, but are not limited to, radioactive isotopes (At 211 , 1 131 , 1 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 and radioactive isotopes of Lu); chemotherapeutic agents; growth inhibitory agents; enzymes and fragments thereof such as nucleolytic enzymes; and toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof.

Exemplary cytotoxic agents can be selected from anti -microtubule agents, platinum coordination complexes, alkylating agents, antibiotic agents, topoisomerase II inhibitors, antimetabolites, topoisomerase I inhibitors, hormones and hormonal analogues, signal transduction pathway inhibitors, non-receptor tyrosine kinase angiogenesis inhibitors, immunotherapeutic agents, proapoptotic agents, inhibitors of LDH-A; inhibitors of fatty acid biosynthesis; cell cycle signaling inhibitors; HD AC inhibitors, proteasome inhibitors; and inhibitors of cancer metabolism.

"Chemotherapeutic agent" includes chemical compounds useful in the treatment of cancer. Examples of chemotherapeutic agents include erlotinib (TARCEVA®, Genentech/OSI Pharm.), bortezomib (VELCADE®, Millennium Pharm.), disulfiram , epigallocatechin gallate , salinosporamide A, carfilzomib, 17-AAG(geldanamycin), radicicol, lactate dehydrogenase A (LDH-A), fulvestrant (FASLODEX®, AstraZeneca), sunitib (SUTENT®, Pfizer/Sugen), letrozole (FEMARA®, Novartis), imatinib mesylate (GLEEVEC®., Novartis), finasunate (VATALANIB®, Novartis), oxaliplatin (ELOXATIN®, Sanofi), 5-FU (5 -fluorouracil), leucovorin, Rapamycin (Sirolimus, RAPAMUNE®, Wyeth), Lapatinib (TYKERB®, GSK572016, Glaxo Smith Kline), Lonafamib (SCH 66336), sorafenib (NEXAVAR®, Bayer Labs), gefitinib (IRESSA®, AstraZeneca), AG1478, alkylating agents such as thiotepa and CYTOXAN® cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; Eiziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, triethylenephosphoramide, tri ethylenethiophosphorami de and trimethylomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including topotecan and irinotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogs); cryptophycins (particularly cryptophycin I and cryptophycin 8); adrenocorticosteroids (including prednisone and prednisolone); cyproterone acetate; 5a-reductases including finasteride and dutasteride); vorinostat, romidepsin, panobinostat, valproic acid, mocetinostat dolastatin; aldesleukin, talc duocarmycin (including the synthetic analogs, KW-2189 and CBI- TM I); eleutherobin; pancrati statin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlomaphazine, chlorophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosoureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin, especially calicheamicin yil and calicheamicin coll (Angew Chem. Inti. Ed. Engl. 1994 33: 183-186); dynemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, ADRIAMYCIN® (doxorubicin), morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino- doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, porfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5 -fluorouracil (5- FU); folic acid analogs such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfomithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidamnol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK® polysaccharide complex (JHS Natural Products, Eugene, Oreg.); razoxane; rhizoxin; sizofuran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxoids, e.g., TAXOL (paclitaxel; Bristol- Myers Squibb Oncology, Princeton, N.J.), ABRAXANE® (Cremophor-free), albumin- engineered nanoparticle formulations of paclitaxel (American Pharmaceutical Partners, Schaumberg, 111.), and TAXOTERE® (docetaxel, doxetaxel; Sanofi -Aventis); chloranmbucil; GEMZAR® (gemcitabine); 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; etoposide (VP- 16); ifosfamide; mitoxantrone; vincristine; NAVELBINE® (vinorelbine); novantrone; teniposide; edatrexate; daunomycin; aminopterin; capecitabine (XELODA®); ibandronate; CPT-I I; topoisomerase inhibitor RFS 2000; difluoromethyl ornithine (DMFO); retinoids such as retinoic acid; and pharmaceutically acceptable salts, acids and derivatives of any of the above.

Chemotherapeutic agent also includes (i) anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including NOLVADEX®; tamoxifen citrate), raloxifene, droloxifene, iodoxyfene , 4-hydroxytamoxifen, trioxifene, keoxifene,LYl 17018, onapristone, and FARESTON® (toremifine citrate); (ii) aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE® (megestrol acetate), AROMASIN® (exemestane; Pfizer), formestanie, fadrozole, RIVISOR® (vorozole), FEMARA® (letrozole; Novartis), and ARIMIDEX® (anastrozole; AstraZeneca); (iii) anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide and goserelin; buserelin, tripterelin, medroxyprogesterone acetate, diethylstilbestrol, premarin, fluoxymesterone, all transretionic acid, fenretinide, as well as troxacitabine (a 1,3-di oxolane nucleoside cytosine analog); (iv) protein kinase inhibitors; (v) lipid kinase inhibitors; (vi) antisense oligonucleotides, particularly those which inhibit expression of genes in signaling pathways implicated in aberrant cell proliferation, such as, for example, PKC-alpha, Ralf and H-Ras; (vii) ribozymes such as VEGF expression inhibitors (e.g., ANGIOZYME®) and HER2 expression inhibitors; (viii) vaccines such as gene therapy vaccines, for example, ALLOVECTIN®, LEUVECTIN®, and VAXID®; PROLEUKIN®, rIL-2; a topoisomerase I inhibitor such as LURTOTECAN®; ABARELIX® rmRH; and (ix) pharmaceutically acceptable salts, acids and derivatives of any of the above.

Chemotherapeutic agent also includes antibodies such as alemtuzumab (Campath), bevacizumab (AVASTIN®, Genentech); cetuximab (ERBITUX®, Imclone); panitumumab (VECTIBIX®, Amgen), rituximab (RITUXAN®, Genentech/Biogen Idee), pertuzumab (OMNITARG®, 2C4, Genentech), trastuzumab (HERCEPTIN®, Genentech), tositumomab (Bexxar, Corixia), and the antibody drug conjugate, gemtuzumab ozogamicin (MYLOTARG®, Wyeth). Additional humanized monoclonal antibodies with therapeutic potential as agents in combination with the compounds of the invention include: apolizumab, aselizumab, atlizumab, bapineuzumab, bivatuzumab mertansine, cantuzumab mertansine, cedelizumab, certolizumab pegol, cidfusituzumab, cidtuzumab, daclizumab, eculizumab, efalizumab, epratuzumab, erlizumab, felvizumab, fontolizumab, gemtuzumab ozogamicin, inotuzumab ozogamicin, ipilimumab, labetuzumab, lintuzumab, matuzumab, mepolizumab, motavizumab, motovizumab, natalizumab, nimotuzumab, nolovizumab, numavizumab, ocrelizumab, omalizumab, palivizumab, pascolizumab, pecfusituzumab, pectuzumab, pexelizumab, ralivizumab, ranibizumab, reslivizumab, reslizumab, resyvizumab, rovelizumab, ruplizumab, sibrotuzumab, siplizumab, sontuzumab, tacatuzumab tetraxetan, tadocizumab, talizumab, tefibazumab, tocilizumab, toralizumab, tucotuzumab celmoleukin, tucusituzumab, umavizumab, urtoxazumab, ustekinumab, visilizumab, and the anti-interleukin- 12 (ABT-874/J695, Wyeth Research and Abbott Laboratories) which is a recombinant exclusively human-sequence, full- length IgGi X antibody genetically modified to recognize interleukin- 12 p40 protein.

Chemotherapeutic agent also includes “EGFR inhibitors,” which refers to compounds that bind to or otherwise interact directly with EGFR and prevent or reduce its signaling activity, and is alternatively referred to as an “EGFR antagonist.” Examples of such agents include antibodies and small molecules that bind to EGFR. Examples of antibodies which bind toEGFR include MAb 579 (ATCC CRL HB 8506), MAb 455 (ATCC CRL HB8507), MAb 225 (ATCC CRL 8508), MAb 528 (ATCC CRL 8509) (see, US Patent No. 4,943, 533, Mendelsohn et al.) and variants thereof, such as chimerized 225 (C225 or Cetuximab; ERBUTIX®) and reshaped human 225 (H225) (see, WO 96/40210, Imclone Systems Inc.); IMC-11F8, a fully human, EGFR-targeted antibody (Imclone); antibodies that bind type II mutant EGFR (US Patent No. 5,212,290); humanized and chimeric antibodies that bind EGFR as described in US Patent No. 5,891,996; and human antibodies that bind EGFR, such as ABX-EGF or Panitumumab (see WO98/50433, Abgenix/ Amgen); EMD 55900 (Stragliotto et al. Eur. J. Cancer 32A:636-640 (1996)); EMD7200 (matuzumab) a humanized EGFR antibody directed against EGFR that competes with both EGF and TGF-alpha for EGFR binding (EMD/Merck); human EGFR antibody, HuMax-EGFR (GenMab); fully human antibodies known as El.l, E2.4, E2.5, E6.2, E6.4, E2.ll, E6. 3 and E7.6. 3 and described in US 6,235,883; MDX-447 (Medarex Inc); and mAb 806 or humanized mAb 806 (Johns et al, J. Biol. Chem. 279(29):30375-30384 (2004)). The anti-EGFR antibody may be conjugated with a cytotoxic agent, thus generating an immunoconjugate (see, e.g., EP659,439A2, Merck Patent GmbH). EGFR antagonists include small molecules such as compounds described in US Patent Nos: 5,616,582, 5,457,105,5,475,001, 5,654,307, 5,679,683, 6,084,095, 6,265,410, 6,455,534, 6,521,620, 6,596,726, 6,713,484, 5,770,599, 6,140,332, 5,866,572, 6,399,602, 6,344,459, 6,602,863, 6,391,874, 6,344,455, 5,760,041, 6,002,008, and 5,747,498, as well as the following PCT publications: W098/14451, W098/50038, W099/09016, and WO99/24037. Particular small molecule EGFRantagonists include OSI-774 (CP-358774, erlotinib, TARCEVA® Genentech/O SI Pharmaceuticals); PD 183805 (Cl 1033, 2-propenamide, N-[4-[(3-chloro-4- fluorophenyl)amino]-7-[3-(4-morpholinyl)propoxy]-6-quinazoli nyl]-, dihydrochloride, Pfizer Inc.); ZD1839, gefitinib (IRESSA®) 4-(3’-Chloro-4’-fluoroanilino)-7-methoxy-6-(3- morpholinopropoxy)quinazoline, AstraZeneca); ZM 105180 ((6-amino-4-(3-methylphenyl- amino)-quinazoline, Zeneca); BIBX-1382 (N8-(3-chloro-4-fluoro-phenyl)-N2-(l-methyl- piperidin-4-yl)-pyrimido[5,4-d]pyrimidine-2,8-diamine, Boehringer Ingelheim); PKI-166 ((R)- 4- [4- [(I -phenyl ethyl)amino] -1 H-pyrrolo[2,3 -d]pyrimidin-6-yl] -phenol); (R)-6-(4- hydroxyphenyl)-4-[(l-phenylethyl)amino]-7H-pyrrolo[2,3-d]pyr imidine); CL-387785 (N-[4-[(3- bromophenyl)amino]-6-quinazolinyl] -2-butynamide); EKB-569 (N- [4- [(3 -chloro-4- fluorophenyl)amino]-3-cyano-7-ethoxy-6-quinolinyl]-4-(dimeth ylamino)-2-butenamide) (Wyeth); AG1478 (Pfizer); AG1571 (SU 5271; Pfizer); dual EGFR/HER2 tyrosine kinase inhibitors such as lapatinib (TYKERB®, GSK572016 or N-[3-chloro-4-[(3 fluorophenyl)methoxy]phenyl]-6[5[[[2methylsulfonyl)ethyl]ami no]methyl]-2-furanyl]-4- quinazolinamine).

Chemotherapeutic agents also include “tyrosine kinase inhibitors” including the EGFR- targeted drugs noted in the preceding paragraph; small molecule FIER2 tyrosine kinase inhibitor such as TAK165 available from Takeda; CP-724,714, an oral selective inhibitor of the ErbB2 receptor tyrosine kinase (Pfizer and OSI); dual-HER inhibitors such as EKB-569 (available from Wyeth) which preferentially binds EGFR but inhibits both HER2 and EGFR- overexpressing cells; lapatinib (GSK572016; available from Glaxo-SmithKline), an oral HER2 and EGFR tyrosine kinase inhibitor; PKI-166 (available from Novartis); pan-HER inhibitors such as canertinib (CI-1033; Pharmacia); Raf-I inhibitors such as antisense agent ISIS-5132 available from ISIS Pharmaceuticals which inhibit Raf-I signaling; non-HER targeted TK inhibitors such as imatinib mesylate (GLEEVEC®, available from Glaxo SmithKline); multitargeted tyrosine kinase inhibitors such as sunitinib (SUTENT®, available from Pfizer); VEGF receptor tyrosine kinase inhibitors such as vatalanib (PTK787/ZK222584, available from Novartis/Schering AG); MAPK extracellular regulated kinase I inhibitor Cl-1040 (available from Pharmacia); quinazolines, such as PD 153035, 4-(3-chloroanilino) quinazoline; pyridopyrimi dines; pyrimidopyrimidines; pyrrol opyrimidines, such as CGP 59326, CGP 60261 and CGP 62706; pyrazolopyrimidines, 4-(phenylamino)-7H-pyrrolo[2,3-d] pyrimidines; curcumin (diferuloyl methane, 4,5-bis (4-fluoroanilino)phthalimide); tyrphostines containing nitrothiophene moi eties; PD-0183805 (Wamer-Lamber); antisense molecules (e.g. those that bind to HER-encoding nucleic acid); quinoxalines (US Patent No. 5,804,396); tryphostins (US Patent No. 5,804,396); ZD6474 (Astra Zeneca); PTK-787 (Novartis/Schering AG); pan-HER inhibitors such as CI- 1033 (Pfizer); Affinitac (ISIS 3521; Isis/Lilly); imatinib mesylate (GLEEVEC®); PKI 166 (Novartis); GW2016 (Glaxo SmithKline); CI-1033 (Pfizer); EKB-569 (Wyeth); Semaxinib (Pfizer); ZD6474 (AstraZeneca); PTK-787 (Novartis/Schering AG); INC- IC1 I (Imclone), rapamycin (sirolimus, RAPAMUNE®); or as described in any of the following patent publications: US Patent No. 5,804,396; WO 1999/09016 (American Cyanamid); WO 1998/43960 (American Cyanamid); WO 1997/38983 (Warner Lambert); WO 1999/06378 (Warner Lambert); WO 1999/06396 (Warner Lambert); WO 1996/30347 (Pfizer, Inc); WO 1996/33978 (Zeneca); WO 1996/3397 (Zeneca) and WO 1996/33980 (Zeneca).

Chemotherapeutic agents also include dexamethasone, interferons, colchicine, metoprine, cyclosporine, amphotericin, metronidazole, alemtuzumab, alitretinoin, allopurinol, amifostine, arsenic trioxide, asparaginase, BCG live, bevacuzimab, bexarotene, cladribine, clofarabine, darbepoetin alfa, denileukin, dexrazoxane, epoetin alfa, elotinib, filgrastim, histrelin acetate, ibritumomab, interferon alfa-2a, interferon alfa-2b, lenalidomide, levamisole, mesna, methoxsalen, nandrolone, nelarabine, nofetumomab, oprelvekin, palifermin, pamidronate, pegademase, pegaspargase, pegfilgrastim, pemetrexed disodium, plicamycin, porfimer sodium, quinacrine, rasburicase, sargramostim, temozolomide, VM-26, 6-TG, toremifene, tretinoin, ATRA, valrubicin, zoledronate, and zoledronic acid, and pharmaceutically acceptable salts thereof.

Chemotherapeutic agents also include hydrocortisone, hydrocortisone acetate, cortisone acetate, tixocortol pivalate, triamcinolone acetonide, triamcinolone alcohol, mometasone, amcinonide, budesonide, desonide, fluocinonide, fluocinolone acetonide, betamethasone, betamethasone sodium phosphate, dexamethasone, dexamethasone sodium phosphate, fluocortolone, hydrocorti sone- 17-butyrate, hydrocortisone-17-valerate, aclometasone dipropionate, betamethasone valerate, betamethasone dipropionate, prednicarbate, clobetasone- 17-butyrate, clobetasol-17-propionate, fluocortolone caproate, fluocortolone pivalate and fluprednidene acetate; immune selective anti-inflammatory peptides (ImSAIDs) such as phenylalanine-glutamine-glycine (PEG) and its D-isomeric form (feG) (IMULAN BioTherapeutics, LLC); anti-rheumatic drugs such as azathioprine, ciclosporin (cyclosporine A), D-penicillamine, gold salts, hydroxychloroquine, leflunomideminocycline, sulfasalazine, tumor necrosis factor alpha (TNFa) blockers such as etanercept (Enbrel), infliximab (Remicade), adalimumab (Humira), certolizumab pegol (Cimzia), golimumab (Simponi), Interleukin I (IL-I) blockers such as anakinra (Kineret), T cell costimulation blockers such as abatacept (Orencia), Interleukin 6 (IL-6) blockers such as tocilizumab (ACTEMERA®); Interleukin 13 (IL-13) blockers such as lebrikizumab; Interferon alpha (IFN) blockers such as Rontalizumab; Beta 7 integrin blockers such as rhuMAb Beta7; IgE pathway blockers such as Anti-Ml prime; Secreted homotrimeric LTa3 and membrane bound heterotrimer LTa I/|32 blockers such as Anti-lymphotoxin alpha (LTa); radioactive isotopes (e.g., At 211 , 1 131 , 1 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 and radioactive isotopes of Lu); miscellaneous investigational agents such as thioplatin, PS-341, phenylbutyrate, ET-18- OCH3, or famesyl transferase inhibitors (L-739749, L-744832); polyphenols such as quercetin, resveratrol, piceatannol, epigallocatechine gallate, theaflavins, flavanols, procyanidins, betulinic acid and derivatives thereof; autophagy inhibitors such as chloroquine; delta-9-tetrahydrocannabinol (dronabinol, MARINOL®); beta-lapachone; lapachol; colchicines; betulinic acid; acetyl camptothecin, scopolectin, and 9-aminocamptothecin); podophyllotoxin; tegafur (UFTORAL®); bexarotene (TARGRETIN®); bisphosphonates such as clodronate (for example, BONEFOS® or OSTAC®), etidronate (DIDROCAL®), NE-58095, zoledronic acid/zoledronate (ZOMETA®), alendronate (FOSAMAX®), pamidronate (AREDIA®), tiludronate (SKELID®), or risedronate (ACTQNEL®); and epidermal growth factor receptor (EGF-R); vaccines such as THERATOPE® vaccine; perifosine, COX-2 inhibitor (e.g. celecoxib or etoricoxib), proteosome inhibitor (e.g. PS341); CCI-779; tipifamib (R11577); orafenib, ABT510; Bcl-2 inhibitor such as oblimersen sodium (GENASENSE®); pixantrone; famesyltransferase inhibitors such as lonafamib (SCH 6636, SARAS AR™); and pharmaceutically acceptable salts, acids or derivatives of any of the above; as well as combinations of two or more of the above such as CHOP, an abbreviation for a combined therapy of cyclophosphamide, doxorubicin, vincristine, and prednisolone; and FOLFOX, an abbreviation for a treatment regimen with oxaliplatin (ELOXATIN™) combined with 5-FU and leucovorin.

In another embodiment, compounds of formula (I) can be co-formulated with an immunooncology agent. Immuno-oncology agents include, for example, a small molecule drug, antibody, or other biologic or small molecule. Examples of biologic immuno-oncology agents include, but are not limited to, cancer vaccines, antibodies, and cytokines. In one aspect, the antibody is a monoclonal antibody. In another aspect, the monoclonal antibody is humanized or human. In another aspect, the antibody is a bispecific antibody.

In one aspect, the immuno-oncology agent is (i) an agonist of a stimulatory (including a co-stimulatory) receptor or (ii) an antagonist of an inhibitory (including a co-inhibitory) signal on T cells, both of which result in amplifying antigen-specific T cell responses (often referred to as immune checkpoint regulators).

Certain of the stimulatory and inhibitory molecules are members of the immunoglobulin super family (IgSF). One important family of membrane-bound ligands that bind to costimulatory or co-inhibitory receptors is the B7 family, which includes B7-1, B7-2, B7-H1 (PD- Ll), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6. Another family of membrane bound ligands that bind to co-stimulatory or co-inhibitory receptors is the TNF family of molecules that bind to cognate TNF receptor family members, which includes CD40 and CD40L, OX-40, OX-40L, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/ Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fnl4, TWEAK, BAFFR, EDAR, XEDAR, TACI, APRIL, BCMA, LTfiR, LIGHT, DcR3, HVEM, VEGI/TL1A, TRAMP/DR3, EDAR, EDAI, XEDAR, EDA2,TNFR1, Lymphotoxin a/TNPp, TNFR2, TNF a, LT R, Lymphotoxin a ip2, FAS, FASL, RELT, DR6, TROY, NGFR.

In one aspect, T cell responses can be stimulated by a combination of a compound of formula (I) and one or more of (i) an antagonist of a protein that inhibits T cell activation (e.g., immune checkpoint inhibitors) such as CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, TIM-3, Galectin 9, CEACAM-1, BTLA, CD69, Galectin-1, TIGIT, CD113, GPR56, VISTA, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1, and TIM-4, and (ii) an agonist of a protein that stimulates T cell activation such as B7-1, B7-2, CD28, 4-1BB (CD137), 4-1BBL, ICOS, ICOS-L, 0X40, OX40L, GITR, GITRL, CD70, CD27, CD40, DR3 and CD28H.

Other agents that can be combined with compounds of formula (I) for the treatment of cancer include antagonists of inhibitory receptors on NK cells or agonists of activating receptors on NK cells. For example, compounds of formula (I) can be combined with antagonists of KIR, such as lirilumab.

Yet other agents for combination therapies include agents that inhibit or deplete macrophages or monocytes, including but not limited to CSF-1R antagonists such as CSF-1R antagonist antibodies including RG7155 or FPA-008.

In another aspect, compounds of formula (I) can be used with one or more of agonistic agents that ligate positive costimulatory receptors, blocking agents that attenuate signaling through inhibitory receptors, antagonists, and one or more agents that increase systemically the frequency of anti-tumor T cells, agents that overcome distinct immune suppressive pathways within the tumor microenvironment (e.g., block inhibitory receptor engagement (e.g., PD-L1/PD- 1 interactions), deplete or inhibit Tregs (e.g., using an anti-CD25 monoclonal antibody (e.g., daclizumab) or by ex vivo anti-CD25 bead depletion), inhibit metabolic enzymes such as IDO, or reverse/prevent T cell anergy or exhaustion) and agents that trigger innate immune activation and/or inflammation at tumor sites.

In some embodiments, the immuno-oncology agent is a CTLA-4 antagonist, such as an antagonistic CTLA-4 antibody. Suitable CTLA-4 antibodies include, for example, YERVOY (ipilimumab) or tremelimumab. In another aspect, the immuno-oncology agent is a PD-1 antagonist, such as an antagonistic PD-1 antibody. Suitable PD-1 antibodies include, for example, OPDIVO (nivolumab), KEYTRUDA (pembrolizumab), or MEDI-0680 (AMP-514; WO2012/145493). The immuno-oncology agent may also include pidilizumab (CT-011), though its specificity for PD-1 binding has been questioned. Another approach to target the PD-1 receptor is the recombinant protein composed of the extracellular domain of PD-L2 (B7-DC) fused to the Fc portion of IgGl, called AMP-224

In another aspect, the immuno-oncology agent is a PD-L1 antagonist, such as an antagonistic PD-L1 antibody. Suitable PD-L1 antibodies include, for example, TECENTRIQ (atezolizumab) (RG7446; W02010/077634), durvalumab (MEDI4736), BMS-936559 (W02007/005874), and MSB0010718C (WO2013/79174).

In another aspect, the immuno-oncology agent is a LAG-3 antagonist, such as an antagonistic LAG-3 antibody. Suitable LAG3 antibodies include, for example, BMS-986016 (W02010/19570, W02014/08218), or IMP-731 or IMP-321 (W02008/132601,

WO2009/44273).

In another aspect, the immuno-oncology agent is a CD137 (4- IBB) agonist, such as an agonistic CD137 antibody. Suitable CD137 antibodies include, for example, urelumab and PF- 05082566 (WO2012/32433).

In another aspect, the immuno-oncology agent is a GITR agonist, such as an agonistic GITR antibody. Suitable GITR antibodies include, for example, BMS-986153, BMS-986156, TRX-518 (W02006/105021, W02009/009116) and MK-4166 (WO2011/028683).

In another aspect, the immuno-oncology agent is an IDO antagonist. Suitable IDO antagonists include, for example, INCB-024360 (W02006/122150, WO2007/75598, WO2008/36653, WO2008/36642), indoximod, or NLG-919 (W02009/73620,

W02009/1156652, WO2011/56652, WO2012/142237).

In another aspect, the immuno-oncology agent is an 0X40 agonist, such as an agonistic 0X40 antibody. Suitable 0X40 antibodies include, for example, MEDL6383 or MEDL6469. In another aspect, the immuno-oncology agent is an OX40L antagonist, such as an antagonistic 0X40 antibody. Suitable OX40L antagonists include, for example, RG-7888 (WO06/029879). In another aspect, the immuno-oncology agent is a CD40 agonist, such as an agonistic CD40 antibody. In yet another embodiment, the immuno-oncology agent is a CD40 antagonist, such as an antagonistic CD40 antibody. Suitable CD40 antibodies include, for example, lucatumumab or dacetuzumab.

In another aspect, the immuno-oncology agent is a CD27 agonist, such as an agonistic CD27 antibody. Suitable CD27 antibodies include, for example, varlilumab.

In another aspect, the immuno-oncology agent is MGA271 (to B7H3) (W02011/109400).

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

1) Preparative examples

All reaction examples and intermediates were prepared under an argon atmosphere if not specified otherwise.

1.1) General procedures

• Reduction of nitro-group: General procedure 2

To a solution of an intermediate of formula (IV) (23 mmol) in EtOH (112 mL) and water (18.7 mL) was added IN aqueous hydrogen chloride solution (2.3 mL). The reaction mixture was heated to 50°C and iron (8.99 g, 161 mmol) was added to the hot and stirred solution. The temperature was raised to 80°C and stirred for 3.5 h. The reaction mixture was cooled to RT and filtered over celite, washed with ethyl acetate and concentrated under reduced pressure. The desired product (V) was used crude in the next step.

• Cyclization: General procedure 3 To a solution of of an intermediate of formula (V) (5.74 mmol) in DMF (15 mL) were added DIPEA (1.85 g, 2.51 mL, 14.4 mmol, 2.5 eq) and 2,4,6-tripropyl-l,3,5,2,4,6-trioxatriphosphinane 2,4,6- trioxide 50% solution in EtOAc (7.31 g, 6.76 mL). The reaction mixture was stirred at RT for 3 h. The reaction mixture was quenched with water and extracted twice with DCM, washed with IM aqueous NaOH solution, IM aqueous HC1 solution and sat. aqueous NaCl solution, dried over Na2SO4, filtered and concentrated in vacuo. The desired product (VI) was used crude in the next step or was purified by flash column chromatography on silica gel or by reverse phase preparative HPLC.

• Alkylation: General procedure 4

To a solution of an intermediate of formula (VI) (2.74 mmol) in DMSO (10 mL) were added at RT potassium carbonate (1.14 g, 8.23 mmol), potassium iodide (228 mg, 1.37 mmol) and a reagent of formula (VII) (3.29 mmol). The reaction was stirred at RT for 2 h, quenched with water and extracted twice with DCM. The combined organic layers were washed with water, saturated aqueous NaCl solution, dried over Na2SO4, filtered and the solvent was evaporated under reduced pressure. The desired product (VIII) was used crude in the next step or was purified by flash column chromatography on silica gel or by reverse phase preparative HPLC.

• Oxidation: General procedure 5

A solution of an intermediate of formula (VIII) (2.74 mmol) and m-CPBA (1.18 g, 6.85 mmol) in DCM (10 mL) was stirred at RT for 1 day. The reaction was diluted with ethyl acetate and THF, washed with 2N aqueous sodium hydroxide solution, IN aqueous HC1 solution and saturated aqueous NaCl solution, dried over Na2SO4, filtered and the solvent was removed under reduced pressure. The desired product (IX) was used crude in the next step or was purified by flash column chromatography on silica gel or by reverse phase preparative HPLC.

• Boc de-protection: General procedure 6a

To a solution of an intermediate of formula (IX) (0.250 mmol) in EtOAc (4 mL) was added HClZEtOAc (4.0 mL, 16 mmol, 63 eq) at 0 °C. The reaction mixture was stirred at 20°C for 3 h and then concentrated in vacuo. The remaining residue was purified by prep-HPLC and dried by lyophilization to obtain the desired product (I).

• Boc de-protection: General procedure 6b A solution of an intermediate of formula (IX) (22.7 pmol) in l,l,l,3,3,3-hexafluoropropan-2-ol (1.5 mL) was stirred at reflux for 5 days. The solvent was evaporated and the remaining residue was dried under high vacuum to yield desired product (I).

• Boc de-protection: General procedure 6c

To a solution of an intermediate of formula (IX) (0.250 mmol) in l,l,l,3,3,3-hexafluoropropan-2-ol (4 mL) was added HCl/dioxane or HCl/Et2O (0.5 mmol, 2 eq) at 0 °C. The reaction mixture was stirred at 20°C for 2 h. The solvent was evaporated and the resulting solid taken up in DCM and concentrated again to remove trace l,l,l,3,3,3-hexafluoropropan-2-ol. This process was repeated two times followed by drying under high vacuum to obtain the desired product (I).

• Hydrazide Coupling: General procedure 7

To a solution of an intermediate of formula (XIII) (0.5 mmol) in THF (5 ml) was added a carboxylic acid of formula RIOCO2H (0.5 mmol), DIPEA (1.5 mmol) and T3P (50% in EtOAc, 1.5 mmol). The resulting solution was stirred at 60 °C for 2 h. The reaction was then cooled to RT and diluted with water. The mixture was extracted three times with EtOAc. The combined organic layers were washed with brine and then dried over sodium sulfate, filtered and the solvent evaporated under reduced pressure. The desired product (XIV) was used crude in the next step or purified by flash column chromatography on silica gel or by reverse phase preparative HPLC.

• L3,4-Oxadiazole cyclization: General procedure 8

To a solution of an intermediate of formula (XIV) (0.3 mmol) in THF (3 ml) was added Burgess reagent (0.9 mmol). The resulting solution was stirred at RT overnight. Water was added and the mixture was extracted three times with EtOAc. The combined organic layers were washed with brine and then dried over sodium sulfate, filtered and the solvent evaporated under reduced pressure. The desired product (XV) was used crude in the next step or purified by flash column chromatography on silica gel or by reverse phase preparative HPLC.

• Amidoxime Coupling with RCO2H: General procedure 10a

To a solution of an intermediate of formula (XVII, wherein R is H) (1.0 mmol) in THF (8.5 ml) was added a carboxylic acid of formula RIOCO2H (0.12 mmol), DIPEA (2.0 mmol) and HATU (0.15 mmol) and the reaction was stirred at RT for 4 h. Water and EtOAc were added and the layers were separated. The aqueous phase was extracted twice with EtOAc. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate, filtered and the solvent evaporated under reduced pressure. The desired product (XVII, wherein R is -CO(R10)) was used crude in the next step or purified by flash column chromatography on silica gel or by reverse phase preparative HPLC.

• Amidoxime Coupling with RCO2H: General procedure 10b

To solution of an intermediate of formula (XVII, wherein R is H) (0.2 mmol) in DMF (1.5 ml) was added a carboxylic acid of formula RIOCO2H (0.24 mmol), EDC hydrochloride (0.4 mmol), DIPEA (0.6 mmol) and HOBt (0.3 mmol) and the resulting mixture was stirred at RT for 16 h. Water and EtOAc were added, the layers were separated and the aqueous phase extracted twice with EtOAc. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate, filtered and the solvent evaporated under reduced pressure. The desired product (XVII, wherein R is -CO(RIO)) was used crude in the next step or purified by flash column chromatography on silica gel or by reverse phase preparative HPLC.

• L2,4-Oxadiazole cyclization: General procedure 11

A solution of an intermediate of formula (XVII, wherein R is -CO(RIO)) (0.15 mmol) in toluene (1 ml) was heated to 120 °C for 16 h. The solvent was then evaporated under reduced pressure. The desired product (XVIII) was used crude in the next step or purified by flash column chromatography on silica gel or by reverse phase preparative HPLC.

• Amidoxime formation: General procedure 12

To a solution of an intermediate of formula (VIII) (0.3 mmol) in EtOH (2.5 ml) was added solid NaHCCL (1.5 mmol) and hydroxylamine hydrochloride (0.6 mmol). The resulting suspension was heated to 80 °C for 90 min and then allowed to cool to RT. The suspension was filtered and the filter cake washed with EtOH and DCM. The filtrate was concentrated under reduced pressure and the remaining solid was dissolved in DCM and washed with water and brine, dried over anhydrous sodium sulfate, filtered and the solvent evaporated under reduced pressure. The desired product (XVII) was used crude in the next step or purified by flash column chromatography on silica gel or by reverse phase preparative HPLC. • Saponification: General procedure 13

To a solution of an intermediate of formula (VIII) (4 mmol) in THF (18 ml), MeOH (3 ml) and water (6 ml) was added LiOH hydrate (8 mmol) and stirred at RT for 2 h. IN aqueous HC1 was added and the resulting suspension extracted three times with EtOAc. The combined organic layers were washed with brine and then dried over sodium sulfate, filtered and the solvent evaporated under reduced pressure. The desired product (XII) was used crude in the next step or purified by flash column chromatography on silica gel or by reverse phase preparative HPLC.

• Hydrazide formation: General procedure 14

To a solution of an intermediate of formula (XII) (4.5 mmol) in THF (20 ml) was added CDI (5.7 mmol) and stirred at RT for 90 min. To this solution was then added a mixture of hydrazine hydrate (13.5 mmol) in THF (3.3 ml) and stirred for 1 h. The reaction mixture was diluted with water and EtOAc. The layers were separated and the aqueous phase washed twice with EtOAc. The combined organic layers were washed with brine and then dried over sodium sulfate, filtered and the solvent evaporated under reduced pressure. The desired product (XIII) was used crude in the next step or purified by flash column chromatography on silica gel or by reverse phase preparative HPLC.

Example 19

(3R)-3-amino-5-[[4-(l-ethylpropoxy)phenyl]methyl]-7-[5-(l -methyl-l-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lk6,5-benzothiaz epin-4-one

Background of the invention . 1

Detailed description of the invention . 7

Co-administration of compounds of formula (I) and other agents . 43

4-(bromomethyl)-4'-(trifluorom ethyl)- 1,1 '-biphenyl . 73 (cas 613241-14-8) . 73

Claims . 167

Abstract . 180

Step a) (3R)-3-(tert-butoxycarbonylamino)-4-oxo-3,5-dihydro-2H-l,5-b enzothiazepine-7 -carboxylic acid

To a solution of methyl (3R)-3-(tert-butoxycarbonylamino)-4-oxo-3,5-dihydro-2H-l,5- benzothiazepine-7-carboxylate (30.0 g, 85.1 mmol, 1 eq) (CAS: 2089150-62-7) in THF (300 mL) was added a solution of NaOH (5.11 g, 127.7 mmol, 1.5 eq) in water (300 mL) at 0 °C. The mixture was stirred at RT for 7 h. At 0 °C, 0.5 M aqueous HC1 was added to the reaction mixture which was then allowed to warm to RT. EtOAc was added and the layers were separated. The aqueous phase was extracted with EtOAc (2x 200 mL). The combined organic layer were washed with brine (3x200 mL), dried over sodium sulfate, filtered and concentrated to afford the title compound (29.4 g, 86.8 mmol, 91% yield) as yellow solid. MS (ESI): 283.0 [M+H-isobutene] + .

Step b) tert-butyl N-[(3R)-7-(hydrazinecarbonyl)-4-oxo-3,5-dihydro-2H-l,5-benzo thiazepin-3- yl] carbamate

To a solution of (3R)-3-(tert-butoxycarbonylamino)-4-oxo-3,5-dihydro-2H-l,5-b enzothiazepine-7- carboxylic acid (29.4 g, 86.8 mmol, 1 eq) and DIPEA (45.4 mL, 260.6 mmol, 3 eq) in DCM (400 mL) was added T3P (50 wt% in EtOAc, 66.8 mL, 130.3 mmol, 1.5 eq) at RT and the reaction was stirred for 30 min. The reaction mixture was then transferred via dropping funnel to a solution of hydrazine hydrate (17.2 mL, 347.5 mmol, 4 eq) in DCM (190 mL). The reaction mixture was stirred for 3 h at RT and a yellow precipitate formed. The mixture was concentrated and the crude purified by prep- HPLC to afford the title compound (6 g, 17.0 mmol, 19% yield) as white solid. MS (ESI): 297.1 [M+H-i sobutene] + .

Step c) tert-butyl N-[(3R)-7-[[(2-methyl-2-methylsulfonyl-propanoyl)amino] carbamoyl] -4-oxo-3, 5- dihydro-2H-l , 5-benzothiazepin-3-yl carbamate

To a solution of tert-butyl N-[(3R)-7-(hydrazinecarbonyl)-4-oxo-3,5-dihydro-2H-l,5-benzo thiazepin- 3-yl]carbamate (5.0 g, 14.2 mmol, 1 eq), 2-methyl-2-methylsulfonyl-propanoic acid (3.3 g, 19.8 mmol, 1.4 eq), and DIPEA (12.5 mL, 71.7 mmol, 5.06 eq) in THF (100 mL) was added T3P (50 wt% in EtOAc, 19.0 g, 29.8 mmol, 2.1 eq). The mixture was stirred at 60 °C for 3 h. The solution was poured into water (300 mL) and extracted with EtOAc (2 x 200 mL). The combined organic layers were washed with brine (300 mL), dried over sodium sulfate and concentrated. The crude residue was purified by prep-HPLC to give the title compound (5.6 g, 11.1 mmol, 77% yield) as an orange solid. MS (ESI): 445.1 [M+H-isobutene] + .

Step d) tert-butyl N-[ (3R)~ 7-[5-( I -me thy l-l -methylsulf onyl-e thy l)-l, 3, 4-oxadiazol-2-yl ]-4-oxo-3, 5- dihydro-2H-l , 5-benzothiazepin-3-yl carbamate

The title compound was prepared from tert-butyl N-[(3R)-7-[[(2-methyl-2-methylsulfonyl- propanoyl)amino]carbamoyl]-4-oxo-3,5-dihydro-2H-l,5-benzothi azepin-3-yl]carbamate (1.8 g, 3.6 mmol) in analogy to general procedure 8 and was obtained as yellow solid (1.6 g, 89% yield). MS (ESI): 427.0 [M+H-isobutene] + . Step e) tert-butyl N-[(3R)-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiaz ol-2-yl]-4-oxo-3,5- dihydro-2H-l , 5-benzothiazepin-3-yl carbamate

The title compound as prepared from tert-butyl N-[(3R)-7-[5-(l-methyl-l-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-4-oxo-3,5-dihydro-2H-l,5-benzothiazepi n-3-yl]carbamate (800.0 mg, 1.66 mmol) in analogy to general procedure 5 and was obtained as yellow solid (690 mg, 1.34 mmol, 78% yield). MS (ESI): 459.1 [M+H-isobutene] + .

Step f) tert-butyl N-[(3R)-5-[[4-(l-ethylpropoxy)phenyl]methyl]-7-[5-(l-methyl- l-methylsulfonyl- ethyl)-l, 3, 4-oxadiazol-2-yl] -1 , 1, 4-trioxo-2, 3-dihydro-l/.6, 5-benzothiazepin-3-yl] carbamate

To a suspension of tert-butyl N-[(3R)-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiaz ol-2-yl]-

4-oxo-3,5-dihydro-2H-l,5-benzothiazepin-3-yl]carbamate (50.0 mg, 0.1 mmol, 1 eq), potassium iodide (10.0 mg, 0.06 mmol, 0.62 eq) and potassium carbonate (40.0 mg, 0.29 mmol, 2.98 eq) in DMF (1.5 mL) was added l-(chloromethyl)-4-(l-ethylpropoxy)benzene (27.0 mg, 0.13 mmol, 1.31 eq, CAS 248262-55-7) and stirred at RT for 12 h. The mixture was then poured into water (40 mL) and extracted with EtOAc (2x30 mL). The combined organic layers were washed with brine (30 mL), dried over sodium sulfate, filtered and concentrated in vacuum. The remaining residue was purified by preparative TLC (PE/EtOAc= 1 : 1) to afford desired title compound (50 mg, 0.07 mmol, 75% yield) as a colorless oil. MS (ESI): 635.3 [M+H-isobutene] + . Step g) (3R)-3-amino-5-[ [4-( l-ethylpropoxy)phenyl] methyl] -7- [5-(l -methyl- 1-methylsulfony l-e thy l)- 1, 3, 4-oxadiazol-2-yl]-l, l-dioxo-2, 3 -dihydro- 1 6, 5-benzothiazepin-4-one

The title compound was prepared from tert-butyl N-[(3R)-5-[[4-(l-ethylpropoxy)phenyl]methyl]-7- [5-(l -methyl- 1-methylsulfonyl-ethyl)- 1,3, 4-oxadiazol-2-yl]-l,l,4-tri oxo-2, 3-dihydro- 1X6, 5- benzothiazepin-3-yl]carbamate (50.0 mg, 0.07 mmol) in analogy to general procedure 6a and was obtained as white solid, as hydrochloride salt (23.8 mg, 0.04 mmol, 52% yield). MS (ESI): 591.1 [M+H] + .

The examples of the following table were prepared in analogy to Example 19, using the appropriate benzyl halide building block.

(*) as hydrochloride salt

Example 15 (3R)-3-amino-5-[[4-[benzyl(methyl)amino]phenyl]methyl]-7-[5- (l-methyl-l-methylsulfonyl- ethyl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lk6,5-ben zothiazepin-4-one

Step a) tert-butyl N-[(3R)-5-[[4-[benzyl(methyl)amino]phenyl]methyl]-7-[5-(l-me thyl-l- methylsulfonyl-ethyl)-l, 3, 4-oxadiazol-2-yl -l, 1, 4-trioxo-2, 3-dihydro-l 6, 5-benzothiazepin-3- yl] carbamate

To a solution tert-butyl N-[(3R)-7-[5-(l -methyl- l-methylsulfonyl-ethyl)-l, 3, 4-oxadiazol-2-yl]-4- oxo-3, 5-dihydro-2H-l,5-benzothiazepin-3-yl]carbamate (50 mg, 0.1 mmol, 1 eq, Example 19, step e) in toluene (1.5 mL) was added [4-[benzyl(methyl)amino]phenyl]methanol (55.2 mg, 0.24 mmol, 2.5 eq, CAS 131719-59-0), PhsP (0.05 mL, 0.19 mmol, 2 eq) and di-tert-butyl azodicarboxylate (44.7 mg, 0.19 mmol, 2 eq). The mixture was degassed with nitrogen and stirred at 50 °C for 3 h. The reaction was diluted with water (5 mL) and EtOAc (5 ml). The layers were separated and the aqueous phase extracted with EtOAc (3x5 mL). The combined organic extracts were dried over sodium sulfate, filtered and concentrated. The remaining crude was purified using column chromatography on silica gel (60% EtOAc in PE) to obtain the title compound (36 mg, 0.05 mmol, 51% yield) as a light yellow solid. MS (ESI): 724.0 [M+H-isobutene] + . Step b) ( 3R)-3-amino-5-[[ 4-[benzyl(methyl)amino ] phenyl] methyl]- 7-[5-( I -methyl- 1 -me thylsulfonyl- ethyl)-l, 3, 4-oxadiazol-2-yl -l, l-dioxo-2, 3 -di hydro- 1/.6, 5-benzothiazepin-4-one

The title compound was prepared from tert-butyl N-[(3R)-5-[[4- [benzyl(methyl)amino]phenyl]methyl]-7-[5-(l-methyl-l-methyls ulfonyl-ethyl)-l,2,4-oxadiazol-3- yl]-l,l,4-trioxo-2,3-dihydro-lk6,5-benzothiazepin-3-yl]carba mate (36 mg, 0.05 mmol) in analogy to general procedure 6a and was obtained as light yellow solid (7.4 mg, 0.01 mmol, 23% yield). MS (ESI): 624.1 [M+H] + .

Example 18 of the following table were prepared in analogy to Example 15, using the appropriate benzylic alcohol building block.

Example 17 (3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-ox adiazol-2-yl]-l,l-dioxo-5-[[4-[5-

(trifluoromethyl)-l,2,4-oxadiazol-3-yl]phenyl]methyl]-2,3 -dihydro-lk6,5-benzothiazepin-4-one

Step a) tert-butyl N-[(3R)-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiaz ol-2-yl]-l,l,4-trioxo- 5-[[ 4-[5-(trifluoromethyl)-l, 2, 4-oxadiazol-3-yl] phenyl] methyl] -2, 3-dihydro-176, 5-benzothiazepin-

3-yl carbamate

To a solution of tert-butyl N-[(3R)-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-oxadiaz ol-2-yl]-4- oxo-3, 5-dihydro-2H-l,5-benzothiazepin-3-yl]carbamate (40 mg, 0.08 mmol, 1 eq, Example 19, step e), KI (6.45 mg, 0.04 mmol, 0.5 eq) and potassium carbonate (32.23 mg, 0.23 mmol, 3 eq) in DMF (1 mL) was added a solution of 3-[4-(bromomethyl)phenyl]-5-(trifluoromethyl)-l,2,4-oxadiazo le (28.6 mg, 0.09 mmol, 1.2 eq) in DMF (0.5 mL) at 20 °C. The reaction mixture was stirred at that temperature for 2 h. The reaction was diluted with water (5 ml) and extracted with EtOAc (8 ml x 3). The combined organic layers were dried over sodium sulfate, filtered and concentrated in vacuo. The remaining residue was purified by preparative TLC (60% EtOAc in PE) to give the title compound (45 mg, 0.06 mmol, 78% yield) as white solid. MS (ESI): 685.2 [M+H-isobutene] + . Step b) (3R)-3-amino-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3,4-ox adiazol-2-yl]-l,l-dioxo-5-[[4-

[5-(trifluoromethyl)-l ,2,4-oxadiazol-3-yl]phenyl]methyl] -2, 3-dihydro-126,5-benzothiazepin-4-one

The title compound was prepared form tert-butyl N-[(3R)-7-[5-(l-methyl-l-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l,4-trioxo-5-[[4-[5-(trifhroromethyl )-l,2,4-oxadiazol-3-yl]phenyl]methyl]- 2,3-dihydro-lk6,5-benzothiazepin-3-yl]carbamate (40 mg, 0.05 mmol) in analogy to general procedure 6a and was obtained as white solid, as hydrochloride salt (34.3 mg, 0.05 mmol, 93% yield). MS (ESI): 641.2 [M+H] + .

The examples of the following table were prepared in analogy to Example 17, using the appropriate carboxylic acid building block.

(*) as hydrochloride salt

Example 48

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo- 5-[(6-phenoxy-3-pyridyl)methyl]-2,3-dihydro-lk6,5-benzothiaz epin-4-one

Step a tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2, 4-oxadiazol-3-yl]~ 1, 1, 4-trioxo-3, 5-dihydro-2H-l/.6, 5-benzothiazepin-3-yl] carbamate

The title compound was prepared in analogy to general procedure 5 from tert-butyl N-[(3R)-8-fluoro-

7-[5-(l -methyl- 1 -methyl sulfonyl-ethyl)- 1,2, 4-oxadiazol-3-yl]-4-oxo-3,5-dihydro-2H- 1,5- benzothiazepin-3-yl]carbamate (Example 43, Step c), 54 mg, 0.108 mmol) and was obtained as white powder (28.5 mg, 50% yield). MS (ESI): 477.2 [M-isobutene+H] + .

Step b) tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2, 4-oxadiazol-3-yl]-

1, 1, 4-trioxo-5-[ ( 6-phenoxy-3-pyridyl)methyl -2, 3-dihydro-lX6, 5-benzothiazepin-3-yl carbamate

The title compound was prepared in analogy to general procedure 4 from tert-butyl N-[(3R)-8-fluoro- 7-[5-( I -methyl- l-methylsulfonyl-ethyl)-l, 2, 4-oxadiazol-3-yl]-l, 1, 4-trioxo-3, 5-dihydro-2H-l/.6, 5- benzothiazepin-3-yl] carbamate (9 mg, 0.017 mmol, 1 eq) using 5-(chloromethyl)-2-phenoxy-pyridine (CAS 5795855-1, 5.57 mg, 0.025 mmol, 1.5 eq) and was obtained as white powder (4.4 mg, 36% yield). MS (ESI): 716.4 [M+H] + .

Step c) (3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl) -l,2,4-oxadiazol-3-yl]-l,l- dioxo-5-[ ( 6-phenoxy-3-pyridyl )methyl -2, 3 -dihydro- 1 6, 5-benzothiazepin-4-one

The title compound was prepared in analogy to general procedure 6c from tert-butyl N-[(3R)-8- fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2,4-oxadiazo l-3-yl]-l, 1 ,4-trioxo-5-[(6-phenoxy-3- pyridyl)methyl] -2, 3-dihydro-126,5-benzothiazepin-3-yl] carbamate (4.4mg, 0.006 mmol) and was obtained as white powder, as hydrochloride salt (4 mg, 94% yield). MS (ESI): 660.4 [M+HCOO]' .

Example 50 of the following table was prepared in analogy to Example 48, step a-c), using the appropriate building block.

* as 2, 2, 2, -trifluoroacetic acid salt

Example 43

(3R)-3-amino-5-[(4-chlorophenyl)methyl]-8-fluoro-7-[5-(l- methyl-l-methylsulfonyl-ethyl)- l,2,4-oxadiazol-3-yl]-l,l-dioxo-2,3-dihydro-lk6,5-benzothiaz epin-4-one

Step a tert-butyl N-[(3R)-8-fhioro-7-[(Z)-N'-hydroxycarbamimidoyl]-4-oxo-3,5-d ihydro-2H-l,5- benzothiazepin-3-yl carbamate To a solution of tert-butyl N-[(3R)-7-cyano-8-fluoro-4-oxo-3,5-dihydro-2H-l,5-benzothiaz epin- 3- yl]carbamate(2R)-3-(2-amino-4-cyano-5-fluoro-phenyl)sulfanyl -2-(tert- butoxycarbonylamino)propanoic acid (200 mg, 0.59 mmol) in MeOH (2 mL) were added hydroxylamine hydrochloride (63.7 mg, 0.89 mmol, 1.5 eq) and sodium bicarbonate (249 mg, 2.96 mmol, 5 eq). The mixture was stirred for 16 h at 70°C, cooled to RT, filtered and the filter cake was washed with DCM. The combined filtrates were concentrated in vacuo. The reaction was diluted with DCM and washed with water and brine. The organic layer was then dried over sodium sulfate, filtered and concentrated under reduced pressure to afford the title compound (444 mg, 1.19 mmol, 74% yield) as yellow solid. MS (ESI): 315.1 [M-isobutene+H] + .

Step b) [ (Z)-[amino-[(3R)-3-(tert-butoxycarbonylamino)-8-fluoro-4-oxo -3, 5-dihydro-2H-l, 5- benzothiazepin- 7-yl methylene ] amino ] 2-methyl-2-methylsulfonyl-propanoate The title compound was prepared in analogy to general procedure 10a from tert-butyl N-[(3R)-8- fluoro-7-[(Z)-N' -hydroxy carbamimidoyl]-4-oxo-3,5-dihydro-2H-l,5-benzothiazepin-3-yl] carbamate (440 mg, 1.03 mmol) and was obtained as a light yellow solid (520 mg, 97% yield). MS (ESI): 463.2 [M-i sobutene+H] + . Step c) tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2, 4-oxadiazol-3-yl]-4- oxo-3, 5-dihydro-2H-l, 5-benzothiazepin-3-yl carbamate

The title compound was prepared in analogy to general procedure 11 from tert-butyl N-[(3R)-8-fluoro- 7-[(Z)-N'-(l-methyl-l-methylsulfonyl-ethoxy)carbamimidoyl]-4 -oxo-3,5-dihydro-2H-l,5- benzothiazepin-3-yl] carbamate (520 mg, 1 mmol, 1 eq) and was obtained as a light yellow solid (250 mg, 47% yield). MS (ESI): 445.2 [M-isobutene+H] + .

Step d) tert-butyl N-[(3R)-5-[(4-chlorophenyl)methyl]-8-fluoro-7-[5-(l-methyl-l -methylsulfonyl- ethyl)-l, 2, 4-oxadiazol-3-yl ]-4-oxo-2, 3-dihydro-l, 5-benzothiazepin-3-yl carbamate The title compound was prepared in analogy to general procedure 4 from tert-butyl N-[(3R)-8-fluoro- 7-[5-(l -methyl- 1 -methyl sulfonyl-ethyl)- 1,2, 4-oxadiazol-3-yl]-4-oxo-3,5-dihydro-2H- 1,5- benzothiazepin-3-yl]carbamate (30 mg, 0.057 mmol, 1 eq) using l-(bromomethyl)-4-chloro-benzene (17.55 mg, 0.085 mmol, Eq: 1.5) and was obtained as an off-white powder (18 mg, 49% yield). MS (ESI): 569.3 [M-i sobutene+H] + . Step e) tert-butyl N-[(3R)-5-[(4-chlorophenyl)methyl]-8-fluoro-7-[5-(l-methyl-l -methylsulfonyl- ethyl)-l, 2, 4-oxadiazol-3-yl] -1 , 1, 4-trioxo-2, 3-dihydro-lX6, 5-benzothiazepin-3-yl] carbamate

The title compound was prepared in analogy to general procedure 5 from tert-butyl N-[(3R)-5-[(4- chlorophenyl)methyl]-8-fluoro-7-[5-(l -methyl- l-methylsulfonyl-ethyl)-l, 2, 4-oxadiazol-3-yl]-4-oxo-

2,3-dihydro-l,5-benzothiazepin-3-yl]carbamate (18 mg, 0.028 mmol, Eq: l) and was obtained as an off-white powder (20.4 mg, 108% yield). MS (ESI): 601.2 [M-isobutene+H] + .

Step f) (3R)-3-amino-5-[(4-chlorophenyl)methyl]-8-fluoro-7-[5-(l-met hyl-l-methylsulfonyl-ethyl)-

1, 2, 4-oxadiazol-3-yl]-l, l-dioxo-2, 3 -dihydro- 1 6, 5-benzothiazepin-4-one

The title compound was prepared in analogy to general procedure 6c from tert-butyl N-[(3R)-5-[(4- chlorophenyl)methyl ]-8-fluoro- 7-[5-( I -methyl-l-methylsulfonyl-ethyl)-l, 2, 4-oxadiazol-3-yl -l, 1, 4- trioxo-2,3-dihydro-lX6,5-benzothiazepin-3-yl] carbamate (20.4 mg, 0.031 mmol) and was obtained as white solid, as hydrochloride salt (15 mg, 80% yield). MS (ESI): 557.3 [M+H] + . The examples of the following table were prepared in analogy to Example 43 step d-f, using the appropriate building block.

Example 30

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo- 5-[(4-phenoxyphenyl)methyl]-2,3-dihydro-lk6,5-benzothiazepin -4-one

Step a) tert-butyl N-[(3R)-7-cyano-8-fluoro-4-oxo-5-[(4-phenoxyphenyl)methyl] -2, 3-dihydro-l, 5- benzothiazepin-3-yl carbamate The title compound was prepared in analogy to general procedure 4 from tert-butyl N-[(3R)-7-cyano- 8-fluoro-4-oxo-3,5-dihydro-2H-l,5-benzothiazepin-3-yl]carbam ate (Example 53 step d) 200 mg, 0.593 mmol) using l-(bromomethyl)-4-phenoxy-benzene (CAS 36881-42-2, 156 mg, 0.593 mmol) and was obtained as white solid (210 mg, 67% yield) MS (ESI): 542.0 [M+Na] + .

Step b) tert-butyl N-[(3R)-8-fluoro-7-[(Z)-N'-hydroxycarbamimidoyl]-4-oxo-5-[(4 - phenoxyphenyl)methyl] -2, 3-dihydro-l, 5-benzothiazepin-3-yl] carbamate

The title compound was prepared in analogy to general procedure 12 from tert-butyl N-[(3R)-7-cyano- 8-fluoro-4-oxo-5-[ ( 4-phenoxyphenyl )methyl]-2, 3-dihydro-l, 5-benzothiazepin-3-yl carbamate (210 mg, 0.404 mmol) using and was obtained as light yellow solid (125 mg, 49% yield) MS (ESI): 553.0 [M+H] + . Step c) [ (Z)-[amino-[(3R)-3-(tert-butoxycarbonylamino)-8-fluoro-4-oxo -5-[(4- phenoxyphenyl)methyl] -2, 3-dihydro-l, 5-benzothiazepin- 7 -y I] methylene ] amino ] 2-methyl-2- methylsulfonyl-propanoate The title compound was prepared in analogy to general procedure 10b from tert-butyl N-[(3R)-8- fluoro- 7-[ (Z)-N'-hydroxycarbamimidoyl ]-4-oxo-5-[ ( 4-phenoxyphenyl)methyl -2, 3-dihydro-l, 5- benzothiazepin-3-yl] carbamate (125 mg, 0.226 mmol) and was obtained as light yellow gum (154 mg, 93% yield) MS (ESI): 644.9 [M-isobutene+H] + .

Step d) tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2, 4-oxadiazol-3-yl]-4- oxo-5-[ (4-phenoxyphenyl)methyl ]-2, 3-dihydro-l, 5-benzothiazepin-3-yl ] carbamate

The title compound was prepared in analogy to general procedure 11 from [(Z)-[amino-[(3R)-3-(tert- butoxycarbonylamino)-8-fluoro-4-oxo-5-[(4-phenoxyphenyl)meth yl]-2, 3 -dihydro- 1,5- benzothiazepin-7-yl] methylene] amino] 2-methyl-2-methylsulfonyl-propanoate (154 mg, 0.220 mmol) and was obtained as light brown gum (140 mg, 73% yield) MS (ESI): 627.0 [M-isobutene+H] + .

Step e) tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2, 4-oxadiazol-3-yl]- 1, 1, 4-trioxo-5-[ ( 4-phenoxyphenyl)methyl -2, 3-dihydro-l .6, 5-benzothiazepin-3-yl carbamate

The title compound was prepared in analogy to general procedure 5 from tert-butyl N-[(3R)-5-[(4- chlorophenyl)methyl]-8-fluoro-7-[5-(2-methyl-2-methylsulfony l-propyl)-l,2,4-oxadiazol-3-yl]-4- oxo-2, 3-dihydro-l ,5-benzothiazepin-3-yl] carbamate (140 mg, 0.205 mmol) and was obtained as a white solid (130 mg, 74% yield) MS (ESI): 659.1 [M-isobutene+H] + .

Step f) (3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl) -l,2,4-oxadiazol-3-yl]-l,l- dioxo-5-[ ( 4-phenoxyphenyl)methyl -2, 3-dihydro-l/.6, 5-benzothiazepin-4-one

The title compound was prepared in analogy to general procedure 6a tert-butyl N-[(3R)-8-fluoro-7- [5-(l -methyl- 1-methylsulfonyl-ethyl)- 1,2, 4-oxadiazol-3-yl]-l, 1,4-tri oxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-3-yl] carbamate (130 mg, 0.182 mmol) and was obtained as a white solid, as a hydrochloride acid (45.9 mg, 38% yield) MS (ESI): 615.0 [M+H] + .

Example 44 (3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl) -l,2,4-oxadiazol-3-yl]-l,l-dioxo-

5-[[4-(trifluoromethoxy)phenyl]methyl]-2,3-dihydro-lk6,5- benzothiazepin-4-one

Step a) [(Z)-[amino-[(3R)-3-(tert-butoxycarbonylamino)-8-fluoro-4-ox o-3,5-dihydro-2H-l,5- benzothiazepin- 7-yl methylene ] amino ] 2-methyl-2-methylsulfonyl-propanoate The title compound was prepared in analogy to general procedure 10a from tert-butyl N-[(3R)-8- fluoro-7-[(Z)-N' -hydroxy carbamimidoyl]-4-oxo-3,5-dihydro-2H-l,5-benzothiazepin-3-yl] carbamate (Example 43, step a) (440 mg, 1.03 mmol, 1 eq) and 2-methyl-2-methylsulfonyl-propanoic acid (CAS 25841-43-4) and was obtained as a light yellow solid (520 mg, 97% yield). MS (ESI): 463.2 [M- isobutene+H] + . Step b) tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2, 4-oxadiazol-3-yl]-4- oxo-3, 5-dihydro-2H-l, 5-benzothiazepin-3-yl carbamate The title compound was prepared in analogy to general procedure 11 from [(Z)-[amino-[(3R)-3-(tert- butoxycarbonylamino)-8-fluoro-4-oxo-3,5-dihydro-2H-l,5-benzo thiazepin-7-yl]methylene]amino] 2-methyl-2-methylsulfonyl-propanoate (520 mg, 1 mmol, 1 eq) and was obtained as a white solid (250 mg, 47% yield). MS (ESI): 499.3 [M-H]’. Step c) tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2, 4-oxadiazol-3-yl]-4- oxo-5-[ [ 4-(trifluoromethoxy)phenyl methyl -2, 3-dihydro-l, 5-benzothiazepin-3-yl carbamate

The title compound was prepared in analogy to general procedure 4 from tert-butyl N-[(3R)-8-fluoro- 7-[5-(l -methyl- 1 -methyl sulfonyl-ethyl)- 1,2, 4-oxadiazol-3-yl]-4-oxo-3,5-dihydro-2H- 1,5- benzothiazepin-3-yl]carbamate (30 mg, 0.057 mmol, 1 eq) and a l-(bromomethyl)-4- (trifluoromethoxy)benzene (CAS 50824-05-0) and was obtained as an off-white solid (31 mg, 79% yield). MS (ESI): 619.3 [M-isobutene+H] + .

Step d) tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2, 4-oxadiazol-3-yl]- 1, 1, 4-trioxo-5-[[ 4-(trifluoromethoxy)phenyl] methyl] -2, 3-dihydro-l/.6, 5-benzothiazepin-3- yl] carbamate The title compound was prepared in analogy to general procedure 5 from tert-butyl N-[(3R)-8-fluoro- 7-[5-(l -methyl- 1 -methyl sulfonyl-ethyl)- 1,2, 4-oxadiazol-3-yl]-4-oxo-5-[[4- (trifluoromethoxy)phenyl]methyl]-2,3-dihydro-l,5-benzothiaze pin-3-yl]carbamate (31 mg, 0.046 mmol, 1 eq) and was obtained as an off-white solid (22 mg, 68% yield). MS (ESI): 651.4 [M-H]'. Step e) (3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl) -l,2,4-oxadiazol-3-yl]-l,l- dioxo-5-[[4-(trifluoromethoxy)phenyl methyl -2, 3-dihydro-l/.6, 5-benzothiazepin-4-one

The title compound was prepared in analogy to general procedure 6c from tert-butyl N-[(3R)-8-fluoro- 7-[5-(l -methyl- 1 -methyl sulfonyl-ethyl)- 1, 2, 4-oxadiazol-3-yl]-l, 1,4-tri oxo-5-[[4- (trifluoromethoxy)phenyl]methyl]-2,3-dihydro-lX6,5-benzothia zepin-3-yl]carbamate (22 mg, 0.031 mmol, 1 eq) and was obtained as hydrochloride salt as a white solid (9.9 mg, 48% yield). MS (ESI): 607.3 [M+H] + .

Example 2 (3R)-3-amino-5-[(4-chlorophenyl)methyl]-8-fluoro-7-[5-(l-met hyl-l-methylsulfonyl-ethyl)- l,3,4-oxadiazol-2-yl]-l,l-dioxo-2,3-dihydro-lk6,5-benzothiaz epin-4-one

Step a) methyl (3R)-3-(tert-butoxycarbonylamino)-5-[(4-chlorophenyl)methyl] -8-fluoro-4-oxo-2,3- dihydro-1, 5-benzothiazepine- 7 -carboxylate

The title compound was prepared in analogy to general procedure 4 from methyl (3R)-3-(tert- butoxycarbonylamino)-8-fluoro-4-oxo-3,5-dihydro-2H-l,5-benzo thiazepine-7-carboxylate (CAS:2002449-38-7) (5.67 g, 13.77 mmol, 1 eq) and was obtained as light yellow solid (2699 mg, 36%). MS (ESI): 439.3 [M-isobutene+H] + .

Step b) ( 3R)-3-(tert-butoxycarbonylamino)-5-[ ( 4-chlorophenyl)methyl ]-8-fluoro-4-oxo-2, 3-dihydro- 1, 5-benzothiazepine- 7 -carboxylic acid

The title compound was prepared in analogy to general procedure 13 from methyl (3R)-3-(tert- butoxycarbonylamino)-5-[(4-chlorophenyl)methyl]-8-fluoro-4-o xo-2,3-dihydro-l,5- benzothiazepine-7-carboxylate (300 mg, 606 pmol, Eq: 1) and was obtained as yellow oil (280 mg, 512 pmol, 85 % yield). MS (ESI): 425.2 [M-isobutene+H] + .

Step c) tert-butyl N-[(3R)-5-[(4-chlorophenyl)methyl]-8-fluoro-7-(hydrazinecarb onyl)-4-oxo-2,3- dihydro-1, 5-benzothiazepin-3-yl carbamate

The title compound was prepared in analogy to general procedure 14 from (3R)-3-(tert- butoxycarbonylamino)-5-[(4-chlorophenyl)methyl]-8-fluoro-4-o xo-2,3-dihydro-l,5- benzothiazepine-7-carboxylic acid (2.9 g, 6.03 mmol, Eq: 1) and was obtained as light yellow solid (1.9 g, 64%). MS (ESI): 439.3 [M-isobutene+H] + .

Step d) tert-butyl N-[(3R)-5-[(4-chlorophenyl)methyl]-8-fhioro-7-[5-(l-methyl-l -methylsulfonyl- ethyl)-l, 3, 4-oxadiazol-2-yl]-4-oxo-2, 3-dihydro-l, 5-benzothiazepin-3-yl] carbamate tert-butyl N-[(3R)-5-[(4-chlorophenyl)methyl]-8-fluoro-7-(hydrazinecarb onyl)-4-oxo-2,3-dihydro- l,5-benzothiazepin-3-yl]carbamate (100 mg, 0.20 mmol, 1.0 eq) was dissolved in THF (2. mL) and 2-mesyl-2-methyl-propionic acid (36.9 mg, 0.22 mmol, 1.1 eq), HATU (84.5 mg, 0.22 mmol, 1.1 eq) and DIEA (52.2 mg, 70.6 uL, 0.40 mmol, 2.0 eq) were added to give a yellow solution. The mixture was stirred for 45 minutes at RT. Burgess Reagent (240.7 mg, 1.01 mmol, 5 eq) was added and the reaction mixture stirred for 60 min at RT. The crude material was concentrated under reduced pressure and the remaining solid purified by flash chromatography on silica gel (0-50% EtOAc in heptane) to give the title compound (44 mg, 31%) as light yellow solid. MS (ESI): 569.2 [M-isobutene+H] + .

Step e) (3R)-3-amino-5-[ (4-chlorophenyl)methyl]-8-fluoro-7-[5-(l-methyl-l-methylsulf onyl-ethyl)-

1, 3, 4-oxadiazol-2-yl]-l, l-dioxo-2, 3 -dihydro- 1 6, 5-benzothiazepin-4-one The title compound was prepared in analogy to general procedure 6a from tert-butyl N-[(3R)-5-[(4- chlorophenyl)methyl]-8-fluoro-7-[5-(l-methyl-l-methylsulfony l-ethyl)-l,3,4-oxadiazol-2-yl]-4-oxo- 2,3-dihydro-l,5-benzothiazepin-3-yl]carbamate (49. mg, 0.075 mmol, 1 eq) and was obtained as light yellow solid, as hydrochloride salt (12 mg, 26%). MS (ESI): 557.2 [M+H] +

The example of the following table was prepared in analogy to Example 2, using the appropriate carboxylic acid building block. Example 53

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-

5-[(4-tetrahydropyran-4-ylphenyl)methyl]-2,3-dihydro-lk6, 5-benzothiazepin-4-one Step a) 4- [4-(chloromethyl)phenyl] tetrahydropyran

To a solution of (4-tetrahydropyran-4-ylphenyl)methanol (CAS: 1276024) (300 mg, 1.56 mmol, 1 eq) in DCM (10 mL) was added thionyl chloride (0.4 mL, 5.65 mmol, 3.6 eq) via syringe at RT. The solution was stirred at RT for 12 h. The mixture was poured into water (50 mL) and the pH was adjusted to around pH 9 by progressively adding solid NaHCCL. The solution was extracted with DCM (100 mL). The organic phase was washed with brine (100 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give the title compound (330 mg, 1.57 mmol, 95% yield) as light yellow oil. MS (ESI): 211.2 [M+H] +

Step b) (2R)-2-(tert-butoxycarbonylamino)-3-(4-cyano-5-fluoro-2-nitr o-phenyl)sulfanyl-propanoic acid

To a solution of 2,4-difhioro-5-nitro-benzonitrile (9.4 g, 50 mmol) and (tert-butoxycarbonyl)-L- cysteine (11.07 g, 50 mmol) in DCM (157 mL) was added DIPEA (17.48 mL, 100 mmol, Eq: 2). The reaction mixture was stirred for 24 hours at 22°C, diluted with DCM (40 mL) and washed once with IN aqueous HC1 solution and extracted twice with DCM. The combined organic layers were washed with brine solution, dried over sodium sulfate, filtrated and concentrated in vacuo to yield a yellow solid (23.5 g) containing the title compound. MS (ESI): 286.1 [M-isobutene+H] + .

Step c) (2R)-3-(2-amino-4-cyano-5-fluoro-phenyl)sulfanyl-2-(tert-but oxycarbonylamino)propanoic acid

The title compound was prepared in analogy to general procedure 2 from (2R)-2-(tert- butoxycarbonylamino)-3-(4-cyano-5-fluoro-2-nitro-phenyl)sulf anyl-propanoic acid (23 g, 57.9 mmol) and was obtained as a black solid (23 g, 48.1 mmol, 80% yield). MS (ESI): 300.1 [M- isobutene+H] + . Step d) tert-butyl N-[ (3R)~ 7-cyano-8-fluoro-4-oxo-3, 5-dihydro-2H-l, 5-benzothiazepin-3- yl] carbamate

The title compound was prepared in analogy to general procedure 3 from (2R)-3-(2-amino-4-cyano- 5-fluoro-phenyl)sulfanyl-2-(tert-butoxycarbonylamino)propano ic acid (23 g, 48.1 mmol) and was obtained as a light yellow solid (8.4 g, 24.9 mmol, 48% yield). MS (ESI): 282.1 [M-isobutene+H] + . Step e) tert-butyl N-[ (3R - 7-cyano-8-fluoro-4-oxo-5-[ ( 4-tetrahydropyran-4-ylphenyl)methyl -2, 3- dihydro-1, 5-benzothiazepin-3-yl carbamate

To a solution of 4-[4-(chloromethyl)phenyl]tetrahydropyran (Example 53 step a) (224.83 mg, 1.07 mmol, 1.2 eq), K2CO3 (245.8 mg, 1.78 mmol, 2.0 eq) and KI (73.8 mg, 0.44 mmol, 0.5 eq) in DMF (6 mL) was added dropwise a solution of tert-butyl N-[(3R)-7-cyano-8-fluoro-4-oxo-3,5-dihydro-2H- l,5-benzothiazepin-3-yl]carbamate (300.0 mg, 0.89 mmol, 1.0 eq) in DMF (3 mL) at RT and the mixture was stirred for 12 h. The reaction mixture was poured into water and extracted with EtOAc (3x). The combined organic phase was washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The remaining residue was purified by column chromatography on silica gel (20% EtOAc in petroleum ether) to afford the title compound (500 mg, 0.97 mmol, 100% yield) as light yellow oil. MS (ESI): 412.2 [M-isobutene+H] +

Step f) tert-butyl N-[(3R)-8-fluoro-7-[(Z)-N'-hydroxycarbamimidoyl]-4-oxo-5-[(4 -tetrahydropyran- 4-ylphenyl)methyl] -2, 3-dihydro-l ,5-benzothiazepin-3-yl] carbamate

To a solution of tert-butyl N-[(3R)-7-cyano-8-fluoro-4-oxo-5-[(4-tetrahydropyran-4- ylphenyl)methyl]-2,3-dihydro-l,5-benzothiazepin-3-yl]carbama te (400 mg, 0.78 mmol, 1 eq) and DIPEA (0.53 mL, 3.13 mmol, 4.0 eq) inEtOH (5 mL) was added hydroxylamine hydrochloride (165.3 mg, 2.35 mmol, 3 eq) and sirred at 50 °C for 3h. The reaction mixture was concentrated under reduced pressure. EtOAc and water were added and the layers were separated. The aqueous phase was extracted with EtOAc (2x). The combined organic phase was washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure to afford the title compound (400 mg, 0.73 mmol, 83% yield) as white solid. MS (ESI): 545.3 [M+H] + Step g) [(Z)-[amino-[(3R)-3-(tert-butoxycarbonylamino)-8-fluoro-4-ox o-5-[(4-tetrahydropyran-4- ylphenyl)methyl -2, 3-dihydro-l, 5-benzothiazepin- 7-yl methylene ] amino ] 2-methyl-2- methylsulfonyl-propanoate

The title compound was prepared in analogy to general procedure 10a from tert-butyl N-[(3R)-8- fluoro-7-[(Z)-N' -hydroxy carbamimidoyl]-4-oxo-5-[(4-tetrahydropyran-4-ylphenyl)methyl ]-2, 3- dihydro-l,5-benzothiazepin-3-yl]carbamate (350 mg, 0.6 mmol) and was obtained as light yellow solid (481 mg, 92% yield). MS (ESI): 693.2 [M+H] + . Step h) tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2, 4-oxadiazol-3-yl]-4- oxo-5-[(4-tetrahydropyran-4-ylphenyl)methyl]-2, 3-dihydro-l, 5-benzothiazepin-3-yl] carbamate

The title compound was prepared in analogy to general procedure 11 from [(Z)-[amino-[(3R)-3-(tert- butoxycarbonylamino)-8-fluoro-4-oxo-5-[(4-tetrahydropyran-4- ylphenyl)methyl]-2, 3-dihydro-l, 5- benzothiazepin-7-yl]methylene]amino] 2-methyl-2-methylsulfonyl-propanoate (500 mg, 0.7 mmol, 1 eq) and was obtained as light yellow solid (136 mg, 28% yield). MS (ESI): 575.4 [M+H] + .

Step i) tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2, 4-oxadiazol-3-yl]- 1, 1, 4-trioxo-5-[(4-tetrahydropyran-4-ylphenyl)methyl]-2, 3-dihydro-l/.6, 5-benzothiazepin-3- yl] carbamate

The title compound was prepared in analogy to general procedure 5 from tert-butyl N-[(3R)-8-fluoro- 7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2,4-oxadiazol-3-yl] -4-oxo-5-[(4-tetrahydropyran-4- ylphenyl)methyl]-2, 3-dihydro-l, 5-benzothiazepin-3-yl]carbamate (130 mg, 0.2 mmol, 1 eq) and was obtained as an off-white solid (120 mg, 88% yield). MS (ESI): 607.3 [M-isobutene+H] + . Step j ) (3R)-3-amino-8-fluoro- 7-[ 5-( 1 -methyl- l-methylsulfonyl-ethyl)-l, 2, 4-oxadiazol-3-yl]-l, 1- dioxo-5-[(4-tetrahydropyran-4-ylphenyl)methyl]-2, 3-dihydro-l/.6, 5-benzothiazepin-4-one

The title compound was prepared from tert-butyl N-[(3R)-8-fhroro-7-[5-(l-methyl-l-methylsulfonyl- ethyl)-l,2,4-oxadiazol-3-yl]-l,l,4-trioxo-5-[(4-tetrahydropy ran-4-ylphenyl)methyl]-2,3-dihydro- lk6,5-benzothiazepin-3-yl]carbamate (50.0 mg, 0.1 mmol) in analogy to general procedure 6a and was obtained as white solid, as hydrochloride salt (30.6 mg, 0.05 mmol, 70% yield). MS (ESI): 607.3 [M+H] + .

The example of the following table was prepared in analogy to Example 53, using the appropriate benzyl halide building block.

Example 35

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,3,4-oxadiazol-2-yl]-l,l-dioxo-

5- [[4- [4-(trifluoromethyl)phenyl] phenyl] methyl]-2,3-dihydro-lk6,5-benzothiazepin-4-one

Step a) (3R)-3-(tert-butoxycarbonylamino)-8-fluoro-4-oxo-3,5-dihydro -2H-l,5-benzothiazepine-7- carboxylic acid

To the solution of methyl (3R)-3-(tert-butoxycarbonylamino)-8-fluoro-4-oxo-3,5-dihydro -2H-l,5- benzothiazepine-7-carboxylate (CAS:2002449-38-7) (8.5 g, 22.9 mmol, 1 eq) in THF (150 mL) was added a solution of NaOH (1220 mg, 30.5 mmol, 1.33 eq) in water (150 mL) at 0 °C. Then the mixture was stirred at RT for 9 h. The reaction mixture was added dropwise into 0.5 M aqueous HC1 (70 mL). The mixture was extracted with EtOAc (3x). The combined organic layer was washed with brine and dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford the title compound (5700 mg, 15.99 mmol, 57% yield) as white solid. MS (ESI): 301.0 [M+H] + . Step b) tert-butyl N-[(3R)-8-fluoro-7-(hydrazinecarbonyl)-4-oxo-3,5-dihydro-2H- l,5- benzothiazepin-3-yl carbamate

(3R)-3-(tert-butoxycarbonylamino)-8-fluoro-4-oxo-3,5-dihy dro-2H-l,5-benzothiazepine-7- carboxylic acid (202.6 mg, 0.55 mmol, 1 eq) and CDI (116.2 mg, 0.72 mmol, 1.3 eq) were dissolved in THF (2.4 mL) and stirred at RT for 30 min. This solution was then dropwise added to a solution of hydrazine monohydrate (53.0 mg, 51.5 uL, 1.65 mmol, 3 eq) in THF (0.8 mL) and stirred for 30 min. The reaction mixture was partially concentrated under reduced pressure and diluted with EtOAC. This solution was washed with water and brine, dried over magnesium sulfate and concentrated in vacuo to yield the title compound (214.7 mg, 100%) as light yellow solid. MS (ESI): 369.3 [M-H]

Step c) tert-butyl N-[(3R)-8-fluoro-7-[ [(2-methyl-2-methylsulfonyl-propanoyl)amino] carbamoyl] -4- oxo-3, 5-dihydro-2H-l, 5-benzothiazepin-3-yl carbamate

To a mixture of 2-methyl-2-methylsulfonyl-propanoic acid (0.27 g, 1.62 mmol, 1.2 eq), tert-butyl N- [(3R)-8-fluoro-7-(hydrazinecarbonyl)-4-oxo-3,5-dihydro-2H-l, 5-benzothiazepin-3-yl]carbamate (0.5 g, 1.35 mmol, 1.0 eq) and N,N-diisopropylethylamine (0.59 mL, 3.37 mmol, 2.5 eq) in THF (10 mL) was added T3P in EtOAc (1.81 g, 2.84 mmol, 2.1 eq) atRT and stirred for 16 h. The solution was poured into water (10 mL) and extracted with EtOAc (10 mL). The organic phase was washed with brine (30 mL), dried over sodium sulfate and concentrated under reduced pressure. The remaining residue was purified by column chromatography on silica gel (0-100% EtOAc in heptane) to give the title compound (570 mg, 1.1 mmol, 80% yield) as white solid. MS (ESI): 445.1 [M-isobutene+H] + . Step d) tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3, 4-oxadiazol-2-yl]-4- oxo-3, 5-dihydro-2H-l, 5-benzothiazepin-3-yl carbamate

The title compound was prepared from tert-butyl N-[(3R)-8-fluoro-7-[[(2-methyl-2-methylsulfonyl- propanoyl)amino]carbamoyl]-4-oxo-3,5-dihydro-2H-l,5-benzothi azepin-3-yl]carbamate (1800 mg, 3.5 mmol) in analogy to general procedure 8 in 1,4-di oxane and was obtained as yellow solid (1000 mg, 2 mmol, 54% yield). MS (ESI): 445.1 [M-isobutene+H] + .

Step e) tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3, 4-oxadiazol-2-yl]- 1, 1, 4-trioxo-3, 5-dihydro-2H-l/.6, 5-benzothiazepin-3-yl] carbamate

The title compound was prepared from tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl- ethyl)-l,3,4-oxadiazol-2-yl]-4-oxo-3,5-dihydro-2H-l,5-benzot hiazepin-3-yl]carbamate (1000 mg, 2 mmol) in analogy to general procedure 5 and was obtained as white solid (1000 mg, 1.88 mmol, 85% yield). MS (ESI): 477.0 [M-isobutene+H] + . Step f) tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,3, 4-oxadiazol-2-yl]- 1, 1, 4-trioxo-5-[[ 4-[ 4-(trifluoromethyl)phenyl] phenyl] methyl] -2, 3-dihydro-l/.6,5-benzolhiazepin-3- yl] carbamate

The title compound was prepared from tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl- ethyl)-l,3,4-oxadiazol-2-yl]-l,l,4-trioxo-3,5-dihydro-2H-lk6 ,5-benzothiazepin-3-yl]carbamate (50 mg, 0.1 mmol) in analogy to general procedure 4 and was obtained as white solid (50 mg, 0.07 mmol, 63% yield). MS (ESI): 710.8 [M-isobutene+H] + .

Step g) (3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl) -l, 3, 4-oxadiazol-2-yl]-l, 1- dioxo-5-[[4-[4-(trifluoromethyl)phenyl] phenyl] methyl] -2, 3-dihydro-126,5-benzothiazepin-4-one

The title compound was prepared from tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl- ethyl)-l,3,4-oxadiazol-2-yl]-l,l,4-trioxo-5-[[4-[4-(trifluor omethyl)phenyl]phenyl]methyl]-2,3- dihydro-lX6,5-benzothiazepin-3-yl]carbamate (45 mg, 0.1 mmol) in analogy to general procedure 6a and was obtained as white solid, as hydrochloride salt (14.9 mg, 0.02 mmol, 35% yield). MS (ESI): 667.2 [M+H] + .

The examples of the following table were prepared in analogy to Example 35, using the appropriate benzyl halide building block.

Example 59

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,2,4-oxadiazol-3-yl]-l,l-dioxo-

5-[[4-[[3-(trifluoromethyl)-l,2,4-oxadiazol-5-yl]methoxy] phenyl]methyl]-2,3-dihydro-lk6,5- benzothiazepin-4-one

Step a) tert-butyl N-[(3R)-7-cyano-8-fluoro-4-oxo-5-[(4-triisopropylsilyloxyphe nyl)methyl]-2,3- dihydro-1, 5-benzothiazepin-3-yl carbamate

The title compound was prepared in analogy to general procedure 4 from tert-butyl N-[(3R)-7-cyano- 8-fluoro-4-oxo-3,5-dihydro-2H-l,5-benzothiazepin-3-yl]carbam ate (Example 53 step d) (100 mg, 0.3 mmol) and was obtained as colorless oil (100 mg, 0.17 mmol, 52% yield). MS (ESI): 622.2 [M+Na] + . Step b) tert-butyl N-[ ( 3R)-8-fluoro- 7-[ (Z)-N'-hydroxycarbamimidoyl ]-4-oxo-5-[ ( 4- triisopropylsilyloxyphenyl)methyl] -2, 3-dihydr o-l, 5-benzothiazepin-3-yl] carbamate

The title compound was prepared in analogy to general procedure 12 from tert-butyl N-[(3R)-7-cyano- 8-fluoro-4-oxo-5-[(4-triisopropylsilyloxyphenyl)methyl]-2,3- dihydro-l,5-benzothiazepin-3- yl]carbamate (470 mg, 0.8 mmol) and was obtained as colorless oil (390 mg, 0.62 mmol, 71% yield). MS (ESI): 633.3 [M+H] + .

Step c) tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2, 4-oxadiazol-3-yl]-4- oxo-5-[ (4-triisopropylsilyloxyphenyl)methyl -2, 3-dihydro-l, 5-benzothiazepin-3-yl carbamate

To a solution of tert-butyl N-[(3R)-8-fluoro-7-[(Z)-N'-hydroxycarbamimidoyl]-4-oxo-5-[(4 - triisopropylsilyloxyphenyl)methyl]-2,3-dihydro-l,5-benzothia zepin-3-yl]carbamate (390 mg, 0.62 mmol, 1.0 eq), DIEA (106.4 mg, 0.92 mmol, 1.5 eq) and 2-methyl-2-methylsulfonyl-propanoic acid (112.6 mg, 0.68 mmol, 1.1 eq) in toluene (5 mL) was added T3P in EtOAc (784.0 mg, 1.23 mmol, 2.0 eq) at 100°C and stirred for 12 h. The reaction was concentrated under reduced pressure and the remaining crude was purified by chromatography on silica gel (20% EtOAc in petroleum ether) to give the title compound (320 mg, 0.42 mmol, 54% yield) as colorless oil. MS (ESI): 785.2 [M+Na] + .

Step c) tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2, 4-oxadiazol-3-yl]~ 1,1, 4-trioxo-5-[ ( 4-triisopropylsilyloxyphenyl )methyl]-2, 3-dihydro-l 6, 5-benzothiazepin-3- yl] carbamate

The title compound was prepared in analogy to general procedure 5 from tert-butyl N-[(3R)-8-fluoro- 7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2,4-oxadiazol-3-yl] -4-oxo-5-[(4-tetrahydropyran-4- ylphenyl)methyl]-2,3-dihydro-l,5-benzothiazepin-3-yl]carbama te (200 mg, 0.3 mmol) and was obtained as colorless oil (180 mg, 0.23 mmol, 81% yield). MS (ESI): 817.1 [M+Na] + . Step d) tert-butyl N-[(3R)-8-fluoro-5-[(4-hydroxyphenyl)methyl]-7-[5-(l-methyl- l-methylsulfonyl- ethyl)-l, 2, 4-oxadiazol-3-yl] -1 , 1, 4-trioxo-2, 3-dihydro-l/.6, 5-benzothiazepin-3-yl] carbamate

To a mixture of tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2, 4-oxadiazol- 3-yl]- l,l,4-trioxo-5-[(4-triisopropylsilyloxyphenyl)methyl]-2, 3-dihydro- 1X6, 5-benzothiazepin-3- yl]carbamate (175 mg, 0.22 mmol, 1.0 eq) in DMF (2 mL) was added CsF (100.3 mg, 0.66 mmol, 3.0 eq) at RT and stirred for 12 h. The reaction was quenched upon addition of water and extracted with EtOAc (3x). The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to give the title compound (120 mg, 0.19 mmol, 85% yield) as colorless oil. MS (ESI): 639.2 [M+H] + .

Step e) tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2, 4-oxadiazol-3-yl]-

1, l,4-trioxo-5-[[4-[[3-(trifluoromethyl)-l,2,4-oxadiazol-5-yl] methoxy]phenyl]methyl]-2, 3-dihydro- 1/.6, 5-benzothiazepin-3-yl carbamate To a mixture of tert-butyl N-[(3R)-8-fluoro-5-[(4-hydroxyphenyl)methyl]-7-[5-(l-methyl- l- methylsulfonyl-ethyl)-l,2,4-oxadiazol-3-yl]-l,l,4-trioxo-2,3 -dihydro-lX6,5-benzothiazepin-3- I l l yl]carbamate (70.0 mg, 0.11 mmol, 1.0 eq) and K2CO3 (30.2 mg, 0.22 mmol, 2.0 eq) in acetonitrile (1 mL) was added 5-(chloromethyl)-3-(trifluoromethyl)-l,2,4-oxadiazole (CAS: 1000442-49-7) (20.4 mg, 0.11 mmol, 1.0 eq) and KI (18.2 mg, 0.11 mmol, 1.0 eq) at 50°C and stirred for 1 h. The reaction was quenched upon addition of water and extracted with EtOAc (3x). The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The remaining residue was purified by Prep-HPLC to give the title compound (20 mg, 0.03 mmol, 23% yield) as colorless oil upon lyophilization. MS (ESI): 789.2 [M+H] + .

Step f) (3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl) -l,2,4-oxadiazol-3-yl]-l,l- dioxo-5-[[4-[ [ 3-(trifluoromethyl)-l , 2, 4-oxadiazol-5-yl methoxy] phenyl] methyl -2, 3 -di hydro- 1/.6, 5- benzothiazepin-4-one

The title compound was prepared in analogy to general procedure 6a from tert-butyl N-[(3R)-8-fluoro- 7-[5-(l -methyl- l-methylsulfonyl-ethyl)-l, 2, 4-oxadiazol-3-yl]-l,l,4-trioxo-5-[[4-[[3- (trifluoromethyl)-l, 2, 4-oxadiazol-5-yl]methoxy]phenyl]methyl]-2,3-dihydro- 1X6, 5-benzothi azepin- 3-yl]carbamate (12 mg, 0.02 mmol) and was obtained as white solid as a hydrochloride salt (6 mg, 0.01 mmol, 51.2% yield). MS (ESI): 689.2 [M+H] +

Example 51 and 52

(3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-eth yl)-l,2,4-oxadiazol-3-yl]-l-oxo-5-

[(4-phenoxyphenyl)methyl]-2,3-dihydro-lk4,5-benzothiazepi n-4-one [epimer A] and (3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl) -l,2,4-oxadiazol-3-yl]-l-oxo-5-

[(4-phenoxyphenyl)methyl]-2,3-dihydro-lk4,5-benzothiazepi n-4-one [epimer B]

Step a) tert-butyl N-[(3R)-7-cyano-8-fluoro-4-oxo-5-[(4-phenoxyphenyl)methyl]-2 ,3-dihydro-l,5- benzothiazepin-3-yl ] carbamate

The title compound was prepared in analogy to general procedure 4 from tert-butyl N-[(3R)-7- cyano-8-fluoro-4-oxo-3,5-dihydro-2H-l,5-benzothiazepin-3-yl] carbamate (Example 53 step d) (500 mg, 1.5 mmol) and was obtained as white solid (570 mg, 1.1 mmol, 73% yield). MS (ESI): 542.2 [M+Na] + .

Step b) tert-butyl N-[(3R)-8-fluoro-7-[(Z)-N'-hydroxycarbamimidoyl]-4-oxo-5-[(4 - phenoxyphenyl)methyl] -2, 3-dihydr o-l, 5-benzothiazepin-3-yl] carbamate The title compound was prepared in analogy to general procedure 12 from tert-butyl N-[(3R)-7-cyano- 8-fluoro-4-oxo-5-[(4-phenoxyphenyl)methyl]-2,3-dihydro-l,5-b enzothiazepin-3-yl]carbamate (200 mg, 0.34 mmol) and was obtained as white solid (210 mg, 95% yield). MS (ESI): 553.4 [M+H] + .

Step c) [ (Z)-[amino-[(3R)-3-(tert-butoxycarbonylamino)-8-fluoro-4-oxo -5-[(4- phenoxyphenyl)methyl] -2, 3-dihydro-l, 5-benzothiazepin- 7 -y I] methylene ] amino ] 2-methyl-2- methylsulfonyl-propanoate

The title compound was prepared in analogy to general procedure 10a from tert-butyl N-[(3R)-8- fluoro-7-[(Z)-N' -hydroxy carbamimidoyl]-4-oxo-5-[(4-tetrahydropyran-4-ylphenyl)methyl ]-2, 3- dihydro-1, 5-benzothiazepin-3-yl]carbamate (112.3 mg, 0.17 mmol) and was obtained as white powder (119.5 mg, 91% yield). MS (ESI): 559.3 [M-isobutene+H] + .

Step d) tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2, 4-oxadiazol-3-yl]-4- oxo-5-[(4-phenoxyphenyl)methyl]-2, 3-dihydro-l, 5-benzothiazepin-3-yl] carbamate The title compound was prepared in analogy to general procedure 11 from ) [(Z)-[amino-[(3R)-3-(tert- butoxycarbonylamino)-8-fluoro-4-oxo-5-[(4-phenoxyphenyl)meth yl]-2, 3-dihydro-l, 5- benzothiazepin-7-yl]methylene]amino] 2-methyl-2-methylsulfonyl-propanoate (119.5 mg, 0.16 mmol) and was obtained as white powder (89 mg, 81% yield). MS (ESI): 627.3 [M-isobutene+H] + .

Step e) tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2, 4-oxadiazol-3-yl]-

1, 4-dioxo-5-[ ( 4-phenoxyphenyl)methyl] -2, 3-dihydro-l 7. 4 , 5-benzothiazepin-3-yl carbamate (epimer A and tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl)-l,2, 4-oxadiazol-3-yl]-l,4- dioxo-5-[ ( 4-phenoxyphenyl)methyl -2, 3-dihydro-l . 4 , 5-benzothiazepin-3-yl] carbamate (epimer B)

The title compounds were prepared from tert-butyl N-[(3R)-8-fhroro-7-[5-(l -methyl- 1- methylsulfonyl-ethyl)-l, 2, 4-oxadiazol-3-yl]-4-oxo-5-[(4-phenoxyphenyl)methyl]-2, 3-dihydro-l, 5- benzothiazepin-3-yl]carbamate (89 mg, 0.13 mmol) in analogy to general procedure 5 using m-CPBA (28.6 mg, 0.13 mmol, 1 eq). The two epimers were separated and obtained as white powder (45.9 mg, 50% yield, epimer A) and white powder (21.9 mg, 24% yield, epimer B). MS (ESI): 643.2 [M+H- isobutene] + for both products.

Step fl) (3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl) -l,2,4-oxadiazol-3-yl]-l-oxo- 5-[(4-phenoxyphenyl)methyl]-2, 3-dihydro-l/. 4 , 5-benzothiazepin-4-one [epimer A ] The title compound was prepared from tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl- ethyl)-l,2,4-oxadiazol-3-yl]-l,4-dioxo-5-[(4-phenoxyphenyl)m ethyl]-2,3-dihydro-lX 4 ,5- benzothiazepin-3-yl]carbamate [epimer A] (45.9 mg, 0.07 mmol) in analogy to general procedure 6c and was obtained as white powder, as hydrochloride salt (39.7 mg, 88% yield). MS (ESI): 599.2

[M+H] + .

Step f2) (3R)-3-amino-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl-ethyl) -l,2,4-oxadiazol-3-yl]-l-oxo- 5-[ ( 4-phenoxyphenyl)methyl -2, 3-dihydro-l/. 4 , 5-benzothiazepin-4-one [ epimer B ]

The title compound was prepared from tert-butyl N-[(3R)-8-fluoro-7-[5-(l-methyl-l-methylsulfonyl- ethyl)-l,2,4-oxadiazol-3-yl]-l,4-dioxo-5-[(4-phenoxyphenyl)m ethyl]-2,3-dihydro-lX 4 ,5- benzothiazepin-3-yl]carbamate [epimer B] (21.9 mg, 0.03 mmol) in analogy to general procedure 6c and was obtained as white powder, as hydrochloride salt (20.2 mg, 94% yield). MS (ESI): 599.2 [M+H] + .

Example 63 (3R)-3-amino-5-[[4-(4-methoxyphenyl)phenyl]methyl]-7-[5-(l-m ethyl-l-methylsulfonyl-ethyl)- l,2,4-oxadiazol-3-yl]-l,l-dioxo-2,3-dihydro-lk6,5-benzothiaz epin-4-one

Step a) (2R)-2-(tert-butoxycarbonylamino)-3-(4-cyano-2-nitro-phenyl) sulfanyl-propanoic acid

To a solution of 4-fluoro-3 -nitrobenzonitrile (10.0 g, 60.2 mmol) in DCM (200 mL) was added (2R)- 2-(tert-butoxycarbonylamino)-3-sulfanyl-propanoic acid (14.6 g, 66.2 mmol, 1.1 eq) and DIPEA (20.5 mL, 120.4 mmol, 2 eq) and the mixture was stirred at RT for 16 h. The mixture was concentrated under reduced pressure and the remaining residue dissolved in EtOAc (200 mL), washed with 1 N aqueous HC1 (50 mL), water (100 mL), and brine (50 mL x 2). The organic phase was dried over sodium sulfate, filtered and concentrated under reduced pressure to give the title compound (24 g, 65.3 mmol, 77% yield) as a yellow solid. MS (ESI): 390.1 [M+Na] + .

Step b) (2R)-3-(2-amino-4-cyano-phenyl)sulfanyl-2-(tert-butoxycarbon ylamino)propanoic acid

The title compound was prepared from (2R)-2-(tert-butoxycarbonylamino)-3-(4-cyano-2-nitro- phenyl)sulfanyl-propanoic acid (10.0 g, 27.2 mmol) in analogy to general procedure 2 and was obtained as-brown solid (10 g, 87% yield). MS (ESI): 338.2 [M+H] + . Step c) tert-butyl N-[(3R)-7-cyano-4-oxo-3,5-dihydro-2H-l,5-benzothiazepin-3-yl ] carbamate

The title compound was prepared from (2R)-3-(2-amino-4-cyano-phenyl)sulfanyl-2-(tert- butoxycarbonylamino)propanoic acid (11.3 g, 33.5 mmol) in analogy to general procedure 3 and was obtained as-white solid (5 g, 45% yield). MS (ESI): 263.9 [M-isobutene+H] + .

Step d) tert-butyl N-[ (3R)~ 7-cyano-5-[ [4-( 4-methoxyphenyl )phenyl / methyl ]-4-oxo-2, 3-dihydro-l, 5- benzothiazepin-3-yl carbamate

To a solution of tert-butyl tert-butyl N-[(3R)-7-cyano-4-oxo-3,5-dihydro-2H-l,5-benzothiazepin-3- yl]carbamate (300.0 mg, 0.89 mmol, 1.0 eq), potassium iodide (0.01 g, 0.18 mmol, 0.2 eq), sodium carbonate (237 mg, 2.24 mmol, 2.5 eq) in DMF (2 mL) was added a solution of l-(chloromethyl)-4- (4-methoxyphenyl)benzene (218.5 mg, 0.94 mmol, 1.05 eq) in DMF (2 mL) at RT and the mixture was stirred at 40 °C for 12 h. The solution was poured into water and extracted with EtOAc (3x). The combined organic phase was washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The remaining residue was purified by column chromatography on silica gel (0-50% EtOAc in heptane) to give the title compound (460 mg, 0.89 mmol, 69% yield) as yellow solid. MS (ESI): 460.0 [M-isobutene+H] + .

Step e) tert-butyl N-[(3R)-7-cyano-5-[[4-(4-methoxyphenyl)phenyl]methyl]-l,l,4- trioxo-2,3- dihydro-1/.6, 5-benzothiazepin-3-yl] carbamate

The title compound was prepared from tert-butyl N-[(3R)-7-cyano-5-[[4-(4- methoxyphenyl)phenyl]methyl]-4-oxo-2,3-dihydro-l,5-benzothia zepin-3-yl]carbamate (450 mg, 0.9 mmol) in analogy to general procedure 5 and was obtained as-yellow solid (360 mg, 0.66 mmol, 44.4% yield). MS (ESI): 548.1 [M+H] + .

Step f) tert-butyl N-[(3R)-7-[(Z)-N'-hydroxycarbamimidoyl]-5-[[4-(4- methoxyphenyl)phenyl methyl -l, 1, 4-trioxo-2, 3-dihydro-l/.6, 5-benzothiazepin-3-yl carbamate

The solution of tert-butyl N-[(3R)-7-cyano-5-[[4-(4-methoxyphenyl)phenyl]methyl]-l,l,4- trioxo- 2,3-dihydro-lX6,5-benzothiazepin-3-yl]carbamate (350 mg, 0.64 mmol, 1.0 eq), sodium acetate (157.2 mg, 1.92 mmol, 3.0 eq) and hydroxylamine hydrochloride (66.6 mg, 0.96 mmol, 1.5 eq) in EtOH (5 m ) was stirred at 50 °C for 12 h. The reaction was concentrated in vacuum to give a suspension that was poured into water. The aqueous phase was extracted with EtOAc (3x). The combined organic phase was washed with brine, dried with anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the title compound (360 mg, 0.62 mmol, 97% yield) as yellow solid. MS (ESI): 581.4 [M+H] + . Step g) [(Z)-[amino-[(3R)-3-(tert-butoxycarbonylamino)-5-[ [4-(4-methoxyphenyl)phenyl] methyl] - 1, 1, 4-trioxo-2, 3-dihydro-l/.6, 5-benzothiazepin- 7 -yl] methylene ] amino] 2-methyl-2-methylsulfonyl- propanoate The title compound was prepared from tert-butyl N-[(3R)-7-[(Z)-N'-hydroxycarbamimidoyl]-5-[[4- (4-methoxyphenyl)phenyl]methyl]-l, 1,4-tri oxo-2, 3-dihydro- 1X6, 5-benzothiazepin-3-yl]carbamate (10 mg, 0.02 mmol, leq.) in analogy to general procedure 10a and was obtained as-yellow oil (10 mg, 0.01 mmol, 80% yield). MS (ESI): 729.0 [M+H] + .

Step h) tert-butyl N-[(3R)-5-[[4-(4-methoxyphenyl)phenyl]methyl]-7-[5-(l-methyl -l-methylsulfonyl- ethyl)- 1, 2, 4-oxadiazol-3-yl] -1 , 1, 4-trioxo-2, 3-dihydro-126, 5-benzothiazepin- 3-yl] carbamate

The title compound was prepared from [(Z)-[amino-[(3R)-3-(tert-butoxycarbonylamino)-5-[[4-(4- methoxyphenyl)phenyl]methyl]-l, 1,4-tri oxo-2, 3-dihydro-l X6,5-benzothi azepin-7- yl]methylene]amino] 2-methyl-2-methylsulfonyl-propanoate (280 mg, 0.4 mmol, leq.) in analogy to general procedure 11 and was obtained as-yellow solid (100 mg, 0.14 mmol, 37% yield). MS (ESI): 733.2 [M+Na] + .

Step i) ( 3R)-3-amino-5-[[ 4-(4-methoxyphenyl )phenyl methyl]- 7-[5-( I -methyl- 1-methylsulfonyl- ethyl)-l, 2, 4-oxadiazol-3-yl -l, l-dioxo-2, 3 -di hydro- 1/.6, 5-benzothiazepin-4-one

The title compound was prepared from tert-butyl N-[(3R)-5-[[4-(4-methoxyphenyl)phenyl]methyl]- 7-[5-(l -methyl- 1 -methyl sulfonyl-ethyl)- 1,2, 4-oxadiazol-3-yl]-l, 1,4-tri oxo-2, 3-dihydro- 1X6,5- benzothiazepin-3-yl]carbamate (95 mg, 0.13 mmol, leq.) in analogy to general procedure 6a and was obtained as-yellow solid, as hydrochloride salt (85.6 mg, 0.13 mmol, 96% yield). MS (ESI): 611.3 [M+H] + .

Example 68

(3R)-3-amino-7-[5-(l-methylsulfonylcyclobutyl)-l,3,4-oxad iazol-2-yl]-l,l-dioxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-4-one Step a) methyl (3R)-3-(tert-butoxycarbonylamino)-4-oxo-5-[(4-phenoxyphenyl) methyl]-2,3-dihydro-

1, 5-benzothiazepine- 7 -carboxylate

The title compound was prepared from methyl (3R)-3-(tert-butoxycarbonylamino)-4-oxo-3,5- dihydro-2H-l,5-benzothiazepine-7-carboxylate (1.0 g, 2.84 mmol, 1.0 eq) in analogy to general procedure 4 and was obtained as-white solid (1.38 g, 87% yield). MS (ESI): 479.2 [M-isobutene+H] + . Step b) (3R)-3-(tert-butoxycarbonylamino)-4-oxo-5-[(4-phenoxyphenyl) methyl]-2, 3-dihydro-l,5- benzothiazepine- 7 -carboxylic acid

The title compound was prepared from methyl (3R)-3-(tert-butoxycarbonylamino)-4-oxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-l,5-benzothiazepine-7-carb oxylate (1.38 g, 2.58 mmol, 1.0 eq) in analogy to general procedure 13 and was obtained as-yellow foam (1.54 g, 97% yield). MS (ESI):

465.1 [M-i sobutene+H] + .

Step c) (3R)-3-(tert-butoxycarbonylamino)-l, 1, 4-trioxo-5-[(4-phenoxyphenyl)methyl]-2, 3-dihydro-

I 6, 5-benzothiazepine- 7 -carboxylic acid

Sodium periodate (1.11 g, 5.19 mmol, 2.34 eq) was dissolved in water (8.5 mL) and the mixture was cooled to 0 °C under Ar. RuCh.SEEO (5.81 mg, 0.022 mmol, 0.01 eq) was added and the reaction stirred for 5 min. A solution of (3R)-3-(tert-butoxycarbonylamino)-4-keto-5-(4-phenoxybenzyl) -2,3- dihydro-1, 5-benzothiazepine-7-carboxylic acid (1.36 g, 2.22 mmol, 1.0 eq) in acetonitrile (8.54 mL) was then added in one portion and stirred for 3 h. The reaction was quenched upon addition of iPrOH (2 ml), EtOAc and 2M aqueous HC1. The grey suspension was filtered over a plug of celite and the filtrate extracted with EtOAc. The combined organic phase was washed with sat. aq. Na2S20s, dried over sodium sulfate and concentrated under reduced pressure to afford the title compound (1.3 g, 99%) as yellow solid. MS (ESI): 497.1 [M-isobutene+H] + .

Step d) tert-butyl N-[(3R)-7-(hydrazinecarbonyl)-l,l,4-trioxo-5-[(4-phenoxyphen yl)methyl]-2,3- dihydro-1/.6, 5-benzothiazepin-3-yl] carbamate

The title compound was prepared from (3R)-3-(tert-butoxycarbonylamino)-l,l,4-trioxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepine-7-ca rboxylic acid (1.3 g, 2.19 mmol, 1.0 eq) in analogy to general procedure 14 and was obtained as-white solid (980 mg, 79% yield). MS (ESI): 511.1 [M-isobutene+H] + . Step d) tert-butyl N-[(3R)-7-[[(l-methylsulfonylcyclobutanecarbonyl)amino]carba moyl]-l,l,4- trioxo-5-[(4-phenoxyphenyl)methyl]-2, 3-dihydro-l/.6, 5-benzothiazepin-3-yl] carbamate

The title compound was prepared from tert-butyl N-[(3R)-7-(hydrazinecarbonyl)-l,l,4-trioxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-3-yl] carbamate (500 mg, 0.88 mmol, 1.0 eq) in analogy to general procedure 7 (CAS 1250528-75-6) and was obtained as-yellow solid (260 mg, 41% yield). MS (ESI): 727.1 [M+H] + .

Step e) tert-butyl N-[(3R)-7-[5-(l-methylsulfonylcyclobutyl)-l,3,4-oxadiazol-2- yl]-l,l,4-trioxo-5- [ ( 4-phenoxyphenyl )methyl -2, 3-dihydro-l/.6, 5-benzothiazepin-3-yl carbamate

The title compound was prepared from tert-butyl N-[(3R)-7-[[(l- methylsulfonylcyclobutanecarbonyl)amino]carbamoyl]-l,l,4-tri oxo-5-[(4-phenoxyphenyl)methyl]- 2,3-dihydro-lX6,5-benzothiazepin-3-yl]carbamate (260 mg, 0.36 mmol) in analogy to general procedure 8 and was obtained as-yellow oil (120 mg, 0.17mmol, 45% yield). MS (ESI): 653.1 [M- isobutene+H] + . Step e) (3R)-3-amino-7-[ 5-(l -methylsulfonylcyclobutyl)-!, 3, 4-oxadiazol-2-yl] -1 , l-dioxo-5-[(4- phenoxyphenyl)methyl] -2, 3-dihydro-l/.6, 5-benzothiazepin-4-one

The title compound was prepared from tert-butyl N-[(3R)-7-[5-(l-methylsulfonylcyclobutyl)-l,3,4- oxadiazol-2-yl]-l,l,4-trioxo-5-[(4-phenoxyphenyl)methyl]-2,3 -dihydro-lk6,5-benzothiazepin-3- yl]carbamate (50 mg, 0.07 mmol) in analogy to general procedure 6a and was obtained as-off-white solid, as hydrochloride salt (35 mg, 0.05mmol, 64% yield). MS (ESI): 609.2 [M+H] + .

The examples of the following table can be prepared in a similar manner to Example 68, using the appropriate carboxylic acid building block. as a hydrochloride salt

Example 65

2- [5- [(3R)-3-amino- 1, 1 ,4-trioxo-5- [ [4- [4-(trifluoromethyl)phenyl] phenyl] methyl] -2,3-dihydro- lk6,5-benzothiazepin-7-yl]-l,3,4-oxadiazol-2-yl]-N,N-dimethy l-propane-2-sulfonamide

Step a) methyl (3R)-3-(tert-butoxycarbonylamino)-4-oxo-5-[[4-[4-

(trifhioromethyl)phenyl] phenyl] methyl] -2, 3-dihydro-l,5-benzothiazepine-7 -carboxylate

The title compound was prepared from methyl (3R)-3-(tert-butoxycarbonylamino)-4-oxo-3,5- dihydro-2H-l,5-benzothiazepine-7-carboxylate (CAS:2089150-62-7) (400 mg, 1.14 mmol) in analogy to general procedure 4 and was obtained as light brown oil (420 mg, 0.72 mmol, 60% yield). MS (ESI): 587.3 [M+H] + .

Step b) ( 3R)-3-(tert-butoxycarbonylamino)-4-oxo-5-[ [ 4-[ 4-(trifluoromethyl )phenyl phenyl] methyl /- 2, 3-dihydro-l , 5 -benzothiazepine-7 -carboxylic acid

The title compound was prepared from methyl (3R)-3-(tert-butoxycarbonylamino)-4-oxo-5-[[4-[4- (trifluoromethyl)phenyl]phenyl]methyl]-2,3-dihydro-l,5-benzo thiazepine-7-carboxylate (400 mg, 0.68 mmol, 1.0 eq) in analogy to general procedure 13 and was obtained as yellow solid (350 mg, 0.61 mmol, 86% yield). MS (ESI): 517.2 [M-isobutene+H] + .

Step c) (3R)-3-(tert-butoxycarbonylamino)-l, 1, 4-trioxo-5-[[ 4-[ 4-

(trijluoromethyl)phenyl] phenyl] methyl] -2, 3-dihydro-l/.6, 5-benzothiazepine-7 -carboxylic acid

To a solution of sodium periodate (821.8 mg, 3.84 mmol, 2.0 eq) in water (8 mL) was added RuCL (3.98 mg, 0.02 mmol, 0.01 eq) at 0 °C and stirred for 10 min. A solution of (3R)-3-(tert- butoxycarbonylamino)-4-oxo-5-[[4-[4-(trifluoromethyl)phenyl] phenyl]methyl]-2,3-dihydro-l,5- benzothiazepine-7-carboxylic acid (1100 mg, 1.92 mmol, 1.0 eq) in acetonitrile (12 mL) was added to the mixture at 0 °C and stirred at RT for 2 h. The reaction mixture was quenched with slow addition of isopropanol (4 mL) under stirring. Then EtOAc (50 mL) was added to the mixture, which was then carefully acidified with 0.5 N aqueous HC1 to pH = 4-5. The grey suspension was filtered through a plug of celite and the filtrate extracted with EtOAc (3x). The combined organic extracts were washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure to give the title compound (1150 mg, 1.9 mmol, 87% yield) as light green solid. MS (ESI): 603.1 [M-H]

Step d) tert-butyl N-[(3R)-7-(hydrazinecarbonyl)-l,l,4-trioxo-5-[[4-[4-

(trifluoromethyl)phenyl phenyl methyl] -2, 3-dihydro-l 6, 5-benzothiazepin-3-yl] carbamate The title compound was prepared from (3R)-3-(tert-butoxycarbonylamino)-l,l,4-trioxo-5-[[4-[4- (trifluoromethyl)phenyl]phenyl]methyl]-2,3-dihydro-lX6,5-ben zothiazepine-7-carboxylic acid (2.3 g, 3.8 mmol) in analogy to general procedure 14 and was obtained as light brown solid (2.2 g, 3.56 mmol, 76% yield). MS (ESI): 563.2 [M-isobutene+H] + .

Step e) tert-butyl N-[(3R)-7-[ [ [2-(dimethylsulfamoyl)-2-methyl-propanoyl] amino] carbamoyl] -1 , 1 ,4- trioxo-5-[[ 4-[ 4-(trifhioromethyl )phenyl phenyl methyl -2, 3-dihydro-l/.6, 5-benzothiazepin-3- yl] carbamate

To a solution of 2-(dimethylsulfamoyl)-2-methyl-propanoic acid (132.55 mg, 0.68 mmol, 2.1 eq) and N,N-diisopropylethylamine (83.57 mg, 0.65 mmol, 2.0 eq) in DMF (2 mL) was added HATU (91.3 mg, 0.39 mmol, 1.2 eq) at RT. The mixture was stirred 25 min and then a solution of tert-butyl N- [(3R)-7-(hydrazinecarbonyl)-l,l,4-trioxo-5-[[4-[4-(trifluoro methyl)phenyl]phenyl]methyl]-2,3- dihydro-lX6,5-benzothiazepin-3-yl]carbamate (200 mg, 0.32 mmol, 1.0 eq) in DMF (2 mL) was added and stirred for 5h at RT. The reaction mixture was poured into water, EtOAc was added and the layers were separated. The aqueous phase was extracted with EtOAc (3x). The combined organic extracts were washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The remaining residue was purified by preparative TLC (100% EtOAc) to give the title compound (110 mg, 0.14 mmol, 36% yield) as light yellow oil. MS (ESI): 740.2 [M-isobutene+H] + .

Step f) tert-butyl N-[ (3R)-7-[5-[ I -(dimethylsulfamoyl)-l-methyl-ethyl]-l, 3, 4-oxadiazol-2-yl]-l , 1, 4- trioxo-5-[[ 4-[ 4-(trifluoromethyl )phenyl phenyl methyl -2, 3-dihydro-l .6, 5-benzothiazepin-3- yl] carbamate

The title compound was prepared from tert-butyl N-[(3R)-7-[[[2-(dimethylsulfamoyl)-2-methyl- propanoyl]amino]carbamoyl]-l,l,4-trioxo-5-[[4-[4-(trifluorom ethyl)phenyl]phenyl]methyl]-2,3- dihydro-lX6,5-benzothiazepin-3-yl]carbamate (100 mg, 0.13 mmol) in analogy to general procedure 8 in toluene and was obtained as light yellow oil (55 mg, 0.07 mmol, 56%). MS (ESI): 722.2 [M- isobutene+H] + .

Step f) tert-butyl N-[ (3R)-7-[5-[ I -(dimethylsulfamoyl)-l-methyl-ethyl]-l, 3, 4-oxadiazol-2-yl]-l, 1, 4- trioxo-5-[[ 4-[ 4-(trifluoromethyl )phenyl phenyl methyl -2, 3-dihydro-l/.6, 5-benzothiazepin-3- yl] carbamate

The title compound was prepared from tert-butyl N-[(3R)-7-[5-[l -(dimethyl sulfamoyl)- 1 -methylethyl]-!, 3, 4-oxadiazol-2-yl]- 1,1, 4-trioxo-5-[[4-[4-(tri fluoromethyl )phenyl]phenyl]methyl]-2, 3- dihydro-lX6,5-benzothiazepin-3-yl]carbamate (50.0 mg, 0.06 mmol) in analogy to general procedure 6a and was obtained as white solid (19.2 mg, 0.03 mmol, 41% yield). MS (ESI): 678.2 [M+H] + . The example of the following table can be prepared in a similar manner to Example 65, using the appropriate carboxylic acid building block.

Example 54 (3R)-3-amino-8-fluoro-7-[2-(methylsulfonylmethyl)tetrazol-5- yl]-l,l-dioxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-lk6,5-benzothiazepin-4-one

Step a) tert-butyl N-[(3R)-7-cyano-8-fluoro-l, 1, 4-trioxo-5-[(4-phenoxyphenyl)methyl]-2, 3-dihydro- /z 6, 5-benzothiazepin-3-yl carbamate

The title compound was prepared from tert-butyl N-[(3R)-7-cyano-8-fluoro-4-oxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-l,5-benzothiazepin-3-yl]ca rbamate (Example 51 and 52, step a) (0.57 g, 1.1 mmol) in analogy to general procedure 5 and was obtained as white solid (600 mg, 1.09 mmol, 98% yield). MS (ESI): 574.3 [M+Na] + .

Step b) tert-butyl N-[(3R)-8-fluoro-l,l,4-trioxo-5-[(4-phenoxyphenyl)methyl]-7- (2H-tetrazol-5-yl)- 2, 3 -dihydro- 1 6, 5-benzothiazepin-3-yl] carbamate

To a solution of tert-butyl N-[(3R)-7-cyano-8-fluoro-l,l,4-trioxo-5-[(4-phenoxyphenyl)me thyl]-2,3- dihydro-lX6,5-benzothiazepin-3-yl]carbamate (500.0 mg, 0.91 mmol, 1.0 eq) in toluene (10 mL) was added azido(trimethyl)silane (0.48 mL, 3.63 mmol, 4.0 eq) and dibutyltin oxide (50 mg, 0.2 mmol, 0.22 eq, CAS 818-08-6). The mixture was degassed with N2 (3x) and then stirred at 100 °C for 3 h. After cooling to RT, the mixture was filtered and the filter cake was washed with toluene (5 mL) and petroleum ether (5 mL). The filtrate was concentrated under reduced pressure to obtain the title compound (530 mg, 0.89 mmol, 90% yield) as grey solid. MS (ESI): 617.3 [M+Na] + .

Step c) tert-butyl N-[(3R)-8-fluoro-7-[2-(methylsulfanylmethyl)tetrazol-5-yl]-l ,l,4-trioxo-5-[(4- phenoxyphenyl)methyl] -2, 3-dihydro-l 6, 5-benzothiazepin-3-yl carbamate and tert-butyl N-[ ( 3R)-8- fluoro- 7-[ I -(methylsulfanylmethyl) tetrazol-5-yl ]-l, 1, 4-trioxo-5-[ ( 4-phenoxyphenyl)methyl]-2, 3- di hydro- 1/.6, 5-benzothiazepin-3-yl] carbamate

To a solution of tert-butyl N-[(3R)-8-fluoro-l,l,4-trioxo-5-[(4-phenoxyphenyl)methyl]-7- (2H- tetrazol-5-yl)-2,3-dihydro-lX6,5-benzothiazepin-3-yl]carbama te (200 mg, 0.34 mmol, 1.0 eq), K2CO3 (92.8 mg, 0.67 mmol, 2.0 eq) and KI (40.0 mg, 0.24 mmol, 0.71 eq) in DMF (4 mL) was added chloromethyl methyl sulfide (0.03 mL, 0.37 mmol, 1.1 eq) at RT and the mixture was stirred for 16 h. The mixture was diluted with EtOAc (5 mL) and washed with brine (5 mL x 3), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The remaining residue was purified by column chromatography on silica gel (50% EtOAc in petroleum ether) to give tert-butyl N-[(3R)-8- fluoro-7-[2-(methylsulfanylmethyl)tetrazol-5-yl]-l,l,4-triox o-5-[(4-phenoxyphenyl)methyl]-2,3- dihydro-lX6,5-benzothiazepin-3-yl]carbamate (90 mg, 0.14 mmol, 41% yield) as white solid and tertbutyl N-[(3R)-8-fluoro-7-[l-(methylsulfanylmethyl)tetrazol-5-yl]-l ,l,4-trioxo-5-[(4- phenoxyphenyl)methyl]-2,3-dihydro-lX6,5-benzothiazepin-3-yl] carbamate (16 mg, 0.02 mmol, 7% yield) as white solid. MS (ESI): 699.2 [M-isobutene+H] + for both products.

Step d) tert-butyl N-[(3R)-8-fluoro-7-[2-(methylsulfonylmethyl)tetrazol-5-yl]-l ,l,4-trioxo-5-[(4- phenoxyphenyl)methyl] -2, 3-dihydro-l/.6, 5-benzothiazepin-3-yl (carbamate The title compound was prepared from tert-butyl N-[(3R)-8-fluoro-7-[2- (methylsulfanylmethyl)tetrazol-5-yl]-l,l,4-trioxo-5-[(4-phen oxyphenyl)methyl]-2,3-dihydro-lX6,5- benzothiazepin-3-yl]carbamate (80 mg, 0.15 mmol, 1.0 eq) in analogy to general procedure 5 and was obtained as white solid (90 mg, 0.13 mmol, 78% yield). MS (ESI): 631.2 [M-isobutene+H] + . Step e) (3R)-3-amino-8-fluoro-7-[2-(methylsulfonylmethyl)tetrazol-5- yl]-l,l-dioxo-5-[(4- phenoxyphenyl)methyl] -2, 3-dihydro-l/.6, 5-benzothiazepin-4-one

The title compound was prepared from tert-butyl N-[(3R)-8-fluoro-7-[2- (methyl sulfanyl methyl )tetrazol-5-yl]-l ,l , 4-trioxo-5-[(4-phenoxyphenyl)methyl]-2, 3 -dihydro- IX6, 5- benzothiazepin-3-yl]carbamate (90 mg, 0.13 mmol, 1.0 eq) in analogy to general procedure 6a and was obtained as white solid, as hydrochloride salt (40.2 mg, 0.06 mmol, 49% yield). MS (ESI): 587.3 [M+H] + .

The example of the following table was prepared in a similar manner to Example 54 (step d and e) from the indicated starting material.

* as hydrochloride salt

Intermediate 1 l-(bromomethyl)-4-(2-methoxy-l,l-dimethyl-ethoxy)benzene Step a) ethyl 2-methyl-2-(4-methylphenoxy)propanoate

A mixture of p-cresol (3.5 g, 32.3 mmol, 1.26 eq), ethyl 2-bromoisobutyrate (5 g, 25.6 mmol, 1.0 eq) and potassium carbonate (10 g, 72.3 mmol, 2.8 eq) in DMF (20 mL) was stirred at RT for 12 h. The mixture was poured into water (100 mL) and extracted with EtOAc (200 mL). The organic phase was washed with brine (100 mL), dried over sodium sulfate and concentrated under reduced pressure. The remaining residue was purified by column chromatography (2-20% EtOAc in petroleum ether) to give the title compound (2.5 g, 11.2 mmol, 42% yield) as colorless oil. MS (ESI): 223.1 [M+H] +

Step b) 2-methyl-2-(4-methylphenoxy)propan-l-ol

To a solution of ethyl 2-methyl-2-(4-methylphenoxy)propanoate (1.0 g, 4.5 mmol, 1.0 eq) in THF (10 mL) was added LiAlFLj (550.0 mg, 14.5 mmol, 3.2 eq) slowly at 0 C under argon. Upon complete addition, the solution was allowed to warm to RT and stirred for 2 h. The solution was poured into water (100 mL), and extracted with EtOAc (300 mL). The organic phase was washed with brine (200 mL), dried over sodium sulfate and concentrated under reduced pressure to give the title compound (680 mg, 3.77 mmol, 80% yield) as colorless oil. 'H NMR (400 MHz, DMSO-d 6 ) 8 = 7.07 (d, 2H), 6.88 (d, 2H), 4.85 (s, 1H), 2.25 (s, 3H), 1.15 (s, 6H).

Step c) l-(2-methoxy-l, 1 -dimethyl-ethoxy) -4-methyl-benzene

To a solution of 2-methyl-2-(4-methylphenoxy)propan-l-ol (1500 mg, 8.32 mmol, 1.0 eq) in THF (15 mL) was added NaH (900 mg, 22.5 mmol, 2.7 eq) slowly at 0 C under nitrogen atmosphere. The solution was stirred at RT for 30min. Then iodomethane (1.1 mL, 17.6 mmol, 2.12 eq) was added to the mixture and stirred at RT for 4 h. The mixture was poured into water (20 mL) and extracted with EtOAc (50 mL). The organic phase was washed with brine (50 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give the title compound (1.5 g, 7.72 mmol, 91% yield) as yellow oil. X H NMR (400 MHz, CDC1 3 ) 6 = 7.07 (d, 2H), 6.91 (d, 2H), 4.13 (q, J= 7.1 Hz, 1H), 3.44 (s, 3H), 3.36 (s, 2H), 2.32 (s, 3H), 1.28 (s, 6H).

Step d) l-(bromomethyl)-4-(2-methoxy-l,l-dimethyl-ethoxy)benzene

A solution of N-bromosuccinimide (1400 mg, 7.87 mmol, 1.02 eq), 1 -(2 -methoxy- 1,1 -dimethyl - ethoxy)-4-methyl-benzene (1500 mg, 7.72 mmol, 1.0 eq) and AIBN (170.0 mg, 1.04 mmol, 0.13 eq) in CCU (15 mL) was stirred at 80°C for 3 h. The mixture was poured into saturated aq. NaHCCL (30 mL) and extracted with DCM (100 mL). The organic phase was washed with brine (100 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The remaining crude was purified by prep-TLC (0-10% EtOAc in petroleum ether) to give the title compound (1.8 g, 6.59 mmol, 85% yield) as brown oil. 'H NMR (400 MHz, CDCI3) 8 = 7.22 (d, 2H), 6.92 (d, 2H), 4.42 (s, 2H), 3.35 (s, 3H), 3.28 (s, 2H), 1.23 (s, 6H).

Intermediate 6 l-(chloromethyl)-4-[3-(difluoromethoxy)phenoxy]benzene

Step a) 4-[3-(difluoromethoxy)phenoxy]benzaldehyde To a mixture of 3-(difhioromethoxy)phenylboronic acid (461.69 mg, 2.46 mmol, 1.5 eq) and 4- hydroxybenzaldehyde (200 mg, 1.64 mmol, 1 eq) in 1,4-dioxane (15 mL) were added pyridine (0.53 mL, 6.55 mmol, 4 eq) and copper diacetate (735.69 mg, 3.68 mmol, 2.25 eq) at RT. The mixture was then heated to 80 °C for 12 h. The reaction was allowed to cool to RT and diluted with water (3 mL). The resulting solution was extracted with EtOAc (3x5 mL) and the combined organic extracts were dried over sodium sulfate, filtered and concentrated under reduced pressure. The remaining residue was purified by column chromatography on silica gel (4-40% EtOAc in petroleum ether) to afford the title compound (90 mg, 0.34 mmol, 15% yield) as yellow oil. MS (ESI): 264.9 [M+H] + .

Step b) [4-[3-(difluoromethoxy)phenoxy]phenyl]methanol

To a mixture of 4-[3-(difluoromethoxy)phenoxy]benzaldehyde (140 mg, 0.5 mmol, 1 eq) in THF (4 mL) was added Li AIH4 (27.75 mg, 0.7 mmol, 1.38 eq) at 0 °C under nitrogen atmosphere. The reaction was stirred for 1 h at 0 °C. The mixture was quenched by addition of water (0.24 mL), IM aqueous NaOH (0.04 mL), and water (0.12 ml). The mixture was filtered and the filtrate concentrated under reduced pressure to afford a yellow oil (110 mg) containing the title product which was used in the next step without further purification. MS (ESI): 248.9 [M-0H] + .

Step c) l-(chloromethyl)-4-[3-(difluoromethoxy)phenoxy] benzene

To a solution of [4-[3-(difluoromethoxy)phenoxy]phenyl]methanol (110 mg, 0.410 mmol, 1 eq) in DCM (10 mL) was added thionyl chloride (0.18 mL, 2.48 mmol, 6 eq) and stirred at RT for 16 h. The mixture was concentrated and the remaining residue was dissolved in THF (2 mL) and concentrated again under reduced pressure. This process was repeated twice to give the title compound (110 mg, 0.39 mmol, 69% yield) as yellow oil. MS (ESI): 334.0 [M+H+piperidine] + .

Intermediate 7 l-(chloromethyl)-4-[3-(trifluoromethoxy)phenoxy] benzene

Step a) 4-[3-(trifluoromethoxy)phenoxy]benzaldehyde

To a mixture of 3-(trifluoromethoxy)phenol (1.44 g, 8.06 mmol, 1 eq) and 4-fluoro-benzaldehyde (0.86 mL, 8.06 mmol, 1 eq) in DMF (50 mL) was added cesium carbonate (3.94 g, 12.09 mmol, 1.5 eq) at RT. The reaction was heated to 100 °C for 12 h. The mixture was quenched by addition of water (150 mL) and extracted with EtOAc (4x50 mL). The combined organic layers were washed with brine (100 mL), dried over dry sodium sulfate, filtered and concentrated under reduced pressure. The remaining residue was purified by column chromatography on silica gel (5-90% EtOAc in PE) to afford the title compound (2 g, 7.09 mmol, 87% yield) as yellow oil. MS (ESI): 282.9 [M+H] + .

Step b) [4-[3-(trifluoromethoxy)phenoxy]phenyl]methanol

To a mixture of 4-[3-(trifluoromethoxy)phenoxy]benzaldehyde (1.0 g, 3.54 mmol, 1 eq) in THF (15 mL) was added LiAlEL (185.57 mg, 4.89 mmol, 1.38 eq) at 0 °C und nitrogen and stirred for 1 h. The mixture was quenched by addition of water (0.05 mL) and IM aqueous NaOH (0.05 mL) and water (0.15 ml). The mixture was filtered and the filtrate concentrated under reduced pressure to afford a yellow oil (1 g) containing the title product which was used in the next step without further purification. MS (ESI): 266.9 [M-0H] + .

Step c) l-(chloromethyl)-4-[3-(trifluoromethoxy)phenoxy]benzene

To a solution of [4-[3-(trifluoromethoxy)phenoxy]phenyl]methanol (800.0 mg, 2.81 mmol, 1 eq) in DCM (15 mL) was added thionyl chloride (1.23 mL, 16.89 mmol, 6 eq) and the mixture was stirred at 20 °C for 16 h. The mixture was concentrated and the remaining residue was dissolved in THF (2 mL) and concentrated again under reduced pressure. This process was repeated twice to give the title compound (780 mg, 2.58 mmol, 68% yield) as yellow oil. MS (ESI): 352.0 [M+H+piperidine] + .

Intermediate 10 l-[4-(chloromethyl)phenyl]-4-methoxy-2-methyl-benzene

Step a) 4-(4-methoxy-2-methyl-phenyl)benzaldehyde

To a mixture of 4-formylphenylboronic acid (1.64 g, 10.9 mmol, 1.1 eq) and 4-bromo-3-methylanisole (2 g, 9.95 mmol, 1 eq) in toluene (15 mL) were added 2M aq. Na2COs (4.97 ml, 9.95 mmol, 1 eq) and Pd(PPhs)4 (344.85 mg, 0.3 mmol, 0.03 eq) in EtOH (25 ml) at RT under nitrogen. The mixture was then stirred for 16 h. The reaction was diluted with water (15 mL). The resulting solution was extracted with EtOAc (3x15 mL) and the combined organic extracts were dried over sodium sulfate, filtered and concentrated. The remaining residue was purified by column chromatography on silica gel (5- 60% EtOAc in PE) to afford the title compound (1.6 g, 7.07 mmol, 65% yield) as yellow oil. MS (ESI): 226.9 [M+H] + . Step b) [4-(4-methoxy-2-methyl-phenyl)phenyl] methanol

To a mixture of 4-(4-methoxy-2-methyl-phenyl)benzaldehyde (600 mg, 2.27 mmol, 1 eq) in THF (10 mL) was added LiAlTLj (118.93 mg, 3.13 mmol, 1.38 eq) at 0 °C under nitrogen atmosphere. The reaction was stirred for 1 h at 0 °C. The mixture was quenched by addition of water (0.24 mL) and IM aqueous NaOH (0.24 mL) and water (0.75 ml). The mixture was filtered and the filtrate concentrated under vacuum to afford a yellow oil (640 mg) containing the title product which was used in the next step without further purification. MS (ESI): 211.0 [M-0H] + .

Step c) l-[4-(chloromethyl)phenyl]-4-methoxy-2-methyl-benzene

To a solution of [4-(4-methoxy-2-methyl-phenyl)phenyl]methanol (640 mg, 2.8 mmol, 1 eq) in DCM (10 mL) was added thionyl chloride (1.22 mL, 16.8 mmol, 6 eq) and stirred atRT for 16 h. The mixture was concentrated and the remaining residue was dissolved in THF (2 mL) and concentrated again. This process was repeated twice to give the title compound (600 mg, 2.43 mmol, 78% yield) as yellow oil. MS (ESI): 296.0 [M+H+piperidine] + .

Intermediate 8 and 9 of the following table were prepared in analogy to Intermediate 10, using the appropriate benzyl halide building block.

Intermediate 11

2-[4-(bromomethyl)phenyl]-5-(trifluoromethoxy)pyridine Step a) 2-(p-tolyl)-5-(trifluoromethoxy)pyridine

A mixture of p-tolylboronic acid (9.4 g, 69.1 mmol, 1 eq) and K2CO3 (28.6 g, 207 mmol, 3 eq) in 1,4- di oxane (244 mL) and water (24.4 mL) was degassed with argon for 10 minutes. Then 1,1- bis(diphenylphosphino)ferrocene dichloro palladium(II) (1.02 g, 1.38 mmol, 0.02 eq) was added at room temperature. The reaction mixture was heated to 80°C and stirred for 7 hours, partitioned between ethyl acetate (450 ml) and water (225 ml) and the layers were separated. The aqueous layer was extracted with 150 ml portion of ethyl acetate. The combined organic layers were washed with 150 ml portion of brine dried over MgSC and concentrated in vacuo. The crude material was purified by flash chromatography (SiCh, 330g, 0-10% EtOAc in heptane) to yield the title compound as white solid (13 g, 68%, 92 % purity). MS (ESI): 254.1 [M+H] + .

Step b) 2-[4-(bromomethyl)phenyl]-5-(trifluoromethoxy)pyridine

To a solution of 2-(p-tolyl)-5-(trifluoromethoxy)pyridine (1015 mg, 4 mmol, 1 eq) in acetonitrile, extra dry (17 m ) was added N-bromsuccinimide (749 mg, 4.21 mmol, 1.05 eq) and AIBN (197 mg, 1.2 mmol, 0.3 eq). The reaction mixture was stirred at 80°C overnight, poured into brine and extracted 3 times with EtOAc. The combined organic phases were dried over Na2SO4, filtered and concentrated. The crude product was purified using flash chromatography (SiO2, 40g, 0-10% EtOAc in heptane) to yield the title compound as white solid (971 mg, 62%, 86 % purity). MS (ESI): 332.0 [M+H] + .

2) Biological examples

2.1) In vitro DGK Inhibition Assays

DGK a and C, kinases use ATP to phosphorylate the substrate 1,2-dilauroyl-sn-glycerol (DLG, incorporated in the liposomes). ATP is converted to ADP as a result of this enzymatic reaction.

After the kinase reaction, an ATP-depletion reagent is added to terminate the kinase reaction and deplete any remaining ATP, leaving only ADP. Second, a detection reagent is added to simultaneously convert ADP to ATP and allow the newly synthesized ATP to be converted to light using a coupled luciferase/luciferin reaction.

Reagents and Material

Buffer Ingredients (solutions & salts)

Protein / Substrates / Tracer

Full length DGKA and Z were expressed in Sf21 insect cells by infecting the cells with the baculovirus stock at MOI of 2. Purification of both enzymes was performed as previously described by Takahashi et al., PeerJ, 2018 (Takahashi, D.; Sakane, F. Expression and purification of human diacylglycerol kinase alpha from baculovirus-infected insect cells for structural studies. PeerJ 2018, 6, No. e5449).

Hardware Assay Buffer (30ml)

Assay procedure A concentrated liposome solution was prepared in assay buffer without DTT and BSA: 2mM of DLG in 21 mM of total liposome (2 mM DLG / 8 mM PS / 11 mM PC). The reaction mixtures contain the assay buffer with a final DLG concentration of 125uM ATP concentrations of 25pM (for DGKA assay) or 50 pM (for DGKZ assay). The reactions were started by addition of DGK a and C, kinases at 4 nM and 2 nM final concentrations, respectively. After 1 hour reaction, the amount of ADP formed was detected with the ADP-Glo kinase assay (Promega) according to the manufacturer instructions.

Compounds were added in 11-points dose response, starting at lOmM, 1 :3 dilutions, with a final DMSO concentration of 2%. The multidrop combi was used as a liquid handler and luminescence was read with 0.5 s by the envision reader (PE). Results

In vitro DGK Inhibition Assays (ADP Gio)

DGK a and C, kinases use ATP to phosphorylate the substrate 1,2-dilauroyl-sn-glycerol (DLG). ATP is converted to ADP as a result of this enzymatic reaction.

After the kinase reaction, an ATP-depletion reagent is added to terminate the kinase reaction and deplete any remaining ATP, leaving only ADP. Second, a detection reagent is added to simultaneously convert ADP to ATP and allow the newly synthesized ATP to be converted to light using a coupled luciferase/luciferin reaction. Experimental Procedure, Reagents and Material DGK a and C, kinase ADP Gio assays were ran by Reaction Biology Corp., 1 Great Valley Parkway, Suite 2, Malvern, 19355, PA, USA. Information provided by the service provider are the following: DGK a and C, kinases were used at 2 nM final concentration. Reactions were carried out at 50 pM ATP. 500 uM of the substrate DLG (Dilauroyl-sn-glycerol) was used. Compounds were received at 10 mM DMSO stock solution and were tested in 10-dose IC50 duplicate with 3 -fold serial dilution starting at 1 pM. A control compound, Calphostin C, was tested in 10-dose IC50 with 3-fold serial dilution starting at 100 pM.

Results:

2.2) IL2 secretion measurements

As readouts for T-cell activation, IL2 secretion after 24 hours and proliferation after 5 days was measured. Increases in IL2 secretion and proliferation upon compound treatment were assessed as the % of the maximum of reference compound Al. WO 2016/139181 discloses reference compound Al as example 70. As a counter screen and to make sure that no unwanted TCR- independent activation was triggered, PBS conditions were run for all compounds.

Reagents and Material

Cell Culture

Expanded primary human T-cells were thawed and cultured in RPMI 1640 (Gibco, #61870-010) + 5% human serum (HS, Sigma, #H3667) + ImM Sodium Pyruvate (Gibco, #11360-039) + 50pM 2- mercaptoethanol (Gibco, #31350-010) and lx Pen-Strep (Life Technologies, #15140122) medium at density of 2 Mio/ml for 3 hours in 5% CO2, 37°C and 95% humidity. For coating of plates, PBS++ with PBS— or PBS++ with CD3 antibody (concentration depending on donor and determined by CD3 titrations) was added lOOpl/well to Poly-D Lysine coated 96-well plates. Plates were sealed and incubated at room temperature for 3 hours on a table-top rocking platform. After incubation, plates were washed once with PBS— and filled with 40pl/well culture medium only. Compounds were then added (see next section) to medium only plates. After 3 hours of culturing the T-cells, cells were filtered through a cell strainer (Miltenyi Biotech, #130-041-407), counted again and concentration was adjusted to 1.25 Mio/ml.

Cells were then seeded 80pl/well to the 40pl/well including dispensed compounds according to plate layout. By adding cells, compounds were further diluted 1 :3, and resulting in 100k cells/120pl/well. After 24 hours 40pl of supernatant was collected carefully from the top while not disturbing the cells and transferred into a round bottom 96well plate. Collected and frozen supernatant was used for detection of IL2 using the IL-2 Human ProQuantum Immunoassay Kit (Invitrogen) or using the Human IL-2 ELISA Kit (Thermo Fisher).

Compound treatment Compounds were added in a 5 or 6pt dose response with the Tecan D300e Digital Dispenser, all conditions 3 times more concentrated than the end-concentration, since cells are added afterwards (80pl cells to 40pl prepared medium with treatment). The DR was starting at 20pM or lOpM final top concentration and a dilution factor of 3.333. The positive control was the reference compound Al that was added in a dose response as well, additionally to 3 wells of only 20pM representing the positive stimulator control. All wells were normalized with DMSO to a final concentration of 0.6% (0.2% end-concentration).

I 1.2 ProQuantum Immunoassay

The immunoassay is done following the manufacturer’s manual (Invitrogen, # A35603).

Additional information: For the immunoassay, MicroAmp™ EnduraPlate™ Optical 384-Well plates are used. Frozen supernatant is thawed and centrifuged for 5 minutes at lOOOxg, both steps at 4°C. After centrifugation, required sample amount is taken from the top, and in a separate LightCycler V- bottom plate (working plate) diluted with assay dilution buffer, dilution factor depending on the PBS or CD3 condition but at least 1 :3. IL-2 standard and blanks are prepared in the same V-bottom plate, standard with a range of 0.0128-5000pg/ml (extended version). After preparation, 5 pl of sample dilutions or standard/blanks are transferred to the optical 384-well plate (assay plate) and the lOpl reaction protocol is being followed. For measurement, the QuantStudio 12K Flex system is used. Raw data is extracted and IL-2 concentrations are calculated with the Thermo Fisher online app (app s .thermofi sher . com/ app s/ proquantum) .

IL2 Elisa

ELISA is done following the manufacturer’s manual (Thermo Fisher Scientific, #88-7025-88). Additional information: For the ELISA Nunc MaxiSorp 96 well plates are used. Frozen supernatant is thawed and centrifuged for 5 minutes at lOOOxg, both steps at 4°C. After that, required sample amount is taken from the top, and in a separate V-bottom plate diluted with ELISA diluent, dilution factor depending on the PBS or CD3 condition. IL-2 standard and blanks are prepared in the same V-bottom plate. After preparation, 50pl of sample dilutions and lOOpl of standard or blanks are transferred to the Nunc plates.

Calculations and data reporting CD3 and PBS plates were analysed separately in Genedata Screener using Roche Normalization PCT POS CTRL with DMSO set as Neutral Control and 20pM of the reference compound Al set as Stimulator Control/100%.

For CD3 conditions EC50 and Emax of the fitted sigmoidal curve were reported. If no curve could be fitted, the EC50 was reported as blank field and the Emax was based on individual data points.

The Emax did not always correspond to the highest concentration tested. Compounds which activate unstimulated cells or compounds which negatively affected viability (see proliferation assay) were flagged.

Results 2,4) Proliferation assay

Reagents and Material

Expanded primary human T-cells are thawed and cultured in RPMI 1640 (Gibco, #61870-010) + 5% human serum (HS, Sigma, #H3667) + ImM Sodium Pyruvate (Gibco, #11360-039) + 50pM 2- mercaptoethanol (Gibco, #31350-010) and lx Pen-Strep (Life Technologies, #15140122) medium at density of 2 Mio/ml for 3 hours in 5% CO2, 37°C and 95% humidity. For coating of plates, PBS++ only or PBS++ with CD3 antibody (concentration depending on donor and determined by CD3 titrations) is added lOOpl/well to Poly-D Lysine coated 96-well plates. Plates are sealed and incubated at room temperature for 3 hours on a table-top rocking platform. After incubation, plates are washed once with PBS— and filled with 40pl/well culture medium only. Compounds are then added (see next section) to medium only plates. After 3 hours of culturing the T-cells, cells are filtered through a cell strainer (Miltenyi Biotech, #130-041-407), counted again and concentration is adjusted to 1.25 Mio/ml.

Cells are then seeded 80pl/well to the 40pl/well including dispensed compounds according to plate layout. By adding cells, compounds are further diluted 1 :3, and resulting in 100k cells/120pl/well. After 48 hours 40pl of supernatant is collected carefully from the top while not disturbing the cells. Cells are assessed for proliferation 5 days later by measuring ATP consumption using CellTiterGlo (Promega).

Compound treatment

Compounds were added in a 5 or 6pt dose response with the Tecan D300e Digital Dispenser, all conditions 3 times more concentrated than the end-concentration, since cells are added afterwards (80pl cells to 40pl prepared medium with treatment). The DR was starting at 20pM or lOpM final top concentration and a dilution factor of 3.333. The positive control was the reference compound Al that was added in a dose response as well, additionally to 3 wells of only 20pM representing the positive stimulator control. All wells were normalized with DMSO to a final concentration of 0.6% (0.2% end-concentration).

Cell Titer Gio Measurements

After 5 days, for detection of ATP which is directly proportional to the number of cells present per well, the CellTiter-Glo® 2.0 Reagent is used. After visual control for toxicity or precipitations of the tested compounds, the plates are equilibrated to room temperature for 45 minutes. CellTiter-Glo® 2.0 Reagent is equilibrated to room temperature as well. After equilibration, an equal amount of CellTiter-Glo reagent is added to the cells (80pl/well) with an electronic multichannel pipette. Plates are placed on a rocking platform for 15 minutes at room temperature. After incubation, the bottom of the plates is sealed with backing tape. Luminescence is measured with PHERAstar FSX (interval time 0.5sec, gain 3000, focal height 15mm) and exported as CSV file for analysis in Genedata screener.

Calculations and data reporting CD3 and PBS plates were analysed separately in Genedata Screener using Roche Normalization PCT POS CTRL with DMSO set as Neutral Control and 20pM of the reference compound Al set as Stimulator Control/100%.

For CD3 conditions EC50 and Emax of the fitted sigmoidal curve were reported. If no curve could be fitted, the EC50 was reported as blank field and the Emax was based on individual data points.

The Emax did not always correspond to the highest concentration tested. Compounds which activate unstimulated cells (see IL2 measurements) or compounds which negatively affected viability were flagged. Results 2,4) T-cell - TCB - MV3 killing assays Reagents and Material

MACS filter (cell strainer) Miltenyi Biotech, #130- 041-407

Cell Culture

All culturing steps are executed at 5% C02, 37°C and 95% humidity.

MV-3 RFP cells are cultured in MV-3 medium (DMEM + 10% FBS, lx PenStrep and 0.5 pg/mL Puromycin) for at least 3 weeks. Cultured MV-3 cells at 80% confluency are washed once with PBS- - and trypsinized until detached. Cells are then counted and resuspended to 1*105 cells/mL in T-cell medium (RPMI 1640 + 5% human serum + ImM Sodium Pyruvate + 50pM 2-mercaptoethanol and lx Pen-Strep). Cells are seeded with 100 pL/well into a 96-well plate (TTP, #92696), and placed for 40 minutes without moving at room temperature in order to achieve evenly distributed attachment of cells. Plates are then incubated until further use.

On the next day, expanded primary human T-cells are thawed and resuspended in T-cell medium to 4*106 cells/mL. For 3 hours, they are cultured in a 6-well plate with 6 mL per well at maximum. After culturing the T-cells, they are filtered through a cell strainer (Miltenyi Biotech, #130-041-407), counted again, and cell concentration is adjusted to 2*106 cells/mL.

Compound treatment

MCSP-TCB or PBS are pre-diluted in T-cell medium (concentration depending on T-cell donor), 4 times more concentrated than the end-concentration. 60 pL/well of pre-dilutions are then distributed into a round bottom plate (Costar, #3799) according to plate layout. Compounds are added in a 9pt dose response with the Tecan D300e Digital Dispenser, as well 4 times more concentrated than the end-concentration. DMSO concentration of all wells is adjusted to 0.8 %, resulting in 0.2 % as final concentration.

60 pL per well of T-cell suspension are added to the prepared round bottom plate and resuspended with a manual multichannel. 100 pL/well of the resuspended T-cell suspension including treatments are then transferred cautiously to the over-night cultured MV-3 cells according to plate layout. 100 pL T-cell medium only is added to the outer MV-3 wells only. Final compound DR is starting at 20 pM with a dilution factor of 3.333. Final TCB concentration is between 1.5 pM to 5 pM and was determined for each T-cell donor individually by running TCB titrations. For each donor, a TCB concentration was chosen which corresponds to 10-20% of MV3 baseline cell killing in the absence of compound treatment. Positive control is the reference compound Al which is added in a DR, as well as additional wells with only 20 pM. 20 pM of reference compound Al represent the positive stimulator control, TCB only (DMSO wells) the neutral control.

Calculations

After transfer of T-cells with treatment pre-dilutions, MV-3 cells are imaged by time-lapse microscopy using IncucyteZOOM™ (Essen BioScience, MI, USA). Imaging is performed every 3 hours for a total of 120 hours (10X objective, phase and red image channels, acquisition time 400 ms, Green/Red 4614 optical module). RFP object count per well is analysed in the IncucyteZOOM™ Software (Version 2019B Rev2) with a mask that was previously created and optimized for MV-3 cells. Raw data is exported as object count/well and values are normalized as % TCL compared to wells with MV-3 only, representing 100% growth and therefore 0% TCL.

RFP measurements

Calculated % TCL values are analysed in Genedata Screener using Roche Normalization PCT POS CTRL with MCSP-TCB only set as Neutral Control and 20 pM of the reference compound Al set as Stimulator Control/100%.

EC50 and Emax values were provided in the table below.

Induced TCL by compounds without TCB treatment or toxicity (observed in the PBS condition) were be flagged.

Results