Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CYCLOPROPYL FUSED THIAZINE DERIVATIVES AS BETA-SECRETASE INHIBITORS AND METHODS OF USE
Document Type and Number:
WIPO Patent Application WO/2018/112086
Kind Code:
A1
Abstract:
The present disclosure provides a new class of compounds useful for the modulation of beta-secretase enzyme (BACE) activity. The compounds have a general Formula (I) wherein variables A, R1, R2, R2', R3, R4, and R5 of Formula (I) are defined herein. This disclosure also provides pharmaceutical compositions comprising the compounds, and uses of the compounds and compositions for treatment of disorders and/or conditions related to Aβ plaque formation and deposition, resulting from the biological activity of BACE. Such BACE mediated disorders include, for example, Alzheimer's Disease, cognitive deficits, cognitive impairments, and other central nervous system conditions.

Inventors:
ALLEN JENNIFER R (US)
AMEGADZIE ALBERT (US)
BOURBEAU MATTHEW P (US)
CHEN JIAN J (US)
FROHN MICHAEL J (US)
HARRINGTON PAUL E (US)
LOW JONATHAN D (US)
MA VU V (US)
NGUYEN THOMAS T (US)
PICKRELL ALEXANDER J (US)
REEVES COREY (US)
Application Number:
PCT/US2017/066184
Publication Date:
June 21, 2018
Filing Date:
December 13, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
AMGEN INC (US)
International Classes:
C07D417/10; A61K31/5415; A61P25/28; C07D279/08; C07D417/14
Domestic Patent References:
WO2014013076A12014-01-23
WO2000017369A22000-03-30
WO2014098831A12014-06-26
WO2014099794A12014-06-26
WO2014099788A12014-06-26
WO2014097038A12014-06-26
WO2014093190A12014-06-19
WO2014066132A12014-05-01
WO2014065434A12014-05-01
WO2014062553A12014-04-24
WO2014062549A12014-04-24
WO2014045162A12014-03-27
WO2014013076A12014-01-23
WO2013182638A12013-12-12
WO2013164730A12013-11-07
WO2013030713A12013-03-07
WO2013028670A12013-02-28
WO2013004676A12013-01-10
WO2012162334A12012-11-29
WO2012162330A12012-11-29
WO2012147762A12012-11-01
WO2012139425A12012-10-18
WO2012138734A12012-10-11
WO2011029803A12011-03-17
WO2011005738A12011-01-13
WO2010013794A12010-02-04
WO2010013302A12010-02-04
WO2009151098A12009-12-17
WO2009134617A12009-11-05
WO2012146762A12012-11-01
WO2016022724A12016-02-11
WO2011069934A12011-06-16
Foreign References:
US20160046618A12016-02-18
US5441870A1995-08-15
US5712130A1998-01-27
US5942400A1999-08-24
US20120245157A12012-09-27
US20120245154A12012-09-27
US20120238557A12012-09-20
US20110152253A12011-06-23
US20100160290A12010-06-24
US20100075957A12010-03-25
US20090209755A12009-08-20
US20090082560A12009-03-26
EP2703401A12014-03-05
EP1942105A12008-07-09
Other References:
YAN ET AL., LANCETNEUROL., vol. 13, no. 3, 2014, pages 319 - 329
JOACHIM ET AL., ALZHEIMER. DIS. ASSOC. DISORD., vol. 6, no. 1, 1992, pages 7 - 34
SELKOE, NEURON, vol. 6, no. 4, 1991, pages 487 - 498
SEUBERT ET AL., NATURE, vol. 359, 1992, pages 325 - 327
CITRON, TRENDS PHARMACOL. SCI., vol. 25, no. 2, 2004, pages 92 - 97
SHANKAR ET AL., NAT. MED., vol. 14, no. 8, 2008, pages 837 - 842
SINHA ET AL., NATURE, vol. 402, 1999, pages 537 - 540
SABBAGH ET AL., ALZHEIMER'S DISEASE REVIEW, vol. 3, 1997, pages 1 - 19
COLE ET AL., MOLECULAR NEURODEGENERATION, vol. 2, no. 22, 2007, pages 1 - 25
LUO ET AL., NAT. NEUROSCI., vol. 4, no. 3, 2001, pages 231 - 232
HENLEY ET AL., EXPERT OPIN. PHARMACOTHER., vol. 10, no. 10, 2009, pages 1657 - 1664
SIEMERS ET AL., CLIN. NEUROPHARMACOL., vol. 30, no. 6, 2007, pages 317 - 325
SIEMERS ET AL., NEUROLOGY, vol. 66, no. 4, 2006, pages 602 - 604
YAN ET AL., LANCET NEUROLOGY, vol. 13, no. 3, 2014, pages 319 - 329
CATHEPSIN B; L. KOIKE ET AL., MOL. CELL NEUROSCI., vol. 22, no. 2, 2003, pages 146 - 161
SHACKA ET AL., AUTOPHAGY, vol. 3, no. 5, 2007, pages 474 - 476
FOLIO ET AL., PLOS ONE, vol. 6, no. 7, 2011, pages e21908
"Martin Citron's presentation at the AD/PD Conference 3-2011", ALZHEIMER'S RESEARCH FORUM NEWS, 31 March 2011 (2011-03-31)
JACQUES ET AL.: "Enantiomers, Racemates and Resolutions", 1981, WILEY-INTERSCIENCE
WILEN ET AL., TETRAHEDRON, vol. 33, pages 2725
ELIEL: "Stereochemistry of Carbon Compounds", 1962, MCGRAW-HILL
WILEN: "Tables of Resolving Agents and Optical Resolutions", 1972, UNIV. OF NOTRE DAME PRESS, pages: 268
BERGE ET AL., J. PHARM. SCI., vol. 66, no. 1, 1977, pages 1 - 19
STAHL ET AL.: "Pharmaceutical Salts: Properties, Selection, and Use", 2011
R. LAROCK: "Comprehensive Organic Transformations", 1989, VCH PUBLISHERS
T.W. GREENE; P.G.M. WUTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY AND SONS
L. FIESER; M. FIESER: "Fieser and Fieser's Reagents for Organic Synthesis", 1994, JOHN WILEY AND SONS
A. KATRITZKY; A. POZHARSKI, HANDBOOK OF HETEROCYCLIC CHEMISTRY, 2001
M. BODANSZKY; A. BODANSZKY: "The Practice of Peptide Synthesis", 1984, SPRINGER-VERLAG
J. SEYDEN-PENNE: "Reductions by the Alumino- and Borohydrides in Organic Synthesis", 1997, WILEY-VCH
"Encyclopedia of Reagents for Organic Synthesis", 1995, JOHN WILEY AND SONS
GULNIK ET AL., FEBS LETT., vol. 413, no. 2, 1997, pages 379 - 384
YASUDA ET AL., J BIOCHEM., vol. 125, no. 6, 1999, pages 1137 - 1143
HSIAO ET AL., SCIENCE, vol. 274, 1996, pages 99 - 102
DOVEY ET AL., J NEUROCHEM., vol. 76, no. 1, 2001, pages 173 - 181
BEST ET AL., J. PHARMACOL. EXP. THER., vol. 313, no. 2, 2005, pages 902 - 908
DE MEYER ET AL.: "re-affirm the putative role that the accumulation of A(3 peptide in cerebral spinal fluid (CSF) in a subject plays in the progression of symptoms, initially revealed as mild cognitive impairment, which ultimately leads to AD", ARCH NEUROL., vol. 67, no. 8, 2010, pages 949 - 956
TANZI ET AL., CELL, vol. 120, no. 4, 2005, pages 545 - 555
WALSH ET AL., NEURON, vol. 44, no. 1, 2004, pages 181 - 193
PALOP ET AL., NAT. NEUROSCI., vol. 13, no. 7, 2010, pages 812 - 818
SELKOE, BEHAV. BRAIN RES., vol. 192, no. 1, 2008, pages 106 - 113
GAMES ET AL., NATURE, vol. 373, 1995, pages 523 - 527
GOTZ ET AL., MOL. PSYCHIATRY, vol. 9, no. 7, 2004, pages 664 - 683
HSIA ET AL., PROC. NATL. ACADEMY OF SCIENCE, vol. 96, 1999, pages 3228 - 3233
HARRIS ET AL., NEURON, vol. 68, 2010, pages 428 - 441
AD. VASSAR ET AL., LANCET NEUROL., vol. 13, 2014, pages 319 - 329
YAN, TRANSL. NEURODEGENER., vol. 5, no. 13, 2016, pages 1 - 11
VASSAR ET AL., LANCET NEUROL., vol. 13, 2014, pages 319 - 329
Attorney, Agent or Firm:
BERGAUER, Markus (US)
Download PDF:
Claims:
What is claimed is:

1. A compound of Formula I

I

or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein

A is N, CH, or CR4;

R1 is H, -CN, Cwalkyl, Ca-ealkenyl, -Ci-4alkyl-C(0)NR1 Rr, -Ci-4alkyl-C(0)- heterocycloalkyl, -(HC=CH)-C(0)NR1'R1', -(HC=CH)-C(0)-heterocycloalkyl, -C(0)NR1 R1', or -C(0)-heterocycloalkyl, wherein the Ci-ealkyl and the C2-ealkenyl are (i) optionally substituted with one to three fluoro substituents or (ii) optionally substituted with -CN, OH, methoxy, or a 5-membered nitrogen-containing heteroaryl, wherein the 5-membered nitrogen-containing heteroaryl is optionally substituted with

R1 is, independently, H or

R2 and R2 are independently H or halogen;

R3 is Ci-4alkyl, wherein the is optionally substituted with one to three fluoro substituents;

R4 is halogen;

R5 is H, C3-6Cycloalkyl, phenyl, or 5- or 6-membered heteroaryl, wherein the phenyl or heteroaryl is optionally substituted with one to three substituents independently selected from halogen, -CN, Cwalkyl, 2-propynyloxy, 2-butynyloxy, or oxazolylmethoxy.

2. The compound according to Claim 1, or a tautomer thereof, or a

pharmaceutically acceptable salt of said compound or tautomer, wherein the compound of Formula I is a compound of Formula II

II.

3. The compound according to Claim 1 , or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein the compound of Formula I is a compound of Formula III

III.

4. The compound according to Claim 1, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein the compound of Formula I is a compound of Formula ΙΙΓ

ΙΙΓ .

5. The compound according to any one of Claims 1-4, or a tautomer thereof,

6. The compound according to any one of Claims 1-5, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein R2 and R2 are H.

7. The compound according to any one of Claims 1-5, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein R2 and R2 are F.

8. The compound according to any one of Claims 1-7, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein R3 is methyl, - CH2F, or CHF2.

9. The compound according to any one of Claims 1-7, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein R3 is methyl or- CH2F. - no

10. The compound according to any one of Claims 1-9, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein R4 is F.

1 1. The compound according to any one of Claims 1-10, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein R5 is H, cyclopropyl, phenyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, or pyrazinyl, wherein the phenyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, or pyrazinyl is optionally substituted with one or two substituents independently selected from F, CI, -CN, methyl, 2-propynyloxy, 2-butynyloxy, or 2-oxazolylmethoxy.

12. The compound according to any one of Claims 1-1 1, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein

- I l l -

13. The compound of Claim 1 or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, selected from

6-((3-((lS,5S,6S)-3-amino-5-(fluoromethyl)-l-(hydroxymethyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

6-((3 -(( 1 S,5 S,6S)-3 -amino- 1 ,5 -bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 - en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

(lS,5S,6S)-5-(5-((5-chloropyridin-2-yl)ethynyl)-2-fluorophenyl)-l,5- bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-3 -amine ;

5 -((3 -(( 1 S,5 S,6S)-3 -amino- 1 ,5 -bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 - en-5-yl)-4-fluorophenyl)ethynyl)pyrazine-2-carbonitrile;

6-((3 -(( 1 S,5 S,6S)-3 -amino- 1 ,5 -bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 - en-5-yl)-4-fluorophenyl)ethynyl)-5-methylnicotinonitrile;

(lS,5S,6S)-5-(5-((5-(but-2-yn-l-yloxy)pyridin-2-yl)ethynyl)-2-fluorophenyl)-l,5- bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-3 -amine ;

6-((3-((lR,5S,6S)-3-amino-l-(cyanomethyl)-5-(fluoromethyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

6-((3-((lR,5S,6S)-3-amino-l-((E)-2-cyanovinyl)-5-(fluoromethyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

(lS,5S,6S)-5-(2-fluoro-5-((5-(oxazol-2-ylmethoxy)pyridin-2-yl)ethynyl)phenyl)-l,5- bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-3 -amine ;

2-((3 -(( 1 S,5 S,6S)-3 -amino- 1 ,5 -bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 - en-5-yl)-4-fluorophenyl)ethynyl)thiazole-5-carbonitrile;

6-((3-((lS,5S,6R)-3-amino-7,7-difluoro-l-(hydroxymethyl)-5-methyl-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile; 6-((3 -(( 1 R,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -((E)-3 -morpholino-3 -oxoprop- 1 -en- 1 - yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

(E)-3-((lR,5S,6S)-3-amino-5-(5-((5-cyanopyridin-2-yl)ethynyl)-2-fluorophenyl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)-N,N-dimethylacrylamide;

2-(( 1 R,5 S,6S)-3 -amino-5 -(5 -((5 -cyanopyridin-2-yl)ethynyl)-2-fluorophenyl)-5 - (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)acetamide;

6-((5-((lS,5S,6S)-3-amino-l-(fluoromethyl)-5-methyl-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-6-fluoropyridin-3-yl)ethynyl)nicotinonitrile;

6-((3-((lS,5S,6S)-3-amino-5-(fluoromethyl)-l-((4-methyl-lH-l,2,3-triazol-l- yl)methyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

6-((3 -(( 1 R,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -((E)-3 -morpholino-3 -oxoprop- 1 -en- 1 - yl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-5 -yl)-4-fluorophenyl)ethynyl)-5 - methylnicotinonitrile ;

(E)-3-((lR,5S,6S)-3-amino-5-(5-((5-chloropyrimidin-2-yl)ethynyl)-2-fluorophenyl)- 5 -(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en- 1 -yl)- 1 -morpholinoprop-2-en- 1 -one;

(E)-3-((lR,5S,6S)-3-amino-5-(2-fluoro-5-((3-memylisothiazol-5-yl)ethynyl)phenyl)- 5 -(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en- 1 -yl)- 1 -morpholinoprop-2-en- 1 -one;

6-((3 -(( 1 S,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -(3 -ιηοφ1ιο1ίηο-3 -oxopropyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

6-((3 -(( 1 S,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -(3 -morpholino-3 -oxopropyl)-2-thia-4- azabicyclo [4.1.0]hept-3 -en-5 -yl)-4-fluorophenyl)ethynyl)-5 -methylnicotinonitrile;

2-((3 -(( 1 R,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -((E)-3 -ιηοφ1ιο1ίηο-3 -oxoprop- 1 -en- 1 - yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)pyrimidine-5- carbonitrile;

4-((3 -(( 1 R,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -((E)-3 -ιηοφ1ιο1ίηο-3 -oxoprop- 1 -en- 1 - yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)benzonitrile;

(lS,5S,6S)-3-amino-5-(5-((5-cyano-3-methyl-2-pyridinyl)ethynyl)-2-fluorophenyl)- N,N,5-trimethyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxamide;

6-((3 -(( 1 R,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -((E)-3 -moφholino-3 -oxoprop- 1 -en- 1 - yl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-5 -yl)-4-fluorophenyl)ethynyl)-5 -fluoronicotinonitrile; 6-((3-((lS,5S,6S)-l-((lH-imidazol-l-yl)methyl)-3-amino-5-(fluoromethyl)-2-thia-4- azabicyclo [4.1.0]hept-3 -en-5 -yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

(lS,5S,6S)-3-amino-5-(5-((5-cyano-3-methylpyridin-2-yl)ethynyl)-2-fluorophenyl)- 5-methyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carbonitrile;

(lS,5S,6S)-3-amino-5-(5-((5-cyano-3-methylpyridin-2-yl)ethynyl)-2-fluorophenyl)- 5 -(fluoromethyl)-N,N-dimethyl-2-thia-4-azabicyclo [4.1.0]hept-3 -ene- 1 -carboxamide ;

6-((3 -(( 1 S,5 S,6S)-3 -amino-5 -methyl- 1 -(pyrrolidine- 1 -carbonyl)-2-thia-4- azabicyclo [4.1.0]hept-3 -en-5 -yl)-4-fluorophenyl)ethynyl)-5 -methylnicotinonitrile;

(lS,5S,6S)-3-amino-5-(5-((4-cyanophenyl)ethynyl)-2-fluorophenyl)-N,N,5- trimethyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxamide;

(lS,5S,6S)-3 -amino-5 -(2-fluoro-5 -((5 -(2-propyn- 1 -yloxy)-2- pyridinyl)ethynyl)phenyl)-N,N,5-trimethyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l- carboxamide;

(lS,5S,6S)-3-amino-5-(2-fluoro-5-((3-methyl-5-(oxazol-2-ylmethoxy)pyridin-2- yl)ethynyl)phenyl)-N,N,5 -trimethyl-2-thia-4-azabicyclo [4.1.0]hept-3 -ene- 1 -carboxamide ;

(lS,5S,6S)-3-amino-5-(5-(cyclopropylethynyl)-2-fluorophenyl)-N,N,5-trimethyl-2- thia-4-azabicyclo[4.1.0]hept-3-ene- 1 -carboxamide;

6-((3-((lS,5S)-3-amino-l-(methoxymethyl)-5-methyl-2-thia-4-azabicyclo[4.1.0]hept 3-en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

( 1 S,5 S)-5 -(5 -((5 -chloro-3 -methylpyridin-2-yl)ethynyl)-2 -fluorophenyl)- 1 - (methoxymethyl)-5-methyl-2-thia-4-azabicyclo[4.1.0]hept-3-en-3-amine;

6-((3-((lS,5S)-3-amino-l-(methoxymethyl)-5-methyl-2-thia-4-azabicyclo[4.1.0]hept 3 -en-5 -yl)-4-fluorophenyl)ethynyl)-5 -methylnicotinonitrile;

6-((5-((lS,5S)-3-amino-5-methyl-l-(moφholine-4-carbonyl)-2-thia-4- azabicyclo [4.1.0]hept-3 -en-5-yl)-6-fluoropyridin-3 -yl)ethynyl)-5 -methylnicotinonitrile;

6-((5 -(( 1 S,5 S)-3 -amino-5 -methyl- 1 -(pyrrolidine- 1 -carbonyl)-2-thia-4- azabicyclo [4.1.0]hept-3 -en-5-yl)-6-fluoropyridin-3 -yl)ethynyl)-5 -methylnicotinonitrile;

(lS,5S,6S)-5-(5-((4-chloropyridin-2-yl)ethynyl)-2-fluorophenyl)-l,5- bis(fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-3-amine; or

(lS,5S,6S)-3-amino-5-(5-ethynyl-2-fluorophenyl)-N,N,5-trimethyl-2-thia-4- azabicyclo[4.1.0]hept-3-ene-l-carboxamide.

14. The compound according to any one of Claims 1-10, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein R5 is phenyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, or pyrazinyl, wherein the phenyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, or pyrazinyl is optionally substituted with one or two substituents independently selected from -CN, methyl, 2-propynyloxy, 2-butynyloxy, or 2- oxazolylmethoxy .

15. The compound according to any one of Claims 1-1 1, or a tautomer thereof,

16. The compound of Claim 1, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, selected from 6-((3-((lS,5S,6S)-3-amino-5-(fluoromethyl)-l-(hydroxymethyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

6-((3 -(( 1 S,5 S,6S)-3 -amino- 1 ,5 -bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 - en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

(lS,5S,6S)-5-(5-((5-chloropyridin-2-yl)ethynyl)-2-fluorophenyl)-l,5- bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-3 -amine ;

5 -((3 -(( 1 S,5 S,6S)-3 -amino- 1 ,5 -bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 - en-5-yl)-4-fluorophenyl)ethynyl)pyrazine-2-carbonitrile;

6-((3 -(( 1 S,5 S,6S)-3 -amino- 1 ,5 -bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 - en-5-yl)-4-fluorophenyl)ethynyl)-5-methylnicotinonitrile;

(lS,5S,6S)-5-(5-((5-(but-2-yn-l-yloxy)pyridin-2-yl)ethynyl)-2-fluorophenyl)-l,5- bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-3 -amine ;

6-((3-((lR,5S,6S)-3-amino-l-(cyanomethyl)-5-(fluoromethyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

6-((3-((lR,5S,6S)-3-amino-l-((E)-2-cyanovinyl)-5-(fluoromethyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

(lS,5S,6S)-5-(2-fluoro-5-((5-(oxazol-2-ylmethoxy)pyridin-2-yl)ethynyl)phenyl)-l,5- bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-3 -amine ;

2-((3 -(( 1 S,5 S,6S)-3 -amino- 1 ,5 -bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 - en-5-yl)-4-fluorophenyl)ethynyl)thiazole-5-carbonitrile;

6-((3-((lS,5S,6R)-3-amino-7,7-difluoro-l-(hydroxymethyl)-5-methyl-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

6-((3 -(( 1 R,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -((E)-3 -morpholino-3 -oxoprop- 1 -en- 1 - yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

(E)-3-((lR,5S,6S)-3-amino-5-(5-((5-cyanopyridin-2-yl)ethynyl)-2-fluorophenyl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)-N,N-dimethylacrylamide;

2-(( 1 R,5 S,6S)-3 -amino-5 -(5 -((5 -cyanopyridin-2-yl)ethynyl)-2-fluorophenyl)-5 - (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)acetamide;

6-((5-((lS,5S,6S)-3-amino-l-(fluoromethyl)-5-methyl-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-6-fluoropyridin-3-yl)ethynyl)nicotinonitrile; 6-((3-((lS,5S,6S)-3-amino-5-(fluoromethyl)-l-((4-methyl-lH-l,2,3-triazol-l- yl)methyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

6-((3 -(( 1 R,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -((E)-3 -ιηοφ1ιο1ίηο-3 -oxoprop- 1 -en- 1 yl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-5 -yl)-4-fluorophenyl)ethynyl)-5 - methylnicotinonitrile ;

(E)-3-((lR,5S,6S)-3-amino-5-(2-fluoro-5-((3-memylisothiazol-5-yl)ethynyl)phenyl)- 5 -(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en- 1 -yl)- 1 -morpholinoprop-2-en- 1 -one;

6-((3 -(( 1 S,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -(3 -ιηοφ1ιο1ίηο-3 -oxopropyl)-2-thia-4 azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

6-((3 -(( 1 S,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -(3 -ιηοφ1ιο1ίηο-3 -oxopropyl)-2-thia-4 azabicyclo [4.1.0]hept-3 -en-5 -yl)-4-fluorophenyl)ethynyl)-5 -methylnicotinonitrile;

2-((3 -(( 1 R,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -((E)-3 -ιηοφ1ιο1ίηο-3 -oxoprop- 1 -en- 1 yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)pyrimidine-5- carbonitrile;

4-((3 -(( 1 R,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -((E)-3 -ιηοφ1ιο1ίηο-3 -oxoprop- 1 -en- 1 yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)benzonitrile;

(lS,5S,6S)-3-amino-5-(5-((5-cyano-3-methyl-2-pyridinyl)ethynyl)-2-fluorophenyl)- N,N,5-trimethyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxamide;

6-((3-((lS,5S,6S)-l-((lH-imidazol-l-yl)methyl)-3-amino-5-(fluoromethyl)-2-thia-4- azabicyclo [4.1.0]hept-3 -en-5 -yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

(lS,5S,6S)-3-amino-5-(5-((5-cyano-3-methylpyridin-2-yl)ethynyl)-2-fluorophenyl)- 5-methyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carbonitrile;

(lS,5S,6S)-3-amino-5-(5-((5-cyano-3-methylpyridin-2-yl)ethynyl)-2-fluorophenyl)- 5 -(fluoromethyl)-N,N-dimethyl-2-thia-4-azabicyclo [4.1.0]hept-3 -ene- 1 -carboxamide ;

6-((3 -(( 1 S,5 S,6S)-3 -amino-5 -methyl- 1 -(pyrrolidine- 1 -carbonyl)-2-thia-4- azabicyclo [4.1.0]hept-3 -en-5 -yl)-4-fluorophenyl)ethynyl)-5 -methylnicotinonitrile;

(lS,5S,6S)-3-amino-5-(5-((4-cyanophenyl)ethynyl)-2-fluorophenyl)-N,N,5- trimethyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxamide;

( 1 S,5 S,6S)-3 -amino-5 -(2-fluoro-5 -((5 -(2-propyn- 1 -yloxy)-2- pyridinyl)ethynyl)phenyl)-N,N,5-trimethyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l- carboxamide; (lS,5S,6S)-3-amino-5-(2-fluoro-5-((3-methyl-5-(oxazol-2-ylmethoxy)pyridin-2- yl)ethynyl)phenyl)-N,N,5 -trimethyl-2-thia-4-azabicyclo [4.1.0]hept-3 -ene- 1 -carboxamide ;

6-((3-((lS,5S)-3-amino-l-(methoxymethyl)-5-methyl-2-thia-4-azabicyclo[4.1.0]hept- 3-en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

( 1 S,5 S)-5 -(5 -((5 -chloro-3 -methylpyridin-2-yl)ethynyl)-2 -fluorophenyl)- 1 - (methoxymethyl)-5-methyl-2-thia-4-azabicyclo[4.1.0]hept-3-en-3-amine;

6-((3-((lS,5S)-3-amino-l-(methoxymethyl)-5-methyl-2-thia-4-azabicyclo[4.1.0]hept- 3 -en-5 -yl)-4-fluorophenyl)ethynyl)-5 -methylnicotinonitrile;

6-((5-((lS,5S)-3-amino-5-methyl-l-(moφholine-4-carbonyl)-2-thia-4- azabicyclo[4.1 ]hept-3-en-5-yl)-6-fluoropyridin-3-yl)ethynyl)-5-methylnicotinonitrile; or

6-((5 -(( 1 S,5 S)-3 -amino-5 -methyl- 1 -(pyrrolidine- 1 -carbonyl)-2-thia-4- azabicyclo [4.1.0]hept-3 -en-5-yl)-6-fluoropyridin-3 -yl)ethynyl)-5 -methylnicotinonitrile .

17. A pharmaceutical composition comprising the compound according to any one of Claims 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, and a pharmaceutically acceptable excipient.

18. A compound according to any one of Claims 1 - 16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, or the pharmaceutical composition according to Claim 17 for use as a medicament.

19. A compound according to any one of Claims 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, or the pharmaceutical composition according to Claim 17 for use in reducing beta amyloid peptide levels in the cerebral spinal fluid of a subject.

20. A compound according to any one of Claims 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, or the pharmaceutical composition according to Claim 17 for use in treating Alzheimer's disease, cognitive impairment, or a combination thereof in a subject.

21. A compound according to any one of Claims 1 - 16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, or the pharmaceutical composition according to Claim 17 for use in treating a neurological disorder selected from mild cognitive impairment, Down's syndrome, hereditary cerebral hemorrhage with Dutch- type amyloidosis, cerebral amyloid angiopathy, degenerative dementia, dementia associated with Parkinson's disease, dementia associated with supranuclear palsy, dementia associated with cortical basal degeneration, diffuse Lewy body type of Alzheimer's disease, or a combination thereof in a subject.

22. A compound according to any one of Claims 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, or the pharmaceutical composition according to Claim 17 for use in reducing formation of plaque on the brain of a subject.

23. Use of the compound according to any one of Claims 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, or the pharmaceutical composition according to Claim 17 in the preparation of a medicament for reducing beta amyloid peptide levels in the cerebral spinal fluid of a subject.

24. Use of the compound according to any one of Claims 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, or the pharmaceutical composition according to Claim 17 in the preparation of a medicament for treating Alzheimer's disease, cognitive impairment, or a combination thereof in a subject.

25. Use of the compound according to any one of Claims 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, or the pharmaceutical composition according to Claim 17 in the preparation of a medicament for the treatment of a neurological disorder selected from mild cognitive impairment, Down's syndrome, hereditary cerebral hemorrhage with Dutch-type amyloidosis, cerebral amyloid angiopathy, degenerative dementia, dementia associated with Parkinson's disease, dementia associated with supranuclear palsy, dementia associated with cortical basal degeneration, diffuse Lewy body type of Alzheimer's disease, or a combination thereof in a subject.

26. Use of a compound according to any one of Claims 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, or the pharmaceutical composition according to Claim 17 in the preparation of a medicament for the reduction of formation of plaque on the brain of a subject.

27. A method of reducing beta amyloid peptide levels in the cerebral spinal fluid of a subject in need thereof, the method comprising administering to the subject a

therapeutically effective amount of the compound according to any one of Claims 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer.

28. A method of treating Alzheimer's disease, cognitive impairment or a combination thereof in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound according to any one of Claims 1- 16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer.

29. A method of treating a neurological disorder selected from mild cognitive impairment, Down's syndrome, hereditary cerebral hemorrhage with Dutch-type amyloidosis, cerebral amyloid angiopathy, degenerative dementia, dementia associated with Parkinson's disease, dementia associated with supranuclear palsy, dementia associated with cortical basal degeneration, diffuse Lewy body type of Alzheimer's disease, or a combination thereof in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound according to any one of Claims 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer.

30. A method of reducing the formation of plaque on the brain of a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound according to any one of Claims 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer.

Description:
CYCLOPROPYL FUSED THIAZINE DERIVATIVES AS BETA-SECRETASE INHIBITORS AND METHODS OF USE

CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims benefit of U.S. Provisional Patent Application No.

62/434,721, filed December 15, 2016, which is incorporated by reference herein in its entirety.

FIELD

The present disclosure relates generally to pharmaceutically active compounds and pharmaceutical compositions thereof for the modulation of beta site amyloid precursor protein cleaving enzyme (BACE) activity. Provided herein are uses of these compounds and pharmaceutical compositions thereof for treatment of disorders and/or conditions related to beta-amyloid plaque formation and deposition, resulting from the biological activity of BACE. Such BACE mediated disorders include, for example, Alzheimer's disease, cognitive deficits, cognitive impairments, and other central nervous system conditions. BACKGROUND

Alzheimer's disease (AD) affects greater than 12 million aging people worldwide, and, importantly, the number affected continues to grow. AD accounts for the majority of dementias clinically diagnosed after the age of 60. AD is generally characterized by the progressive decline of memory, reasoning, judgement and orientation. As the disease progresses, motor, sensory, and vocal abilities are affected until there is global impairment of multiple cognitive functions. The loss of cognitive function occurs gradually. Patients with severe cognitive impairment and/or diagnosed as end-stage AD are generally bedridden, incontinent, and dependent on custodial care. The AD patient eventually dies in about nine to ten years, on average, after initial diagnosis. Due to the incapacitating, generally humiliating and ultimately fatal effects of AD, there is a need to treat AD effectively upon diagnosis.

AD is characterized by two major physiological changes in the brain. The first change, beta amyloid plaque formation, supports the "amyloid cascade hypothesis" which conveys the thought that AD is caused by the formation of characteristic beta amyloid (Αβ) peptide deposits in the brain (commonly referred to as Αβ "plaques" or "plaque deposits") and in cerebral blood vessels (beta amyloid angiopathy). A wealth of evidence suggests that Αβ and accompanying amyloid plaque formation is central to the pathophysiology of AD and is likely to play an early role in this intractable neurodegenerative disorder. Yan et al. , Lancet Neurol. 13(3):319-329 (2014). The second change in AD is the formation of intraneuronal tangles, consisting of an aggregate form of the microtubule -binding protein tau. Besides being found in patients with AD, intraneuronal tangles are also found in other dementia-inducing disorders. Joachim et al , Alzheimer. Dis. Assoc. Disord. 6(l):7-34 (1992).

Several lines of evidence indicate that progressive cerebral deposition of Αβ peptide plays a seminal role in the pathogenesis of AD and can precede cognitive symptoms by years or even decades. Selkoe, Neuron 6(4):487-498 (1991). Release of Αβ peptide from neuronal cells grown in culture and the presence of Αβ peptide in cerebrospinal fluid (CSF) of both normal individuals and AD patients has been demonstrated. Seubert et al. , Nature 359:325- 327 (1992). Autopsies of AD patients have revealed large numbers of lesions comprising Αβ and tau peptides in areas of the human brain believed to be important for memory and cognition.

Smaller numbers of these lesions in a more restricted anatomical distribution are found in the brains of most aged humans who do not have clinical AD. Amyloid containing plaques and vascular amyloid angiopathy were also found in the brains of individuals with Down's Syndrome, Hereditary Cerebral Hemorrhage with Amyloidosis of the Dutch-type (HCHWA-D), and other neurodegenerative disorders.

It has been hypothesized that Αβ peptide formation is a causative precursor or factor in the development of AD. More specifically, deposition of Αβ peptide in areas of the brain responsible for cognition is believed to be a major factor in the development of AD. Αβ plaques are primarily composed of Αβ peptide. Αβ peptide is derived from the proteolytic cleavage of a large transmembrane amyloid precursor protein (APP), and is a peptide comprised of about 39-42 amino acid residues. Αβ 1-42 (42 amino acids long) is thought to be the major component of these plaque deposits in the brains of AD patients. Citron, Trends Pharmacol. Sci. 25 (2): 92-97 (2004).

Similar plaques appear in some variants of Lewy body dementia and in inclusion body myositis, a muscle disease. Αβ peptides also form aggregates coating cerebral blood vessels in cerebral amyloid angiopathy. These plaques are composed of fibrillar Αβ aggregates that display a characteristic β-sheet structure, a protein fold shared by other peptides such as prions associated with protein misfolding diseases. Research on laboratory rats suggest that the dimeric, soluble form of the peptide is a causative agent in the development of AD and is the smallest synaptotoxic species of soluble amyloid beta oligomer. Shankar et al, Nat. Med. 14(8):837-842 (2008).

Several aspartyl proteases, including β-secretase and γ-secretase, are involved in the processing or cleavage of APP, resulting in the formation of Αβ peptide. β-Secretase (BACE, also commonly referred to as memapsin) is the first to cleave APP to generate two fragments: (1) a first N-terminus fragment (βΑΡΡβ) and (2) a second C-99 fragment, which is subsequently cleaved by γ-secretase to generate the Αβ peptide. APP has also been found to be cleaved by a-secretase to produce sAPPa, a secreted form of APP that does not result in Αβ plaque formation. This alternate pathway precludes the formation of Αβ peptide. A description of the proteolytic processing fragments of APP is found, for example, in U.S. Patent Nos. 5,441,870, 5,712, 130 and 5,942,400.

BACE is an aspartyl protease enzyme comprising 501 amino acids and responsible for processing APP at the β-secretase specific cleavage site. BACE is present in two forms, BACE 1 and BACE 2, designated as such depending upon the specific cleavage site of APP. β-Secretase is described in Sinha et al, Nature 402:537-540 (1999) and International Patent Application Publication No. WO2000/017369. It has been proposed that Αβ peptide accumulates as a result of APP processing initiated by BACE. Moreover, in vivo processing of APP at the β-secretase cleavage site is thought to be a rate-limiting step in Αβ peptide production. Sabbagh et al, Alzheimer 's Disease Review 3: 1-19 (1997). Thus, inhibition of the BACE enzyme activity is desirable for the treatment of AD.

Studies have shown that the inhibition of BACE may be linked to the treatment of

AD. The BACE enzyme is essential for the generation of Αβ peptide. BACE knockout mice do not produce Αβ peptide and are free from AD associated pathologies including neuronal loss and certain memory deficits. Cole et al. , Molecular Neurodegeneration 2:22, pages 1-25 (2007). When crossed with transgenic mice that over express APP, the progeny of BACE deficient mice show reduced amounts of Αβ peptide in brain extracts as compared with control animals. Luo et al., Nat. Neurosci. 4(3):231-232 (2001). The fact that BACE initiates the formation of Αβ peptide, and the observation that BACE levels are elevated in this disease provide direct and compelling reasons to develop therapies directed at BACE inhibition, thus, reducing Αβ peptide formation and its associated toxicities. To this end, inhibition of β-secretase activity and a corresponding reduction of Αβ peptide in the brain should provide a therapeutic method for treating AD and other Αβ peptide or plaque related disorders.

Consequently, the approach of regulating or reducing Αβ peptide formation and deposition as a potential treatment for AD has received tremendous attention, support and commitment from both researchers and investors alike. A small molecule γ-secretase inhibitor, LY450139 ("Semagacestat"), an Αβ peptide lowering agent, advanced to phase III clinical trials for the treatment of AD. The pharmacokinetics of semagacestat in plasma, as well as the plasma and cerebral spinal fluid (CSF) Αβ peptide levels as pharmacodynamic responses to semagacestat administration were evaluated in healthy human subjects in single and multiple doses, and pharmacokinetic and pharmacodynamic changes were also assessed in mild to moderate AD patients in two (2) clinical trials (Henley et al. , Expert Opin.

Pharmacother . 10(10): 1657-1664 (2009); Siemers et al, Clin. Neuropharmacol . 30(6): 317- 325 (2007); and Siemers et al, Neurology 66(4):602-604 (2006)). Additional approaches have been taken in attempts to treat AD and plaque-related disorders. See, for example, Yan et al , Lancet Neurology 13(3):319-329 (2014).

Furthermore, each of the following exemplary patent application publications describes inhibitors of BACE, useful for treating AD and other β-secretase mediated disorders: WO2014/098831, WO2014/099794, WO2014/099788, WO2014/097038, WO2014/093190, WO2014/066132, WO2014/065434, WO2014/062553, WO2014/062549, WO2014/045162, WO2014/013076, WO2013/182638, WO2013/164730, WO2013/030713, WO2013/028670, WO2013/004676, WO2012/162334, WO2012/162330, WO2012/147762, WO2012/139425, WO2012/138734, US2012/0245157, US2012/0245154, US2012/0238557, WO2011/029803, WO2011/005738, US2011/0152253, WO2010/013794, WO2010/013302, US2010/0160290, US2010/0075957, WO2009/151098, WO2009/134617, US2009/0209755, US2009/0082560, EP2703401 (equivalent of WO2012/ 146762) and EP1942105.

The lysosomal aspartic protease Cathepsin D (CatD) is ubiquitously expressed in eukaryotic organisms. CatD activity is essential to accomplish the acid-dependent extensive or partial proteolysis of protein substrates within endosomal and lysosomal compartments therein delivered via endocytosis, phagocytosis or autophagocytosis. CatD may also act at physiological pH on small-size substrates in the cytosol and in the extracellular milieu. Mouse and fruit fly CatD knock-out models have highlighted the multi-pathophysiological roles of CatD in tissue homeostasis and organ development.

Inhibition of protein CatD has been implicated in undesirable side effects. For instance, the inhibition of CatD is believed to be linked to adverse retinal development and retinal atrophy. Particularly, in mice it was found that CatD is essential for the metabolic maintenance of retinal photoreceptor cells and that its deficiency induces apoptosis of the cells, while the loss of inner nuclear layer (INL) neurons is mediated by nitric oxide release from microglial cells. However, in the very same mice, it was also found that no atrophic change was detected in the retina of mice deficient in Cathepsin B or L. Koike et al. , Mol. Cell Neurosci. 22(2): 146-161 (2003). Further, animal models of CatD deficiency are characterized by a progressive and relentless neurodegenerative phenotype similar to that observed in Neuronal Ceroid Lipofuscinoses (NCL), a group of pediatric neurodegenerative diseases known collectively as Batten Disease. It has been shown that the targeted deletion of the pro-apoptotic molecule Bax prevents apoptotic markers, but not neuronal cell death and neurodegeneration induced by CatD deficiency, which suggests that alterations in the macroautophagy-lysosomal degradation pathway can mediate neuronal cell death in

NCL/Batten Disease in the absence of apoptosis. Shacka et al. , Autophagy 3(5):474-476

(2007). Finally, an adverse effect of the inhibition of CatD is evident from the data presented in Folio et al., PLoS One 6(7):e21908 (2011). The authors of the PLoS One paper found that knock-down of CatD affects the retinal pigment epithelium, impairs swim-bladder ontogenesis and causes premature death in zebrafish. The main phenotypic alterations produced by CatD knock-down in zebrafish were: 1. abnormal development of the eye and of retinal pigment epithelium; 2. absence of the swim-bladder; 3. skin hyper-pigmentation; 4. reduced growth and premature death. Rescue experiments confirmed the involvement of CatD in the developmental processes leading to these phenotypic alterations.

Moreover, such toxicity findings which, in view of the literature, may have played a role in the termination of a human BACE-mediated AD clinical trial. Eli Lilly terminated a phase I clinical trial of LY 2811376 after rat toxicology studies showed that a higher compound dose given for three months damaged the pigment epithelium of the rat's eye. The retinal layer had inclusions and extensive damage. The Phase I dosing trial was terminated and people brought in for eye assessments did not show any abnormalities. (Alzheimer's Research Forum News, 3-31-2011 reporting on Martin Citron's presentation at the AD/PD Conference 3-2011 in Barcelona, Spain).

Hence, it is desirable to provide compounds which modulate the activity of and are selective for BACE, while not suffering from undesirable side effects possibly due to intervention with or the reduction and/or direct or indirect inhibition of the expression and/or function of other proteins or biological pathways.

SUMMARY

The compounds disclosed herein are useful for the modulation of β-secretase activity, and as treatment of AD. Particularly, the compounds provided herein are useful for the regulation or reduction of the formation of Αβ peptide and, consequently, the regulation and/or reduction of formation of Αβ plaque both in the brain, as well as in the CNS. To this end, the compounds are useful for the treatment of AD and other β-secretase and/or plaque- related and/or mediated disorders. For example, the compounds are useful for the prophylaxis and/or treatment, acute and/or chronic, of AD and other diseases or conditions involving the deposition or accumulation of Αβ peptide, and formation of plaque, in the brain.

First, provided herein is A com ound of Formula I

I

or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein

A is N, CH, or CR 4 ; R 1 is H, -CN, Ci-ealkyl, Ca-ealkenyl, -Ci- 4 alkyl-C(0)NR 1 R r , -Ci- 4 alkyl-C(0)- heterocycloalkyl, -(HC^H CC NR 1 R 1 , -(HC=CH)-C(0)-heterocycloalkyl, -C(0)NR 1 R 1' , or -C(0)-heterocycloalkyl, wherein the Ci-ealkyl and the C2-ealkenyl are (i) optionally substituted with one to three fluoro substituents or (ii) optionally substituted with -CN, OH, methoxy, or a 5-membered nitrogen-containing heteroaryl, wherein the 5-membered nitrogen-containing heteroaryl is optionally substituted with Ci- 4 alkyl;

R 1 is, independently, H or Ci- 4 alkyl;

R 2 and R 2 are independently H or halogen;

R 3 is Ci-4alkyl, wherein the Ci-4alkyl is optionally substituted with one to three fluoro substituents;

R 4 is halogen;

R 5 is H, C3- 6 cycloalkyl, phenyl, or 5- or 6-membered heteroaryl, wherein the phenyl or heteroaryl is optionally substituted with one to three substituents independently selected from halogen, -CN, Ci- 4 alkyl, 2-propynyloxy, 2-butynyloxy, or oxazolylmethoxy.

Second, provided herein are pharmaceutical compositions comprising a compound of

Formula I and a pharmaceutically acceptable excipient.

Third, provided herein are compounds of Formula I or pharmaceutical compositions thereof for use as a medicament.

Fourth, provided herein are compounds of Formula I or pharmaceutical compositions thereof for use in reducing beta amyloid peptide levels in the cerebral spinal fluid of a subject.

Fifth, provided herein are compounds of Formula I or pharmaceutical compositions thereof for use in treating Alzheimer's disease, cognitive impairment, or a combination thereof in a subject. In addition, provided herein are compounds of Formula I or

pharmaceutical compositions thereof for treating a neurological disorder selected from mild cognitive impairment, Down's syndrome, hereditary cerebral hemorrhage with Dutch-type amyloidosis, cerebral amyloid angiopathy, degenerative dementia, dementia associated with Parkinson's disease, dementia associated with supranuclear palsy, dementia associated with cortical basal degeneration, diffuse Lewy body type of Alzheimer's disease, or a combination thereof in a subject. Sixth, provided herein are compounds of Formula I or pharmaceutical compositions thereof for use in reducing formation of plaque in the brain of a subject.

Reference will now be made in detail to embodiments of the present disclosure. While certain embodiments of the present disclosure will be described, it will be understood that it is not intended to limit the embodiments of the present disclosure to those described embodiments. To the contrary, reference to embodiments of the present disclosure is intended to cover alternatives, modifications, and equivalents as may be included within the spirit and scope of the embodiments of the present disclosure as defined by the appended claims. DETAILED DESCRIPTION

Provided herein as Embodiment 1 is a com ound of Formula I

or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein

A is N, CH, or CR 4 ;

R 1 is H, -CN, Cwalkyl, C 2 -6alkenyl, -Ci- 4 alkyl-C(0)NR 1 R r , -Ci- 4 alkyl-C(0)- heterocycloalkyl, -(HC=CH)-C(0)NR 1 R 1' , -(HC=CH)-C(0)-heterocycloalkyl, -C(0)NR 1 R 1' or -C(0)-heterocycloalkyl, wherein the Ci-ealkyl and the C2-ealkenyl are (i) optionally substituted with one to three fluoro substituents or (ii) optionally substituted with -CN, OH, methoxy, or a 5-membered nitrogen-containing heteroaryl, wherein the 5-membered nitrogen-containing heteroaryl is optionally substituted with Cwalkyl;

R 1 is, independently, H or Cwalkyl;

R 2 and R 2 are independently H or halogen; R 3 is Ci-4alkyl, wherein the Ci-4alkyl is optionally substituted with one to three fluoro substituents;

R 4 is halogen;

R 5 is H, C3-6Cycloalkyl, phenyl, or 5- or 6-membered heteroaryl, wherein the phenyl or heteroaryl is optionally substituted with one to three substituents independently selected from halogen, -CN, Ci-4alkyl, 2-propynyloxy, 2-butynyloxy, or oxazolylmethoxy.

Provided herein as Embodiment 2 is the compound according to Embodiment 1, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein the compound of Formula I is a com ound of Formula II

Provided herein as Embodiment 3 is the compound according to Embodiment 1, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein the compound of Formula I is a com ound of Formula III

III.

Provided herein as Embodiment 4 is the compound according to Embodiment 1, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein the compound of Formula I is a compound of Formula ΙΙΓ

Provided herein as Embodiment 5 is the compound according to any one of Embodiments 1-4, or a tautomer thereof, or a pharmaceutically acceptable salt of said

Provided herein as Embodiment 6 is the compound according to any one of Embodiments 1-5, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein R 2 and R 2 are H. Provided herein as Embodiment 7 is the compound according to any one of

Embodiments 1-5, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein R 2 and R 2 are F.

Provided herein as Embodiment 8 is the compound according to any one of

Embodiments 1-7, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein R 3 is methyl, -CH 2 F, or CHF 2 .

Provided herein as Embodiment 9 is the compound according to any one of

Embodiments 1-7, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein R 3 is methyl or-CFhF.

Provided herein as Embodiment 10 is the compound according to any one of Embodiments 1-9, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein R 4 is F.

Provided herein as Embodiment 11 is the compound according to any one of Embodiments 1-10, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein R 5 is H, cyclopropyl, phenyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, or pyrazinyl, wherein the phenyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, or pyrazinyl is optionally substituted with one or two substituents independently selected from F, CI, -CN, methyl, 2-propynyloxy, 2-butynyloxy, or 2-oxazolylmethoxy.

Provided herein as Embodiment 12 is the compound according to any one of Embodiments 1-11, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein

Provided herein as Embodiment 13 is the compound of Embodiment 1 or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, selected from

6-((3-((lS,5S,6S)-3-amino-5-(fluoromethyl)-l-(hydroxymeth yl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile;

6-((3 -(( 1 S,5 S,6S)-3 -amino- 1 ,5 -bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 - en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

(lS,5S,6S)-5-(5-((5-chloropyridin-2-yl)ethynyl)-2-fluorophen yl)-l,5- bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-3 -amine ;

5 -((3 -(( 1 S,5 S,6S)-3 -amino- 1 ,5 -bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 - en-5-yl)-4-fluorophenyl)ethynyl)pyrazine-2-carbonitrile;

6-((3 -(( 1 S,5 S,6S)-3 -amino- 1 ,5 -bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 - en-5-yl)-4-fluorophenyl)ethynyl)-5-methylnicotinonitrile;

(lS,5S,6S)-5-(5-((5-(but-2-yn-l-yloxy)pyridin-2-yl)ethynyl)- 2-fluorophenyl)-l,5- bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-3 -amine ;

6-((3-((lR,5S,6S)-3-amino-l-(cyanomethyl)-5-(fluoromethyl)-2 -thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile; 6-((3-((lR,5S,6S)-3-amino-l-((E)-2-cyanovinyl)-5-(fluorometh yl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile;

(lS,5S,6S)-5-(2-fluoro-5-((5-(oxazol-2-ylmethoxy)pyridin-2-y l)ethynyl)phenyl)-l,5 bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-3 -amine ;

2-((3 -(( 1 S,5 S,6S)-3 -amino- 1 ,5 -bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 - en-5-yl)-4-fluorophenyl)ethynyl)thiazole-5-carbonitrile;

6-((3-((lS,5S,6R)-3-amino-7,7-difluoro-l-(hydroxymethyl)-5-m ethyl-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile;

6-((3 -(( 1 R,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -((E)-3 -morpholino-3 -oxoprop- 1 -en- 1 yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl )ethynyl)nicotinonitrile;

(E)-3-((lR,5S,6S)-3-amino-5-(5-((5-cyanopyridin-2-yl)ethynyl )-2-fluorophenyl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)-N,N -dimethylacrylamide;

2-(( 1 R,5 S,6S)-3 -amino-5 -(5 -((5 -cyanopyridin-2-yl)ethynyl)-2-fluorophenyl)-5 - (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)acet amide;

6-((5-((lS,5S,6S)-3-amino-l-(fluoromethyl)-5-methyl-2-thia-4 - azabicyclo[4.1.0]hept-3-en-5-yl)-6-fluoropyridin-3-yl)ethyny l)nicotinonitrile;

6-((3-((lS,5S,6S)-3-amino-5-(fluoromethyl)-l-((4-methyl-lH-l ,2,3-triazol-l- yl)methyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluor ophenyl)ethynyl)nicotinonitrile;

6-((3 -(( 1 R,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -((E)-3 -morpholino-3 -oxoprop- 1 -en- 1 yl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-5 -yl)-4-fluorophenyl)ethynyl)-5 - methylnicotinonitrile ;

(E)-3-((lR,5S,6S)-3-amino-5-(5-((5-chloropyrimidin-2-yl)ethy nyl)-2-fluorophenyl)- 5 -(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en- 1 -yl)- 1 -morpholinoprop-2-en- 1 -one;

(E)-3-((lR,5S,6S)-3-amino-5-(2-fluoro-5-((3-memylisothiazol- 5-yl)ethynyl)phenyl)- 5 -(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en- 1 -yl)- 1 -morpholinoprop-2-en- 1 -one;

6-((3 -(( 1 S,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -(3 -morpholino-3 -oxopropyl)-2-thia-4 azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile;

6-((3 -(( 1 S,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -(3 -morpholino-3 -oxopropyl)-2-thia-4 azabicyclo [4.1.0]hept-3 -en-5 -yl)-4-fluorophenyl)ethynyl)-5 -methylnicotinonitrile; 2-((3 -(( 1 R,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -((E)-3 -morpholino-3 -oxoprop- 1 -en- 1 - yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl )ethynyl)pyrimidine-5- carbonitrile;

4-((3 -(( 1 R,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -((E)-3 -ιηοφ1ιο1ίηο-3 -oxoprop- 1 -en- 1 - yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl )ethynyl)benzonitrile;

(lS,5S,6S)-3-amino-5-(5-((5-cyano-3-methyl-2-pyridinyl)ethyn yl)-2-fluorophenyl)- N,N,5-trimethyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carbo xamide;

6-((3 -(( 1 R,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -((E)-3 -moφholino-3 -oxoprop- 1 -en- 1 - yl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-5 -yl)-4-fluorophenyl)ethynyl)-5 -fluoronicotinonitrile;

6-((3-((lS,5S,6S)-l-((lH-imidazol-l-yl)methyl)-3-amino-5-(fl uoromethyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile;

(lS,5S,6S)-3-amino-5-(5-((5-cyano-3-methylpyridin-2-yl)ethyn yl)-2-fluorophenyl)- 5-methyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carbonitrile ;

(lS,5S,6S)-3-amino-5-(5-((5-cyano-3-methylpyridin-2-yl)ethyn yl)-2-fluorophenyl)- 5 -(fluoromethyl)-N,N-dimethyl-2-thia-4-azabicyclo [4.1.0]hept-3 -ene- 1 -carboxamide ;

6-((3 -(( 1 S,5 S,6S)-3 -amino-5 -methyl- 1 -(pyrrolidine- 1 -carbonyl)-2-thia-4- azabicyclo [4.1.0]hept-3 -en-5 -yl)-4-fluorophenyl)ethynyl)-5 -methylnicotinonitrile;

(lS,5S,6S)-3-amino-5-(5-((4-cyanophenyl)ethynyl)-2-fluorophe nyl)-N,N,5- trimethyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxamide ;

( 1 S,5 S,6S)-3 -amino-5 -(2-fluoro-5 -((5 -(2-propyn- 1 -yloxy)-2- pyridinyl)ethynyl)phenyl)-N,N,5-trimethyl-2-thia-4-azabicycl o[4.1.0]hept-3-ene-l- carboxamide;

(lS,5S,6S)-3-amino-5-(2-fluoro-5-((3-methyl-5-(oxazol-2-ylme thoxy)pyridin-2- yl)ethynyl)phenyl)-N,N,5 -trimethyl-2-thia-4-azabicyclo [4.1.0]hept-3 -ene- 1 -carboxamide ;

(lS,5S,6S)-3-amino-5-(5-(cyclopropylethynyl)-2-fluorophenyl) -N,N,5-trimethyl-2- thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxamide;

6-((3-((lS,5S)-3-amino-l-(methoxymethyl)-5-methyl-2-thia-4-a zabicyclo[4.1.0]hept- 3-en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

( 1 S,5 S)-5 -(5 -((5 -chloro-3 -methylpyridin-2-yl)ethynyl)-2 -fluorophenyl)- 1 - (methoxymethyl)-5-methyl-2-thia-4-azabicyclo[4.1.0]hept-3-en -3-amine; 6-((3-((lS,5S)-3-amino-l-(methoxymethyl)-5-methyl-2-thia-4-a zabicyclo[4.1.0]hept- 3 -en-5 -yl)-4-fluorophenyl)ethynyl)-5 -methylnicotinonitrile;

6-((5-((lS,5S)-3-amino-5-methyl-l-(moφholine-4-carbonyl)-2- thia-4- azabicyclo [4.1.0]hept-3 -en-5-yl)-6-fluoropyridin-3 -yl)ethynyl)-5 -methylnicotinonitrile;

6-((5 -(( 1 S,5 S)-3 -amino-5 -methyl- 1 -(pyrrolidine- 1 -carbonyl)-2-thia-4- azabicyclo [4.1.0]hept-3 -en-5-yl)-6-fluoropyridin-3 -yl)ethynyl)-5 -methylnicotinonitrile ;

(lS,5S,6S)-5-(5-((4-chloropyridin-2-yl)ethynyl)-2-fluorophen yl)-l,5- bis(fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-3-amin e; or

(lS,5S,6S)-3-amino-5-(5-ethynyl-2-fluorophenyl)-N,N,5-trimet hyl-2-thia-4- azabicyclo[4.1.0]hept-3-ene-l-carboxamide.

Provided herein as Embodiment 14 is the compound according to any one of Embodiments 1-10, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein R 5 is phenyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, or pyrazinyl, wherein the phenyl, thiazolyl, isothiazolyl, pyridyl, pyrimidinyl, or pyrazinyl is optionally substituted with one or two substituents independently selected from -CN, methyl, 2-propynyloxy, 2-butynyloxy, or 2-oxazolylmethoxy.

Provided herein as Embodiment 15 is the compound according to any one of Embodiments 1-11, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, wherein

Provided herein as Embodiment 16 is the compound of Embodiment 1, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, selected from

6-((3-((lS,5S,6S)-3-amino-5-(fluoromethyl)-l-(hydroxymeth yl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile;

6-((3 -(( 1 S,5 S,6S)-3 -amino- 1 ,5 -bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 - en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

(lS,5S,6S)-5-(5-((5-chloropyridin-2-yl)ethynyl)-2-fluorophen yl)-l,5- bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-3 -amine ;

5 -((3 -(( 1 S,5 S,6S)-3 -amino- 1 ,5 -bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 - en-5-yl)-4-fluorophenyl)ethynyl)pyrazine-2-carbonitrile;

6-((3 -(( 1 S,5 S,6S)-3 -amino- 1 ,5 -bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 - en-5-yl)-4-fluorophenyl)ethynyl)-5-methylnicotinonitrile;

(lS,5S,6S)-5-(5-((5-(but-2-yn-l-yloxy)pyridin-2-yl)ethynyl)- 2-fluorophenyl)-l,5- bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-3 -amine ;

6-((3-((lR,5S,6S)-3-amino-l-(cyanomethyl)-5-(fluoromethyl)-2 -thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile;

6-((3-((lR,5S,6S)-3-amino-l-((E)-2-cyanovinyl)-5-(fluorometh yl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile;

(lS,5S,6S)-5-(2-fluoro-5-((5-(oxazol-2-ylmethoxy)pyridin-2-y l)ethynyl)phenyl)-l,5- bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-3 -amine ;

2-((3 -(( 1 S,5 S,6S)-3 -amino- 1 ,5 -bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 - en-5-yl)-4-fluorophenyl)ethynyl)thiazole-5-carbonitrile;

6-((3-((lS,5S,6R)-3-amino-7,7-difluoro-l-(hydroxymethyl)-5-m ethyl-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile; 6-((3 -(( 1 R,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -((E)-3 -morpholino-3 -oxoprop- 1 -en- 1 yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl )ethynyl)nicotinonitrile;

(E)-3-((lR,5S,6S)-3-amino-5-(5-((5-cyanopyridin-2-yl)ethynyl )-2-fluorophenyl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)-N,N -dimethylacrylamide;

2-(( 1 R,5 S,6S)-3 -amino-5 -(5 -((5 -cyanopyridin-2-yl)ethynyl)-2-fluorophenyl)-5 - (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)acet amide;

6-((5-((lS,5S,6S)-3-amino-l-(fluoromethyl)-5-methyl-2-thia-4 - azabicyclo[4.1.0]hept-3-en-5-yl)-6-fluoropyridin-3-yl)ethyny l)nicotinonitrile;

6-((3-((lS,5S,6S)-3-amino-5-(fluoromethyl)-l-((4-methyl-lH-l ,2,3-triazol-l- yl)methyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluor ophenyl)ethynyl)nicotinonitrile;

6-((3 -(( 1 R,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -((E)-3 -morpholino-3 -oxoprop- 1 -en- 1 yl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-5 -yl)-4-fluorophenyl)ethynyl)-5 - methylnicotinonitrile ;

(E)-3-((lR,5S,6S)-3-amino-5-(2-fluoro-5-((3-memylisothiazol- 5-yl)ethynyl)phenyl)- 5 -(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en- 1 -yl)- 1 -morpholinoprop-2-en- 1 -one;

6-((3 -(( 1 S,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -(3 -morpholino-3 -oxopropyl)-2-thia-4 azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile;

6-((3 -(( 1 S,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -(3 -morpholino-3 -oxopropyl)-2-thia-4 azabicyclo [4.1.0]hept-3 -en-5 -yl)-4-fluorophenyl)ethynyl)-5 -methylnicotinonitrile;

2-((3 -(( 1 R,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -((E)-3 -morpholino-3 -oxoprop- 1 -en- 1 yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl )ethynyl)pyrimidine-5- carbonitrile;

4-((3 -(( 1 R,5 S,6S)-3 -amino-5 -(fluoromethyl)- 1 -((E)-3 -morpholino-3 -oxoprop- 1 -en- 1 yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl )ethynyl)benzonitrile;

(lS,5S,6S)-3-amino-5-(5-((5-cyano-3-methyl-2-pyridinyl)ethyn yl)-2-fluorophenyl)- N,N,5-trimethyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carbo xamide;

6-((3-((lS,5S,6S)-l-((lH-imidazol-l-yl)methyl)-3-amino-5-(fl uoromethyl)-2-thia-4- azabicyclo [4.1.0]hept-3 -en-5 -yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

(lS,5S,6S)-3-amino-5-(5-((5-cyano-3-methylpyridin-2-yl)ethyn yl)-2-fluorophenyl)- 5-methyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carbonitrile ; (lS,5S,6S)-3-amino-5-(5-((5-cyano-3-methylpyridin-2-yl)ethyn yl)-2-fluorophenyl)- 5 -(fluoromethyl)-N,N-dimethyl-2-1hia-4-azabicyclo [4.1.0]hept-3 -ene- 1 -carboxamide ;

6-((3 -(( 1 S,5 S,6S)-3 -amino-5 -methyl- 1 -(pyrrolidine- 1 -carbonyl)-2-thia-4- azabicyclo [4.1.0]hept-3 -en-5 -yl)-4-fluorophenyl)ethynyl)-5 -methylnicotinonitrile;

(lS,5S,6S)-3-amino-5-(5-((4-cyanophenyl)ethynyl)-2-fluorophe nyl)-N,N,5- trimethyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxamide ;

(lS,5S,6S)-3 -amino-5 -(2-fluoro-5 -((5 -(2-propyn- 1 -yloxy)-2- pyridinyl)ethynyl)phenyl)-N,N,5-trimethyl-2-thia-4-azabicycl o[4.1.0]hept-3-ene-l- carboxamide;

(lS,5S,6S)-3-amino-5-(2-fluoro-5-((3-methyl-5-(oxazol-2-ylme thoxy)pyridin-2- yl)ethynyl)phenyl)-N,N,5 -trimethyl-2-thia-4-azabicyclo [4.1.0]hept-3 -ene- 1 -carboxamide ;

6-((3-((lS,5S)-3-amino-l-(methoxymethyl)-5-methyl-2-thia-4-a zabicyclo[4.1.0]hept- 3-en-5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile;

( 1 S,5 S)-5 -(5 -((5 -chloro-3 -methylpyridin-2-yl)ethynyl)-2 -fluorophenyl)- 1 - (methoxymethyl)-5-methyl-2-thia-4-azabicyclo[4.1.0]hept-3-en -3-amine;

6-((3-((lS,5S)-3-amino-l-(methoxymethyl)-5-methyl-2-thia-4-a zabicyclo[4.1.0]hept- 3 -en-5 -yl)-4-fluorophenyl)ethynyl)-5 -methylnicotinonitrile;

6-((5-((lS,5S)-3-amino-5-methyl-l-(moφholine-4-carbonyl)-2- thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-6-fluoropyridin-3-yl)ethyny l)-5-methylnicotinonitrile; or 6-((5 -(( 1 S,5 S)-3 -amino-5 -methyl- 1 -(pyrrolidine- 1 -carbonyl)-2-thia-4- azabicyclo [4.1.0]hept-3 -en-5-yl)-6-fluoropyridin-3 -yl)ethynyl)-5 -methylnicotinonitrile .

Provided herein as Embodiment 17 is a pharmaceutical composition comprising the compound according to any one of Embodiments 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, and a pharmaceutically acceptable excipient.

Provided herein as Embodiment 18 is a compound according to any one of

Embodiments 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, or the pharmaceutical composition according to Embodiment 17 for use as a medicament.

Provided herein as Embodiment 19 is a compound according to any one of

Embodiments 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, or the pharmaceutical composition according to Embodiment 17 for use in reducing beta amyloid peptide levels in the cerebral spinal fluid of a subject.

Provided herein as Embodiment 20 is a compound according to any one of

Embodiments 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, or the pharmaceutical composition according to Embodiment 17 for use in treating Alzheimer's disease, cognitive impairment, or a combination thereof in a subject.

Provided herein as Embodiment 21 is a compound according to any one of

Embodiments 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, or the pharmaceutical composition according to Embodiment 17 for use in treating a neurological disorder selected from mild cognitive impairment, Down's syndrome, hereditary cerebral hemorrhage with Dutch-type amyloidosis, cerebral amyloid angiopathy, degenerative dementia, dementia associated with Parkinson's disease, dementia associated with supranuclear palsy, dementia associated with cortical basal degeneration, diffuse Lewy body type of Alzheimer's disease, or a combination thereof in a subject.

Provided herein as Embodiment 22 is a compound according to any one of

Embodiments 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, or the pharmaceutical composition according to Embodiment 17 for use in reducing formation of plaque on the brain of a subject.

Provided herein as Embodiment 23 is a use of the compound according to any one of

Embodiments 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, or the pharmaceutical composition according to Embodiment 17 in the preparation of a medicament for reducing beta amyloid peptide levels in the cerebral spinal fluid of a subject.

Provided herein as Embodiment 24 is a use of the compound according to any one of

Embodiments 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, or the pharmaceutical composition according to Embodiment 17 in the preparation of a medicament for treating Alzheimer's disease, cognitive impairment, or a combination thereof in a subject.

Provided herein as Embodiment 25 is a use of the compound according to any one of

Embodiments 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, or the pharmaceutical composition according to Embodiment 17 in the preparation of a medicament for the treatment of a neurological disorder selected from mild cognitive impairment, Down's syndrome, hereditary cerebral hemorrhage with Dutch- type amyloidosis, cerebral amyloid angiopathy, degenerative dementia, dementia associated with Parkinson's disease, dementia associated with supranuclear palsy, dementia associated with cortical basal degeneration, diffuse Lewy body type of Alzheimer's disease, or a combination thereof in a subject.

Provided herein as Embodiment 26 is a use of the compound according to any one of Embodiments 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer, or the pharmaceutical composition according to Embodiment 17 in the preparation of a medicament for the reduction of formation of plaque on the brain of a subject.

Provided herein as Embodiment 27 is a method of reducing beta amyloid peptide levels in the cerebral spinal fluid of a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound according to any one of Embodiments 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer.

Provided herein as Embodiment 28 is a method of treating Alzheimer's disease, cognitive impairment or a combination thereof in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound according to any one of Embodiments 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer.

Provided herein as Embodiment 29 is a method of treating a neurological disorder selected from mild cognitive impairment, Down's syndrome, hereditary cerebral hemorrhage with Dutch-type amyloidosis, cerebral amyloid angiopathy, degenerative dementia, dementia associated with Parkinson's disease, dementia associated with supranuclear palsy, dementia associated with cortical basal degeneration, diffuse Lewy body type of Alzheimer's disease, or a combination thereof in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound according to any one of Embodiments 1-16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer. Provided herein as Embodiment 30 is a method of reducing the formation of plaque on the brain of a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound according to any one of Embodiments 1- 16, or a tautomer thereof, or a pharmaceutically acceptable salt of said compound or tautomer.

The foregoing merely summarizes certain aspects of this disclosure and is not intended, nor should it be construed, as limiting the disclosure in any way.

DEFINITIONS

The following definitions are provided to assist in understanding the scope of this disclosure.

Unless otherwise indicated, all numbers expressing quantities of ingredients, reaction conditions, and so forth used in the specification and claims are to be understood as being modified in all instances by the term "about." Accordingly, unless indicated to the contrary, the numerical parameters set forth in the following specification and attached claims are approximations that may vary depending upon the standard deviation found in their respective testing measurements.

As used herein, if any variable occurs more than one time in a chemical formula, its definition on each occurrence is independent of its definition at every other occurrence. If the chemical structure and chemical name conflict, the chemical structure is determinative of the identity of the compound.

Stereoisomers

The compounds of the present disclosure may contain, for example, double bonds, one or more asymmetric carbon atoms, and bonds with a hindered rotation, and therefore, may exist as stereoisomers, such as double-bond isomers (i.e., geometric isomers (E/Z)), enantiomers, diastereomers, or atropoisomers. Accordingly, the scope of the instant disclosure is to be understood to encompass all possible stereoisomers of the illustrated compounds including the stereoisomerically pure form (for example, geometrically pure, enantiomerically pure, diastereomerically pure, and atropoisomerically pure) and stereoisomeric mixtures (for example, mixtures of geometric isomers, enantiomers, diastereomers, and atropoisomers) of any chemical structures disclosed herein (in whole or in part). This disclosure also encompasses the pharmaceutical compositions comprising stereoisomerically pure forms and the use of stereoisomerically pure forms of any compounds disclosed herein. Further, this disclosure also encompasses pharmaceutical compositions comprising mixtures of stereoisomers of any compounds disclosed herein and the use of said pharmaceutical compositions or mixtures of stereoisomers. These stereoisomers or mixtures thereof may be synthesized in accordance with methods well known in the art and methods disclosed herein. Mixtures of stereoisomers may be resolved using standard techniques, such as chiral columns or chiral resolving agents. See, for example, Jacques et al , Enantiomers, Racemates and Resolutions (Wiley-Interscience, New York, 1981); Wilen et al , Tetrahedron 33 :2725; Eliel, Stereochemistry of Carbon

Compounds (McGraw-Hill, NY, 1962); and Wilen, Tables of Resolving Agents and Optical Resolutions, page 268 (Eliel, Ed., Univ. of Notre Dame Press, Notre Dame, IN, 1972).

The term "stereoisomer" or "stereoisomerically pure" compound as used herein refers to one stereoisomer (for example, geometric isomer, enantiomer, diastereomer and atropoisomer) of a compound that is substantially free of other stereoisomers of that compound. For example, a stereoisomerically pure compound having one chiral center will be substantially free of the mirror image enantiomer of the compound and a

stereoisomerically pure compound having two chiral centers will be substantially free of other enantiomers or diastereomers of the compound. A typical stereoisomerically pure compound comprises greater than about 80% by weight of one stereoisomer of the compound and less than about 20% by weight of other stereoisomers of the compound, greater than about 90% by weight of one stereoisomer of the compound and less than about 10% by weight of the other stereoisomers of the compound, greater than about 95% by weight of one stereoisomer of the compound and less than about 5% by weight of the other stereoisomers of the compound, or greater than about 97% by weight of one stereoisomer of the compound and less than about 3% by weight of the other stereoisomers of the compound. If the stereochemistry of a structure or a portion of a structure is not indicated with, for example, bold or dashed lines, the structure or portion of the structure is to be interpreted as encompassing all stereoisomers of it. A bond drawn with a wavy line indicates that both stereoisomers are encompassed. This is not to be confused with a wavy line drawn perpendicular to a bond which indicates the point of attachment of a group to the rest of the molecule.

Tautomers

As known by those skilled in the art, certain compounds disclosed herein may exist in one or more tautomeric forms. Because one chemical structure may only be used to represent one tautomeric form, it will be understood that for convenience, referral to a compound of a given structural formula includes other tautomers of said structural formula. For exam le, the following is illustrative of tautomers of the compounds of Formula I:

Accordingly, the scope of the instant disclosure is to be understood to encompass all tautomeric forms of the compounds disclosed herein.

Isotopicallv-Labelled Compounds

Further, the scope of present disclosure includes all pharmaceutically acceptable isotopically-labelled compounds of the compounds disclosed herein, such as the compounds of Formula I, wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes suitable for inclusion in the compounds disclosed herein include isotopes of hydrogen, such as 2 H and 3 H, carbon, such as U C, 13 C and 14 C, chlorine, such as 36 CI, fluorine, such as 18 F, iodine, such as 123 I and 125 I, nitrogen, such as 13 N and 15 N, oxygen, such as 15 0, 17 0 and 18 0, phosphorus, such as 32 P, and sulphur, such as 35 S. Certain isotopically-labelled 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 ( 3 H) and carbon- 14 ( 14 C) are particularly useful for this purpose in view of their ease of incorporation and ready means of detection. Substitution with isotopes such as deuterium ( 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be advantageous in some circumstances. Substitution with positron emitting isotopes, such as U C, 18 F, 15 0 and 13 N, can be useful in Positron Emission Topography (PET) studies, for example, for examining target occupancy. Isotopically- labelled compounds of the compounds disclosed herein can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying General Synthetic Schemes and Examples using an appropriate isotopically-labelled reagents in place of the non-labelled reagent previously employed. Solvates

As discussed above, the compounds disclosed herein and the stereoisomers, tautomers and isotopically-labelled forms thereof or a pharmaceutically acceptable salt of any of the foregoing may exist in solvated or unsolvated forms.

The term "solvate" as used herein refers to a molecular complex comprising a compound or a pharmaceutically acceptable salt thereof as described herein and a stoichiometric or non-stoichiometric amount of one or more pharmaceutically acceptable solvent molecules. If the solvent is water, the solvate is referred to as a "hydrate."

Accordingly, the scope of the instant disclosure is to be understood to encompass all solvents of the compounds disclosed herein and the stereoisomers, tautomers and isotopically-labelled forms thereof or a pharmaceutically acceptable salt of any of the foregoing.

Amorphous and Crystalline Forms

In certain embodiments, the compounds described herein and the stereoisomers, tautomers, isotopically-labelled forms thereof or pharmaceutically acceptable salts of any of the foregoing or solvates of any of the foregoing may exist in different forms, such as amorphous forms and crystalline forms (polymorphs). Accordingly, the scope of the instant disclosure is to be understood to encompass all such forms. Miscellaneous Definitions

This section will define additional terms used to describe the scope of the compounds, compositions and uses disclosed herein.

The term "C x - y alkyl" as used herein refers to a straight or branched chain

hydrocarbon containing from x to y carbon atoms, for example, 1 to 4 and 1 to 6 carbon atoms. Representative examples of Ci-4alkyl include, but are not limited to, methyl, ethyl, n- propyl, iso-propyl, n-butyl, sec-butyl, iso-butyl, and tert-butyl. Representative examples of Ci-ealkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec- butyl, iso-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, and n-hexyl.

The term "C x - y alkenyl" as used herein refers to a straight or branched chain hydrocarbon containing from x to y carbon atoms, wherein the chain hydrocarbon has at least at least one carbon-carbon double bond. Representative examples of C2-ealkenyl include, but are not limited to, ethenyl, propenyl, allyl, butenyl and 4-methylbutenyl. The term "alkenyl" embraces chain hydrocarbons having "cis" and "trans" orientations, or alternatively, "E" and "Z" orientations, as appreciated by those of ordinary skill in the art.

The term "cycloalkyl" as used herein refers to a carbocyclic substituent obtained by removing hydrogen from a saturated carbocyclic molecule wherein the cyclic framework has, for example, 3 to 8 carbons (C3-8Cycloalkyl) or 3 to 6 carbons (C3-ecycloalkyl). A

"cycloalkyl" may be a monocyclic ring, examples of which include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.

The term "halogen" as used herein refers to -F, -CI, -Br, or -I.

The term "heteroaryl," as used herein refers to a monocyclic heteroaryl. The monocyclic heteroaryl is a 5 or 6 membered ring. The 5 membered ring consists of two double bonds and one, two, three or four nitrogen atoms and/or optionally one oxygen or sulfur atom. The 6 membered ring consists of three double bonds and one, two, three or four nitrogen atoms. The 5 or 6 membered heteroaryl is connected to the parent molecular moiety through any carbon atom or any nitrogen atom contained within the heteroaryl.

Representative examples of monocyclic heteroaryl include, but are not limited to, furyl, imidazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, oxazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, pyrazolyl, pyrrolyl, tetrazolyl, thiadiazolyl, thiazolyl, thienyl, triazolyl, and triazinyl. The term "5-membered nitrogen-containing heteroaryl" as used herein refers to a 5 membered heteroaryl ring as defined above, wherein one to four of the ring atoms are nitrogen and the remaining ring atoms are carbon. Examples of 5-membered nitrogen- containing heteroaryls include, but are not limited to, pyrrolyl, imidazolyl, pyrazolyl, and triazolyl.

The term "heterocycloalkyl" as used herein refers to a 3 to 8 membered cycloalkyl as defined above, wherein at least one of the ring carbon atoms is replaced with a heteroatom selected from nitrogen, oxygen or sulfur. Examples of six membered heterocycloalkyl include, but are not limited to, pyrrolidine, piperidine, piperazine, and morpholine.

The term "pharmaceutically acceptable" as used herein refers to generally recognized for use in subjects, particularly in humans.

The term "pharmaceutically acceptable salt" as used herein refers to a salt of a compound that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound. Such salts include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid,

methanesulfonic acid, and the like; or (2) salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, for example, an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, N-methylglucamine, dicyclohexylamine, and the like. Additional examples of such salts can be found in Berge et al. , J. Pharm. Sci.

66(1): 1-19 (1977). See also Stahl et al , Pharmaceutical Salts: Properties, Selection, and Use, 2 nd Revised Edition (2011).

The term "pharmaceutically acceptable excipient" as used herein refers to a broad range of ingredients that may be combined with a compound or salt disclosed herein to prepare a pharmaceutical composition or formulation. Typically, excipients include, but are not limited to, diluents, colorants, vehicles, anti-adherants, glidants, disintegrants, flavoring agents, coatings, binders, sweeteners, lubricants, sorbents, preservatives, and the like. The term "subject" as used herein refers to humans and mammals, including, but not limited to, primates, cows, sheep, goats, horses, dogs, cats, rabbits, rats, and mice. In one embodiment the subject is a human.

The term "treating" as used herein refers not only to treating a subject to relieve the subject of one or more signs and symptoms of a disease or condition or to eliminate one or more such signs and symptoms, but also to prophylactically treating an asymptomatic subject to prevent the onset of the disease or condition or preventing, slowing or reversing the progression of the disease or condition.

The term "therapeutically effective amount" as used herein refers to that amount of a compound disclosed herein that will elicit the biological or medical response of a tissue, a system, or subject that is being sought by a researcher, veterinarian, medical doctor or other clinician. The term also encompasses the amount of compound disclosed herein that will prevent or reduce the risk of occurrence of the biological or medical event that is sought to be prevented in a tissue, a system, or subject by a researcher, veterinarian, medical doctor or other clinician.

GENERAL SYNTHETIC PROCEDURES

The compounds provided herein can be synthesized according to the procedures described in this and the following sections. The synthetic methods described herein are merely exemplary, and the compounds disclosed herein may also be synthesized by alternate routes utilizing alternative synthetic strategies, as appreciated by persons of ordinary skill in the art. It should be appreciated that the general synthetic procedures and specific examples provided herein are illustrative only and should not be construed as limiting the scope of the present disclosure in any manner.

Generally, the compounds of Formula I can be synthesized according to the following schemes. Any variables used in the following schemes are the variables as defined for Formula I, unless otherwise noted. All starting materials are either commercially available, for example, from Sigma-Aldrich Chemical Company, Inc., St. Louis, MO, USA, or known in the art and may be synthesized by employing known procedures using ordinary skill. Starting material may also be synthesized via the procedures disclosed herein. Scheme

Compounds of Formula I may be synthesized as shown in Scheme 1. Compound i, wherein X is CI, Br, or I, may be synthesized by methods known to the person of ordinary skill in the art or as disclosed in WO2016022724, which is herein incorporated by reference in its entirety. Alkyne iii is obtained by reacting compound i with a metallated alkyne ii, wherein W is, for example, a trialkylstannyl, such as tributylstannyl, under Stille coupling conditions, using a Pd(0) catalyst, such as bis(tri-t-butylphosphine)palladium(0) in a suitable solvent, such as 1,4-dioxane. Alternatively, alkyne iii is obtained by reacting compound i with a silyl substituted alkyne ii, wherein W is, for example, trimethylsilyl, under

Sonogashira coupling conditions using, for example, copper(I) iodide, a base, such as trimethylamine, and a palladium catalyst, such as bis(triphenylphosphine)palladium(II) in a suitable solvent, such as tetrahydrofurane.

A compound of Formula I is obtained by reacting alkyne iii with compound iv under Sonogashira coupling conditions, such as the coupling conditions mentioned in the foregoing pharagraph. Alternatively, a compound of Formula I is obtained by reacting compound i with alkyne v under Sonogashira coupling conditions, such as the coupling conditions mentioned in the foregoing paragraph. As can be appreciated by the skilled artisan, the above synthetic schemes and representative examples are not intended to comprise a comprehensive list of all means by which the compounds described and claimed in this application may be synthesized. Further methods will be evident to those of ordinary skill in the art. Additionally, the various synthetic steps described above may be performed in an alternate sequence or order to give the desired compounds.

For example, in these procedures, the steps may be preceded, or followed, by additional protection/deprotection steps as necessary. Particularly, if one or more functional groups, for example carboxy, hydroxy, amino, or mercapto groups, are or need to be protected in preparing the compounds disclosed herein, because they are not intended to take part in a specific reaction or chemical transformation, various known conventional protecting groups may be used. For example, protecting groups typically utilized in the synthesis of natural and synthetic compounds, including peptides, nucleic acids, derivatives thereof and sugars, having multiple reactive centers, chiral centers and other sites potentially susceptible to the reaction reagents and/or conditions, may be used.

Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the compounds described herein are known in the art and include, for example, those such as described in R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, 3 rd edition, John Wiley and Sons (1999); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); A.

Katritzky and A. Pozharski, Handbook of Heterocyclic Chemistry, 2 nd edition (2001); M. Bodanszky, A. Bodanszky, The Practice of Peptide Synthesis, Springer- Verlag, Berlin Heidelberg (1984); J. Seyden-Penne, Reductions by the Alumino- and Borohydrides in Organic Synthesis, 2 nd edition, Wiley-VCH, (1997); and L. Paquette, editor, Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995).

All synthetic procedures described herein can be carried out under known reaction conditions, advantageously under those described herein, either in the absence or in the presence (usually) of solvents. As appreciated by those of ordinary skill in the art, the solvents should be inert with respect to, and should be able to dissolve, the starting materials and other reagents used. Solvents should be able to partially or wholly solubilize the reactants in the absence or presence of catalysts, condensing agents or neutralizing agents, for example ion exchangers, typically cation exchangers for example in the H + form. The ability of the solvent to allow and/or influence the progress or rate of the reaction is generally dependent on the type and properties of the solvent(s), the reaction conditions including temperature, pressure, atmospheric conditions such as in an inert atmosphere under argon or nitrogen, and concentration, and of the reactants themselves.

Suitable solvents for conducting reactions to synthesize the compounds provided herein include, but are not limited to, water; esters, including lower alkyl-lower alkanoates, for example, EtOAc; ethers including aliphatic ethers, for example, Et20 and ethylene glycol dimethylether or cyclic ethers, for example, THF; liquid aromatic hydrocarbons, for example, benzene, toluene and xylene; alcohols, for example, MeOH, EtOH, 1-propanol, iPrOH, n- and t-butanol; nitriles, for example, CH3CN; halogenated hydrocarbons, for example,

CH 2 CI 2 , CHCI3 and CC ; acid amides, for example, DMF; sulfoxides, for example, DMSO; bases, including heterocyclic nitrogen bases, for example, pyridine; carboxylic acids, for example, lower alkanecarboxylic acids, for example, AcOH; inorganic acids, for example, HC1, HBr, HF, and H2SO4; carboxylic acid anhydrides, for example, lower alkane acid anhydrides, for example, acetic anhydride; cyclic, linear, or branched hydrocarbons, for example, cyclohexane, hexane, pentane, and isopentane; and mixtures of any of these solvents, such as purely organic solvent combinations, or water-containing solvent combinations, for example, aqueous solutions. These solvents and solvent mixtures may also be used in "working-up" the reaction as well as in processing the reaction and/or isolating the reaction product(s), such as in chromatography.

Purification methods are known in the art and include, for example, crystallization, chromatography (for example, liquid and gas phase), extraction, distillation, trituration, and reverse phase HPLC. Reactions conditions such as temperature, duration, pressure, and atmosphere (inert gas, ambient) are known in the art and may be adjusted as appropriate for the reaction.

The disclosure further encompasses "intermediate" compounds, including structures produced from the synthetic procedures described, whether isolated or generated in-situ and not isolated, prior to obtaining the finally desired compound. Structures resulting from carrying out steps from a transient starting material, structures resulting from divergence from the described method(s) at any stage, and structures forming starting materials under the reaction conditions are all "intermediates" included in the scope of this disclosure.

Further, processes for making and further reacting these intermediates are also understood to be encompassed in the scope of this disclosure.

Also provided herein are new starting materials and/or intermediates, as well as processes for the preparation thereof. In select embodiments, such starting materials are used and reaction conditions so selected as to obtain the desired compound(s). Starting materials are either known, commercially available, or can be synthesized in analogy to or according to methods that are known in the art. Many starting materials may be prepared according to known processes and, in particular, can be prepared using processes described in the examples. In synthesizing starting materials, functional groups may be protected with suitable protecting groups when necessary. Protecting groups, their introduction and removal are described above.

EXAMPLES

This section provides specific examples of compounds of Formula I and methods of making the same.

List of Abbreviations

Table 1

Boc tert-butylcarbonyl

CDI carbonyldiimidazole

DCM dichloromethane

DIAD diisopropyl azodicarboxylate

Ghosez's reagent l-chloro-N,N,2-trimethylpropenylamine

HATU 1 - [bis(dimethylamino)methy lene] - 1 H- 1 ,2, 3 -triazolo [4,5- b]pyridinium 3-oxid hexafluorophosphate

PTSA p-toluene sulfonic acid

SEM [2-(tTrimethylsilyl)ethoxy]methyl

T3P 1-propanephosphonic anhydride

TFA trifluoroacetic acid

THF tetrahydrofuran TMS trimethylsilyl

TsCl p-tolunesulfonyl chloride

General Analytical and Purification Methods

Provided in this section are descriptions of the general analytical and purification methods used to prepare the specific compounds provided herein.

Chromatography :

Unless otherwise indicated, crude product-containing residues were purified by passing the crude material or concentrate through either a Biotage or Isco brand silica gel column (pre-packed or individually packed with S1O2) and eluting the product off the column with a solvent gradient as indicated. For example a description of (330 g S1O2, 0-40% EtOAc/hexane) means the product was obtained by elution from the column packed with 330grams of silica, with a solvent gradient of 0% to 40% EtOAc in hexanes.

Preparative HPLC Method:

Where so indicated, the compounds described herein were purified via reverse phase HPLC using one of the following instruments: Shimadzu, Varian, Gilson; utilizing one of the following two HPLC columns: (a) a Phenomenex Luna or (b) a Gemini column (5 micron or 10 micron, C18, 150x50 mm)

A typical run through the instrument included: eluting at 45 mL/min with a linear gradient of 10% (v/v) to 100% MeCN (0.1% v/v TFA) in water (0.1% TFA) over 10 minutes; conditions can be varied to achieve optimal separations.

Proton NMR Spectra:

Unless otherwise indicated, all l NMR spectra were collected on a Bruker NMR

Instrument at 300 MHz or 400 MHz. Where so characterized, all observed protons are reported as parts-per-million (ppm) downfield from tetramethylsilane (TMS) or other internal reference in the appropriate solvent indicated. F NMR Spectra:

Unless otherwise indicated, all 19 F NMR spectra were run on a Bruker NMR

Instrument at 376 MHz. All observed protons are reported as parts-per-million (ppm) downfield.

Mass Spectra (MS)

Unless otherwise indicated, all mass spectral data for starting materials, intermediates and/or exemplary compounds are reported as mass/charge (m/z), having an (M+H + ) molecular ion. The molecular ion reported was obtained by electrospray detection method (commonly referred to as an ESI MS) utilizing a PE SCIEX API 150EX MS instrument or an Agilent 1100 series LC/MSD system. Compounds having an isotopic atom, such as bromine and the like, are generally reported according to the detected isotopic pattern, as appreciated by those skilled in the art.

Compound Names

The compounds disclosed and described herein have been named using either (1) the naming convention provided with Chem-Draw Ultra 12.0.3. software, available in Chem Office, or (2) by the ISIS database software (Advanced Chemistry Design Labs or ACD software).

Specific Examples

Provided in this section are the procedures to synthesize specific examples of the compounds provided herein. All starting materials are either commercially available from Sigma-Aldrich Chemical Company, Inc., St. Louis, MO, USA, unless otherwise noted, or known in the art and may be synthesized by employing known procedures using ordinary skill.

Preparation of ((lS,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5-(fluorometh yl)-2- thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)methanol (100b).

To a solution of 100a (prepared according to the procedures reported in WO

2016022724) (0.74 g, 1.90 mmol) in THF (8 mL) at room temperature was added lithium borohydride (2.0 M in THF, 3.0 mL, 6.0 mmol) and methanol (0.62 mL, 15.30 mmol). The reaction mixture was stirred at room temperature for 1 h and quenched slowly with sat'd aqueous NH4CI. After the bubbling ceased, the mixture was transferred to a separatory funnel and diluted with water and EtOAc. The aqueous phase was extracted with EtOAc (2 x) and the combined organic extracts were washed with brine (lx), dried over MgSO/t, filtered, and concentrated to give a yellow oil. Purification by silica gel chromatography (40 to 100% EtOAc in heptane) gave ((lS,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5-(fluorometh yl)- 2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)methanol (100b, 0.45 g, 1.24 mmol, 66% yield) as a white solid. LC/MS (ESI + ) m/z = 362.9/364.9 [M+H] + . 'H NMR (400 MHz, CDC1 3 ) δ 0.69 (t, J= 6.3 Hz, 1H), 1.04 (dd, J = 9.6, 5.9 Hz, 1H), 1.75 (s br, 1H), 1.80 (t, J= 8.8 Hz, 1H), 3.59 (d, J = 11.7 Hz, 1H), 3.75 (d, J= 11.9 Hz, 1H), 4.62 (dd, J = 47.0, 8.22 Hz, 1H), 4.65 (s br, 2H), 4.84 (dd, J= 47.0, 8.4 Hz, 1H), 6.95 (dd, J= 11.5, 8.6 Hz, 1H), 7.38 (ddd, J= 8.6, 4.2, 2.7 Hz, 1H), 7.77 (dd, J= 6.9, 2.5 Hz, 1H). Preparation of ((lS,5S)-3-amino-5-(2-fluoro-5-((trimethylsilyl)ethynyl)phen yl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)meth anol (100c).

To a mixture of dichlorobis(triphenylphosphine)palladium (II) (Strem Chemicals Inc., Newburyport, MA, USA) (122 mg, 0.17 mmol), copper(I) iodide (Sigma-Aldrich, St. Louis, MO, USA) (45 mg, 0.24 mmol), and ((lS,5S)-3-amino-5-(5-bromo-2-fluorophenyl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)meth anol (100b, 446 mg, 1.23 mmol) was added THF (3 mL), trimethylsilylacetylene (Sigma-Aldrich, St. Louis, MO, USA) (1.70 mL, 12.0 mmol), and triethylamine (4.0 mL, 29 mmol). The reaction mixture was degassed by bubbling nitrogen through the solution for 5 minutes and the reaction mixture was heated at 60 °C in a sealed vial for 22 hours. The reaction mixture was cooled to room temperature and diluted with EtOAc. The organic phase was washed with sat'd aqueous NH4CI (1 x), brine (1 x), dried over MgS04, filtered, and concentrated. Purification by silica gel chromatography (10 to 90% EtOAc in hexane) gave ((lS,5S)-3-amino-5-(2-fluoro-5- ((trimethylsilyl)ethynyl)phenyl)-5 -(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en- 1 - yl)methanol (100c, 437 mg, 1.15 mmol, 94% yield) as a light brown foam. LC/MS (ESI + ) m/z = 381.0 [M+H] + . ¾ ΝΜΡν (400 MHz, CDCI3) δ 0.25 (s, 9H), 0.68 (t, J= 6.3 Hz, 1H), 1.01 - 1.08 (m, 1H), 1.66 (s br, 1H), 1.78 - 1.85 (m, 1H), 3.60 (d, J= 11.9 Hz, 1H), 3.76 (d, J= 11.9 Hz, 1H), 4.60 (dd, J= 47.3, 8.6 Hz, 1H), 4.61 (s br, 2H), 4.87 (dd, J= 47.3, 8.4 Hz, 1H), 7.00 (dd, J= 11.9, 8.4 Hz, 1H), 7.35 - 7.42 (m, 1H), 7.75 (dd, J = 7.6, 2.0 Hz, 1H). Preparation of ((lS,5S,6S)-3-amino-5-(5-ethynyl-2-fluorophenyl)-5-(fluorome thyl)-2- thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)methanol (lOOd).

To a solution of ((lS,5S,6S)-3-amino-5-(2-fluoro-5-((trimethylsilyl)ethynyl)p henyl)- 5-(fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)me thanol (100c, 437 mg, 1.15 mmol) in MeOH (5 mL) at room temperature was added potassium carbonate (396 mg, 2.87 mmol). The reaction mixture was heated at 50 °C for 15 minutes and cooled to RT. It was diluted with EtOAc, filtered through a fine frit, and concentrated. Purification by flash column chromatography on silica gel (24 g, 10% to 90% EtOAc (10% MeOH) in heptane) gave ((lS,5S,6S)-3-amino-5-(5-ethynyl-2-fluorophenyl)-5-(fluorome thyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-l-yl)methanol (lOOd, 252 mg, 0.82 mmol, 71% yield) as a pale yellow foam. LC/MS (ESI + ) m/z = 309.0 [M+H] + . Preparation of 6-((3-((lS,5S,6S)-3-amino-5-(fluoromethyl)-l-(hydroxymethyl) -2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile (100).

To a mixture of 2-bromo-5-cyanopyridine (Sigma-Aldrich, St. Louis, MO, USA) (241 mg, 1.32 mmol), ((lS,5S,6S)-3-amino-5-(5-ethynyl-2-fluorophenyl)-5-(fluorome thyl)- 2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)methanol (lOOd, 252 mg, 0.817 mmol), dichlorobis(triphenylphosphine)palladium (II) (80 mg, 0.11 mmol), and copper(I) iodide (23 mg, 0.12 mmol) was added THF (2.7 mL) and triethylamine (0.23 mL, 1.65 mmol). The reaction mixture was degassed by bubbling nitrogen through the solution for 5 minutes and the reaction mixture was heated at 60 °C for 2.5 hours then cooled to room temperature and diluted with EtOAc. The mixture was filtered through a medium glass frit and the filtrate was concentrated. Purification of the residue by flash column chromatography on silica gel (10 to 90% EtOAc (5% MeOH) in hexanes) gave 6-((3-((lS,5S,6S)-3-amino-5-(fluoromethyl)-l- (hydroxymethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4- fluorophenyl)ethynyl)nicotinonitrile (Example 100, 172 mg, 0.42 mmol, 51% yield) as a white solid. LC/MS (ESI + ) m/z = 4U .O [M+H] + . ¾ NMR (400 MHz, CDCI 3 ) δ 0.70 (t, J= 6.26 Hz, 1H), 1.06 (dd, J = 9.7, 5.8 Hz, 1H), 1.68 (s br, 1H), 1.82 (t, J = 7.9 Hz, 1H), 3.61 (d, J= 11.9 Hz, 1H), 3.76 (d, J= 11.9 Hz, 1H), 4.65 (s br, 2H), 4.66 (dd, J= 47.3, 8.6 Hz, 1H), 4.87 (dd, J= 47.3, 8.6 Hz, 1H), 7.09 (dd, J= 11.7, 8.4 Hz, 1H), 7.51 - 7.57 (m, 1H), 7.60 (d, J= 8.2 Hz, 1H), 7.94 (d, J= 8.0 Hz, 2H), 8.87 (s, 1H). Example 101: 6-((3-((lS,5S,6S)-3-amino-l,5-bis(fluoromethyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile.

101d 101 Preparation of (lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-l,5-bis(fluoromethyl)- 2-thia-4- azabicyclo[4.1.0]hept-3-en-3-amine (101b).

A mixture of fert-butyl ((lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-l,5- bis(fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-3-yl)( (2- (trimethylsilyl)ethoxy)methyl)carbamate (101a, prepared according to the procedures reported in WO 2016022724) (1.30 g, 2.18 mmol) in sulfuric acid (8 mL, 150 mmol) was stirred at room temperature for 15 min. Ice was added, and the mixture was diluted with EtOAc. 10 M aqueous NaOH was added slowly until the mixture was at approximately pH 7. The organic layer was separated, and the aqueous layer was extracted twice more with EtOAc. The combined organic extracts were washed with brine, dried over MgSO/t, filtered, and concentrated in vacuo. Chromatographic purification of the residue (silica gel, 0 to 30% EtOAc in heptane) gave (lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-l,5-bis(fluoromethyl)- 2- thia-4-azabicyclo[4.1.0]hept-3-en-3-amine (101b, 565 mg, 1.55 mmol, 71% yield) as a colorless oil that partially solidified upon standing. LC/MS (ESI + ) m/z = 365.0/367.0

[M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 7.73 (dd, J = 7.04, 2.54 Hz, 1H) 7.39 (ddd, J = 8.61, 4.30, 2.54 Hz, 1H) 6.95 (dd, J = 11.54, 8.61 Hz, 1H) 4.58 - 4.88 (m, 2H) 4.47 (dd, J = 35.80, 9.98 Hz, 1H) 4.35 (dd, J = 35.21, 10.17 Hz, 1H) 1.82 - 1.90 (m, 1H) 1.18 (dd, J = 9.68, 5.97 Hz, 1H) 0.78 (td, J = 6.41, 3.62 Hz, 1H). NH 2 peak was not observed. Preparation of (lS,5S,6S)-5-(2-fluoro-5-((trimethylsilyl)ethynyl)phenyl)-l, 5- bis(fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-3-amin e (101c).

101b (565 mg, 1.55 mmol), trans-dichlorobis(triphenylphosphine)palladium (II) (163 mg, 0.23 mmol), and copper(I) iodide (58.9 mg, 0.309 mmol) were mixed in a round bottom flask and placed under a nitrogen atmosphere. THF (4 mL), (trimethylsilyl)acetylene (2.19 mL, 15.47 mmol), and triethylamine (5.0 mL, 35.9 mmol) were added, and the reaction mixture was stirred at 50 °C for 15 hours. Additional trans- dichlorobis(triphenylphosphine)palladium (II) (163 mg, 0.23 mmol), copper(I) iodide (58.9 mg), and (trimethylsilyl)acetylene (2.19 ml) were added, and the reaction was stirred at 50 °C for another 6 hours. The reaction mixture was cooled to room temperature and diluted with EtOAc. The mixture was washed with sat'd aqueous NH4CI followed by brine, dried over MgSOzi, filtered, and concentrated in vacuo. Chromatographic purification of the residue (silica gel, 0 to 25% EtOAc in heptane) gave (lS,5S,6S)-5-(2-fluoro-5- ((trimethylsilyl)ethynyl)phenyl)-l,5-bis(fluoromethyl)-2-thi a-4-azabicyclo[4.1.0]hept-3-en-3- amine (101c, 472 mg, 1.23 mmol, 80 % yield) as an orange solid. LC/MS (ESI + ) m/z = 383.0 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 7.70 (dd, J = 7.63, 1.96 Hz, 1H) 7.36 - 7.41 (m, 1H) 6.99 (dd, J = 11.93, 8.41 Hz, 1H) 4.55 - 4.89 (m, 2H) 4.27 - 4.54 (m, 2H) 1.86 (t, J = 8.22 Hz, 1H) 1.18 (dd, J = 9.59, 6.06 Hz, 1H) 0.73 - 0.80 (m, 1H) 0.24 (s, 9 H). N¾ peak was not observed.

Preparation of (lS,5S,6S)-5-(5-ethynyl-2-fluorophenyl)-l,5-bis(fluoromethyl )-2-thia-4- azabicyclo[4.1.0]hept-3-en-3-amine (101 d).

Potassium carbonate (426 mg, 3.08 mmol) was added to a stirred solution of 101c

(472 mg, 1.23 mmol) in methanol (6 mL) at room temperature. The reaction mixture was warmed to 50 °C and stirred for 20 minutes. The reaction mixture was cooled to room temperature, diluted with EtOAc, and filtered. The filtrate was concentrated in vacuo.

Chromatographic purification of the residue (silica gel, 0 to 50% EtOAc in heptane) gave (lS,5S,6S)-5-(5-ethynyl-2-fluorophenyl)-l,5-bis(fluoromethyl )-2-thia-4- azabicyclo[4.1.0]hept-3-en-3-amine (101d, 59 mg, 0.19 mmol, 15% yield) as an off-white solid. LC/MS (ESI + ) m/z = 311.1 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 7.72 (dd, J = 7.63, 2.15 Hz, 1H) 7.42 (ddd, J = 8.36, 4.74, 2.15 Hz, 1H) 7.02 (dd, J = 11.74, 8.41 Hz, 1H) 4.58 - 4.90 (m, 2H) 4.28 - 4.54 (m, 2H) 3.03 (s, 1H) 1.87 (t, J = 8.22 Hz, 1H) 1.20 (dd, J = 9.68, 5.97 Hz, 1H) 0.77 (td, J = 6.41, 3.62 Hz, 1H). NH 2 peak was not observed.

Preparation of 6-((3-((lS,5S,6S)-3-amino-l,5-bis(fluoromethyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile (101).

lOld (59 mg, 0.19 mmol), 2-bromo-5-cyanopyridine (52 mg, 0.28 mmol), trans- dichlorobis(triphenylphosphine)palladium (II) (14 mg, 0.02 mmol), and copper(I) iodide (6 mg, 0.03 mmol) were mixed in a round bottom flask and placed under a nitrogen atmosphere. THF (1 mL) and triethylamine (0.053 mL, 0.380 mmol) were added, and the reaction mixture was stirred at 60 °C for 1.5 hours. The reaction mixture was cooled to room temperature and diluted with EtOAc. The mixture was washed with sat'd aqueous NH4CI followed by brine, dried over MgSO/i, filtered, and concentrated in vacuo. Chromatographic purification of the residue (silica gel, 0 to 50% EtOAc in heptane) gave 6-((3-((lS,5S,6S)-3-amino-l,5- bis(fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)- 4- fluorophenyl)ethynyl)nicotinonitrile (101, 33 mg, 0.08 mmol, 42% yield) as an off-white solid. LC/MS (ESI + ) m/z = 413.1 [M+H] + . ¾ NMR (400 MHz, DMSO-d6) δ ppm 9.05 (d, J = 1.37 Hz, 1H) 8.38 (dd, J = 8.22, 2.15 Hz, 1H) 7.96 (dd, J = 7.63, 2.15 Hz, lH) 7.85 (d, J = 8.22 Hz, 1H) 7.67 (ddd, J = 8.31, 4.50, 2.25 Hz, 1H) 7.35 (dd, J = 11.93, 8.41 Hz, 1H) 6.54 (s, 2H) 4.58 - 4.77 (m, 2H) 4.38 - 4.58 (m, 2H) 1.85 (t, J = 8.12 Hz, 1H) 1.19 (dd, J = 9.68, 5.38 Hz, 1H) 0.58 - 0.64 (m, 1H).

Example 102: (lS,5S,6S)-5-(5-((5-chloropyridin-2-yl)ethynyl)-2-fluorophen yl)-l,5- bis(fl

101 d 102

This compound (17 mg, 0.040 mmol, 38% yield) as an off-white solid was prepared in a fashion similar to that described for Example 101, here using lOld (32 mg, 0.10 mmol) and 2-bromo-5-chloropyridine (30 mg, 0.16 mmol) as starting materials. LC/MS (ESI + ) m/z = 422.0 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 8.56 (d, J = 2.35 Hz, 1H) 7.85 (dd, J = 7.53, 2.05 Hz, 1H) 7.66 (dd, J = 8.41, 2.54 Hz, 1H) 7.51 (ddd, J = 8.41, 4.69, 2.15 Hz, 1H) 7.45 (d, J = 8.41 Hz, 1H) 7.07 (dd, J = 11.74, 8.41 Hz, 1H) 4.59 - 4.91 (m, 2H) 4.28 - 4.55 (m, 2H) 1.87 (t, J = 8.31 Hz, 1H) 1.20 (dd, J = 9.68, 5.97 Hz, 1H) 0.78 (td, J = 6.36, 3.52 Hz, 1H). N¾ peak was not observed.

Example 103: 5-((3-((lS,5S,6S)-3-amino-l,5-bis(fluoromethyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)pyra zine-2-carbonitrile

This compound (18 mg, 45% yield) as a tan solid was prepared in a fashion similar to that described for Example 101, here using lOld (30 mg, 0.10 mmol) and 2-bromo-5- cyanopyrazine (Apollo Scientific Ltd, Manchester, UK, 27 mg, 0.15 mmol) as starting materials. LC/MS (ESI + ) m/z = 414.0 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 8.87 (d, J = 1.37 Hz, 1H) 8.80 (d, J = 1.37 Hz, 1H) 7.94 (dd, J = 7.43, 2.15 Hz, 1H) 7.59 (ddd, J = 8.41, 4.69, 2.15 Hz, 1H) 7.13 (dd, J = 11.74, 8.41 Hz, 1H) 4.62 - 4.90 (m, 2H) 4.30 - 4.54 (m, 2H) 1.89 (t, J = 8.22 Hz, 1H) 1.21 (dd, J = 9.59, 6.06 Hz, 1H) 0.80 (td, J = 6.36, 3.72 Hz, 1H). N¾ peak was not observed.

Example 104: 6-((3-((lS,5S,6S)-3-amino-l,5-bis(fluoromethyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)-5-m ethylnicotinonitrile

This compound (16 mg, 0.04 mmol, 39% yield) as a white solid was prepared in a fashion similar to that described Example 101, here using lOld (30 mg, 0.10 mmol) and 2- bromo-5-cyano-3-picoline (29 mg, 0.15 mmol) as starting materials. LC/MS (ESI + ) m/z = 427.1 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 8.70 (d, J = 1.37 Hz, 1H) 7.89 (dd, J = 7.53, 2.05 Hz, 1H) 7.81 (d, J = 1.17 Hz, 1H) 7.55 (ddd, J = 8.31, 4.60, 2.15 Hz, 1H) 7.10 (dd, J = 11.74, 8.41 Hz, 1H) 4.62 - 4.91 (m, 2H) 4.29 - 4.54 (m, 2H) 2.56 (s, 3H) 1.88 (t, J = 8.31 Hz, 1H) 1.21 (dd, J = 9.59, 6.06 Hz, 1H) 0.76 - 0.82 (m, 1H). NH 2 peak was not observed. Example 105: (lS,5S,6S)-5-(5-((5-(but-2-yn-l-yloxy)pyridin-2-yl)ethynyl)- 2-

Preparation of 2-bromo-5-(but-2-yn-l-yloxy)pyridine (105a).

2-Bromo-5-hydroxypyridine (348 mg, 2.00 mmol), l-bromo-2-butyne (1064 mg,

8.00 mmol), and potassium carbonate (691 mg, 5.00 mmol) were mixed in acetonitrile (8 mL) in a sealed vial. The reaction mixture was heated to 80 °C for 3 hours. The reaction mixture was cooled to room temperature, diluted with EtOAc, and quenched with sat'd aqueous NH4CI. The organic layer was separated, washed with brine, dried over MgSO/t, filtered, and concentrated in vacuo. Chromatographic purification of the residue (silica gel, 0 to 40% EtOAc in heptane) gave 2-bromo-5-(but-2-yn-l-yloxy)pyridine (105a, 345 mg, 1.53 mmol, 76% yield) as a white solid. LC/MS (ESI + ) m/z = 226.0/228.0 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 8.14 (d, J = 3.13 Hz, 1H) 7.38 (d, J = 8.80 Hz, 1H) 7.18 (dd, J = 8.71, 3.23 Hz, 1H) 4.68 (q, J = 2.28 Hz, 2H) 1.85 (t, J = 2.35 Hz, 3H). Preparation of (lS,5S,6S)-5-(5-((5-(but-2-yn-l-yloxy)pyridin-2-yl)ethynyl)- 2- fluorophenyl)-l,5-bis(fluoromethyl)-2-thia-4-azabicyclo[4.1. 0]hept-3-en-3-amine (105).

This compound (8 mg, 0.018 mmol, 18% yield) as an off-white solid was prepared in a fashion similar to that described for Example 101, here using lOld (30 mg, 0.10 mmol) and 105a (33 mg, 0.15 mmol) as starting materials. LC/MS (ESI + ) m/z = 456.0 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 8.36 (d, J = 2.74 Hz, 1H) 7.78 (dd, J = 7.53, 2.05 Hz, 1H) 7.48 - 7.53 (m, 1H) 7.48 (d, J = 8.80 Hz, 1H) 7.25 - 7.29 (m, 1H) 7.06 (dd, J = 11.74, 8.41 Hz, 1H) 4.61 - 4.94 (m, 2H) 4.73 (q, J = 2.22 Hz, 2H) 4.28 - 4.55 (m, 2H) 1.92 (t, J = 8.41 Hz, 1H) 1.86 (t, J = 2.35 Hz, 3H) 1.25 (dd, J = 9.59, 6.06 Hz, 1H) 0.81 (td, J = 6.31, 3.42 Hz, 1H). N¾ peak was not observed.

Example 106: 6-((3-((lR,5S,6S)-3-amino-l-(cyanomethyl)-5-(fluoromethyl)-2 -thia-4-

Preparation of 2-((lR,5S,6S)-3-amino-5-(2-fluoro-5-((trimethylsilyl)ethynyl )phenyl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)acet onitrile (106b).

To a solution of 2-((lR,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)acet onitrile (106a, prepared according to the procedures reported in WO 2016022724) (260 mg, 0.70 mmol) in THF (2.0 mL) was added (trimethylsilyl)-acetylene (1.97 mL, 13.97 mmol), copper(I) iodide (27 mg, 0.14 mmol), triethylamine (0.29 mL, 2.09 mmol), and dichlorobis(triphenyl- phosphine)palladium (II) (98 mg, 0.14 mmol). The resulting mixture was purged with nitrogen for 3 min, then capped with automatically release pressure cap, and stirred at 60 °C overnight. The mixture was quenched with sat'd aqueous NH4CI (10 mL) and extracted with EtOAc (2 x 30 mL). The combined organic extracts were dried over MgSC>4 and concentrated in vacuo. The residue was purified by silica gel chromatography (0 to 100% EtOAc in heptane) to give 2-((lR,5S,6S)-3-amino-5-(2-fluoro-5-((trimethylsilyl)ethynyl )phenyl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)acet onitrile (106b, 272 mg) as a yellow oil. LC/MS (ESI + ) m/z = 390.0 [M+H] + . Preparation of 6-((3-((lR,5S,6S)-3-amino-l-(cyanomethyl)-5-(fluoromethyl)-2 -thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile (106).

To a solution of 2-((lR,5S,6S)-3-amino-5-(2-fluoro-5- ((trimethylsilyl)ethynyl)phenyl)-5 -(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en- 1 - yl)acetonitrile (106b, 272 mg, 0.70 mmol) in methanol (3 mL) was added potassium carbonate (241 mg, 1.74 mmol). The resulting mixture was stirred at 50 °C for 15 min, then cooled to room temperature and diluted with EtOAc (10 mL). The mixture was washed with sat'd aqueous NaHCC , dried over MgSO/t, and concentrated in vacuo to provide 2- ((lR,5S,6S)-3-amino-5-(5-ethynyl-2-fluorophenyl)-5-(fluorome thyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-l-yl)acetonitrile (106c) as a yellow oil. LC/MS (ESI + ) m/z = 318.0 [M+H] + . To the resulting yellow oil 106c in THF (4 mL) was added 2-bromo-5- cyanopyridine (192 mg, 1.05 mmol), copper(I) iodide (27 mg, 0.14 mmol), bis(triphenyl- phosphine)palladium (II) (98 mg, 0.14 mmol), and triethylamine (0.20 mL, 1.40mmol). The mixture was heated at 60 °C for 1 hour, then cooled to room temperature and quenched with sat'd aqueous NH4CI (20 mL). The mixture was extracted with EtOAc (2 x 20 mL). The combined organic extracts were dried over MgS04 and concentrated in vacuo. The residue was purified by silica gel chromatography (0 to 100% EtOAc in heptane) to give 6-((3- ((lR,5S,6S)-3-amino-l-(cyanomemyl)-5-(fluoromethyl)-2-thia-4 -azabicyclo[4.1.0]hept-3-en- 5-yl)-4-fluorophenyl)ethynyl)nicotinonitrile (Example 106, 47 mg, 0.11 mmol, 16% yield for 2 steps) as a yellow solid. LC/MS (ESI + ) m/z = 420.0 [M+H] + . ¾ NMR (400 MHz,

CHLOROFORM-d) δ 8.87 (br s, 1H), 7.95 (br d, J = 8.22 Hz, 1H), 7.80 (br d, J = 5.87 Hz, 1H), 7.64 (br d, J = 7.43 Hz, 1H), 7.57-7.61 (m, 1H), 7.13 (dd, J = 8.22, 11.54 Hz, 1H), 4.63- 5.02 (m, 2H), 2.82 (s, 2H), 2.04 (br s, 1H), 1.28-1.35 (m, 1H), 0.86 (br t, J = 6.55 Hz, 1H). NH2 peak was not observed.

Example 107: 6-((3-((lR,5S,6S)-3-amino-l-((E)-2-cyanovinyl)-5-(fluorometh yl)-2-thia-4-

107c 107

Preparation of tert-butyi ((lR,5S,6S)-5-(5-bromo-2-fluorophenyl)-l-((E)-2-cyanovinyl)- 5-(fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-3-yl)(( 2- (trimethylsilyl)ethoxy)methyl)carbamate (107b).

To a stirred solution of diethyl cyanomethylphosphonate (0.72 mL, 4.06 mmol) in THF (5 mL) was added potassium fert-butoxide (1.0 M solution in THF, 4.06 mL, 4.06 mmol). The mixture was stirred for 30 minutes then added to a stirred solution of fert-butyl ((lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-5-(fluoromethyl)-l-fo rmyl-2-thia-4- azabicyclo[4.1.0]hept-3-en-3-yl)((2-(trimethylsilyl)ethoxy)m ethyl)carbamate (107a, prepared according to the procedures reported in WO 2016022724) (2.07 g, 3.38 mmol) in 20 mL of THF. The resulting mixture was stirred for 1 hour, then quenched with saturated aqueous NH4CI and extracted with EtOAc (3 x). The extracts were dried over Na2SO/t, concentrated and the residue was purified on a silica gel column (5 to 10% EtOAc in hexanes) to give tert- butyl ((lR,5S,6S)-5-(5-bromo-2-fluorophenyl)-l-((E)-2-cyanovinyl)- 5-(fluoromethyl)-2-thia- 4-azabicyclo[4.1.0]hept-3-en-3-yl)((2-(trimethylsilyl)ethoxy )methyl)carbamate (107b, 0.97 g, 47% yield). LC/MS (ESI+) m/z = 638.0/640.0 [M+H] + . Preparation of (E)-3-((lR,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5-(fluo romethyl)- 2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)acrylonitrile (107c).

To a flask containing 107b (0.79 g, 1.28 mmol) at 0 °C was added TFA/H 2 S0 4 (9: 1, 10 mL). The mixture was stirred at 0 °C for 30 minutes, and then concentrated to remove excess of TFA. The residue was cooled with an ice bath and basified by the dropwise addition of sat'd aqueous Na2C03 until pH >9. The mixture was extracted with DCM (3 x). The extracts were dried over Na2S04 and concentrated to give (E)-3-((lR,5S,6S)-3-amino-5- (5 -bromo-2-fluorophenyl)-5 -(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en- 1 - yl)acrylonitrile (107c, 0.38 g, 76% yield). LC/MS (ESI+) m/z = 386.0/388.0 [M+H] + .

Preparation of 6-((3-((lR,5S,6S)-3-amino-l-((E)-2-cyanovinyl)-5-(fluorometh yl)-2-thia- 4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)ni cotinonitrile (107).

This compound (7 mg, 16.2 μιηοΐ, 6% yield) was prepared in a manner similar to that described for Example 106, here starting with 107c (E)-3-((lR,5S,6S)-3-amino-5-(5-bromo- 2-fluorophenyl)-5 -(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en- 1 -yl)acrylonitrile (100 mg, 0.26 mmol). LC/MS (ESI+) m/z = 432.0 [M+H] + . ¾ NMR (DMSO-d6) δ 9.05-9.09 (m, 1H), 8.40 (dd, J = 8.2, 2.2 Hz, 1H), 7.91 (dd, J = 7.6, 2.2 Hz, 1H), 7.86 (dd, J = 8.2, 0.8 Hz, 1H), 7.65-7.73 (m, 1H), 7.38 (dd, J = 11.9, 8.6 Hz, 1H), 6.80 (d, J = 16.2 Hz, 1H), 6.69 (s, 2H), 5.76 (d, J = 16.2 Hz, 1H), 4.70-4.80 (m, 1H), 4.59-4.68 (m, 1H), 2.15 (t, J = 8.5 Hz, 1H), 1.59 (dd, J = 9.7, 5.2 Hz, 1H), 0.93-1.04 (m, 1H).

Example 108: (lS,5S,6S)-5-(2-fluoro-5-((5-(oxazol-2-ylmethoxy)pyridin-2-

Preparation of 2-(((6-bromopyridin-3-yl)oxy)methyl)oxazole (108a).

Triphenylphosphine (211 mg, 0.80 mmol) and 2-bromo-5-hydroxypyridine (100 mg,

0.57mmol) were mixed in THF (2 inL) at 0 °C. 2-Oxazolemethanol (Scientific, Matrix Columbia, SC, USA) (68 mg, 0.69 mmol) and diisopropyl azodicarboxylate (0.16 mL, 0.80 mmol) were added, and the reaction mixture was stirred at 0 °C for 30 minutes before being warmed to room temperature and stirred for 16 hours. The reaction mixture was concentrated in vacuo. Chromatographic purification of the residue (silica gel, 0 to 50% EtOAc in heptane) gave 2-(((6-bromopyridin-3-yl)oxy)methyl)oxazole (108a, 152 mg, 0.60 mmol, 104% yield) as a white solid. LC/MS (ESI + ) m/z = 255.1/257.1 [M+H] + . 1 H NMR (400 MHz, CHLOROFORM-d) δ ppm 8.18 (d, J = 2.93 Hz, 1H) 7.70 (s, 1H) 7.39 (d, J = 8.80 Hz, 1H) 7.24 - 7.28 (m, 1H) 7.17 (s, 1H) 5.19 (s, 2H). Preparation of (lS,5S,6S)-5-(2-fluoro-5-((5-(oxazol-2-ylmethoxy)pyridin-2- yl)ethynyl)phenyl)-l,5-bis(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3-en-3-amine (108).

This compound (5 mg, 0.01 mmol, 11% yield) as a white solid was prepared in a fashion similar to that described for Example 101, here using lOld (29 mg, 0.10 mmol) and 108a (36 mg, 0.14 mmol) as starting materials. LC/MS (ESI + ) m/z = 485.1 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 8.40 (d, J = 2.93 Hz, 1H) 7.80 (dd, J = 7.53, 2.05 Hz, 1H) 7.71 (s, 1H) 7.48 - 7.52 (m, 1H) 7.47 (d, J = 8.61 Hz, 1H) 7.34 (dd, J = 8.71, 3.03 Hz, lH) 7.18 (s, 1H) 7.06 (dd, J = 11.74, 8.41 Hz, 1H) 5.23 (s, 2H) 4.60 - 4.93 (m, 2H) 4.28 - 4.55 (m, 2H) 1.90 (t, J = 8.02 Hz, 1H) 1.23 (dd, J = 9.59, 6.06 Hz, 1H) 0.79 (td, J = 6.36, 3.72 Hz, 1H). N¾ peak was not observed.

Example 109: 2-((3-((lS,5S,6S)-3-amino-l,5-bis(fluoromethyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)thia zole-5-carbonitrile.

This compound (6 mg, 0.014 mmol, 17% yield) as an off-white solid was prepared in a fashion similar to that described for Example 101, here using lOld (26 mg, 0.084 mmol) and 2-bromo-5-cyanothiazole (Combi-Blocks, San Diego, California, USA) (24 mg, 0.126 mmol) as starting materials. LC/MS (ESI + ) m/z = 419.0 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 8.27 (s, 1H) 7.89 (dd, J = 7 A3, 1.96 Hz, 1H) 7.57 (ddd, J = 8.36, 4.65, 2.05 Hz, 1H) 7.13 (dd, J = 11.64, 8.51 Hz, 1H) 4.63 - 4.92 (m, 2H) 4.30 - 4.54 (m, 2H) 1.92 (t, J = 8.12 Hz, 1H) 1.24 (dd, J = 9.59, 6.06 Hz, 1H) 0.81 (td, J = 6.31, 3.81 Hz, 1H). N¾ peak was not observed.

Example 110: 6-((3-((lS,5S,6R)-3-amino-7,7-difluoro-l-(hydroxymethyl)-5-m ethyl-2- thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethyn yl)nicotinonitrile.

110c 110

Preparation of ((lS,5S,6R)-3-amino-7,7-difluoro-5-(2-fluoro-5- ((trimethylsilyl)ethynyl)phenyl)-5-methyl-2-thia-4-azabicycl o[4.1.0]hept-3-en-l- yl)methano (110b).

A sealable vial was charged with dichlorobis(triphenylphosphine)palladium(II) (0.07 g, 0.10 mmol), copper(I) iodide (0.02 g, 0.10 mmol), and ((lS,5S,6R)-3-amino-5-(5-bromo- 2-fluorophenyl)-7,7-difluoro-5-methyl-2-thia-4-azabicyclo[4. 1.0]hept-3-en-l-yl)methanol (110a, prepared according to the procedures reported in WO 2016022724) (0.26 g, 0.68 mmol). THF (2.7 mL) was added followed by (trimethylsilyl)-acetylene (0.94 mL, 6.63 mmol) and triethylamine (2.17 mL, 15.57 mmol). The reaction mixture was degassed by bubbling nitrogen through the solution for 5 minutes. The vial was sealed and the reaction was heated to 60 °C for 22 hours. Additional dichlorobis(triphenylphosphine)palladium (II) (0.07 g, 0.10 mmol), copper(I) iodide (0.02 g, 0.10 mmol) and (trimethylsilyl)-acetylene

(0.94 mL, 6.63 mmol) were added. Nitrogen was bubbled through the reaction mixture for 5 minutes then the reaction was heated at 60 °C for 16 hours. The reaction mixture was transferred to a separatory funnel containing water and EtOAc. The phases were mixed and the organic layer was separated, washed sequentially with sat'd aqueous NH4CI and brine, then dried over magnesium sulfate and concentrated under reduced pressure. The crude residue was purified via silica gel flash chromatography (10 - 90% EtOAc in hexanes) to afford ((lS,5S,6R)-3-amino-7,7-difluoro-5-(2-fluoro-5-((trimethylsi lyl)ethynyl)phenyl)-5- methyl-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)methanol (110b, 0.17 g, 0.43 mmol, 63% yield). LC/MS (ESI + ) m/z = 399.0/401.0 [M+H] + . ¾ NMR (300 MHz, CHLOROFORM-d) δ ppm 0.26 (s, 9 H) 1.65 (s, 3H) 2.43 - 2.55 (m, 1H) 3.88 - 4.03 (m, 2H) 7.01 (dd, J = 12.06, 8.40 Hz, 1H) 7.38 (ddd, J = 8.40, 4.75, 2.19 Hz, 1H) 7.80 (dd, J = 7.82, 2.12 Hz, 1H). NH 2 and OH peaks were not observed.

Preparation of ((lS,5S,6R)-3-amino-5-(5-ethynyl-2-fluorophenyl)-7,7-difluor o-5-methyl- 2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)methanol (110c).

To a solution of ((lS,5S,6R)-3-amino-7,7-difluoro-5-(2-fluoro-5- ((trimethylsilyl)ethynyl)phenyl)-5-methyl-2-thia-4-azabicycl o[4.1.0]hept-3-en-l-yl)methanol (110b, 0.17 g, 0.43 mmol) in methanol (2.2 mL) was added potassium carbonate (0.15 g, 1.07 mmol). The reaction was heated at 50 °C for 15 minutes. It was cooled to room temperature and filtered. The filtrate was concentrated under reduced pressure to afford 110c which was used in the next reaction without further purification (assuming the theoretical yield). LC/MS (EST) m/z = 327.0 [M+H] + .

Preparation of 6-((3-((lS,5S,6R)-3-amino-7,7-difluoro-l-(hydroxymethyl)-5-m ethyl-2- thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethyn yl)nicotinonitrile (110).

A sealable vial was charged with ((lS,5S,6R)-3-amino-5-(5-ethynyl-2-fluorophenyl)- 7,7-difluoro-5-methyl-2-thia-4-azabicyclo[4.1.0]hept-3-en-l- yl)methanol (110c, 0.14 g, 0.43 mmol), bis(triphenylphosphine)palladium(II) dichloride (0.045 g, 0.06 mmol), 2-bromo-5- cyanopyridine (Sigma- Aldrich, St. Louis, MO, USA) (0.13 g, 0.69 mmol), and copper(I) iodide (0.01 g, 0.06 mmol). The vial was sealed and evacuated / backfilled with nitrogen three times. THF (2.1 mL) was added followed by triethylamine (0.12 mL, 0.86 mmol). The reaction was heated at 60 °C for 3 hours, then cooled to room temperature and filtered through a pad of celite. The cake was washed with EtOAc and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel flash chromatography (10 - 90% (EtOAc containing 5% MeOH) in hexanes). The product obtained was purified again by silica gel flash chromatography (50 - 100% (EtOAc with 5% MeOH) in hexanes) to afford 6-((3-((lS,5S,6R)-3-amino-7,7-difluoro-l-(hydroxymethyl)-5-m ethyl-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile (110) (26 mg, 0.06 mmol, 14% yield). LC/MS (ESI + ) m/z = 429.0 [M+H] + . 'H NMR (300 MHz,

CHLOROFORM-d) δ ppm 1.71 (s, 3H) 2.45 - 2.58 (m, 1H) 3.88 - 4.04 (m, 2H) 7.12 (dd, J = 11.77, 8.40 Hz, 1H) 7.55 (ddd, J = 8.44, 4.64, 2.27 Hz, 1H) 7.64 (dd, J = 8.18, 0.73 Hz, 1H) 7.89 - 8.03 (m, 1H) 7.94 - 8.02 (m, 1H) 8.89 (dd, J = 2.19, 0.88 Hz, 1H). NH 2 and OH peaks were not observed.

Example 111: 6-((3-((lR,5S,6S)-3-amino-5-(fluoromethyl)-l-((E)-3-morpholi no-3- oxoprop-l-en-l-yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl) -4- fl

Preparation of (E)-ethyl 3-((lR,5S,6S)-3-aniino-5-(5-bromo-2-fluorophenyl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)acry late (111b).

To (E)-ethyl 3-((lR,5S,6S)-5-(5-bromo-2-fluorophenyl)-3-((tert-butoxycarb onyl)((2- (trimethylsilyl)ethoxy)methyl)amino)-5 -(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en- l-yl)acrylate (111a, prepared according to the procedures reported in WO 2016022724) (3.62 g, 5.47 mmol) at 0 °C was added a mixture of TFA/H2SO4 (9: 1, 30 inL) dropwise. After the addition, the mixture was stirred at 0 °C for 30 minutes, and then concentrated in vacuo. The residue was cooled with an ice bath, then basified with sat'd aqueous Na2C03 until pH >9, and extracted with DCM (3 x). The organic extracts were dried over Na2S04 and

concentrated to give (E)-ethyl 3-((lR,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)acry late (111b, 2.36 g, 100% yield) which was used as crude. LC/MS (ESI + ) in : = 433.0/435.0 [M+H] + .

Preparation of (E)-3-((lR,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5-(fluo romethyl)- 2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)acrylic acid (111c).

To a stirred solution of 111b (2.3 g, 5.3 mmol) in EtOH (30 mL) was added lithium hydroxide (26.7 mL of 1 N aqueous solution, 26.7 mmol). The reaction mixture was stirred at room temperature overnight, then concentrated, and acidified with 1 N aqueous HCl until pH = 4. The heterogeneous mixture was filtered and the cake was washed with water. The tan solid was collected and dried to give (E)-3-((lR,5S,6S)-3-amino-5-(5-bromo-2- fluorophenyl)-5-(fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hep t-3-en-l-yl)acrylic acid (111c, 2.3 g, 93% yield) which was used as crude. LC/MS (ESI + ) m/z = 403.0/405.0 [M+H] + . Preparation of (E)-3-((lR,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5-(fluo romethyl)- 2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)-l-morpholinoprop-2 -en-l-one (llld).

To a stirred mixture of 111c (200 mg, 0.50 mmol), (88 μί, 0.50 mmol) and morpholine (95 μί, 1.09 mmol) in DCM (6 mL) was added HATU (226 mg, 0.60 mmol). The reaction was stirred for 2 hours, then treated with water and extracted with DCM (3 x). The organic extracts were dried over Na2S04, concentrated and purified by silica gel chromatography (65% EtOAc in hexanes) to give (E)-3-((lR,5S,6S)-3-amino-5-(5-bromo-2- fluorophenyl)-5 -(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en- 1 -yl)- 1 -morpholinoprop- 2-en-l-one (llld, 159 mg, 68% yield). LC/MS (ESI + ) m/z = 474.0/476.0 [M+H] + . 'H NMR (400 MHz, CDC1 3 ) δ: 7.78 (dd, J = 6.8, 2.5 Hz, 1H), 7.39 (ddd, J = 8.6, 4.3, 2.6 Hz, 1H), 6.96 (dd, J = 11.5, 8.6 Hz, 1H), 6.61 (d, J = 14.7 Hz, 1H), 6.40 (d, J = 14.9 Hz, 1H), 4.72- 4.88 (m, 1H), 4.55-4.72 (m, 1H), 3.70 (d, J = 2.9 Hz, 6H), 3.58 (br. s., 2H), 2.07 (ddd, J = 9.5, 7.5, 1.6 Hz, 1H), 1.37 (dd, J = 9.7, 5.8 Hz, 1H), 1.02-1.10 (m, 1H). NH 2 peak was not observed. Preparation of 6-((3-((lR,5S,6S)-3-amino-5-(fluoromethyl)-l-((E)-3-morpholi no-3- oxoprop-l-en-l-yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl) -4- fluorophenyl)ethynyl)nicotinonitrile (111).

A mixture of llld (130 mg, 0.28 mmol), 6-ethynylnicotinonitrile (Frontier

Scientific, Logan, UT, USA) (53 mg, 0.41 mmol), copper(I) iodide (13 mg, 0.07 mmol), triethylamine (115 μί, 0.83 mmol), and PdCh(t-Bu3P)2 (35 mg, 0.07 mmol) in dioxane (2 mL) was heated at 80 °C for 24 hours. The reaction mixture was cooled to RT, treated with sat'd aqueous NH4CI, and extracted with EtOAc (3 x). The extracts were dried over Na2S04, concentrated and purified by silica gel chromatography (0 - 50% of (EtOAc/EtOH = 3/1) in heptane) to give 6-((3-((lR,5S,6S)-3-amino-5-(fluoromethyl)-l-((E)-3-moφholi no-3- oxoprop- 1 -en- 1 -yl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-5 -yl)-4- fluorophenyl)ethynyl)nicotinonitrile (111, 7 mg, 5% yield). LC/MS (ESI+) m/z = 520.2 [M+H] + . ¾ NMR (DMSO-d6) δ: 9.05 (dd, J = 2.1, 0.9 Hz, 1H), 8.38 (dd, J = 8.2, 2.2 Hz, 1H), 7.95 (dd, J = 7.5, 2.2 Hz, 1H), 7.84 (dd, J = 8.2, 0.8 Hz, 1H), 7.67 (ddd, J = 8.4, 4.5, 2.2 Hz, 1H), 7.35 (dd, J = 11.8, 8.5 Hz, 1H), 6.54-6.63 (m, 3H), 6.42-6.51 (m, 1H), 4.57-4.80 (m, 2H), 3.56 (s, 4H), 3.49-3.55 (s, 4H), 1.98 (t, J = 8.4 Hz, 1H), 1.54 (dd, J = 9.6, 5.3 Hz, 1H), 0.87-0.94 (m, 1H).

Example 112: (E)-3-((lR,5S,6S)-3-amino-5-(5-((5-cyanopyridin-2-yl)ethynyl )-2- fluorophenyl)-5-(fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hep t-3-en-l-yl)-N,N- dimethylacrylamide.

Preparation of (E)-3-((lR,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5-(fluo romethyl)- 2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)-N,N-dimethylacryla mide (112a).

To a stirred mixture of (E)-3-((lR,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)acry lic acid (111c, 325 mg, 0.81 mmol) and dimethylamine (604 μί, 1.21 mmol), and /Pr2NEt (183 μί, 1.05 mmol) in DMF (6 mL) was added HATU (368 mg, 0.97 mmol). The mixture was stirred for 2 hours, then treated with water and extracted with DCM (3 x). The extracts were dried over Na2S04, concentrated and purified by silica gel chromatography (30% EtOAc/hexanes) to give (E)-3- ((lR,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5-(fluorometh yl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-l-yl)-N,N-dimethylacrylamide (112a, 250 mg, 72% yield).

LC/MS (ESI+) m/z = 431.0/433.0 [M+H] + . ¾ NMR (CDC1 3 ) δ: 7.75 (dd, J = 6.8, 2.5 Hz, 1H), 7.40 (ddd, J = 8.6, 4.2, 2.6 Hz, 1H), 6.96 (dd, J = 11.5, 8.6 Hz, 1H), 6.52-6.64 (m, 1H), 6.38-6.51 (m, 1H), 4.54-4.90 (m, 2H), 3.11 (s, 3H), 3.01 (s, 3H), 1.95-2.14 (m, 1H), 1.39 (dd, J = 9.7, 5.8 Hz, 1H), 0.96-1.12 (m, 1H). NH 2 peak was not observed. Preparation of (E)-3-((lR,5S,6S)-3-amino-5-(5-ethynyl-2-fluorophenyl)-5-

(fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)- N,N-dimethylacrylamide (112b).

A mixture of 112a (250 mg, 0.58 mmol), tributyl(ethynyl)stannane (275 mg, 0.87 mmol), and Pd(t-Bu3P)2 (30 mg, 0.06 mmol) in dioxane (5 mL) was heated at 80 °C for 1 h. It was cooled to RT, treated with sat'd aqueous KF and stirred for 15 minutes. The mixture was filtered through a pad of celite, washed with EtOAc, and the filtrate was transferred to a separatory funnel. The layers were separated. The organic layer was dried over Na2S04, concentrated and purified on a silica gel column (60% EtOAc/hexanes) to give (E)-3- ((lR,5S,6S)-3-amino-5-(5-ethynyl-2-fluorophenyl)-5-(fluorome thyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-l-yl)-N,N-dimethylacrylamide (112b, 153 mg, 70% yield).

LC/MS (ESI+) m/z = 376.1 [M+H] + . Preparation of (E)-3-((lR,5S,6S)-3-amino-5-(5-((5-cyanopyridin-2-yl)ethynyl )-2- fluorophenyl)-5-(fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hep t-3-en-l-yl)-N,N- dimethylacrylamide (112).

A mixture of 112b (150 mg, 0.40 mmol), 6-bromonicotinonitrile (110 mg, 0.60 mmol), copper(I) iodide (19 mg, 0.10 mmol), triethylamine (167 μί, 1.20 mmol), and bis(tri- tert-butylphosphine)palladium(O) (20 mg, 0.04 mmol) in dioxane (4 inL) was heated at 80 °C for 1 hour. The reaction mixture was cooled to RT, treated with sat'd aqueous NH4CI and stirred for 30 minutes. The mixture was extracted with EtOAc (3 x). The organic extracts were dried over Na2S04, concentrated and purified on a silica gel column (30 - 60% EtOAc in DCM) to give (E)-3-((lR,5S,6S)-3-amino-5-(5-((5-cyanopyridin-2-yl)ethynyl )-2- fluorophenyl)-5-(fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hep t-3-en- l-yl)-N,N- dimethylacrylamide (112, 65 mg, 34% yield). LC/MS (ESI+) m/z = 478.1 [M+H] + . 'H NMR (DMSO-d6) δ: 9.06 (d, J = 1.4 Hz, 1H), 8.39 (dd, J = 8.1, 2.1 Hz, 1H), 7.97 (dd, J = 7.4, 2.0 Hz, 1H), 7.85 (d, J = 8.0 Hz, 1H), 7.62-7.73 (m, 1H), 7.36 (dd, J = 11.9, 8.4 Hz, 1H), 6.59 (s, 2H), 6.49-6.55 (m, 1H), 6.41-6.48 (m, 1H), 4.76 (s, 1H), 4.64 (s, 1H), 3.05 (s, 3H), 2.88 (s, 3H), 1.99 (t, J = 8.1 Hz, 1H), 1.52 (dd, J = 9.7, 5.2 Hz, 1H), 0.91 (t, J = 6.2 Hz, 1H).

Example 113: 2-((lR,5S,6S)-3-amino-5-(5-((5-cyanopyridin-2-yl)ethynyl)-2-

113d 113 Preparation of 2-((lR,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5-(fluorome thyl)-2- thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)acetamide (113b).

A mixture of 2-((lR,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5-(fluorome thyl)- 2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)acetonitrile (106a, 498 mg, 1.34 mmol) in water (2 mL) and trifluoroacetic acid (5 mL) was heated at 70 °C for 10 hours. LCMS showed about 8% conversion. Heating was continued at 70 °C for additional 48 hours. The mixture was cooled to room temperature, then quenched with sat'd aqueous NaHCC until pH was about 8. The mixture was extracted with EtOAc (2 x 50 mL). The combined organic extracts were dried over MgS04 and concentrated in vacuo. The residue was purified by silica gel chromatography (0 to 100% EtOAc in heptane) to give 113b (206 mg, 0.53 mmol, 39% yield) as a colorless solid. LC/MS (ESf) m/z = 389.9/391.9 [M+H] + .

Preparation of 2-((lR,5S,6S)-3-amino-5-(2-fluoro-5-((trimethylsilyl)ethynyl )phenyl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)acet amide (113c).

To a solution of 2-((lR,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)acet amide (113b, 206 mg, 0.53 mmol) in THF (1.3 mL) was added dichlorobis(triphenyl-phosphine)palladium (II) (74.1 mg, 0.106 mmol), copper(I) iodide (20 mg, 0.11 mmol), triethylamine (0.22 mL, 1.58 mmol) and (trimethylsilyl) -acetylene (1.49 mL, 10.56 mmol). The mixture was stirred at 60 °C overnight, then cooled to room temperature and quenched with sat'd aqueous NH4CI (40 mL). The mixture was extracted with EtOAc (2 x 60 mL). The organic extracts were dried over MgS04 and concentrated in vacuo. The residue was purified by silica gel

chromatography (0 to 100% EtOAc in heptane) to give 2-((lR,5S,6S)-3-amino-5-(2-fluoro-5- ((trimethylsilyl)ethynyl)phenyl)-5 -(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en- 1 - yl)acetamide (113c, 190 mg, 0.46 mmol, 88% yield) as a light yellow solid. LC/MS (ESI + ) m/z = 408.0 [M+H] + .

Preparation of 2-((lR,5S,6S)-3-amino-5-(5-ethynyl-2-fluorophenyl)-5-(fluoro methyl)-2- thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)acetamide (113d).

To a solution of 2-((lR,5S,6S)-3-amino-5-(2-fluoro-5- ((trimethylsilyl)ethynyl)phenyl)-5 -(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en- 1 - yl)acetamide (113c, 190 mg, 0.46 mmol) in methanol (2 mL) was added potassium carbonate (161 mg, 1.16 mmol). The resulting mixture was stirred at 50 °C for 15 minutes, then cooled to room temperature and partitioned between EtOAc (30 mL) and sat'd aqueous NaHCC . The organic layer was dried over MgSO/t, and concentrated in vacuo to give 2-((lR,5S,6S)-3- amino-5-(5-ethynyl-2-fluorophenyl)-5-(fluoromethyl)-2-thia-4 -azabicyclo[4.1.0]hept-3-en- 1- yl)acetamide (113d, 156 mg, 0.46 mmol) as a light brown oil which was used as crude.

LC/MS (ESI + ) m/z = 336.0 [M+H] + .

Preparation of 2-((lR,5S,6S)-3-amino-5-(5-((5-cyanopyridin-2-yl)ethynyl)-2- fluorophenyl)-5-(fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hep t-3-en-l-yl)acetamide

(113).

To a solution of 113d (156 mg, 0.46 mmol) in THF (2.5 mL) was added 2-bromo-5- cyanopyridine (128 mg, 0.70 mmol), bis(triphenyl-phosphine)palladium (II) (65 mg, 0.09 mmol), copper(I) iodide (18 mg, 0.09 mmol), triethylamine (0.13 mL, 0.93 mmol). The resulting mixture was stirred at 60 °C under a N2 atmosphere overnight. After cooling to RT, the reaction was quenched with sat'd aqueous NH 4 CI (40 mL) and extracted with EtOAc (2 x 100 mL). The combined organic extracts were dried over MgSC>4 and concentrated in vacuo. The residue was purified by silica gel chromatography (0 to 100% EtOAc in heptane) to give 2-(( 1 R,5 S,6S)-3 -amino-5 -(5 -((5 -cyanopyridin-2-yl)ethynyl)-2-fluorophenyl)-5 - (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)acet amide (Example 113, 39 mg, 0.09 mmol, 19% yield) as a yellow solid. LC/MS (ESI + ) m/z = 438.0 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ 8.86 (d, J = 1.56 Hz, 1H), 7.94 (dd, J = 2.05, 8.12 Hz, 1H), 7.83 (br d, J = 6.65 Hz, 1H), 7.62 (d, J = 8.22 Hz, 1H), 7.56 (ddd, J = 2.05, 4.65, 8.36 Hz, 1H), 7.11 (dd, J = 8.51, 11.64 Hz, 1H), 5.50-6.27 (m, 2H), 4.89-5.07 (m, 1H), 4.60-4.79 (m, 1H), 2.74 (br d, J = 16.63 Hz, 1H), 2.47 (br d, J = 16.43 Hz, 1H), 1.98 (br t, J = 7.82 Hz, 1H), 1.09 (br dd, J = 5.97, 9.10 Hz, 1H), 0.72 (t, J = 6.16 Hz, 1H). NH 2 peak was not observed.

Example 114: 6-((5-((lS,5S,6S)-3-amino-l-(fluoromethyl)-5-methyl-2-thia-4 - azabicy -3-en-5-yl)-6-fluoropyridin-3-yl)ethynyl)nicotinonitrile.

114a 114b

To a vial containing tert-butyl ((lS,5S,6S)-5-(5-bromo-2-fluoropyridin-3-yl)-l- (fluoromethyl)-5-methyl-2-thia-4-azabicyclo[4.1.0]hept-3-en- 3-yl)((2-

(trimethylsilyl)ethoxy)methyl)carbamate (114a, prepared according to the procedures reported in WO 2016022724) (0.20 g, 0.35 mmol) at room temperature was added cone, sulfuric acid (0.37 mL, 6.91 mmol). The mixture was stirred at room temperature for 6 min, and then poured into ice water (10 mL). The pH was adjusted to 11 by the dropwise addition of NaOH (10 N aqueous solution). The solution was extracted with EtO Ac (2 x). The combined organic extract was concentrated and the residue was purified by silica gel chromatography (0 - 60% EtOAc in heptane) to give (lS,5S,6S)-5-(5-bromo-2-fluoropyridin- 3-yl)-l-(fluoromethyl)-5-methyl-2-thia-4-azabicyclo[4.1.0]he pt-3-en-3-amine (114b, 102 mg, 0.29 mmol, 85% yield). LC/MS (ESI + ) m/z = 350.0/352.0 [M+l+H] + .

A mixture of (lS,5S,6S)-5-(5-bromo-2-fluoropyridin-3-yl)-l-(fluoromethyl) -5- methyl-2-thia-4-azabicyclo[4.1.0]hept-3-en-3-amine (114b, 90 mg, 0.26 mmol), 6- ethynylnicotinonitrile (Combi-Blocks, Inc., San Diego, CA, USA) (66 mg, 0.58 mmol), bis(tri-tert-butylphosphine)palladium (0) (33 mg, 0.065 mmol), copper(I) iodide (12 mg, 0.065 mmol), and triethylamine (1.0 mL, 7.19 mmol) in 1,4-dioxane (2 mL) contained in a vial was flushed with argon, capped, and heated at 80 °C for 1 h. The reaction mixture was cooled to room temperature, concentrated, and the residue was purified on a silica gel column (0 - 80% EtOAc in heptane) to give 6-((5-((lS,5S,6S)-3-amino-l-(fluoromethyl)-5-methyl-2- thia-4-azabicyclo[4.1 ]hept-3-en-5-yl)-6-fluoropyridin-3-yl)ethynyl)nicotinonitril e

(Example 114, 45 mg, 0.11 mmol, 44% yield). LC/MS (ESI + ) m/z = 396.2 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ 8.89 (d, J = 1.37 Hz, 1H), 8.34-8.41 (m, 2H), 7.98 (dd, J = 2.15, 8.02 Hz, 1H), 7.64 (d, J = 7.82 Hz, 1H), 4.21-4.62 (m, 4H), 1.86-1.97 (m, 1H), 1.68-1.73 (m, 3H), 0.98 (dd, J = 6.06, 9.39 Hz, 1H), 0.81 (dt, J = 4.21, 6.41 Hz, 1H). 19 F NMR (376 MHz, CHLOROFORM-d) δ -60.78 (s, IF), -212.97 (s, IF).

Example 115: 6-((3-((lS,5S,6S)-3-amino-5-(fluoromethyl)-l-((4-methyl-lH-l ,2,3-triazol- l-yl)methyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4- fluorophenyl)ethynyl)nicotinonitrile.

SEIVL . Boc

Preparation of ((lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-3-((tert-butoxycarbon yl)((2- (trimethylsilyl)ethoxy)methyl)amino)-5-(fluoromethyl)-2-thia -4-azabicyclo[4.1.0]hept-3- en-l-yl)methyl 4-methylbenzenesulfonate (115b).

To a solution of tert-butyl ((lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-5-(fluoromethyl)- l-(hydroxymethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-3-yl)( (2-

(trimethylsilyl)ethoxy)methyl)carbamate (115a, prepared according to the procedures reported in WO 2016022724) (9.73 g, 16.39 mmol) in DCM (25 mL) under a nitrogen atmosphere at room temperature was added 4-methylbenzenesulfonyl chloride (4.69 g, 24.59 mmol) and triethylamine (3.42 mL, 24.59 mmol). The resulting mixture was stirred at room temperature overnight. It was quenched with sat'd aqueous NaHCC (40 mL) and extracted with EtOAc (2 x 100 mL). The organic solution was dried over MgSC>4 and concentrated in vacuo. The residue was purified by silica gel chromatography (0 to 50% EtOAc in heptane) to give ((lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-3-((tert-butoxycarbon yl)((2- (trimethylsilyl)ethoxy)methyl)amino)-5 -(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en- l-yl)methyl 4-methylbenzenesulfonate (115b, 9.60 g, 12.84 mmol, 78% yield) as a colorless oil. LC/MS (ESI + ) m/z = 747.0/749.0 [M+H] + .

Preparation of tert-butyl ((lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-5-(fluoromethyl)-l- ((4-methyl-lH-l,2,3-triazol-l-yl)methyl)-2-thia-4-azabicyclo [4.1.0]hept-3-en-3-yl)((2- (trimethylsilyl)ethoxy)methyl)carbamate (115c).

A solution of 115b (192 mg, 0.257 mmol) in DMSO (1.5 mL) was treated with sodium azide (20 mg, 0.31 mmol) and the resulting mixture was stirred at room temperature overnight. Then, (+)-sodium L-ascorbate (10 mg, 0.05 mmol), CuSO/t-pentahydrate (64 mg, 0.26 mmol), and l-(trimethylsilyl)-l-propyne (57 mg, 0.51 mmol) were added. The resulting mixture was stirred at room temperature for 4 hours then at 55 °C overnight. After cooling to room temperature, the reaction was quenched with a mixture of sat'd aqueous NH4CI (10 mL) and sat'd aqueous NH4OH (1 mL), then extracted with EtOAc (2 x 20 mL). The organic solution was dried over MgS04 and concentrated in vacuo. The residue was purified by silica gel chromatography (0 to 100% EtOAc in heptane) to give tert-butyl ((lS,5S,6S)-5-(5- bromo-2-fluorophenyl)-5 -(fluoromethyl)- 1 -((4-methyl- 1H- 1 ,2,3 -triazol- 1 -yl)methyl)-2-thia- 4-azabicyclo[4.1.0]hept-3-en-3-yl)((2-(trimethylsilyl)ethoxy )methyl)carbamate (115c, 168 mg, 0.25 mmol, 99% yield) as a colorless solid. LC/MS (ESI + ) m/z = 658.1/660.0 [M+H] + . ¾ NMR (400 MHz, DMSO-d 6 ) δ 7.92 (s, 1H), 7.77 (dd, J = 2.54, 6.85 Hz, 1H), 7.56-7.71 (m, 1H), 7.34 (dd, J = 8.80, 11.93 Hz, 1H), 4.89-5.25 (m, 2H), 4.39-4.86 (m, 4H), 3.61 (dt, J = 2.35, 8.12 Hz, 2H), 2.32-2.40 (m, 1H), 2.30 (s, 3H), 1.47 (s, 9H), 1.40 (dd, J = 5.67, 9.59 Hz, 1H), 0.89-0.93 (m, 2H), 0.75 (t, J = 6.16 Hz, 1H), 0.00 (s, 9H). Preparation of (lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-5-(fluoromethyl)-l-((4 -methyl- lH-l,2,3-triazol-l-yl)methyl)-2-thia-4-azabicyclo[4.1.0]hept -3-en-3-amine (115d).

To a round bottom flask containing tert-butyl ((lS,5S,6S)-5-(5-bromo-2- fluorophenyl)-5 -(fiuoromethyl)- 1 -((4-methyl- 1H- 1 ,2,3 -triazol- 1 -yl)methyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-3-yl)((2-(trimethylsilyl)ethoxy)m ethyl)carbamate (115c, 900 mg, 1.36 mmol) at 0 °C was added cone, sulfuric acid (2.19 mL, 41.00 mmol) dropwise. After the addition, the mixture was stirred at room temperature for 12 minutes, then cooled with an ice bath and quenched with saturated aqueous NaOH solution dropwise until pH >10. The mixture was diluted with EtOAc (200 mL) and stirred for 15 minutes. The layers were separated and the aqueous layer was extracted with EtOAc (200 mL). The combined organic extracts were dried over MgSC>4 and concentrated in vacuo. The residue was purified by silica gel chromatography (0 to 100% EtOAc in heptane) to give (lS,5S,6S)-5-(5-bromo-2- fluorophenyl)-5 -(fiuoromethyl)- 1 -((4-methyl- 1H- 1 ,2,3 -triazol- 1 -yl)methyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-3-amine (115d, 437 mg, 1.02 mmol, 75% yield) as a white solid. LC/MS (ESI + ) m/z = 428.0/430.0 [M+H] + . Preparation of (lS,5S,6S)-5-(2-fluoro-5-((trimethylsilyl)ethynyl)phenyl)-5- (fluoromethyl)-l-((4-methyl-lH-l,2,3-triazol-l-yl)methyl)-2- thia-4- azabicyclo [4.1.0] hept-3-en-3-amine (115e)

To a solution of 115d (435 mg, 1.02 mmol) in THF (2 mL) was added copper(I) iodide (39 mg, 0.20 mmol), bis(triphenyl-phosphine)palladium (II) (143 mg, 0.20 mmol), triethylamine (0.42 mL, 3.05 mmol), and (trimethylsilyl) -acetylene (2.87 mL, 20.31 mmol). The resulting mixture was stirred at 60 °C overnight. After cooling to room temperature, the mixture was quenched with sat'd aqueous NH4CI (20 mL) and extracted with EtOAc (2 x 100 mL). The combined organic extracts were dried over MgS04 and concentrated in vacuo. The residue was purified by silica gel chromatography (0 to 100% EtOAc in heptane) to give (lS,5S,6S)-5-(2-fluoro-5-((trimethylsilyl)ethynyl)phenyl)-5- (fluoromethyl)-l-((4-methyl-lH- l,2,3-triazol-l-yl)methyl)-2-thia-4-azabicyclo[4.1.0]hept-3- en-3-amine (115e, 338 mg, 0.76 mmol, 75% yield) as a light brown solid. LC/MS (ESf) m/z = 446.1 [M+H] + .

Preparation of 6-((3-((lS,5S,6S)-3-amino-5-(fluoromethyl)-l-((4-methyl-lH-l ,2,3- triazol-l-yl)methyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-y l)-4- fluorophenyl)ethynyl)nicotinonitrile (115).

A mixture of 115e (338 mg, 0.76 mmol) and potassium carbonate (262 mg, 1.90 mmol) in methanol (3.5 mL) was heated at 50 °C for 5 minutes. After cooling to RT, the mixture was partitioned between EtOAc (100 mL) and sat'd aqueous NaHCC (10 mL). The organic layer was dried over MgS04 and concentrated in vacuo to give (lS,5S,6S)-5-(5- ethynyl-2-fluorophenyl)-5 -(fluoromethyl)- 1 -((4-methyl- 1H- 1 ,2,3 -triazol- 1 -yl)methyl)-2-thia- 4-azabicyclo[4.1.0]hept-3-en-3-amine (115f) as a light brown oil which was used as crude. LC/MS (ESI + ) m/z = 374.0 [M+H] + .

To a solution of the above obtained crude 115f (283 mg, 0.76 mmol) in THF (3 mL) was added 2-bromo-5-cyanopyridine (208 mg, 1.14 mmol), copper(I) iodide (29 mg, 0.15 mmol), bis(triphenyl-phosphine)palladium (II) (106 mg, 0.15 mmol), and triethylamine (0.21 mL, 1.52 mmol). The resulting mixture was then stirred at 60 °C overnight. The mixture was cooled to room temperature, then quenched with sat'd aqueous NH4CI (20 mL), and extracted with EtOAc (2 x 40 mL). The combined organic extracts were dried over MgSC>4 and concentrated in vacuo. The residue was purified by silica gel chromatography (0 to 100% EtOAc in heptane) to give 6-((3-((lS,5S,6S)-3-amino-5-(fluoromethyl)-l-((4-methyl-lH- 1 ,2,3 -triazol- 1 -yl)methyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-5 -yl)-4- fluorophenyl)ethynyl)nicotinonitrile (Example 115, 120 mg, 0.25 mmol, 33% yield) as a light yellow solid. LC/MS (ESI + ) m/z = 476 A [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ 8.86 (br s, 1H), 7.89-7.99 (m, 1H), 7.55-7.76 (m, 3H), 7.50 (br d, J = 14.67 Hz, 1H), 7.14 (dd, J = 8.41, 11.35 Hz, 1H), 4.38-4.93 (m, 4H), 2.37 (s, 3H), 2.22 (br s, 1H), 1.45 (br s, 1H), 0.87-0.94 (m, 1H). NH 2 peak was not observed. Example 116: 6-((3-((lR,5S,6S)-3-amino-5-(fluoromethyl)-l-((E)-3-morpholi no-3- oxoprop-l-en-l-yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl) -4-fluorophenyl)ethynyl)-5-

Preparation of (E)-3-((lR,5S,6S)-3-amino-5-(5-ethynyl-2-fluorophenyl)-5-

(fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)- l-morpholinoprop-2-en-l-one (116a).

1,4-Dioxane (4 mL) and ethynyltributylstannane (0.22 mL, 0.77 mmol) were added to a flask charged with (E)-3-((lR,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5- (fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3-en- 1 -yl)- 1 -morpholinoprop-2-en- 1 -one

(llld, 245 mg, 0.52 mmol) and bis(tri-t-butylphosphine)palladium(0) (26.5 mg, 0.052 mmol) under an argon atmosphere. The reaction mixture was heated to 80 °C and stirred for 45 minutes. The reaction mixture was cooled to room temperature, diluted with EtOAc and 1 M aqueous potassium fluoride, and stirred for 15 minutes. The biphasic mixture was filtered through celite. The organic layer was separated, washed with brine, dried over MgSO/i, filtered, and concentrated in vacuo. Chromatographic purification of the residue (silica gel, 0 to 100% EtOAc in heptane) gave (E)-3-((lR,5S,6S)-3-amino-5-(5-ethynyl-2-fluorophenyl)-5- (fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en- 1 -yl)- 1 -morpholinoprop-2-en- 1 -one (116a, 142 mg, 0.34 mmol, 65% yield) as a colorless oil. LC/MS (ESI + ) m/z = 418.1

[M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 7.75 (1 H, dd, J = 7.53, 2.05 Hz) 7.43 (1 H, ddd, J = 8.31, 4.69, 2.25 Hz) 7.03 (1 H, dd, J = 11.74, 8.41 Hz) 6.61 (1 H, d, J = 14.87 Hz) 6.42 (1 H, d, J = 14.67 Hz) 4.57 - 4.92 (2 H, m) 3.54 - 3.73 (8 H, m) 3.04 (1 H, s) 2.06 - 2.13 (1 H, m) 1.41 (1 H, dd, J = 9.68, 5.77 Hz) 1.07 (1 H, t, J = 6.55 Hz). NH 2 peak was not observed. Preparation of 6-((3-((lR,5S,6S)-3-amino-5-(fluoromethyl)-l-((E)-3-morpholi no-3- oxoprop-l-en-l-yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl) -4-fluorophenyl)ethynyl)-5- methylnicotinonitrile (116).

A mixture of 116a (46 mg, 0.11 mmol), 2-bromo-5-cyano-3-picoline (33 mg, 0.16 mmol), trans-dichlorobis(triphenylphosphine)palladium (II) (8 mg, 0.01 mmol), copper(I) iodide (4 mg, 0.02 mmol), and triethylamine (0.03 mL, 0.22 mmol) in 1 mL of THF under a nitrogen atmosphere was stirred at 60 °C for 2 hours. The reaction mixture was cooled to room temperature, diluted with EtOAc, and washed with sat'd aqueous NH 4 CI followed by brine. The organic solution was dried over MgSO/i, filtered, and concentrated in vacuo. Chromatographic purification of the residue (silica gel, 0 to 100% EtOAc in heptane) gave 6- ((3-((lR,5S,6S)-3-amino-5-(fluoromethyl)-l-((E)-3-moφholino -3-oxoprop-l-en-l-yl)-2-thia- 4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)-5 -methylnicotinonitrile (116, 26 mg, 0.05 mmol, 44% yield) as an off-white solid. LC/MS (ESI + ) m/z = 534.2 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 8.70 (d, J = 1.57 Hz, 1H) 7.95 (dd, J = 7 A3, 2.15 Hz, 1H) 7.81 (dd, J = 1.96, 0.78 Hz, 1H) 7.56 (ddd, J = 8.41, 4.60, 2.25 Hz, 1H) 7.11 (dd, J = 11.74, 8.41 Hz, lH) 6.62 (d, J = 14.87 Hz, lH) 6.42 (d, J = 14.87 Hz, 1H) 4.60 -

4.91 (m, 2H) 3.53 - 3.76 (m, 8 H) 2.10 (ddd, J = 9.39, 7.53, 1.47 Hz, 1H) 1.39 (dd, J = 9.68, 5.77 Hz, 1H) 1.06 - 1.11 (m, 1H). NH 2 peak was not observed.

Example 117: (E)-3-((lR,5S,6S)-3-amino-5-(5-((5-chloropyrimidin-2-yl)ethy nyl)-2- fluorophenyl)-5-(fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hep t-3-en-l-yl)-l- m

This compound (24 mg, 0.045 mmol, 41% yield) as an off-white solid was prepared in a fashion similar to that described for Example 116, here using 116a (46 mg, 0.110 mmol) and 2-bromo-5-chloropyrimidine (Oakwood Products Inc., Estill, SC, USA) (32 mg, 0.165 mmol) as starting materials. LC/MS (ESI + ) m/z = 530.0 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 8.71 (s, 2H) 8.00 (dd, J = 7 A3, 1.76 Hz, 1H) 7.60 (ddd, J = 8.22, 4.60, 2.05 Hz, 1H) 7.10 (dd, J = 11.74, 8.41 Hz, lH) 6.62 (d, J = 14.67 Hz, lH) 6.42 (d, J = 14.87 Hz, 1H) 4.57 - 4.92 (m, 2H) 3.53 - 3.76 (m, 8 H) 2.10 (t, J = 8.51 Hz, 1H) 1.39 (dd, J = 9.59, 5.87 Hz, 1H) 1.07 (t, J = 6.55 Hz, 1H). NH 2 peak was not observed.

Example 118: (E)-3-((lR,5S,6S)-3-amino-5-(2-fluoro-5-((3-methylisothiazol -5- yl)ethynyl)phenyl)-5-(fluoromethyl)-2-thia-4-azabicyclo[4.1. 0]hept-3-en-l-yl)-l-

This compound (26 mg, 0.051 mmol, 46% yield) as a light orange solid was prepared in a fashion similar to that described for Example 116, here using 116a (46 mg, 0.110 mmol) and 5 -bromo-3 -methyl -isothiazole ( Sigma- Aldrich, 29 mg, 0.16 mmol) as starting materials. LC/MS (ESf) m/z = 515.2 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 7.84 (dd, J = 7.43, 1.96 Hz, 1H) 7.46 (ddd, J = 8.22, 4.69, 2.15 Hz, 1H) 7.05 - 7.12 (m, 2H) 6.62 (d, J = 14.67 Hz, 1H) 6.41 (d, J = 14.87 Hz, 1H) 4.59 - 4.91 (m, 2H) 3.53 - 3.75 (m, 8 H) 2.50 (s, 3H) 2.09 (t, J = 8.41 Hz, 1H) 1.39 (dd, J = 9.78, 5.87 Hz, 1H) 1.08 (t, J = 6.55 Hz, 1H). N¾ peak was not observed.

Example 119: 6-((3-((lS,5S,6S)-3-amino-5-(fluoromethyl)-l-(3-morpholino-3 - oxopropyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-

Preparation of (E)-3-((lR,5S,6S)-3-amino-5-(5-ethynyl-2-fluorophenyl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)-l-m orpholinoprop-2-en-l-one (119a).

Lithium borohydride (2 M solution in THF, 0.52 mL, 1.02 mmol) was added to a stirred solution of (E)-3-((lR,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5-(fluo romethyl)- 2-thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)-l-moφholinoprop-2 -en-l-one (Hid, 245 mg, 0.52 mmol) in THF (5 mL) under a nitrogen atmosphere. The reaction mixture was stirred at room temperature for 72 hours. The reaction mixture was cooled to 0 °C and quenched with sat'd aqueous NH4CI. The mixture was extracted twice with DCM. The combined organic layers were dried over MgSO/i, filtered, and concentrated in vacuo. Chromatographic purification of the residue (silica gel, 0 to 100% EtOAc in heptane) gave (E)-3-((lR,5S,6S)- 3-amino-5-(5-ethynyl-2-fluorophenyl)-5-(fluoromethyl)-2-thia -4-azabicyclo[4.1.0]hept-3-en- l-yl)-l-rrK^holinoprop-2-en-l-one (119a, 122 mg, 0.26 mmol, 50% yield) as a white solid. LC/MS (ESI + ) m/z = 474.0/476.0 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 7.78 (1 H, dd, J = 6.85, 2.54 Hz) 7.37 (1 H, ddd, J = 8.61, 4.21, 2.64 Hz) 6.94 (1 H, dd, J = 11.54, 8.61 Hz) 4.49 - 4.92 (2 H, m) 3.57 - 3.73 (6 H, m) 3.45 - 3.52 (2 H, m) 2.46 - 2.64 (2 H, m) 2.14 (1 H, ddd, J = 15.01, 9.44, 5.67 Hz) 1.72 - 1.85 (2 H, m) 0.91 (1 H, dd, J = 9.59, 5.87 Hz) 0.62 (1 H, t, J = 6.16 Hz). NH 2 peak was not observed.

Preparation of 3-((lS,5S,6S)-3-amino-5-(5-ethynyl-2-fluorophenyl)-5-(fluoro methyl)-2- thia-4-azabicyclo[4.1.0]hept-3-en-l-yl)-l-morpholinopropan-l -one (119b).

1,4-Dioxane (2 mL) and ethynyltributylstannane (0.111 mL, 0.386 mmol) were added to a flask charged with 119a (122 mg, 0.257 mmol) and bis(tri-t- butylphosphine)palladium(O) (13 mg, 0.026 mmol) under an argon atmosphere. The reaction mixture was heated to 80 °C for 50 minutes. The reaction mixture was cooled to room temperature, diluted with EtOAc and 1 M aqueous potassium fluoride, and stirred for 15 minutes. The biphasic mixture was filtered through celite. The organic layer was separated, washed with brine, dried over MgSO/i, filtered, and concentrated in vacuo. Chromatographic purification of the residue (silica gel, 0 to 100% EtOAc in heptane) gave 3-((lS,5S,6S)-3- amino-5-(5-ethynyl-2-fluorophenyl)-5-(fluoromethyl)-2-thia-4 -azabicyclo[4.1.0]hept-3-en- 1- yl)-l-moφholinopropan-l-one (119b, 63 mg, 0.150 mmol, 58% yield) as a light brown solid. LC/MS (ESI + ) m/z = 420.1 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 7.78 (1 H, dd, J = 7.63, 2.15 Hz) 7.40 (1 H, ddd, J = 8.36, 4.65, 2.25 Hz) 7.01 (1 H, dd, J = 11.74, 8.41 Hz) 4.48 - 4.94 (2 H, m) 3.58 - 3.74 (6 H, m) 3.46 - 3.52 (2 H, m) 3.03 (1 H, s) 2.46 - 2.64 (2 H, m) 2.14 (1 H, ddd, J = 15.06, 9.59, 5.67 Hz) 1.71 - 1.84 (2 H, m) 0.91 (1 H, dd, J = 9.39, 5.48 Hz) 0.60 (1 H, t, J = 6.16 Hz). NH 2 peak was not observed.

Preparation of 6-((3-((lS,5S,6S)-3-amino-5-(fluoromethyl)-l-(3-morpholino-3 - oxopropyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4- fluorophenyl)ethynyl)nicotinonitrile (119).

3-((lS,5S,6S)-3-Amino-5-(5-ethynyl-2-fluorophenyl)-5-(fluoro methyl)-2-thia-4- azabicyclo[4.1.0]hept-3-en-l-yl)-l-moφholinopropan-l-one (119b, 31 mg, 0.074 mmol), 2- bromo-5-cyanopyridine (20.29 mg, 0.111 mmol), trans- dichlorobis(triphenylphosphine)palladium (II) (5 mg, 7.4 μπιοΐ), and copper(I) iodide (2 mg, 0.01 mmol) were mixed in a round bottom flask and placed under a nitrogen atmosphere. THF (1 mL), and triethylamine (0.02 mL, 0.15 mmol) were added, and the reaction mixture was stirred at 60 °C for 1 hour. The reaction mixture was cooled to room temperature and diluted with EtOAc. The mixture was washed with saturated aqueous NH4CI, washed with brine, dried over MgSO/t, filtered, and concentrated in vacuo. Chromatographic purification of the residue (silica gel, 0 to 100% EtOAc in heptane) gave 6-((3-((l S,5S,6S)-3-amino-5- (fluoromethyl)- 1 -(3 -morpholino-3 -oxopropyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en-5 -yl)-4- fluorophenyl)ethynyl)nicotinonitrile (119, 14 mg, 0.027 mmol, 36.3 % yield) as a light yellow solid. LC/MS (ESI + ) m/z = 522 A [M+H] + . 'H NMR (400 MHz, CHLOROFORM-d) δ ppm 8.86 (d, J = 1.56 Hz, 1H) 7.92 - 7.97 (m, 2H) 7.60 (d, J = 8.22 Hz, 1H) 7.54 (ddd, J = 8.36, 4.55, 2.15 Hz, 1H) 7.09 (dd, J = 1 1.74, 8.41 Hz, 1H) 4.52 - 4.95 (m, 2H) 3.58 - 3.73 (m, 6H) 3.46 - 3.52 (m, 2H) 2.48 - 2.64 (m, 2H) 2.14 (ddd, J = 14.96, 9.29, 5.87 Hz, 1H) 1.74 - 1.87 (m, 2H) 0.93 (dd, J = 9.59, 5.67 Hz, 1H) 0.63 (t, J = 6.16 Hz, 1H). NH 2 peak was not observed. Example 120: 6-((3-((lS,5S,6S)-3-amino-5-(fluoromethyl)-l-(3-morpholino-3 - oxopropyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluor ophenyl)ethynyl)-5-

This compound (20 mg, 0.037 mmol, 51% yield) as a yellow solid was prepared in a fashion similar to that described for Example 119, here using 119b (31 mg, 0.074 mmol) and 2-bromo-5-cyano-3-picoline (22 mg, 0.1 1 1 mmol) as starting materials. LC/MS (ESI + ) m/z = 536.2 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 8.70 (d, J = 1.56 Hz, 1H) 7.94 (dd, J = 7.43, 1.96 Hz, 1H) 7.81 (d, J = 1.17 Hz, 1H) 7.54 (ddd, J = 8.31, 4.60, 2.15 Hz, 1H) 7.09 (dd, J = 1 1.74, 8.41 Hz, 1H) 4.53 - 4.96 (m, 2H) 3.58 - 3.73 (m, 6H) 3.47 - 3.52 (m, 2H) 2.47 - 2.64 (m, 5H) 2.15 (ddd, J = 15.01, 9.24, 5.87 Hz, 1H) 1.75 - 1.88 (m, 2H) 0.94 (dd, J = 9.49, 5.77 Hz, 1H) 0.64 (t, J = 6.16 Hz, 1H). NH 2 peak was not observed. Example 121 : 2-((3-((lR,5S,6S)-3-amino-5-(fluoromethyl)-l-((E)-3-morpholi no-3- oxoprop-l-en-l-yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl) -4-

This compound (21 mg, 0.040 mmol, 34% yield) as a light orange solid was prepared in a fashion similar to that described for Example 116, here using 116a (50 mg, 0.120 mmol) and 2-chloro-pyrimidine-5-carbonitrile (Synthonix Inc., Wake Forest, NC, USA) (25 mg, 0.180 mmol) as starting materials. LC/MS (ESf) m/z = 521.2 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 8.99 (s, 2H) 8.05 (dd, J = 7 A3, 2.15 Hz, 1H) 7.64 (ddd, J = 8.46, 4.65, 2.15 Hz, 1H) 7.13 (dd, J = 11.74, 8.41 Hz, 1H) 6.62 (d, J = 14.87 Hz, 1H) 6.41 (d, J = 14.87 Hz, 1H) 4.59 - 4.90 (m, 2H) 3.54 - 3.74 (m, 8 H) 2.07 - 2.13 (m, 1H) 1.38 (dd, J = 9.68, 5.77 Hz, 1H) 1.05 - 1.10 (m, 1H). NH 2 peak was not observed.

Example 122: 4-((3-((lR,5S,6S)-3-amino-5-(fluoromethyl)-l-((E)-3-morpholi no-3- oxoprop-l-en-l-yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl) -4- fl

This compound (27 mg, 0.052 mmol, 44% yield) as an off-white solid was prepared in a fashion similar to that described for Example 116, here using 116a (50 mg, 0.120 mmol) and 4-bromobenzonitrile (33 mg, 0.180 mmol) as starting materials. LC/MS (ESI + ) m/z = 519.0 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 7.84 (dd, J = 7.63, 2.15 Hz, 1H) 7.63 (d, J = 8.41 Hz, 2H) 7.58 (d, J = 8.41 Hz, 2H) 7.48 (ddd, J = 8.41, 4.69, 2.15 Hz, 1H) 7.09 (dd, J = 11.74, 8.41 Hz, 1H) 6.62 (d, J = 14.87 Hz, 1H) 6.42 (d, J = 14.87 Hz, 1H) 4.60 - 4.92 (m, 2H) 3.54 - 3.73 (m, 8 H) 2.08 - 2.14 (m, 1H) 1.40 (dd, J = 9.68, 5.77 Hz, 1H) 1.07 - 1.11 (m, 1H). N¾ peak was not observed.

Example 123: (lS,5S,6S)-3-amino-5-(5-((5-cyano-3-methyl-2-pyridinyl)ethyn yl)-2-

Preparation of teri-butyl ((lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-l- (dimethylcarbamoyl)-5-methyl-2-thia-4-azabicyclo[4.1.0]hept- 3-en-3-yl)((2- (trimethylsilyl)ethoxy)methyl)carbamate (123b).

To a stirring solution of(lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-3-((fert- butoxycarbonyl)((2-(trimethylsilyl)ethoxy)methyl)amino)-5-me thyl-2-thia-4- azabicyclo[4.1.0]hept-3-ene-l-carboxylic acid (123a, prepared according to the procedures reported in WO 2016022724) (3.0 g, 5.1 mmol) in THF (20 mL) at 20 °C was added 1, 1'- carbonyldiimidazole (1.2 g, 7.6 mmol) in one portion. The solution was stirred for 1 hour at 20 °C. The solution was chilled to 0 °C and dimethylamine (2.0 M solution in THF, 12.7 mL, 25.4 mmol) was added. The solution was stirred for 1 hour, and then ethyl acetate (30 mL) and HCl (30 mL of 1 M aqueous solution) were added. The organic layer was washed with brine (25 mL), dried over magnesium sulfate, filtered and concentrated under reduced pressure to give fert-butyl ((lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-l-(dimethylcarbamoyl) - 5 -methyl-2-thia-4-azabicyclo [4.1.0]hept-3 -en-3 -yl)((2-

(trimethylsilyl)ethoxy)methyl)carbamate (123b, 3.1 g, 5.0 mmol, 98% yield) as a light yellow oil. LC/MS (ESI + ) m/z = 616/618 [M+H] + . 'H NMR (400 MHz, CHLOROFORM-d) δ ppm 7.74 (dd, J = 6.80, 2.54 Hz, 1H) 7.36 (ddd, J = 8.61, 4.11, 2.74 Hz, 1H) 6.96 (dd, J = 11.54, 8.61 Hz, 1H) 5.30 (d, J = 10.37 Hz, 1H) 5.03 (d, J = 10.37 Hz, 1H) 3.63 - 3.74 (m, 2H) 2.47 (ddd, J = 9.54, 7.48, 1.56 Hz, 1H) 3.01 (br s, 6H) 1.57 (br. s, 3H) 1.53 (s, 9 H) 1.19 - 1.25 (m, 1H) 0.96 - 1.04 (m, 3H) 0.02 (s, 9 H).

Preparation of (lS,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-N,N,5-trimethy l-2-thia- 4-azabicyclo[4.1.0]hept-3-ene-l-carboxamide (123c).

A mixture of fert-butyl ((lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-l- (dimethylcarbamoyl)-5-methyl-2-thia-4-azabicyclo[4.1.0]hept- 3-en-3-yl)((2- (trimethylsilyl)ethoxy)methyl)carbamate (123b, 3.0 g, 4.9 mmol) and cone, sulfuric acid (8.5 mL) was stirred at room temperature for 15 minutes. The mixture was slowly added to a mixture of DCM (200 mL) and ice (100 g). The pH was adjusted to about 7 by the portion- wise addition of potassium phosphate tribasic (34 g). The resulting biphasic mixture was separated and the aqueous layer was extracted with a mixture of DCM/MeOH (10: 1 v/v, 2 x). The combined extracts were dried over magnesium sulfate, filtered, and concentrated in vacuo to give an oil. The oil was purified by silica gel chromatography (30 to 100% ethyl acetate in DCM) to give (lS,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-N,N,5-trimethy l-2- thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxamide (123c, 1.7 g, 4.4 mmol, 91% yield) as a white solid. LC/MS (ESI + ) m/z = 386/388 [M+H] + . 'H NMR (400 MHz, CHLOROFORM- d) δ ppm 7.67 (dd, J = 6.94, 2.64 Hz, 1H) 7.28 - 7.37 (m, 1H) 6.91 (dd, J = 11.54, 8.61 Hz, lH) 3.06 (br. s., 6H) 2.30 (t, J = 8.41 Hz, 1H) 1.79 (s, 3H) 1.30 (dd, J = 9.68, 5.77 Hz, 1H) 0.87 (t, J = 6.36 Hz, 1H). NH 2 peak was not observed.

Preparation of (lS,5S,6S)-3-amino-5-(5-ethynyl-2-fluorophenyl)-N,N,5-trimet hyl-2-thia- 4-azabicyclo[4.1.0]hept-3-ene-l-carboxamide (123d).

1,4-Dioxane (10 mL) and ethynyltributylstannane (0.69 mL, 2.38 mmol) were added to a flask charged with (lS,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-N,N,5-trimethy l-2- thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxamide (123c, 612 mg, 1.58 mmol) and bis(tri-t- butylphosphine)palladium (0) (81 mg, 0.16 mmol) under an argon atmosphere. The reaction mixture was heated to 80 °C for 1 hour. The reaction mixture was cooled to room temperature, diluted with ethyl acetate and 1 M aqueous KF solution, and stirred for 15 minutes. The biphasic mixture was filtered through a pad of celite; the organic layer was separated, washed with brine, dried over magnesium sulfate, filtered, and concentrated in vacuo. The residue was purified via silica gel chromatography (0 to 100% ethyl acetate in heptane) to give (lS,5S,6S)-3-amino-5-(5-ethynyl-2-fluorophenyl)-N,N,5-trimet hyl-2-thia-4- azabicyclo[4.1.0]hept-3-ene-l-carboxamide (123d, 349 mg, 1.05 mmol, 67% yield) as a yellow solid. LC/MS (ESI + ) m/z = 332 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 7.67 (d, J = 7.63 Hz, 1H) 7.32 - 7.39 (m, 1H) 6.95 - 7.03 (m, 1H) 3.01 (m, 7 H) 2.28 (t, J = 8.31 Hz, 1H) 1.81 (s, 3H) 1.32 (dd, J = 9.59, 5.67 Hz, 1H) 0.85 (t, J = 6.36 Hz, 1H). N¾ peak was not observed. Preparation of Example 123.

A mixture of (lS,5S,6S)-3-amino-5-(5-ethynyl-2-fluorophenyl)-N,N,5-trimet hyl-2- thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxamide (123d, 90 mg, 0.27 mmol), 2-bromo-5- cyano-3-picoline (Matrix Scientific, Columbia, SC, USA) (80 mg, 0.41 mmol), bis(triphenyl- phosphine)palladium (II) dichloride (19 mg, 0.027 mmol), and copper(I) iodide (8 mg, 0.041 mmol) were mixed in a round bottom flask and placed under a nitrogen atmosphere. THF (2.5 mL) and triethylamine (0.08 mL, 0.54 mmol) were added, and the reaction mixture was stirred at 60 °C for 1 hour. The reaction mixture was cooled to room temperature and then concentrated in vacuo to give an oil. The oil was purified via silica gel chromatography (30 to 100% ethyl acetate in heptane) to give (lS,5S,6S)-3-amino-5-(5-((5-cyano-3- methylpyridin-2-yl)ethynyl)-2-fluorophenyl)-N,N,5-trimethyl- 2-thia-4-azabicyclo[4.1.0]hept- 3-ene-l-carboxamide (123) ( 74 mg, 0.17 mmol, 61% yield) as a yellow solid. LC/MS (ESI + ) m/z = 448 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM- ) δ ppm 8.70 (d, J = 131 Hz, 1H) 7.85 (d, J = 6.65 Hz, 1H) 7.80 (s, 1H) 7.49 (ddd, J = 8.36, 4.55, 2.15 Hz, 1H) 7.07 (dd, J = 11.54, 8.22 Hz, 1H) 3.07 (br. s., 6H) 2.56 (s, 3H) 2.32 (t, J = 8.02 Hz, 1H) 1.83 (s, 3H) 1.27 - 1.37 (m, 1H) 0.88 (t, J = 6.46 Hz, 1H). NH 2 peak was not observed. Example 124: 6-((3-((lR,5S,6S)-3-amino-5-(fluoromethyl)-l-((E)-3-morpholi no-3- oxoprop-l-en-l-yl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl) -4-fluorophenyl)ethynyl)-5- fluoronicotinonitrile.

This compound (14 mg, 0.026 mmol, 26% yield) as a white solid was prepared in a fashion similar to that described for Example 116, here using 116a (42 mg, 0.101 mmol) and 6-chloro-5-fluoronicotinonitrile (24 mg, 0.151 mmol) as starting materials. LC/MS (ESI + ) m/z = 538.1 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 8.66 (d, J = 2.54 Hz, 1H) 7.97 - 8.02 (m, 1H) 7.70 (dd, J = 7.14, 2.64 Hz, 1H) 7.58 - 7.65 (m, 1H) 7.1 1 (dd, J = 1 1.54, 8.61 Hz, lH) 6.62 (d, J = 14.87 Hz, lH) 6.42 (d, J = 14.87 Hz, 1H) 4.58 - 4.92 (m,

2H) 3.54 - 3.75 (m, 8 H) 2.10 (t, J = 8.31 Hz, 1H) 1.41 (dd, J = 9.59, 5.87 Hz, 1H) 1.08 (t, J = 6.36 Hz, 1H). N¾ peak was not observed.

Example 125: 6-((3-((lS,5S,6S)-l-((lH-imidazol-l-yl)methyl)-3-amino-5-(fl uoromethyl)- 2-thia-4-azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)eth ynyl)nicotinonitrile.

Preparation of (lS,5S,6S)-l-((lH-imidazol-l-yl)methyl)-5-(5-bromo-2-fluorop henyl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-3-amine (125a).

A solution of imidazole (68.3 mg, 1.00 mmol) in THF (1.5 inL) at -5 °C under nitrogen was treated with lithium bis(trimethylsilyl)amide (1.00 mL of 1.0 M solution in THF, 1.00 mmol) dropwise and the mixture stirred at -5 °C for 20 minutes. A solution of ((lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-3-((tert-butoxycarbon yl)((2- (trimethylsilyl)ethoxy)methyl)amino)-5 -(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -en- l-yl)methyl 4-methylbenzenesulfonate (115b, 500 mg, 0.67 mmol) in THF (1.5 mL) was added dropwise to the reaction mixture. After the addition, the mixture was stirred at room temperature overnight, then quenched with sat'd aqueous NH4CI (20 mL) and extracted with EtOAc (2 x 50 mL). The organic solution was dried over MgSC>4 and concentrated in vacuo. The residue was purified by silica gel chromatography (0 to 100% EtOAc in heptane) to give tert-butyl ((lS,5S,6S)-l-((lH-imidazol-l-yl)methyl)-5-(5-bromo-2-fluoro phenyl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-3-yl)((2- (trimethylsilyl)ethoxy)methyl)carbamate (430 mg, 0.67 mmol) as a colorless oil. LC/MS

(ESf) m/z = 643.1/645.0 [M+H] + 'H NMR (CHLOROFORM-d) δ: 7.74-7.83 (m, 2H), 7.41 (ddd, J = 8.7, 4.3, 2.7 Hz, 1H), 7.04-7.18 (m, 2H), 6.97 (dd, J = 11.6, 8.7 Hz, 1H), 5.28 (d, J = 10.4 Hz, 1H), 5.03 (d, J = 10.4 Hz, 1H), 4.45-4.86 (m, 2H), 4.08-4.25 (m, 2H), 3.64 (dd, J = 9.2, 7.4 Hz, 2H), 2.02-2.11 (m, 1H), 1.51 (s, 9H), 1.15 (dd, J = 9.8, 6.1 Hz, 1H), 0.96 (dd, J = 9.2, 7.4 Hz, 2H), 0.86 (t, J = 6.5 Hz, 1H), 0.00 (s, 9H).

To a round bottom flask containing tert-butyl ((lS,5S,6S)-l-((lH-imidazol-l- yl)methyl)-5 -(5-bromo-2-fluorophenyl)-5 -(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 - en-3-yl)((2-(trimethylsilyl)ethoxy)methyl)carbamate (430 mg, 0.67 mmol) at 0 °C was added cone, sulfuric acid (1 mL) dropwise. The mixture was stirred at room temperature for 15 minutes, then cooled with an ice bath and treated with sat'd aqueous NaOH dropwise until pH >10. The mixture was diluted with EtOAc (50 mL) and stirred for 30 minutes. The layers were separated and the aqueous layer was extracted with EtOAc (50 mL). The combined organic extracts were dried over MgS04 and concentrated in vacuo. The residue was purified by silica gel chromatograph (0 to 100% EtOAc in heptane, then 0 to 20% MeOH in EtOAc) to give (lS,5S,6S)-l-((lH-imidazol-l-yl)methyl)-5-(5-bromo-2-fluorop henyl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-3-amine (125a, 253 mg, 0.61 mmol, 92% yield) as a light yellow solid. LC/MS (ESf) m/z = 413.0/415.0 [M+H] + .

Preparation of (lS,5S,6S)-l-((lH-imidazol-l-yl)niethyl)-5-(2-fluoro-5- ((trimethylsilyl)ethynyl)phenyl)-5-(fluoromethyl)-2-thia-4-a zabicyclo [4.1.0] hept-3-en-3- amine (125b).

This compound (100 mg, 0.23 mmol, 38% yield) as a yellow solid was prepared in a manner similar to that described for compound 115e, here starting with 125a (253 mg, 0.61 mmol), bis(triphenyl-phosphine)palladium (II) (86 mg, 0.12 mmol), copper(I) iodide (23 mg, 0.12 mmol), triethylamine (0.25 mL, 1.83 mmol), and (trimethylsilyl)-acetylene (1.73 mL, 12.24 mmol). LC/MS (ESI + ) m z 43 1 . 1 [M+H] + .

Preparation of (lS,5S,6S)-l-((lH-imidazol-l-yl)methyl)-5-(5-ethynyl-2-fluor ophenyl)-5- (fluoromethyl)-2-thia-4-azabicyclo[4.1.0]hept-3-en-3-amine (125c).

This compound (83 mg, 0.23 mmol) as a yellow solid was prepared in a manner similar to that described for compound 115f, here starting with 125b (100 mg, 0.23 mmol) and potassium carbonate (64 mg, 0.46 mmol). LC/MS (ESI + ) m/z = 359.1 [M+H] + .

Preparation of 6-((3-((lS,5S,6S)-l-((lH-imidazol-l-yl)methyl)-3-amino-5- (f luoromethyl)-2-thia-4-azabicyclo [4.1.0] hept-3-en-5-yl)-4- fluorophenyl)ethynyl)nicotinonitrile (125).

This compound (30 mg, 28% yield) as a yellow solid was prepared in a manner similar to that described for Example 115, here starting with 125c (83 mg, 0.23 mmol), 2- bromo-5-cyanopyridine (63 mg, 0.34 mmol), copper(I) iodide (11 mg, 0.06 mmol), triethylamine (0.10 mL, 0.70 mmol), and trans-dichlorobis(triphenyl-phosphine)palladium (II) (41 mg, 0.06 mmol). LC/MS (ESI + ) m/z = 461.0 [M+H] + . ¾ NMR (CHLOROFORM-d) δ: 8.86 (d, J = 1.4 Hz, 1H), 7.94 (dd, J = 8.2, 2.2 Hz, 1H), 7.75 (dd, J = 7.4, 1.8 Hz, 1H), 7.50-7.64 (m, 3H), 7.10 (dd, J = 11.6, 8.5 Hz, 2H), 7.02 (br. s., 1H), 4.50-4.89 (m, 2H), 4.02- 4.21 (m, 2H), 1.98-2.05 (m, 1H), 1.28 (dd, J = 9.5, 6.2 Hz, 1H), 0.82 (t, J = 6.4 Hz, 1H). N¾ peak was not observed. Example 126: (lS,5S,6S)-3-amino-5-(5-((5-cyano-3-methylpyridin-2-yl)ethyn yl)-2-

Preparation of tert-butyl ((lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-l-carbamoyl-5- methyl-2-thia-4-azabicyclo[4.1.0]hept-3-en-3-yl)((2- (trimethylsilyl)ethoxy)methyl)carbamate (126a).

l,l'-Carbonyldiimidazole (0.44 g, 2.69 mmol) was added to a stirred solution of (lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-3-((fert-butoxycarbony l)((2- (trimethylsilyl)ethoxy)methyl)amino)-5 -methyl -2 -thia-4-azabicyclo [4.1.0]hept-3 -ene- 1 - carboxylic acid (123a, 1.06 g, 1.79 mmol) in THF (4 mL). The mixture was stirred at room temperature for 1 hour. The mixture was cooled to 0 °C before ammonia (0.5 M in 1,4- dioxane, 17.91 mL, 8.96 mmol) was added via syringe. The reaction mixture was warmed to room temperature and stirred for 16 hours. Additional Ι,Γ-carbonyldiimidazole (0.22 g, 1.35 mmol) and ammonia (0.5 M in 1,4-dioxane, 8.96 mL, 4.48 mmol) were added, and the reaction mixture was stirred for another 4 hours. The reaction mixture was partitioned between EtOAc and 1 M aqueous HC1. The organic layer was separated, washed with brine, dried over MgSO/t, filtered, and concentrated in vacuo. Chromatographic purification of the residue (silica gel, 0 to 50% EtOAc in heptane) gave tert-butyl ((lS,5S,6S)-5-(5-bromo-2- fluorophenyl)-l-carbamoyl-5-methyl-2-thia-4-azabicyclo[4.1.0 ]hept-3-en-3-yl)((2- (trimethylsilyl)ethoxy)methyl)carbamate (126a, 640 mg, 1.08 mmol, 61% yield) as a colorless oil. LC/MS (ESI + ) m/z = 588.0/590.0 [M+H] + . 'H NMR (400 MHz,

CHLOROFORM-d) δ ppm 7.60 (1 H, dd, J = 7.04, 2.54 Hz) 7.36 (1 H, ddd, J = 8.61, 4.11, 2.54 Hz) 6.97 (1 H, dd, J = 11.44, 8.71 Hz) 5.38 (1 H, d, J = 10.56 Hz) 5.09 (1 H, d, J =

10.37 Hz) 3.70 (2 H, td, J = 8.26, 1.86 Hz) 2.26 - 2.32 (1 H, m) 1.88 (1 H, dd, J = 9.68, 5.18 Hz) 1.81 (3 H, d, J = 0.98 Hz) 1.55 (9 H, s) 1.25 - 1.34 (1 H, m) 1.00 (2 H, ddd, J = 10.07, 6.65, 2.45 Hz) 0.03 (9 H, s).

Preparation of (lS,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5-methyl-2-thi a-4- azabicyclo[4.1.0]hept-3-ene-l-carboxamide (126b).

fert-Butyl ((lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-l-carbamoyl-5-methyl- 2-thia-4- azabicyclo[4.1.0]hept-3-en-3-yl)((2-(trimethylsilyl)ethoxy)m ethyl)carbamate (126a, 635 mg, 1.08 mmol) was stirred in sulfuric acid (2 mL, 37.5 mmol) at room temperature for 1 hour. The reaction mixture was diluted with DCM, and ice was added. Approximately 7 g of K3PO 4 was added, and the mixture was taken to pH 7-8 with 10 N aqueous NaOH. The organic layer was separated, and the aqueous layer was extracted once more with 9: 1 DCM/MeOH. The combined organic extracts were dried over MgSO/t, filtered, and concentrated in vacuo. The resulting crude product was slurried in DCM and filtered to give (lS,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5-methyl-2-thi a-4-azabicyclo[4.1.0]hept-3- ene-l-carboxamide (126b 205 mg, 0.57 mmol, 53% yield) as a white solid. LC/MS (ESI + ) m/z = 358.0/360.0 [M+H] + .

Preparation of (lS,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5-methyl-2-thi a-4- azabicyclo[4.1.0]hept-3-ene-l-carbonitrile (126c).

2,2,2-Trifluoroacetic anhydride (0.466 mL, 3.35 mmol) was added dropwise via syringe to a stirred mixture of (lS,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5-methyl-2- thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxamide (126b, 200 mg, 0.56 mmol) and N,N- diisopropylethylamine (1.46 mL, 8.37 mmol) in THF (4 mL) at -78 °C. The reaction mixture was stirred at -78 °C for 15 minutes. The reaction mixture was quenched with sat'd aqueous NH4CI and extracted with EtOAc. The organic layer was separated, dried over MgSO/t, filtered, and concentrated in vacuo to give crude N-((lS,5S,6S)-5-(5-bromo-2-fluorophenyl)- l-cyano-5-methyl-2-thia-4-azabicyclo[4.1.0]hept-3-en-3-yl)-2 ,2,2-trifluoroacetamide as an orange oil (292 mg) that was used directly without further purification. LC/MS (ESI + ) m/z = 436.0/438.0 [M+H] + .

The crude N-((lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-l-cyano-5-methyl-2- thia-4- azabicyclo[4.1.0]hept-3-en-3-yl)-2,2,2-trifluoroacetamide above was mixed with ammonia (2.0 M solution in methanol, 10 mL, 20 mmol) in a sealed vial. The reaction mixture was stirred at 50 °C for 15 hours. The reaction mixture was concentrated in vacuo.

Chromatographic purification of the residue (silica gel, 0 to 40% EtOAc in heptane) gave (lS,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5-methyl-2-thi a-4-azabicyclo[4.1.0]hept-3- ene-l-carbonitrile (126c, 76 mg, 0.22 mmol, 40% yield) as an opaque white oil. LC/MS

(ESI + ) m/z = 340.0/341.9 [M+H] + . 'H NMR (400 MHz, DMSO-d6) δ ppm 7.67 (1 H, dd, J = 7.04, 2.74 Hz) 7.48 - 7.53 (1 H, m) 7.21 (1 H, dd, J = 11.83, 8.71 Hz) 6.65 (2 H, s) 2.41 (1 H, dd, J = 9.39, 8.22 Hz) 1.82 (1 H, dd, J = 9.78, 6.06 Hz) 1.69 (3 H, s) 1.01 (1 H, t, J = 6.85 Hz). N¾ peak was not observed. Preparation of (lS,5S,6S)-3-amino-5-(5-((5-cyano-3-methylpyridin-2-yl)ethyn yl)-2- fluorophenyl)-5-methyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene- l-carbonitrile (126).

1,4-Dioxane (2 mL) and ethynyltributylstannane (0.094 mL, 0.326 mmol) were added to a flask charged with (lS,5S,6S)-3-amino-5-(5-bromo-2-fluorophenyl)-5-methyl-2- thia-4-azabicyclo[4.1.0]hept-3-ene-l-carbonitrile (126c, 74 mg, 0.22 mmol) and bis(tri-7- butylphosphine)palladium(O) (11 mg, 0.022 mmol) under an argon atmosphere. The reaction mixture was heated to 80 °C and stirred for 1 hour. The reaction mixture was cooled to room temperature, diluted with EtOAc and 1 M aqueous potassium fluoride, and stirred for 15 minutes. The biphasic mixture was filtered through celite. The organic layer was separated, washed with brine, dried over MgSO/t, filtered, and concentrated in vacuo to give crude 126d (64 mg) as a brown oil that was used directly without further purification. LC/MS (ESI + ) m/z = 286.1 [M+H] + .

The crude 126d (64 mg), copper(I) iodide (6 mg, 0.03 mmol), and trans- dichlorobis(triphenylphosphine)palladium (II) (15 mg, 0.02 mmol) were mixed in a round bottom flask and placed under a nitrogen atmosphere. THF (2 mL) and triethylamine (0.06 mL, 0.43 mmol) were added, and the reaction mixture was stirred at 60 °C for 1 hour. The reaction mixture was cooled to room temperature and diluted with EtOAc. The mixture was washed with saturated aqueous NH4CI, dried over MgSO/t, filtered, and concentrated in vacuo. Chromatographic purification of the residue (silica gel, 0 to 50% EtOAc in heptane) gave product of insufficient purity. The impure product was slurried in MeOH and filtered to give (lS,5S,6S)-3 -amino-5 -(5 -((5 -cyano-3 -methylpyridin-2-yl)ethynyl)-2-fluorophenyl)-5 - methyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carbonitrile (126, 10 mg, 0.025 mmol, 11% yield) as a tan solid. LC/MS (ESI + ) m/z = 402.0 [M+H] + . ¾ NMR (400 MHz,

CHLOROFORM-d) δ ppm 8.70 (s, 1H) 7.79 - 7.84 (m, 2H) 7.53 (m, J = 7.24, 3.72 Hz, 1H) 7.10 (dd, J = 11.54, 8.41 Hz, 1H) 2.56 (s, 3H) 2.47 (t, J = 8.71 Hz, 1H) 1.83 (s, 3H) 1.68 (dd, J = 9.68, 6.16 Hz, 1H) 1.14 (t, J = 7.04 Hz, 1H). NH 2 peak was not observed. Example 127: (lS,5S,6S)-3-amino-5-(5-((5-cyano-3-methylpyridin-2-yl)ethyn yl)-2- fluorophenyl)-5-(fluoromethyl)-N,N-dimethyl-2-thia-4-azabicy clo[4.1.0]hept-3-ene-l- carboxamide.

Preparation of teri-butyl ((lS,5S,6S)-5-(5-bromo-2-fluorophenyl)-l- (dimethylcarbamoyl)-5-(fluoromethyl)-2-thia-4-azabicyclo[4.1 .0]hept-3-en-3-yl)((2- (trimethylsilyl)ethoxy)methyl)carbamate (127b).

To a stirred mixture of (5S)-5-(5-bromo-2-fluorophenyl)-3-((fert-butoxycarbonyl)((2- (trimethylsilyl)ethoxy)methyl)amino)-5 -(fluoromethyl)-2-thia-4-azabicyclo [4.1.0]hept-3 -ene- 1-carboxylic acid (127a, prepared according to the procedures reported in WO 2016022724) (1.95 g, 3.21 mmol), dimethylamine (2.41 mL, 4.81 mmol), and iPr 2 NEt (0.84 mL, 4.81 mmol) in DMF (20 mL) was added HATU (1.46 g, 3.85 mmol). The mixture was stirred at room temperature for 1 hour, then treated with H2O and extracted with EtOAc (3 x). The extracts were dried over Na2S04 and concentrated. The residue was purified by silica gel chromatography (30% EtOAc in hexanes) to give fert-butyl ((lS,5S,6S)-5-(5-bromo-2- fluorophenyl)-l-(dimethylcarbamoyl)-5-(fluoromethyl)-2-thia- 4-azabicyclo[4.1.0]hept-3-en- 3-yl)((2-(trimethylsilyl)ethoxy)methyl)carbamate (127b, 0.97 g, 47% yield). LC/MS (ESI+) m/z = 634.0/636.0 [M+H] + . ¾ NMR (CHLOROFORM-d) δ 7.74 (dd, J = 6.8, 2.5 Hz, 1H), 7.40 (ddd, J = 8.7, 4.3, 2.5 Hz, 1H), 6.97 (dd, J = 11.4, 8.7 Hz, 1H), 5.34 (d, J = 10.4 Hz, 1H), 5.09 (d, J = 10.4 Hz, 1H), 4.91-5.02 (m, 1H), 4.79-4.90 (m, 1H), 3.68 (td, J = 8.4, 1.5 Hz, 2H), 3.14 (d, J = 20.0 Hz, 3H), 2.89-3.05 (m, 3H), 2.38 (ddd, J = 9.8, 7.3, 2.4 Hz, 1H), 1.52 (s, 9H), 1.42 (dd, J = 9.9, 5.6 Hz, 1H), 0.98 (dd, J = 9.2, 7.4 Hz, 2H), 0.87 (dd, J = 7.0, 5.9 Hz, 1H), -0.03 (s, 9H).

Preparation of (lS,5S,6S)-3-Amino-5-(5-bromo-2-fluorophenyl)-5-(fluoromethy l)-N,N- dimethyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxamide (127c).

(lS,5S,6S)-3-Amino-5-(5-bromo-2-fluorophenyl)-5-(fluoromethy l)-N,N-dimethyl-2- thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxamide (127c, 0.58 g, 96% yield) was prepared in a manner similar to that described for compound 123c, here starting with 127b (0.96 g, 1.51 mmol). LC/MS (ESI+) m/z = 404.0/406.0 [M+H] + .

Preparation of (lS,5S,6S)-3-Amino-5-(5-ethynyl-2-fluorophenyl)-5-(fluoromet hyl)-N,N- dimethyl-2-thia-4-azabicyclo [4.1.0] hept-3-ene- 1-carboxamide (127d).

(lS,5S,6S)-3-Amino-5-(5-ethynyl-2-fluorophenyl)-5-(fluoromet hyl)-N,N-dimethyl- 2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxamide (127d, 301 mg, 66% yield) was prepared in a manner similar to that described for compound 123d, here starting with 127c (530 mg, 1.31 mmol), tributyl(ethynyl)tin (37.8 μί, 0.13 mmol) and Pd(t-Bu 3 P) 2 (804 mg, 1.57 mmol). LC/MS (ESI+) m/z = 350.2 [M+H] + .

Preparation of (lS,5S,6S)-3-Amino-5-(5-((5-cyano-3-methylpyridin-2-yl)ethyn yl)-2- fluorophenyl)-5-(fluoromethyl)-N,N-dimethyl-2-thia-4-azabicy clo[4.1.0]hept-3-ene-l- carboxamide (127).

This compound (25 mg, 12% yield) was prepared in a manner similar to that described for compound 123d, here starting with 127d (150 mg, 0.43 mmol), 2-bromo-5- cyano-3-picoline (102 mg, 0.52 mmol), copper(I) iodide (25 mg, 0.13 mmol), triethylamine (0.18 mL, 1.29 mmol), and Pd(t-Bu 3 P) 2 (22 mg, 0.04 mmol). LC/MS (ESI+) m/z = 466.1 [M+H] + . 'H NMR (DMSO-d 6 ) δ: 8.88 (d, J = 1.6 Hz, 1H), 8.33 (d, J = 1.2 Hz, 1H), 7.75 (dd, J = 7.4, 2.3 Hz, 1H), 7.69 (ddd, J = 8.3, 4.6, 2.3 Hz, 1H), 7.37 (dd, J = 11.7, 8.4 Hz, 1H), 6.73 (s, 2H), 4.76-4.85 (m, 1H), 4.62-4.73 (m, 1H), 2.97-3.20 (m, 3H), 2.77-2.95 (m, 3H), 2.53 (s, 3H), 2.07 (t, J = 8.1 Hz, 1H), 1.50 (dd, J = 9.6, 5.5 Hz, 1H), 0.67 (t, J = 6.3 Hz, 1H).

Example 128: 6-((3-((lS,5S,6S)-3-amino-5-methyl-l-(pyrrolidine-l-carbonyl )-2-thia-4-

Preparation of (lS,5S,6S)-methyl 3-amino-5-(5-((5-cyano-3-methylpyridin-2- yl)ethynyl)-2-fluorophenyl)-5-methyl-2-thia-4-azabicyclo[4.1 .0]hept-3-ene-l-carboxylate (128c).

(lS,5S,6S)-Methyl 5-(5-bromo-2-fluorophenyl)-3-((fert-butoxycarbonyl)((2- (trimethylsilyl)ethoxy)methyl)amino)-5 -methyl -2 -thia-4-azabicyclo [4.1.0]hept-3 -ene- 1 - carboxylate (128a, prepared according to the procedures reported in WO 2016022724) (1.09 g, 1.81mmol) was stirred in sulfuric acid (3 mL, 56.3 mmol) at room temperature for 20 minutes. The reaction mixture was diluted with DCM, and ice was added. Approximately 11 g of K3PO4 was added, and the mixture was taken to approximately pH 7 with 10 N aqueous NaOH. The organic layer was separated, and the aqueous layer was extracted once more with DCM. The combined organic extracts were dried over MgSO/t, filtered, and concentrated in vacuo. Chromatographic purification of the residue (silica gel, 0 to 40% EtOAc in heptane) gave (lS,5S,6S)-methyl 3-amino-5-(5-bromo-2-fluorophenyl)-5-methyl- 2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxylate (596 mg, 1.60 mmol, 88% yield) as a white solid. LC/MS (ESI + ) m/z = 372.9/374.9 [M+H] + . 'H NMR (400 MHz,

CHLOROFORM-d) δ ppm 7.85 (1 H, dd, J = 7.04, 0.98 Hz) 7.31 - 7.37 (1 H, m) 6.94 (1 H, dd, J = 11.44, 8.70 Hz) 3.79 (3 H, s) 2.55 (1 H, t, J = 8.71 Hz) 1.69 (3 H, s) 1.51 (1 H, dd, J = 9.78, 5.28 Hz) 1.11 (l H, m). NH 2 peak was not observed.

1,4-Dioxane (14 mL) and ethynyltributylstannane (0.691 mL, 2.395 mmol) were added to a flask charged with (lS,5S,6S)-methyl 3-amino-5-(5-bromo-2-fluorophenyl)-5- methyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxylate (596 mg, 1.60 mmol) and bis(tri- t-butylphosphine)palladium(O) (82 mg, 0.16 mmol) under an argon atmosphere. The reaction mixture was heated to 80 °C and stirred for 2 hours. The reaction mixture was cooled to room temperature, diluted with EtOAc and 1 M aqueous potassium fluoride, and stirred for 15 minutes. The biphasic mixture was filtered through celite. The organic layer was separated, washed with brine, dried over MgSO/i, filtered, and concentrated in vacuo to give crude (lS,5S,6S)-methyl 3-amino-5-(5-ethynyl-2-fluorophenyl)-5-methyl-2-thia-4- azabicyclo[4.1.0]hept-3-ene-l-carboxylate (128b, 432 mg) as a brown oil that was used directly without further purification.

The crude 128b (432 mg), 2-bromo-5-cyano-3-picoline (267 mg, 1.36 mmol), copper(I) iodide (4 mg, 0.20 mmol), and trans-dichlorobis(triphenylphosphine)palladium (II) (95 mg, 0.14 mmol) were mixed in a round bottom flask and placed under a nitrogen atmosphere. THF (10 mL) and triethylamine (0.38 mL, 2.71 mmol) were added, and the reaction mixture was stirred at 60 °C for 1 hour. The reaction mixture was cooled to room temperature and diluted with EtOAc. The mixture was washed with saturated aqueous NH4CI, washed with brine, dried over MgSO/t, filtered, and concentrated in vacuo.

Chromatographic purification of the residue (silica gel, 0 to 50% EtOAc in heptane) gave

(lS,5S,6S)-methyl 3-amino-5-(5-((5-cyano-3-methylpyridin-2-yl)ethynyl)-2-fluor ophenyl)-5- methyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxylate (128c, 315 mg, 0.72 mmol, 53% yield) as a yellow solid. LC/MS (ESI + ) m/z = 435.0 [M+H] + . ¾ NMR (400 MHz,

CHLOROFORM-d) δ ppm 8.70 (1 H, s) 7.99 - 8.04 (1 H, m) 7.81 (1 H, s) 7.48 - 7.54 (1 H, m) 7.09 (l H, dd, J = 11.74, 8.41 Hz) 3.80 (3 H, s) 2.52 - 2.59 (4 H, m) 1.73 (3 H, s) 1.53 (1 H, dd, J = 9.78, 5.09 Hz) 1.12 (1 H, dd, J = 6.94, 5.97 Hz). NH 2 peak was not observed. Preparation of 6-((3-((lS,5S,6S)-3-amino-5-methyl-l-(pyrrolidine-l-carbonyl )-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)-5-m ethylnicotinonitrile (128).

A solution of lithium hydroxide monohydrate (5.31 mg, 0.127 mmol) in water (0.500 inL) was added to a stirred solution of (l S,5S,6S)-methyl 3-amino-5-(5-((5-cyano-3- methylpyridin-2-yl)ethynyl)-2-fluorophenyl)-5-methyl-2-thia- 4-azabicyclo[4.1.0]hept-3-ene-

1- carboxylate (128c, 50 mg, 0.115 mmol) in THF (0.75 inL) and methanol (0.50 mL). The reaction mixture was stirred at room temperature for 1.5 hours then quenched with sat'd aqueous NH4CI. The resulting precipitate was filtered and washed with water to give crude

( 1 S,5 S,6S)-3 -amino-5 -(5 -((5 -cyano-3 -methylpyridin-2-yl)ethynyl)-2-fluorophenyl)-5 -methyl - 2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxylic acid as a light orange solid (54 mg) that was used as crude. LC/MS (ESf) in : 42 1. 1 [M+H] + .

Propylphosphonic anhydride (50 wt. % in ethyl acetate, 163 mg, 0.26 mmol) was added to a stirred solution of the crude (lS,5S,6S)-3-amino-5-(5-((5-cyano-3-methylpyridin-

2- yl)ethynyl)-2-fluorophenyl)-5 -methyl -2 -thia-4-azabicyclo [4.1.0]hept-3 -ene- 1 -carboxylic acid and pyrrolidine (0.043 mL, 0.514 mmol) in N, N-dimethylformamide (1 mL) at 0 °C.

The reaction mixture was stirred at 0 °C for 1 hour before being warmed to room temperature and stirred for another 2 hours. The reaction mixture was quenched with sat'd aqueous sodium bicarbonate and extracted with EtOAc. The organic layer was separated, washed with brine, dried over MgSO/t, filtered, and concentrated in vacuo. Chromatographic purification of the residue (silica gel, 0 to 75% acetone in heptane) gave 6-((3-((lS,5S,6S)-3- amino-5-methyl-l-(pyrrolidine-l-carbonyl)-2-thia-4-azabicycl o[4.1.0]hept-3-en-5-yl)-4- fluorophenyl)ethynyl)-5-methylnicotinonitrile (Example 128, 5 mg, 0.01 mmol, 8% yield) as a white solid. LC/MS (ESI + ) m/z = 474.2 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 8.70 (d, J = 1.56 Hz, 1H) 7.77 - 7.82 (m, 2H) 7.49 (ddd, J = 8.31, 4.60, 2.15 Hz, 1H) 7.08 (dd, J = 11.54, 8.41 Hz, 1H) 3.66 (br. s., 2H) 3.47 (br. s., 2H) 2.56 (s, 3H) 2.30 (ddd, J = 9.39, 7.34, 1.08 Hz, 1H) 1.82 - 2.02 (m, 4H) 1.84 (s, 3H) 1.41 (dd, J = 9.68, 5.77 Hz, 1H) 0.82 (t, J = 6.36 Hz, 1H). NH 2 peak was not observed. Example 129: (lS,5S,6S)-3-amino-5-(5-((4-cyanophenyl)ethynyl)-2-fluorophe nyl)-N,N,5- trim hyl-2-thia-4-azabicycl -3-ene-l-carboxamide.

A mixture of 123d (65 mg, 0.20 mmol), copper(I) iodide (5 mg), 4- bromobenzonitrile (Acros Organics) (62 mg, 0.34 mmol), dichlorobis(triphenyl- phosphine)palladium (II) (Strem Chemicals) (14 mg, 0.02 mmol) in 2 mL of THF was stirred under argon for 5 minutes then treated with triethylamine (Sigma-Aldrich, 0.05 mL, 0.39 mmol). The flask was evacuated and purged with argon. The mixture was stirred under argon at 60 °C for 1 hour then concentrated in vacuo. The residue was purified on a silica gel column (0 to 40% EtOAc in heptane) to give Example 129 (31 mg, 36% yield) as a white solid. LC/MS (ESI + ) m/z = 433.1 [M+H] + . 'H NMR (400 MHz, CHLOROFORM-d) δ 7.70- 7.83 (m, 1H), 7.50-7.67 (m, 4H), 7.35-7.51 (m, 1H), 6.95-7.16 (m, 1H), 3.78-5.07 (m, 2H), 2.71-3.40 (m, 6H), 2.24-2.38 (m, 1H), 1.73-1.93 (m, 3H), 1.16-1.39 (m, 2H).

Example 130: (lS,5S,6S)-3-amino-5-(2-fluoro-5-((5-(2-propyn-l-yloxy)-2- pyridinyl)ethynyl)phenyl)-N,N,5-trimethyl-2-thia-4-azabicycl o[4.1.0]hept-3- carboxamide.

123d 130 Preparation of 2-bromo-5-((3-(trimethylsilyl)prop-2-yn-l-yl)oxy)pyridine (130a).

A mixture of 2-bromo-5-hydroxypyridine (Matrix Scientific, Columbia, SC, USA, 1.30 g, 7.47 mmol), 3-bromo-l-(trimethylsilyl)-l-propyne (Sigma-Aldrich, 3.20 mL, 20.42 mmol), and potassium carbonate (2.60 g, 18.81 mmol) in G¾CN (20 mL) were heated at 80 °C for 1 hour. The mixture was cooled to room temperature, and partitioned between 100 mL of ethyl acetate and 10 mL of water. The organic layer was washed brine and evaporated in vacuo. The residue was purified on a silica gel column (0 to 25% ethyl acetate in heptane) to give 130a (1.37 g, 64% yield). LC/MS (ESI + ) m/z = 284.0/286.0 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ 7.96-8.01 (m, 1H), 7.22 (d, J = 8.80 Hz, 1H), 7.01-7.05 (m, 1H), 4.54 (s, 2H), -0.08-0.06 (m, 9H).

Preparation of (lS,5S,6S)-3-amino-5-(2-fluoro-5-((5-(2-propyn-l-yloxy)-2- pyridinyl)ethynyl)phenyl)-N,N,5-trimethyl-2-thia-4-azabicycl o[4.1.0]hept-3-ene-l- carboxamide (130).

A mixture of 123d (61 mg, 0.18 mmol), copper(I) iodide (4 mg),

dichlorobis(triphenyl-phosphine)palladium (II) (13 mg, 0.02 mmol) triethylamine (0.05 mL, 0.36 mmol) in THF (1.5 mL) was purged with argon then heated at 60 °C for 3 hours. The mixture was treated with 1 mL of dioxane then heated to 90 °C for 1.5 hours. After cooling to room temperature, the mixture was diluted with 50 mL of EtOAc and filtered. The filtrate was evaporated in vacuo. The residue was dissolved in 1 mL of MeOH and treated with K2CO3 (10 mg). The mixture was stirred for 15 minutes then concentrated. The residue was partitioned between 25 mL of ethyl acetate and 10 mL of water. The organic layer was concentrated and the residue was purified by preparative reverse phase HPLC (Phenomenx Gemini C18 10 μΜ, 150 x 30 mm column, 10 to 90% (0.1% TFA in CH3CN) in (0.1% TFA in water)). Desired fractions were concentrated and the residue was partitioned between EtOAc (25 mL) and 1 N NaOH (5 mL). The EtOAc layer was washed with brine (3 mL) and concentrated in vacuo to give Example 130 (6 mg, 13% yield) as a solid. LC/MS (ESI + ) m/z = 463.1 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ 8.31-8.41 (m, 1H), 7.66-7.78 (m, 1H), 7.40-7.54 (m, 2H), 7.28-7.33 (m, 1H), 6.96-7.09 (m, 1H), 4.73-4.80 (m, 2H), 2.89- 3.25 (m, 6H), 2.55-2.59 (m, 1H), 1.96-2.05 (m, 1H), 1.80-1.89 (m, 3H), 1.36-1.40 (m, 1H), 0.64-0.75 (m, 1H). NH 2 peak was not observed. Example 131: (lS,5S,6S)-3-amino-5-(2-fluoro-5-((3-methyl-5-(oxazol-2- ylmethoxy)pyridin-2-yl)ethynyl)phenyl)-N,N,5-trimethyl-2-thi a-4-azabicyclo[4.1.0]hept-

3

Preparation of 2-(((6-bromo-5-methylpyridin-3-yl)oxy)methyl)oxazole (131a).

A mixture of 2-chloromethyl-oxazole (J&W Pharmlab LLC, Levittown, PA, USA) (0.36 mL, 3.10 mmol), 2-bromo-5-hydroxy-3-picoline (AOBchem USA, Santa Monica, Ca, USA) (0.54 g, 2.87 mmol), and potassium carbonate (0.21 mL, 3.45 mmol) in 2 mL acetonitrile was heated at 90 °C for 1 hour then stirred at room temperature for 14 hours. It was partitioned between 50 mL of ethyl acetate and 10 mL of water. The organic layer was concentrated and the residue was purified on a silica gel column (0 to 40% EtOAc in heptane) to give 131a (0.44 g, 57% yield) as a white solid. LC/MS (ESI + ) m/z = 269.0/271.0 [M+H] + . 1 H NMR (400 MHz, CHLOROFORM-d) δ 7.97-8.05 (m, 1H), 7.64-7.75 (m, 1H), 7.21-7.24 (m, 1H), 7.13-7.18 (m, 1H), 5.13-5.21 (m, 2H), 2.33-2.40 (m, 3H). Preparation of (lS,5S,6S)-3-amino-5-(2-fluoro-5-((3-methyl-5-(l,3-oxazol-2- ylmethoxy)- 2-pyridinyl)ethynyl)phenyl)-N,N,5-trimethyl-2-thia-4-azabicy clo[4.1.0]hept-3-ene-l- carboxamide (131).

A mixture of 2-(((6-bromo-5-methylpyridin-3-yl)oxy)methyl)oxazole (97 mg, 0.36 mmol) and (lS,5S,6S)-3-amino-5-(5-ethynyl-2-fluorophenyl)-N,N,5-trimet hyl-2-thia-4- azabicyclo[4.1.0]hept-3-ene-l-carboxamide (123d, 0.12 g, 0.36 mmol) in 3 mL of THF was treated with trans-dichlorobis(triphenyl-phosphine)palladium (II) (Strem Chemicals) (25 mg, 0.04 mmol), triethylamine (0.10 mL, 0.72 mmol), and copper(I) iodide (10 mg, 0.05 mmol), then purged with argon for 5 minutes. It was heated at 60 °C for 3 hours, and then concentrated in vacuo. The residue was purified on a silica gel column (0 to 100% EtOAc in heptane then 5% MeOH in EtOAc) to afford a material that contained the desired product (131) and an impurity in a ratio of 84/16 based on HPLC integration. This material was subjected to SFC (20 mM N¾ in MeOH on a pyridine column) to give Example 131.

LC/MS (ESI + ) m/z = 520.0 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ 8.16-8.32 (m, 1H), 7.74-7.84 (m, 1H), 7.65-7.73 (m, 1H), 7.39-7.51 (m, 1H), 7.13-7.24 (m, 2H), 6.94- 7.08 (m, 1H), 5.16-5.27 (m, 2H), 2.86-3.34 (m, 6H), 2.42-2.59 (m, 3H), 2.22-2.35 (m, 1H), 1.77-1.89 (m, 3H), 1.24-1.37 (m, 1H), 0.81-0.94 (m, 1H). NH 2 peak was not observed.

Example 132: (lS,5S,6S)-3-amino-5-(5-(cyclopropylethynyl)-2-fluorophenyl) -N,N,5- trimethyl-2-thia-4-azabicyclo[4.1.0]hept-3-ene-l-carboxamide .

A mixture of 123c (48 mg, 0.12 mmol), tetrakis(triphenylphosphine)palladium (Sigma- Aldrich, 19 mg) and copper(I) iodide (3.2 mg) in a round flask was purged with argon for 5 minutes, then treated sequentially with diisopropylamine (0.17 mL, 1.24 mmol), DMF (0.50 mL) and cyclopropylacetylene (Sigma- Aldrich, 53 μί, 0.62 mmol). The mixture was heated at 60 °C for 2 h then 70 °C for 3 hours. It was cooled to room temperature and partitioned between ethyl acetate (25 mL) and brine (15 mL). The organic layer was dried over Na2S04 and evaporated. The residue was purified on silica gel column (0 to 50% (10% MeOH/EtOAc) in EtOAc) to afford Example 132 (28 mg, 61% yield). LC/MS (ESf) m/z = 372.2 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ 6.71-7.86 (m, 3H), 2.47-4.43 (m, 6H), 1.53-2.35 (m, 3H), 1.05-1.49 (m, 3H), 0.34-0.98 (m, 5H). NH 2 peak was not observed. Example 133: 6-((3-((lS,5S)-3-amino-l-(methoxymethyl)-5-methyl-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile.

133c 133

Preparation of tert-Butyi ((lS,5S)-5-(5-ethynyl-2-fluorophenyl)-l-(methoxymethyl)-5- methyl-2-thia-4-azabicyclo[4.1.0]hept-3-en-3-yl)((2- (trimethylsilyl)ethoxy)methyl)carbamate (133b).

To a round bottom flask was charged with fert-butyl ((lS,5S)-5-(5-bromo-2- fluorophenyl)- 1 -(methoxymethyl)-5 -methyl-2-thia-4-azabicyclo [4.1.0]hept-3 -en-3 -yl)((2- (trimethylsilyl)ethoxy)methyl)carbamate (133a, prepared as previously reported in WO 2016022724) (0.30 g, 0.51 mmol) and bis(tri-i-butylphosphine)palladium(0) (Sigma-Aldrich, St. Louis, MO, USA) (0.026 g, 0.05 mmol) under an argon atmosphere. The flask was evacuated and backfilled with N2 3 times and then 1,4-dioxane (5.09 mL) and

ethynyltributylstannane (Sigma-Aldrich, St. Louis, MO, USA) (0.22 mL, 0.76 mmol) were added. The reaction mixture was heated to 60 °C and stirred for 2 hours. The reaction mixture was then cooled to room temperature, diluted with EtOAc and 1 M aqueous KF, and stirred for 15 minutes. The biphasic mixture was filtered through Celite. The organic layer was collected, washed with brine, dried over MgS04, filtered, and concentrated under reduced pressure to yield 133b as a brown oil (partially solidified upon standing) which was used as crude assuming 100% yield. Preparation of tert-butyi ((lS,5S)-5-(5-((5-cyanopyridin-2-yl)ethynyl)-2-fluorophenyl) -l- (methoxymethyl)-5-methyl-2-thia-4-azabicyclo[4.1.0]hept-3-en -3-yl)((2- (trimethylsilyl)ethoxy)methyl)carbamate (133c).

Crude 133b (90 mg, 0.17 mmol), 6-bromonicotinonitrile (Sigma-Aldrich, St. Louis, MO, USA) (31 mg, 0.168 mmol), copper(I) iodide (Johnson Matthey, West Deptford, NJ, USA) (48 mg, 0.025 mmol), and trans-dichlorobis(triphenylphosphine)palladium(II) (Strem Chemicals Inc., Newburyport, MA, USA) (0.012 g, 0.017 mmol) were combined in a round bottom flask and placed under a nitrogen atmosphere. THF (1.68 mL) and triethylamine (0.047 mL, 0.337 mmol) were added, and the reaction mixture was stirred at 60 °C for 1 hour. The reaction mixture was cooled to ambient temperature and diluted with EtOAc. The mixture was washed with sat'd aqueous NH4CI, washed with brine, dried over MgSO/t, filtered, and concentrated under reduced pressure. The crude residue was purified by silica gel chromatography employing a gradient of 0 to 25% (3: 1 EtOAc/EtOH) in heptane to give tert-butyl ((lS,5S)-5-(5-((5-cyanopyridin-2-yl)ethynyl)-2 -fluorophenyl)- l-(methoxymethyl)- 5-methyl-2-thia-4-azabicyclo[4.1.0]hept-3-en-3-yl)((2-

(trimethylsilyl)ethoxy)methyl)carbamate (133c, 0.088 g, 0.138 mmol, 82% yield). LC/MS (ESI + ) m/z = 537.2 [M+H-Boc] + .

Preparation of 6-((3-((lS,5S)-3-amino-l-(methoxymethyl)-5-methyl-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile (133).

To a mixture of tert-butyl ((lS,5S)-5-(5-((5-cyanopyridin-2-yl)ethynyl)-2- fluorophenyl)- 1 -(methoxymethyl)-5 -methyl-2-thia-4-azabicyclo [4.1.0]hept-3 -en-3 -yl)((2- (trimethylsilyl)ethoxy)methyl)carbamate (133c, 0.087 g, 0.137 mmol) in 1,4-dioxane (1.4 mL) was added >-toluenesulfonic acid monohydrate (Sigma-Aldrich, St. Louis, MO, USA) (0.078 g, 0.410 mmol) and the reaction mixture was heated to 80 °C for 3 hours at which point the starting material was consumed. The mixture was diluted with EtOAc, washed with sat'd aqueous sodium bicarbonate, dried over MgSO/t, and concentrated in vacuo. The residue was purified by silica gel chromatography employing a 0 to 60% (3 : 1 EtOAc/EtOH) gradient in heptane to give 6-((3-((lS,5S)-3-amino-l-(methoxymethyl)-5-methyl-2-thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-4-fluorophenyl)ethynyl)nico tinonitrile (0.022 g, 0.054 mmol, 40% yield) as a light yellow solid. LC/MS (ESI + ) m/z = 407.1 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 8.87 (s, 1H) 7.88 - 8.02 (m, 2H) 7.61 (d, J = 8.22 Hz, 1H) 7.50 (ddd, J = 8.27, 4.55, 2.25 Hz, 1H) 6.98 - 7.16 (m, 1H) 3.60 - 3.70 (m, 1H) 3.42 (s, 3H) 3.35 (d, J = 10.76 Hz, 1H) 1.75 - 1.81 (m, 1H) 1.72 (s, 3H) 1.12 - 1.27 (m, 2H) 0.79 (br t, J = 6.16 Hz, 1H). Note: only one NH proton was observed. 19 F NMR (376 MHz,

CHLOROFORM-d) δ ppm - 106.91 (s).

Example 134 : (1 S,5S)-5-(5-((5-chlor o-3-methylpyridin-2-yl)ethynyl)-2-fluor ophi

This compound (0.023 g, 0.053 mmol, 31% overall yield) as a light yellow solid was prepared via a protocol analogous to that employed for the synthesis of Example 133, here starting with fert-butyl ((l S,5S)-5-(5-ethynyl-2-fluorophenyl)- l-(methoxymethyl)-5-methyl- 2-thia-4-azabicyclo[4.1.0]hept-3-en-3-yl)((2-(trimethylsilyl )ethoxy)methyl)carbamate (133b, 0.09 g, 0.168 mmol) and 2-bromo-5-chloro-3-methylpyridine (Sigma-Aldrich, St. Louis, MO, USA) (0.035 g, 0.168 mmol). LC/MS (ESI + ) m/z = 429.1 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-£ ) δ ppm 8.40 (d, J = 2.15 Hz, 1H) 7.95 (dd, J = 7.82, 2.15 Hz, 1H) 7.52 - 7.60 (m, 1H) 7.43 - 7.51 (m, 1H) 6.97 - 7.10 (m, 1H) 4.22 - 4.64 (m, lH) 3.66 (d, J = 10.37 Hz, 1H) 3.41 (s, 3H) 3.35 (d, J = 10.76 Hz, 1H) 2.51 (s, 3H) 1.72 - 1.80 (m, 1H) 1.66 - 1.72 (m, 3H) 1.39 (d, J = 12.13 Hz, 1H) 0.76 - 0.83 (m, 1H). Only one NH proton observed. 19 F NMR (376 MHz, CHLOROFORM-d) δ ppm - 108.46 (s).

Example 135: 6-((3-((lS,5S)-3-amino-l-(methoxymethyl)-5-methyl-2-thia-4-

This compound (0.033 g, 0.078 mmol, 24% overall yield) was prepared as a white solid using a method similar to that described for the synthesis of 133, here starting with ( 1 S,5 S)-5-(5-ethynyl-2-fluorophenyl)- 1 -(methoxymethyl)-5-methyl-2-thia-4- azabicyclo[4.1.0]hept-3-en-3-amine (133b, 0.099 g, 0.325 mmol) and 6-bromo-5- methylnicotinonitrile (Arkpharm Inc, Libertyville, IL, USA) (0.064 g, 0.325 mmol). LC/MS (ESI + ) m/z = 421.1 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 8.62 - 8.79 (m, 1H) 7.94 - 8.06 (m, 1H) 7.74 - 7.86 (m, 1H) 7.35 - 7.58 (m, 1H) 6.93 - 7.16 (m, 1H) 4.05 - 4.63 (m, 1H) 3.59 - 3.71 (m, 1H) 3.30 - 3.38 (m, 1H) 2.48 - 2.66 (m, 3H) 1.74 - 1.81 (m, 1H) 1.69 - 1.74 (m, 3H) 1.49 - 1.65 (m, 3H) 0.84 - 0.92 (m, 1H) 0.74 - 0.82 (m, 1H). Note: only one NH proton observed.

Example 136: 6-((5-((lS,5S)-3-amino-5-methyl-l-(morpholine-4-carbonyl)-2- thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-6-fluoropyridin-3-yl)ethyny l)-5-methylnicotinonitrile.

Preparation of (5S)-5-(5-bromo-2-fluoropyridin-3-yl)-3-((tert-butoxycarbony l)((2-

(trimethylsilyl)ethoxy)methyl)amino)-5-methyl-2-thia-4-az abicyclo[4.1.0]hept-3-ene-l- carboxylic acid (136b).

To a solution of (5S)-methyl 5-(5-bromo-2-fluoropyridin-3-yl)-3-((tert- butoxycarbonyl)((2-(trimethylsilyl)ethoxy)methyl)amino)-5-me thyl-2-thia-4- azabicyclo[4.1.0]hept-3-ene-l-carboxylate (136a, WO 2016022724) (1.68 g, 2.78 mmol) in THF (7.95 mL) and water (3.18 mL) was added lithium hydroxide (0.15 g, 6.40 mmol). This mixture was allowed to stir for 12 hours at 23 °C. The reaction mixture was then concentrated, acidified to pH 4 using 1 N aqueous HC1, extracted with EtOAc (2 x 30 mL), dried over MgS04 and concentrated to give (5S)-5-(5-bromo-2-fluoropyridin-3-yl)-3-((tert- butoxycarbonyl)((2-(trimethylsilyl)ethoxy)methyl)amino)-5-me thyl-2-thia-4- azabicyclo[4.1.0]hept-3-ene-l-carboxylic acid (136b, 1.64 g, 2.78 mmol, 100% yield) as a yellow oil, which was taken forward without purification.

Preparation of ((lS,5S,6S)-3-amino-5-(5-bromo-2-fluoropyridin-3-yl)-5-methy l-2-thia-4- azabicyclo[4.1.0]hept-3-en-l-yl)(morpholino)methanone (136c).

To a solution of l-chloro-N,N,2-trimethylprop-l -en- 1 -amine (Acros Organics, 0.39 g, 2.90 mmol) in 5 mL of MeCN was added (lS,5S,6S)-5-(5-bromo-2-fluoropyridin-3-yl)-3- ((tert-butoxycarbonyl)((2-(trimethylsilyl)ethoxy)methyl)amin o)-5-methyl-2-thia-4- azabicyclo[4.1.0]hept-3-ene-l-carboxylic acid (136b, 0.57 g, 0.96 mmol) in 15 mL of MeCN. This mixture was stirred at ambient temperature for 15 minutes at which point morpholine (0.26 mL, 2.90 mmol) followed by triethylamine (0.54 mL, 3.87 mmol) were added, both in dropwise fashion. This mixture was allowed to stir for 3 hours. The crude mixture was then concentrated under reduced pressure, diluted with EtOAc, washed with sat'd aqueous NH 4 CI, dried over MgS04, and concentrated under reduced pressure. The crude amide was taken up in 8 mL of 1,4 dioxane and /J>-toluenesulfonic acid monohydrate (0.55 g, 2.90 mmol) was added. The mixture was heated at 80 °C for 12 hours. The crude mixture was diluted with EtOAc, washed with sat'd aqueous NaHC03, the aqueous layer was back extracted with EtOAc then DCM, the combined organic washings were dried over MgS04 and concentrated. The residue was purified via silica gel chromatography employing a 0 to 60 % (3: 1

EtOAc/EtOH) in heptane gradient to give ((lS,5S,6S)-3-amino-5-(5-bromo-2-fluoropyridin- 3 -yl)-5 -methyl -2 -thia-4-azabicyclo [4.1.0]hept-3 -en- 1 -yl)(morpholino)methanone (0.18 g, 0.41 mmol, 43% yield) as a colorless oil. LC/MS (ESI + ) m/z = 429.1/431.1 [M+H] + . Preparation of 6-((5-((lS,5S)-3-amino-5-methyl-l-(morpholine-4-carbonyl)-2- thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-6-fluoropyridin-3-yl)ethyny l)-5-methylnicotinonitrile (136).

1,4-Dioxane (4.14 mL) and ethynyltributylstannane ( Sigma- Aldrich, St. Louis, MO, USA) (0.18 mL, 0.62 mmol) were added to a flask charged with ((lS,5S)-3-amino-5-(5- bromo-2-fluoropyridin-3-yl)-5-methyl-2-thia-4-azabicyclo[4.1 .0]hept-3-en- 1- yl)(moφholino)methanone (136c, 0.18 g, 0.41 mmol) and bis(tri-t- butylphosphine)palladium(O) (Sigma-Aldrich, St. Louis, MO, USA) (0.02 g, 0.04 mmol) under an argon atmosphere. The reaction mixture was heated to 80 °C and stirred for 2 hours. The reaction mixture was then cooled to ambient temperature, diluted with EtOAc and 1 M aqueous KF, and stirred for 15 min. The biphasic mixture was filtered through celite. The organic layer was separated, washed with brine, dried over MgSO/t, filtered, and concentrated to yield 136d as a brown oil that partially solidified upon standing. The crude 136d was taken on directly assuming 100% yield without further purification or

characterization.

Crude alkyne 136d (155 mg, 0.41 mmol), 6-bromo-5-methylnicotinonitrile

(Arkpharm Inc, Libertyville, IL, USA) (82 mg, 0.41 mmol), copper(I) iodide (Alfa Aesar, West Deptford, NJ, USA) (12 mg, 0.062 mmol), and trans- dichlorobis(triphenylphosphine)palladium (II) (Strem Chemicals Inc., Newburyport, MA, USA) (29 mg, 0.041 mmol) were mixed in a round bottom flask and placed under a nitrogen atmosphere. THF (2.07 mL) and triethylamine (0.11 mL, 0.82 mmol) were added, and the reaction mixture was stirred at 60 °C for 1 hour. The reaction mixture was cooled to ambient temperature and diluted with EtOAc. The mixture was washed with sat'd aqueous NH4CI, washed with brine, dried over MgS04, filtered, and concentrated. The residue was purified by silica gel chromatography using a 0 to 60% (EtOAc/EtOH = 3: 1) gradient in heptane to give 6-((5-((lS,5S)-3-amino-5-methyl-l-(mo holine-4-carbonyl)-2-thia-4- azabicyclo [4.1.0]hept-3 -en-5-yl)-6-fluoropyridin-3 -yl)ethynyl)-5 -methylnicotinonitrile (Example 136, 86 mg, 0.175 mmol, 42% yield) as an amorphous off-white solid. LC/MS (ESI + ) m/z = 491.2 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 8.72 (d, J = 1.56 Hz, 1H) 8.23 - 8.46 (m, 2H) 7.85 (d, J = 0.98 Hz, 1H) 4.50 (br s, 1H) 3.50 - 3.80 (m, 8 H) 2.49 - 2.73 (m, 3H) 2.24 - 2.45 (m, 1H) 1.66 - 1.90 (m, 3H) 1.21 - 1.35 (m, 1H) 0.77 -

0.94 (m, 1H). Note: Only one NH proton is observed. 19 F NMR (376 MHz, CHLOROFORM- d) δ ppm -60.07 (s). Example 137: 6-((5-((lS,5S)-3-amino-5-methyl-l-(pyrrolidine-l-carbonyl)-2 -thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-6-fluoropyridin-3-yl)ethyny l)-5-methylnicotinonitrile.

Preparation of ((lS,5S)-3-amino-5-(5-bromo-2-fluoropyridin-3-yl)-5-methyl-2 -thia-4- azabicyclo[4.1.0]hept-3-en-l-yl)(pyrrolidin-l-yl)methanone (137a).

This compound (202 mg, 0.49 mmol, 48% overall yield) was prepared in a manner similar to that described for compound 136c, here starting from 136b (596 mg, 1.01 mmol).

Preparation of 6-((5-((lS,5S)-3-amino-5-methyl-l-(pyrrolidine-l-carbonyl)-2 -thia-4- azabicyclo[4.1.0]hept-3-en-5-yl)-6-fluoropyridin-3-yl)ethyny l)-5-methylnicotinonitrile (137).

This compound (121 mg, 0.26 mmol, 52% overall yield) as an amorphous off-white solid was prepared in a 2-step protocol similar to that described for Example 136, here starting from 137a (202 mg, 0.49 mmol). LC/MS (ESI + ) m/z = 475.2 [M+H] + . 'H NMR (400 MHz, CHLOROFORM-d) δ ppm 8.71 (d, J = 1.56 Hz, 1H) 8.33 (s, 1H) 8.24 (dd, J = 9.19, 2.15 Hz, lH) 7.84 (d, J = 1.17 Hz, 1H) 3.67 (br s, 2H) 3.47 (br s, 2H) 2.49 - 2.63 (m, 3H) 2.26 - 2.38 (m, 1H) 1.83 - 2.03 (m, 4H) 1.83 - 1.84 (m, 1H) 1.80 (s, 3H) 1.27 - 1.38 (m, 1H). NH 2 peak was not observed. 19 F NMR (376 MHz, CHLOROFORM-t ) δ ppm -60.01 (s). Example 138: (lS,5S,6S)-5-(5-((4-chloropyridin-2-yl)ethynyl)-2-fluorophi

bis(fl

This compound (8 mg, 0.02 mmol, 25% yield) as an off-white solid was prepared in a fashion similar to that described for Example 101, here using 101c (24 mg, 0.076 mmol) and 2-bromo-4-chloropyridine (Oakwood Products Inc., Estill, SC, USA) (22 mg, 0.11 mmol) as starting materials. LC/MS (ESI + ) m/z = 422.0 [M+H] + . ¾ NMR (400 MHz, CHLOROFORM-d) δ ppm 8.50 (d, J = 5.09 Hz, 1H) 7.87 (dd, J = 7.53, 1.86 Hz, 1H) 7.47 - 7.55 (m, 2H) 7.24 - 7.28 (m, 1H) 7.07 (dd, J = 11.74, 8.41 Hz, 1H) 4.60 - 4.92 (m, 2H) 4.28 - 4.55 (m, 2H) 1.87 (t, J = 8.22 Hz, 1H) 1.19 (dd, J = 9.39, 6.06 Hz, 1H) 0.78 (td, J = 6.16, 3.91 Hz, 1H). N¾ peak was not observed.

Biological Evaluation

Provided in this section is the biological evaluation of the specific examples provided herein. In particular, Table 2 contains biological activity data. The data presented in Table 2 provides the IC50 (μΜ) for the specific examples obtained in a BACE1 enzyme assay, BACE1 cell assay, BACE2 enzyme assay and CatD assay.

Table 2

Ex. BACE1 Enzyme BACE1 Cell BACE2 Enzyme Cat D Enzyme

No. ICso (μΜ) ICso (μΜ) ICso (μΜ) ICso (μΜ)

100 0.080 0.129 4.550 4866.2

101 0.104 0.271 6.040 2862.1

102 0.182 0.685 0.445 673

103 0.086 3.905 1.590 228.51

104 0.038 0.231 2.280 1303.9

105 0.096 0.746 10.300 500.1

106 0.084 0.623 4.730 614.3

107 0.047 0.372 2.550 238.8

108 0.735 2.020 14.100 783.7 109 0.057 1.1 10 0.092 705.3

110 0.047 0.355 2.080 1 162.2

111 0.019 0.072 0.405 300

112 0.025 0.139 1.590 176

113 0.1 16 0.153 3.760 1421.9

114 0.637 0.677 3.370 > 400.0

115 0.109 0.142 2.180 1033.3

116 0.005 0.018 0.166 172

117 0.021 0.029 0.012 169

118 0.006 0.023 0.017 26.2

119 0.049 0.060 0.126 188

120 0.01 1 0.052 0.179 190

121 0.009 1.900 0.047 143

122 0.010 0.049 0.281 84.3

123 0.003 0.003 0.099 236

124 0.049 0.153 0.037 335.1

125 0.583 0.1 18 8.083 248

126 0.021 0.075 0.617 268

127 0.002 0.022 0.077 968.9

128 0.002 0.002 0.423 507.9

129 0.007 0.058 0.251 289

130 0.161 0.152 0.432 925.6

131 0.041 0.053 2.175 994.8

132 0.020 0.059 0.007 46.8

133 0.097 0.059 2.325 > 400.0

134 0.129 0.951 0.613 > 400.0

135 0.023 0.033 1.465 > 400.0

136 0.037 0.181 0.974 > 400.0

137 0.01 1 0.035 0.474 > 400.0

138 1.123 4.850 0.077 903.3

123d 0.378 0.328 0.142 50.5

The results presented in Table 2 have been generated with the in vitro assays described below. These assays may be used to test any of the compounds described herein to assess and characterize a compound's ability to modulate BACE activity and to regulate the cleavage of Αβ precursor protein, thereby reducing or inhibiting the production of Αβ protein. In Vitro Enzymatic BACEl and BACE2 FRET (Fluorescence Resonance Energy Transfer) Assays

The cDNAs for both human recombinant BACEl and 2 with C-terminal 6-His Tags were cloned into transient protein expression vectors, which were subsequently transfected into mammalian cell lines. These recombinant proteins were further purified using Ni-NTA affinity chromatography (Qiagen). The assay buffer used in these screens was 0.05 M acetate, pH 4.5, 8% DMSO final, 100 μΜ genapol (which is a nonionic detergent, below its Critical Micelle Concentration). The β-secretase enzyme (0.02 nM for BACEl and 0.64 nM for BACE2), which was pre-incubated for one hour with the test compound, typically in about luL of DMSO according to a serial dilution, was added thereto. The assay was effectively started by the addition of FRET substrate (50 nM) and the combination was incubated for one hour. The FRET assay was terminated by the addition of tris buffer, which raised the pH to neutrality, and the fluorescence was determined. The FRET substrate was a peptide with commercially available fluorophore and quencher, on opposite sides of the BACE cleavage site. The specific FRET substrate used in this assay was made by Amgen in- house. Commercially available FRET substrates, for example, the FRET substrate offered with the BACEl FRET Assay Kit sold by ThermoFisher Scientific (Catalog Number P2985), may be used in this assay with the appropriate modifications, which are within the purview of the ability of a person with ordinary skill in the art. Proteolytic cleavage of the FRET substrate released quenching of fluorescence (excitation 488 nm and emission 590 nm).

The in vitro BACE FRET enzyme data for each of the Examples is provided in Table

2.

In Vitro BACEl cell-based assay

The cell-based assay measures inhibition or reduction of Αβ40 in conditioned medium of test compound treated cells expressing amyloid precursor protein. Cells stably expressing Amyloid Precursor Protein (APP) were plated at a density of 45K cells/well in 384 well plates (Corning/BioCoat 354663). The test compounds were then added to cells in 22-point dose response concentrations with the starting concentration being 62.5 μΜ. The compounds were diluted from stock solutions in DMSO and the final DMSO concentration of the test compounds on cells was 0.625%. The cells were cultivated overnight at 37°C and 5% C0 2 in DMEM supplemented with 10% FBS. After 24 h of incubation with the test compounds, the conditioned media was collected and the Αβ40 levels were determined using HTRF (Homogeneous Time Resolved Fluorescence). The IC50 of the compound was calculated from the percent of control or percent inhibition of Αβ 40 as a function of the concentration of the test compound.

The HTRF to detect Αβ40 was performed in 384 well plates (Costar 3658). The antibody pair that were used to detect Αβ 40 from cell supematants were ConfAb40 antibody (Amgen in-house) and biotinylated 6E10 (BIOLEGEND). As an alternative to ConfAb40, a commercially available antibody, Anti -beta Amyloid 1-40 antibody [BDI350] from Abeam, Cambridge, MA 02139-1517 (Product code: ab20068), may be used in this assay. The concentrations were 0.35 μg/mL of ConfAb40 antibody and 1.33 μg/mL of 6E10-biotinylated antibody, as well as 4.5 μg/mL of Streptavidin Allophycocyanin Conjugate (ThermoFisher Scientific) in HTRF Buffer (1M Hepes pH 7.5, 1M NaCl, 1% BSA, 0.5% Tween 20).

The conditioned media was incubated with above antibodies and Streptavidin Allophycocyanin Conjugate for 30-60 minutes at 23°C. The final readout was performed on Envision from PerkinElmer.

The in vitro BACE cell-based data for each of the Examples is provided in Table 2.

In Vitro Enzymatic Cathepsin D (CatD) FRET Assay

Recombinant CatD was expressed in CHO cells. The assay buffer for CatD was 0.05 M citrate pH 3.5, 10% DMSO final, 5 mM CHAPS. The CatD enzyme (9 nM) was pre- incubated for one hour with inhibitors, typically in about luL of DMSO according to a serial dilution, is added thereto. The assays was effectively started by the addition of different FRET substrates (20 nM for CatD) and the combination was incubated for one hour. The FRET assay was terminated with by addition of tris buffer, which raises the pH to neutrality, and the fluorescence was determined. The FRET substrate was a peptide with commercially available fluorophore and quencher, on opposite sides of the CatD cleavage site. The CatD substrate peptide sequence was based on sequence # 1 of Table 1 from Gulnik et al. , FEBS Lett. 413(2):379-384 (1997). Proteolytic cleavage of the FRET substrate released quenching of fluorescence (CatD excitation 500 nm and emission 580 nm). Alternatively, a CatD assay may also be run according to the procedure described in Yasuda et al. , J. Biochem. 125(6): 1137-1143 (1999). In addition, the CatD and Cathepsin E assays are described in International Patent Application Publication No. WO2011069934.

The in vitro CatD FRET assay data for each of the Examples is provided in Table 2, conducted by the first procedure described above. As shown by the high micromolar CatD data (very poorly active or inactive against CatD), the compounds disclosed herein possess the unexpected property of little to no ability to inhibit the activity of CatD. Thus, with this surprising selectivity profile, the compounds provided herein are believed to minimize, reduce or completely eliminate any risk of retinal atrophy and abnormal development of the eye and of the retinal pigmented epithelium as it relates to the normal function and activity of CatD.

In vivo Inhibition of β-Secretase

Several animal models, including mouse, rat, dog, and monkey, may be used to screen for inhibition of β-secretase activity in vivo following administration of a test compound. This procedure may be used to show that the compounds provided herein reduce the formation and/or deposition of Αβ peptide in the cerebrospinal fluid (CSF) as well as in the brain. Animals to be used in this experiment can be wild type, transgenic, or gene knockout animals. For example, the Tg2576 mouse model, prepared and conducted as described in Hsiao et al, Science 274:99-102 (1996), and other non-transgenic or gene knockout animals are useful to analyze in vivo inhibition of Αβ peptide production in the presence of test compounds.

Generally, 2 to 18 month old Tg2576 mice, gene knockout mice or non-transgenic animals are administered test compounds formulated in vehicles, such as cyclodextran, phosphate buffers, hydroxypropyl methylcellulose or other suitable vehicles. One to twenty- four hours following the administration of compound, animals are sacrificed, and brains as well as cerebrospinal fluid (CSF) and plasma are removed for analysis of Αβ levels and test compound concentrations (Dovey et al. , J. Neurochem., 76( 1): 173-181 (2001)) Beginning at time 0, animals are administered by oral gavage, or other means of delivery such as intravenous injection, an inhibitory test compound of up to 100 mg/kg in a standard, conventional formulation, such as 2% hydroxypropyl methylcellulose, 1% Tween80. A separate group of animals receive 2% hydroxypropyl methylcellulose, 1% Tween80 alone, containing no test compound, and serve as a vehicle-control group. At the end of the test period, animals are sacrificed and brain tissues, plasma or cerebrospinal fluid are collected. Brains are either homogenized in 10 volumes (w/v) of 0.2% diethylamine (DEA) in 50 mM NaCl (Best et al., J. Pharmacol. Exp. Ther. 313(2):902-908 (2005)), or in 10 volumes of 0.5% TritonX-100 in Tris-buffered saline (pH at about 7.6). Homogenates are centrifuged at 355,000g, 4°C for 30 minutes. CSF or brain supernatants are then analyzed for the presence of Αβ by specific sandwich ELISA assays based on ECL (Electrochemiluminescence) technology. For example, rat Αβ40 is measured using biotinylated-4G8 (Signet) as a capture antibody and Fab40 (an in-house antibody specific to the C-terminal of Αβ40) as a detection antibody. For example, 4 hours after administration of 30 mg/kg oral dose of the test compound in 2% hydroxypropyl methylcellulose, 1% Tween80 (pH2.2) to 200g male Sprague Dawley rats, Αβ peptide levels are measured for reduction by X% and Y% in cerebrospinal fluid and brain, respectively, when compared to the levels measured in the vehicle-treated or control mice. Alternatively, the antibody sold with the V-PLEX abeta40 Peptide (4G8) Kit, commercially available from Meso Scale Diagnostics (MSD), Rockville, Maryland 20850-3173 (Catalog NO. K150SJE-1) may be used in this assay.

This procedure may be used to show that the compounds provided herein reduce the formation and/or deposition of Αβ peptide in the cerebrospinal fluid (CSF) as well as in the brain of a mouse or rat at either 3mpk, 10 mpk or 30 mpk (mpk = mg compound per kg weight of the animal) dosing concentrations after 4hrs.

METHODS OF USE

According to the amyloid cascade hypothesis, cerebral deposition of amyloid-beta (Αβ) peptide is critical for Alzheimer's disease (AD) pathogenesis. Αβ peptide generation is initiated when β-secretase (BACE1) cleaves the amyloid precursor protein. De Meyer et al. re-affirm the putative role that the accumulation of Αβ peptide in cerebral spinal fluid (CSF) in a subject plays in the progression of symptoms, initially revealed as mild cognitive impairment, which ultimately leads to AD. Arch Neurol. 67(8):949-956 (2010). Αβ peptides generated from amyloid precursor protein (APP) by proteolytic cleavage, such as by aspartyl protease enzymes, including β-secretase (BACE) and γ-secretase, likely play a causal role in AD pathogenesis (Tanzi et al , Cell 120(4):545-555 (2005); Walsh et al, Neuron 44(1): 181- 193 (2004)). Although the precise mechanisms of Αβ toxicity are unclear, oligomeric forms of Αβ may contribute to cognitive decline by altering synaptic structure and function (Palop et al , Nat. Neurosci. 13(7):812-818 (2010); Selkoe, Behav. Brain Res. 192(1): 106-113 (2008); Shankar et al. , Nat. Med. 14(8): 837-842 (2008)). Transgenic mouse models that overexpress mutant APP and produce high levels of Αβ show amyloid plaque deposition, synaptic deficits, learning and memory impairments, and other behavioral abnormalities (Games et al , Nature 373:523-527 (1995); Gotz et al., Mol. Psychiatry 9(7):664-683 (2004); Hsia et al, Proc. Natl. Academy of Science USA (96): 3228-3233, 1999; Hsiao et al , Science (274): 99-102, 1996, citing Harris et al, Neuron (68): 428-441, 2010).

For many years now, BACE1 has been a prime target for designing drugs to prevent or treat AD. Vassar et al , Lancet Neurol. 13:319-329 (2014). Several pharmaceutical companies are presently pursuing BACE1 inhibitors in human clinical trials. Id. at abstract.

For example, MK-8931, a small molecule inhibitor of BACE1, was the first molecule to enter phase I clinical trials. Yan, Transl. Neurodegener. 5(13): 1-11 (2016) at page 4. MK- 8931 was shown to have an excellent safety profile with no immediately noticeable side effects. Id. Merck was able to show that MK-8931 enters the brain and blocks β-secretase by showing that MK-8931 significantly reduced CSF Αβ peptide concentrations in a sustained and dose-dependent manner. Vassar et al. , Lancet Neurol. 13:319-329 (2014) at page 323. MK-8931 is currently evaluated in a phase II/III clinical trial to assess the efficacy and safety of the compound for the treatment of AD patients with amnestic mild cognitive impairment (prodromal AD). Yan, Transl. Neurodegener. 5(13): 1-11 (2016) at page 4.

Further, E2609, a BACE inhibitor identified by Eisai, showed significant reduction in Αβ peptide levels in the CSF and plasma in nonhuman primates. Yan, Transl. Neurodegener. 5(13): 1-11 (2016) at page 7. E2609 did not show clinical significant safety concerns after repeated doses up to 200 mg in a phase I clinical trial. Id. After 14d dosing the Αβ peptide level reduction in the CSF was statistically significant compared to baseline (46.2% (25mg), 61.9% (50 mg), 73.8% (100 mg), 79.9% (200 mg)). Id. In November 2014, Eisai stated that a phase II dose -finding study in patients with mild cognitive impairment (MCI) due to AD or prodromal AD and a positive amyloid PET-scan will be conducted in collaboration with Biogen. Additionally, companies are also developing therapies targeting asymptomatic patients. J J-54861911, which was first developed by Shionogi & Co. in Japan and later in collaboration with Janssen, demonstrated an ability to cross the blood-brain barrier and to dose-dependently reduce Αβ peptide concentrations. Yan, Transl. Neurodegener. 5(13): 1-11 (2016) at pages 5-7. For example, an oral dose of 95 mg once daily achieved Αβ peptide reduction of up to 95% in CSF. Id. In October 2015, Janssen and Shionogi launched a phase II/III trial targeting asymptomatic subjects that are at risk for developing Alzheimer's dementia. Id.

Similarly, Amgen and Novartis announced in late 2015 a collaboration to co-develop Novartis' BACE inhibitor CNP520. Yan, Transl. Neurodegener. 5(13): 1-11 (2016) at page 8. The study is aimed at, inter alia, showing that CNP520 "can slow down the onset and progression of clinical symptoms associated with Alzheimer's disease (AD) in participants at the risk to develop clinical symptoms based on their age and genotype."

https://clinicaltrials.gov/ct2/show/NCT02565511 (last visited October 23, 2016).

The compounds disclosed herein have been shown to modulate, and specifically inhibit the activity of the β-secretase enzymes as shown in Table 2 for specific examples disclosed herein, thereby reducing the generation of Αβ peptide. Accordingly, the compounds provided herein are useful for, for example, the prevention or treatment of β- secretase related diseases, including, but not limited to, AD. The compounds provided herein have the ability to modulate the activity of the β-secretase enzyme, thereby regulating the production of Αβ peptide and reducing the formation and deposition of Αβ peptide in both the cerebral spinal fluid as well as in the brain, resulting in a decrease of Αβ plaque in the brain.

More specifically, provided are the following uses for the compounds disclosed herein:

Provided are the compounds disclosed herein for use in reducing beta amyloid peptide levels in the cerebral spinal fluid of a subject.

Provided are the compounds disclosed herein for use in treating AD, cognitive impairment, or a combination thereof in a subject. In one embodiment, the compounds provided herein are useful for treating various stages and degrees of AD, including without limitation, mild, moderate and severe AD. In another embodiment, the compounds provided herein are useful for treating preclinical AD, mild cognitive impairment (MCI) due to AD, and dementia due to AD. In yet another embodiment, the compounds provided herein may be used to treat prodromal subjects.

Provided are the compounds disclosed herein for use in treating a neurological disorder selected from mild cognitive impairment, Down's syndrome, hereditary cerebral hemorrhage with Dutch-type amyloidosis, cerebral amyloid angiopathy, degenerative dementia, dementia associated with Parkinson's disease, dementia associated with supranuclear palsy, dementia associated with cortical basal degeneration, diffuse Lewy body type of AD, or a combination thereof in a subject.

Provided are the compounds disclosed herein for use in reducing formation of plaque in the brain of a subject.

As previously discussed, in certain embodiments, the compounds described herein are to be understood to include all stereoisomers, tautomers, isotopically-labelled forms thereof or pharmaceutically acceptable salts of any of the foregoing or solvates of any of the foregoing or amorphous and crystalline forms (polymorphs) of any of the foregoing.

Accordingly, the scope of the methods and uses provided in the instant disclosure is to be understood to encompass also methods and uses employing all such forms.

Besides being useful for human treatment, the compounds provided herein may be useful for veterinary treatment of companion animals, exotic animals and farm animals, including mammals, rodents, and the like. For example, animals including horses, dogs, and cats may be treated with compounds provided herein.

DOSAGE, FORMULATION, AND ROUTE OF ADMINISTRATION

The amount of compound(s) which is/are administered and the dosage regimen for treating neurological disorders and β-secretase mediated diseases with the compounds and/or compositions disclosed herein depends on a variety of factors, including the age, weight, sex and medical condition of the subject, the type of disease, the severity of the disease, the route and frequency of administration, and the particular compound employed. A daily dose of about 0.01 to 500 mg/kg, or in some embodiments, between about 0.01 and about 50 mg/kg, and in still other embodiments between about 0.01 and about 30 mg/kg body weight may be appropriate. In yet other embodiments, a daily dose of between about 0.1 and about 10 mg/kg body weight may be appropriate and should be useful for all uses disclosed herein. The daily dose can be administered a number of times a day such as from one to four doses per day.

While it may be possible to administer a compound disclosed herein alone in the uses described, the compound administered normally will be present as an active ingredient in a pharmaceutical composition. Thus, in another embodiment, provided herein is a

pharmaceutical composition comprising a compound disclosed herein in combination with a pharmaceutically acceptable excipient, such as diluents, carriers, adjuvants and the like, and, if desired, other active ingredients. In one embodiment, a pharmaceutical composition may comprise a therapeutically effective amount of a compound disclosed herein.

The compound(s) disclosed herein may be administered by any suitable route in the form of a pharmaceutical composition adapted to such a route and in a dose effective for the treatment intended. The compounds and compositions present herein may, for example, be administered orally, mucosally, topically, rectally, pulmonarily, such as by inhalation spray, or parentally including intravascularly, intravenously, intraperitoneally, subcutaneously, intramuscularly, intrasternally, and by infusion techniques, in dosage unit formulations containing conventional pharmaceutically acceptable excipients such as carriers, adjuvants, and vehicles.

For oral administration, the pharmaceutical composition may be in the form of, for example, a tablet, capsule, suspension or liquid. The pharmaceutical composition is typically made in the form of a dosage unit containing a particular amount of the active ingredient. Examples of such dosage units are tablets or capsules. For example, these may contain an amount of active ingredient from about 1 to 2000 mg, from about 1 to 500 mg, and from about 5 to 150 mg.

For therapeutic purposes, the compounds provided herein are ordinarily combined with one or more diluents or other "excipients" appropriate to the indicated route of administration.

If orally administered on a per dose basis, the compounds provided herein may be admixed with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, gelatin, acacia gum, sodium alginate,

polyvinylpyrrolidone, and/or polyvinyl alcohol, to form the final formulation. For example, the active compound(s) and excipient(s) may be tableted or encapsulated by known and accepted methods for convenient administration. Examples of suitable formulations include, without limitation, pills, tablets, soft and hard-shell gel capsules, troches, orally-dissolvable forms and delayed or controlled-release formulations thereof. Particularly, capsule or tablet formulations may contain one or more controlled-release agents, such as

hydroxypropylmethyl cellulose, as a dispersion with the active compound(s).

Formulations for parenteral administration may be in the form of aqueous or nonaqueous isotonic sterile injection solutions or suspensions. These solutions and suspensions may be prepared from sterile powders or granules using one or more of the carriers or diluents mentioned for use in the formulations for oral administration or by using other suitable dispersing or wetting agents and suspending agents. The compounds may be dissolved in water, polyethylene glycol, propylene glycol, ethanol, corn oil, cottonseed oil, peanut oil, sesame oil, benzyl alcohol, sodium chloride, tragacanth gum, and/or various buffers. Other excipients and modes of administration are well and widely known in the pharmaceutical art. The active ingredient may also be administered by injection as a composition with suitable excipients including saline, dextrose, or water, and optionally comprising one or more of a cosolvent such as propylene glycol or emulsifier such as, for example, Tween 80. Such formulations may also include compounds such as a cyclodextrin (for example, Captisol).

The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed, including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.

The active ingredient may also be administered by injection as a composition with suitable carriers including saline, dextrose, or water. The daily parenteral dosage regimen will be from about 0.1 to about 30 mg/kg of total body weight, and in some embodiments may be from about 0.1 to about 10 mg/kg. For pulmonary administration, the pharmaceutical composition may be administered in the form of an aerosol or with an inhaler including dry powder aerosol.

The pharmaceutical compositions may be subjected to conventional pharmaceutical operations such as sterilization and/or may contain conventional excipients, such as preservatives, stabilizers, wetting agents, emulsifiers, buffers etc. Tablets and pills can additionally be prepared with enteric coatings. Such compositions may also comprise excipients, such as wetting, sweetening, flavoring, and perfuming agents. Accordingly, in yet another embodiment of the present disclosure, there is provided a method of manufacturing a medicament, the method comprising combining an amount of a compound according to Formula I with a pharmaceutically acceptable diluent to manufacture the medicament.

In yet another embodiment, the provided herein is a method of manufacturing a medicament for the treatment of AD, the method comprising combining an amount of a compound provided herein with a pharmaceutically acceptable excipient to manufacture the medicament. COMBINATIONS

While the compounds disclosed herein can be dosed or administered as the sole active pharmaceutical agent, they can also be used in combination with one or more compounds provided herein or in conjunction with other agents. When administered as a combination, the therapeutic agents can be formulated as separate compositions that are administered simultaneously or sequentially at different times, or the therapeutic agents can be given as a single composition.

The phrase "co-therapy" (or "combination-therapy"), in defining use of a compound provided herein and another pharmaceutical agent, is intended to embrace administration of each agent in a sequential manner in a regimen that will provide beneficial effects of the drug combination, and is intended as well to embrace co-administration of these agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of these active agents or in multiple, separate capsules for each agent.

Specifically, the administration of compounds provided herein may be in conjunction with additional therapies known to those skilled in the art in the prevention or treatment of β- secretase, γ-secretase and/or other reagents known in influence the formation and/or deposition of Αβ peptide, otherwise responsible for the formation of plaque in the brain. If formulated as a fixed dose, such combination products employ the compounds disclosed herein within the accepted dosage ranges. The compounds provided herein may also be administered sequentially with other known medicinal agents. This disclosure is not limited in the sequence of administration; compounds provided herein may be administered either prior to, simultaneous with or after administration of the known anti-inflammatory agent.

The foregoing description is merely illustrative and is not intended to limit the disclosure to the described compounds, compositions and methods. Variations and changes, which are obvious to one skilled in the art, are intended to be within the scope and nature of the invention, as defined in the appended claims. From the foregoing description, one skilled in the art can easily ascertain the essential characteristics of this invention, and without departing from the spirit and scope thereof, can make various changes and modifications of the invention to adapt it to various usages and conditions.

All references, for example, a scientific publication or patent application publication, cited herein are incorporated herein by reference in their entirety and for all purposes to the same extent as if each reference was specifically and individually indicated to be incorporated by reference in its entirety for all purposes.