Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SUBSTITUTED HYDROXYETHYL AMINE COMPOUNDS AS BETA-SECRETASE MODULATORS AND METHODS OF USE
Document Type and Number:
WIPO Patent Application WO/2008/147544
Kind Code:
A1
Abstract:
The present invention comprises a new class of compounds useful for the modulation of Beta-secretase enzyme activity and for the treatment of Beta-secretase mediated diseases, including Alzheimer's disease (AD) and related conditions. In one embodiment, the compounds have a general Formula I wherein R1a, R1b, R1c, B, W, R3, R4 and R5 are defined herein. In another embodiment, the invention provides compounds of general Formula II wherein A1, A2, A3, A4, R1a, R1b, R1c, R2, R4, R5, W, X and Z are defined herein. The invention also includes use of these compounds in pharmaceutical compositions for treatment, prophylactic or therapeutic, of disorders and conditions related to the activity of beta-secretase protein. Such disorders include, for example, Alzheimer's Disease (AD), cognitive deficits and impairment, schizophrenia and other similar central nervous system conditions. The invention also comprises further embodiments of Formula II, intermediates and processes useful for the preparation of compounds of Formulas I and II.

Inventors:
ZHONG WENGE (US)
HITCHCOCK STEPHEN (US)
PATEL VINOD F (US)
CROGHAN MICHAEL (US)
DINEEN THOMAS (US)
HORNE DANIEL (US)
KALLER MATTHEW (US)
KREIMAN CHARLES (US)
LOPEZ PATRICIA (US)
MONENSCHEIN HOLGER (US)
NGUYEN THOMAS (US)
PENNINGTON LEWIS (US)
XUE QIUFEN (US)
YANG BRYANT (US)
Application Number:
PCT/US2008/006634
Publication Date:
December 04, 2008
Filing Date:
May 23, 2008
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
AMGEN INC (US)
ZHONG WENGE (US)
HITCHCOCK STEPHEN (US)
PATEL VINOD F (US)
CROGHAN MICHAEL (US)
DINEEN THOMAS (US)
HORNE DANIEL (US)
KALLER MATTHEW (US)
KREIMAN CHARLES (US)
LOPEZ PATRICIA (US)
MONENSCHEIN HOLGER (US)
NGUYEN THOMAS (US)
PENNINGTON LEWIS (US)
XUE QIUFEN (US)
YANG BRYANT (US)
International Classes:
A61K31/438; C07D221/06; A61P25/28; C07D311/96; C07D471/10; C07D491/10; C07D493/10; C07D495/10
Domestic Patent References:
WO2005087714A22005-09-22
WO2005070407A12005-08-04
WO2005095326A22005-10-13
WO2007061670A12007-05-31
WO2007062007A12007-05-31
WO2007061930A12007-05-31
WO2000017369A22000-03-30
WO2003045913A12003-06-05
WO2004043916A12004-05-27
WO2003002122A12003-01-09
WO2003006021A12003-01-23
WO2003002518A12003-01-09
WO2004024081A22004-03-25
WO2003040096A22003-05-15
WO2004050619A12004-06-17
WO2004080376A22004-09-23
WO2004099376A22004-11-18
WO2005004802A22005-01-20
WO2004080459A12004-09-23
WO2004062625A22004-07-29
WO2004042910A22004-05-21
WO2005004803A22005-01-20
WO2005005374A12005-01-20
WO2003106405A12003-12-24
WO2003062209A22003-07-31
WO2003030886A22003-04-17
WO2002002505A22002-01-10
WO2001070671A22001-09-27
WO2003057721A22003-07-17
WO2003006013A12003-01-23
WO2003037325A12003-05-08
WO2004094384A22004-11-04
WO2004094413A12004-11-04
WO2003006423A12003-01-23
WO2003050073A12003-06-19
WO2003029169A22003-04-10
WO2004000821A12003-12-31
Other References:
LARAS, YOUNES ET AL: "Synthesis of new substituted 4,5-dihydro-3H-spiro[1,5]-benzoxazepine-2,4'- piperidine and biological properties", AUSTRALIAN JOURNAL OF CHEMISTRY , 59(11), 812-818 CODEN: AJCHAS; ISSN: 0004-9425, 2006, XP008096337
SINHA ET AL., NATURE, vol. 402, 1999, pages 537 - 554
SABBAGH, M. ET AL., ALZ. DIS. REV., vol. 3, 1997, pages 1 - 19
LUO ET AL., NATURE NEUROSCIENCE, vol. 4, 2001, pages 231 - 232
Attorney, Agent or Firm:
REDDY, G. Prabhakar (Patent Operations / 28-2-CThousand Oaks, California, US)
Download PDF:
Claims:
A-1291-WO-PCT - 161 -

What is claimed' is:

1. A compound of Formula I:

π or a pharmaceutically acceptable salt thereof, wherein each of A 1 and A 2 , independently, is CH or CR 6 ; one of A 3 and A 4 , independently, is CH or CR 3 and the other of A 3 and A 4 , independently, is N, CH or CR 6 ;

R la is H, halo, haloalkyl, C w -alkyl, -NH-C 1-6 -alkyl, - N-di-C 1-6 -alkyl, CN, OH or NH 2 , wherein the portion of -O-C^-alkyl, -S-C 1-6 - alkyl, -NH-Ci -6 -alkyl and -N-di-C^-alkyl are optionally substituted independently with 1-5 substituents of R 7 -;

R lb is H, halo, haloalkyl, C 1-6 -alkyl, -O-d-e-alkyl, -S-C^-alkyl, -NH-C 1-6 -alkyl, - N-di-C^-alkyl, CN, OH or NH 2 , wherein the C 1-6 -alkyl portion of -O-C 1-6 -alkyl, -S-Ci -6 - alkyl, -NH-Ci -6 -alkyl and -N-di-C 1-6 -alkyl are optionally substituted independently with 1-5 substituents of R 7 ; alternatively, R la and R lb taken together with the carbon atom to which they are attached form a partially or fully saturated 3-, A-, 5- or 6-membered ring of carbon atoms optionally including 1-2 heteroatoms selected from O, N, or S, the ring optionally substituted independently with 1-3 substituents of R 7 ;

R lc is H, halo, haloalkyl, -NH-C 1-6 -alkyl, - N-di-C 1-6 -alkyl, CN, OH OrNH 2 ;

W is -C(O)-, -C(=S>, - OC(=O) -, -NHC(=O)-, -S(=O)b- or -NHS(K)V, wherein b is 1 or 2;

R 2 is C 1 -C 3 alkyl, -OCi-C 3 alkyl, C 2 -C 3 alkenyl or C 2 -C 3 alkynyl, wherein each of said C 1 -C 3 alkyl, -OC]-C 3 alkyl, C 2 -C 3 alkenyl and C 2 -C 3 alkynyl is optionally substituted independently with 1-3 substituents of R 7 ;

R 3 is -NH-partially or fully unsaturated 5- or 6-membered monocyclic ring formed of carbon atoms wherein said ring optionally including 1-3

A-1291-WO-PCT - 162 - heteroatoms selected from O, N, or S and optionally substituted with 1-5 substituents of R 7 , -NH-l,3-benzodiox-5-yl optionally substituted with 1-3 substituents of R 7 , 1,3- benzodiox-5-yl optionally substituted with 1-3 substituents of R 7 or partially or fully unsaturated 5- or 6-membered monocyclic ring formed of carbon atoms wherein said ring optionally including 1-3 heteroatoms selected from O, N, or S and optionally substituted with 1-5 substituents of R 7 ;

R 4 is H, halo or CWalkyl;

R 5 is H, halo, haloalkyl, oxo, d-e-alkyl, -NH-C ]-6 alkyl, -N-di-Ci -6 -alkyl, CN, OH or NH 2 , wherein the C^-alkyl and the C 1-6 -alkyl portion -NH-Ci- 6 -alkyl and -N-di-Ci-β-alkyl are optionally substituted independently with 1-5 substituents of R 7 ;

X is CH 2 , CHR 6 , CR 6 R 6 , C(=O), O, NH, NR 6 , or S(O) 0 wherein o is O, 1 or 2; Z is a 3-6 membered spirocyclic ring formed of carbon atoms optionally including 1-3 heteroatoms selected from O, N and S and optionally substituted independently with 1-5 substituents of R 7 ; each R 6 , independently, is halo, haloalkyl, C ]-6 -alkyl, CN, OR 7 , NHR 7 , -S-Ci- 6 -alkyl, -N-di-Ci -6 -alkyl, C 2-6 -alkenyl, C 2-6 -alkynyl, C 3-8 -CyClOaIlCyI or C 4-8 - cycloalkenyl, wherein the C 2-6 -alkenyl, C 2-6 -alkynyl, C 3-8 -CyClOaIlCyI, C 4-8 - cycloalkenyl and the Ci- 6 -alkyl portion and are optionally substituted with 1-5 substituents of R 7 ; or R 6 is a fully saturated or partially or fully unsaturated 5- or 6-membered monocyclic or bicyclic ring formed of carbon atoms, said ring optionally including 1-4 heteroatoms selected from O, N, or S and optionally substituted with one or more substituents of R 7 ; and each R 7 , independently, is H, halo, haloalkyl, CN, OH, NO 2 , NH 2 , acetyl, oxo, C 1- l o-alkyl, C 2- i 0 -alkenyl, C 2- i 0 -alkynyl, C 3- io-cycloalkyl, C 4 .i 0 -cycloalkenyl, C M0 - alkylamino-, Cj.io-dialkylamino-, Ci.io-alkoxyl, Ci.io-thioalkoxyl or a fully saturated or partially or fully unsaturated 3-8 membered monocyclic or a 6-12 membered bicyclic, said ring system formed of carbon atoms optionally including 1-3 heteroatoms if monocyclic or 1-6 heteroatoms if bicyclic, said heteroatoms selected from O, N, or S, wherein each of the C 2- io-alkenyl, C 2- io-alkynyl, C 4-10 - cycloalkenyl, Ci.io-alkylamino-, Ci.io-dialkylamino-, Ci.io-alkoxyl, Ci.io-thioalkoxyl and ring of said ring system is optionally substituted independently with 1-5 substituents of halo, haloalkyl, CN, NO 2 , NH 2 , OH, oxo, methyl, methoxyl, ethyl, ethoxyl, propyl,

A- 1291 -WO-PCT - 163 - propoxyl, isopropyl, isopropoxyl, cyclopropyl, cyclopropylmethoxyl, butyl, butoxyl, isobutoxyl, tert-butoxyl, isobutyl, sec-butyl, tert-butyl, cyclobutyl, pentyl, cyclopentyl, hexyl, cyclohexyl, Ci.io-alkylamino-, Ci-io-dialkylamino-, Ci.i 0 -thioalkoxyl, benzyl or phenyl; provided the compound is not

N-((lS)-l-((lR)-2-(((4 I S)-6'-(2,2-dimethylpropyl)-3',4'-dihydrospiro[cyclobutane- l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-3-methylbutyl)acetamide;

N-((lS)-l-((lR)-2-(((4'S)-6 I -(2,2-dimethylpropyl)-3',4'-dihydrospiro[cyclobutane- 1 ,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l -hydroxyethyl)-3 -methyl-3 -buten-1 - yl)acetamide;

N-(( 1 S)- 1 -(( lR)-2-(((4'S)-6'-(2,2-dimethylpropyl)-3 ',4'-dihydrospiro[cyclobutane- l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-3-pentyn-l-yl)acetamide;

N-((lS)-l-((lR)-2-(((4'S)-6'-(2,2-dimethylpropyl)-3',4 1 -dihydrospiro[cyclobutane- 1 ,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l -hydroxyethyl)-4-penten-l -yl)acetamide; N-((lS)-l-((lR)-2-(((ls,3R,4'S)-6'-(2,2-dimethylpropyl)-3-methyl-3',4 I - dihydrospiro[cyclobutane-l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-4- penten- 1 -yl)acetamide;

N-((lS,2R)-3-(((4S)-6-ethyl-3,4-dihydrospiro[chromene-2,l'-cyclobutan]-4- yl)amino)-2-hydroxy- 1 -((methyloxy)methyl)propyl)acetamide; N-((lS,2R)-3-(((4S)-6-ethyl-3,4-dihydrospiro[chromene-2,l'-cyclobutan]-4- yl)amino)-2-hydroxy- 1 -((propyloxy)methyl)propyl)acetamide;

N-((lS,2R>3-(((4S)-6-ethyl-3,4-dihydrospiro[chromene-2,l'-cyclobutan]-4- yl)amino)-2 -hydroxy- 1 -(((phenylmethyl)oxy)methyl)propyl)acetamide;

N-((lS)-l-((lR)-2-(((4'S)-6 I -(2,2-dimethylpropyl)-3',4 I -dihydrospiro[cyclobutane- l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-3-buten-l-yl)acetamide;

N-((lS)-l-((lS)-2-(((4'S)-6 I -(2,2-dimethylpropyl)-3 1 ,4 l -dihydrospiro[cyclobutane- 1 ,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)- 1 -hydroxyethyl)-3-buten- 1 -yl)acetamide;

N-((lS)-l-((lR)-2-(((4 I S)-6 1 -(2,2-dimethylpropyl)-3 l ,4'-dihydrospiro[cyclobutane- l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-4,4,4-trifluorobutyl)acetamide; N-(( 1 S,3E)- 1 -(( 1 R)-2-(((4'S)-6'-(2,2-dimethylpropyl)-3 1 ,4'- dihydrospiro[cyclobutane-l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-3- penten-1 -yl)acetamide;

N-((lS)-l-((lR)-2-(((4'S)-6'-(2,2-dimethylpropyl)-3 1 ,4'-dihydrospiro[cyclobutane- l,2'-pyrano[2,3-b]pyridin]-4 1 -yl)amino)-l-hydroxyethyl)-2-propen-l-yl)acetamide;

A- 1291 -WO-PCT - 164 -

N-(( 1 S)- 1 -(( 1 R)-2-(((4'S)-6'-(2-fluoro-2-methylpropyl>3 ',4'- dihydrospiro[cyclobutane- 1 ,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)- 1 -hydroxyethyl)-3- buten-l-yl)acetamide; and

N-((lS)-l-((lR)-2-(((4'S)-6 t -(2,2-dimethylpropyl)-3',4'-dihydrospiro[cyclobutane- l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-3-hexyn-l-yl)acetamide.

2. The compound of Claim 1, or a pharmaceutically acceptable salt thereof, wherein R la is C^-alkyl, C 2-6 alkenyl, C 2-6 -alkynyl, -O-C 1-6 -alkyl-, -S-C^-alkyl-, -NH-Ci- 6 -alkyl-, -O-C 1-6 -alkenyl-, -S-C 2-6 -alkenyl-, -NH-Ca^-alkenyl-, -O-C 1-6 -alkynyl-, -S-Ci- 6 alkynyl-, -NH-Ci-e-alkynyK -C M alkyl-O-Ci .,-alkyl-, -C M -alkyl-S-C M -alkyl- or -C 1-4 - alkyl-NH-C M -alkyl-;

R lb is H, halo, haloalkyl, C w -alkyl, -O-Ci -6 -alkyl, -S-C^-alkyl, -NH-C 1-6 -alkyl, - N-di-Cβ-alkyl, CN, OH OrNH 2 ; and

R lc is H.

3. The compound of Claim 1, or a pharmaceutically acceptable salt thereof, wherein R la and R lb taken together with the carbon atom to which they are attached form a partially or fully saturated 4-, 5- or 6-membered ring of carbon atoms optionally including 1-2 heteroatoms selected from O, N, or S, the ring optionally substituted independently with 1-3 substituents of R 7 and R lc is H.

4. The compound of Claim 1, or a pharmaceutically acceptable salt thereof, wherein m is 1 and R 3 is a ring selected from phenyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, triazinyl, thiophenyl, furyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, thiazolyl, oxazolyl, isoxazolyl, isothiazolyl, thiadiazolyl, oxadiazolyl, pyrrolidinyl, oxazolinyl, isoxazolinyl, thiazolinyl, pyrazolinyl, morpholinyl, piperidinyl, piperazinyl and pyranyl, said ring optionally substituted with 1-5 substituents of R 7 .

5. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein X is -O-; and ring Z is

A- 1291 -WO-PCT - 165 -

wherein R 7 is as defined in claim 1; and p is 0, 1, 2, 3 or 4.

6. The compound of Claim 1, or a pharmaceutically acceptable salt thereof, wherein

A 1 is CH;

A 2 is CR 6 ; each of A 3 and A 4 , independently, is CH or CR 6 or N, provided no more than one ofA 3 and A 4 is N;

R la ϊs Ci-e-alkyl, C 2-6 alkenyl, C 2 - 6 -alkynyl, C^alkyl-O-C^-alkyl-, C 1-6 -alkyl-S- C 1-3 -alkyK C 1-6 -alkyl-S(O) 2 -Ci -3 -alkyl-, C L e-alkyl-NH-C^-alkyl, di-(C 1-3 -alkyl)-N-C 1-3 - alkyl, C 2-4 -alkenyl-S-C 1-3 -alkyl-, C 2-4 -alkenyl-NH-Ci -3 -alkyl- or C 2 - 4 -alkynyl-NH-Ci -3 -alkyl-, wherein the alkyl, alkenyl or alkynyl moiety of each is optionally substituted with 1-3 substituents of R 7 ;

R lb is H, halo, haloalkyl, C^-alkyl, -O-C w -alkyl, -S-C^-alkyl, -NH-C^-alkyl, - N-di-d-6-alkyl, CN, OH OrNH 2 ; alternatively, R la and R lb taken together with the carbon atom to which they are attached form a partially or fully saturated 5- or 6-membered ring of carbon atoms optionally including 1-2 heteroatoms selected from O, N, or S, the ring optionally substituted independently with 1-3 substituents of R 7 ;

R lc is H;

W is -C(=O)-;

R 2 is Ci-C 3 alkyl, -OCi-C 3 alkyl, C 2 -alkenyl or C 2 -alkynyl, wherein each of said Ci-C 3 alkyl, C 2 -alkenyl and C 2 -alkynyl is optionally substituted independently with 1-2 substituents of R 7 ;

A-1291-WO-PCT - 166 -

R 3 is a ring selected from phenyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, triazinyl, thiophenyl, furyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, thiazolyl, oxazolyl, isoxazolyl, isothiazolyl, thiadiazolyl, oxadiazolyl, pyrrolidinyl, oxazolinyl, isoxazolinyl, thiazolinyl, pyrazolinyl, morpholinyl, piperidinyl, piperazinyl and pyranyl, said ring optionally substituted with 1-5 substituents of R 7 ;

R 4 is H or C M -alkyl;

R 5 is H, halo, haloalkyl, C, -6 -alkyl, -NH-C 1-6 -alkyl, -N-di-C^-alkyl, CN 3 OH OrNH 2 ;

X is CH 2 , CHR 6 , C(=O) or O; Z is a cyclopropyl, cyclobutyl or cyclopentyl ring wherein 0, 1 or 2 carbon atoms of the ring are, independently, replaced with an oxygen atom and the ring optionally substituted independently with 1-5 substituents of R 7 ; and each R 6 , independently, is halo, haloalkyl, -O-Ci -6 -alkyl, -S-Ci- 6 - alkyl, -NH-Ci -6 -alkyl, -N-di-C 1-6 -alkyl, CN, OR 7 , NHR 7 , C 2 ^-alkenyl, C 2-6 -alkynyl, C 3-8 - cycloalkyl, or a ring selected from phenyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, triazinyl, thiophenyl, furyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, thiazolyl, oxazolyl, isoxazolyl, isothiazolyl, thiadiazolyl, oxadiazolyl, pyrrolidinyl, oxazolinyl, isoxazolinyl, thiazolinyl, pyrazolinyl, morpholinyl, piperidinyl, piperazinyl and pyranyl, wherein the Ci- 6 -alkyl, C 2 ^-alkenyl, C 2 - 6 -alkynyl, C 3-8 - cycloalkyl, C 4- g-cycloalkenyl, ring and the C ^-alkyl portion -S-Q^- alkyl, -NH-Ci -6 -alkyl and -N-di-Ci- 6 -alkyl are optionally substituted with 1-5 substituents ofR 7 .

7. The compound of claim 1, or a pharmaceutically acceptable salt thereof, having a general Formula II-A:

wherein

A-1291-WO-PCT - 167 - one of A 3 and A 4 , independently, is CR 3 and the other of A 3 and A 4 , independently, is N, CH or CR 6 ;

R la is H, halo, haloalkyl, C^-alkyl, -O-C 1-6 -alkyl, -S-C 1-6 -alkyl, -NH-C 1-6 -alkyl, - and the portion of -O- -S-Ci -6 -alkyl, -NH-Ci-β-alkyl and are optionally substituted independently with 1-5 substituents of R 7 ;

R lb is H, halo, haloalkyl, C 1-6 -alkyl, -O-C 1-6 -alkyl, - N-di-C^-alkyl, CN, OH or NH 2 , wherein the C 1-6 -alkyl and the C 1-6 -alkyl portion of -O- Q- 6 -alkyl, -S-C] -6 -alkyl, -NH-C 1-6 -alkyl and -N-di-C ]-6 -alkyl are optionally substituted independently with 1-5 substituents of R 7 ; alternatively, R la and R lb taken together with the carbon atom to which they are attached form a partially or fully saturated A-, 5- or 6-membered ring of carbon atoms optionally including 1-2 heteroatoms selected from O, N, or S, the ring optionally substituted independently with 1-3 substituents of R 7 ; R lc is H, F, Cl, methyl, ethyl, methoxyl, ethyoxyl, CN, OH or NH 2 ;

R 2 is C J -C 3 alkyl, -OC]-C 3 alkyl, C 2 -alkenyl or C 2 -alkynyl, wherein each of said Ci-C 3 alkyl, -OCi-C 3 alkyl, C 2 -alkenyl and C 2 -alkynyl is optionally substituted independently with 1-2 substituents of R 7 ;

R 3 is a ring selected from phenyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, triazinyl, thiophenyl, furyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, thiazolyl, oxazolyl, isoxazolyl, isothiazolyl, thiadiazolyl, oxadiazolyl, pyrrolidinyl, oxazolinyl, isoxazolinyl, thiazolinyl, pyrazolinyl, moφholinyl, piperidinyl, piperazinyl and pyranyl, said ring optionally substituted with 1-5 substituents of R 7 ;

R 4 is H, halo or C M -alkyl; R 5 is H, F, Cl, CF 3 , C^-alkyl, -O-C^-alkyl, -S-C M -alkyl, -NH-d- 4 -alkyl,

CN, OH OrNH 2 , wherein the C M -alkyl and the C M -alkyl portion of -O-C ]-4 -alkyl, -S-Ci- 4 -alkyl and -N-C M -alkyl are optionally substituted independently with 1-3 substituents of halo or OH; each R 6 , independently, is halo, haloalkyl, -S-Ci^- alkyl, -NH-C 1-6 -alkyl, -N-di-C^-alkyl, CN, OR 7 , NHR 7 , C 2-6 -alkenyl, C 2-6 -alkynyl, C 3-8 - cycloalkyl or C 2-6 -alkenyl, C 2-6 -alkynyl, C 3-8 - cycloalkyl, C 4-8 -cycloalkenyl and the portion - and -N-di-Ci- 6 -alkyl are optionally substituted with 1-5 substituents of R 7 ;

A-1291-WO-PCT - 168 - each R 7 , independently, is H, halo, haloalkyl, CN, OH, NO 2 , NH 2 , acetyl, oxo, Ci- 10 -alkyl, C 2- io-alkenyl, C 2- io-alkynyl, C 3 -io-cycloalkyl, C 4 . 10 -cycloall.enyl, Ci.jo- alkylamino-, Ci.io-dialkylamino-, Ci.io-alkoxyl, Ci.io-thioalkoxyl or a fully saturated or partially or fully unsaturated 3-8 membered monocyclic or a 6-12 membered bicyclic, said ring system formed of carbon atoms optionally including 1-3 heteroatoms if monocyclic or 1-6 heteroatoms if bicyclic, said heteroatoms selected from O, N, or S, wherein each of the Ci.io-alkyl, C 2- io-alkenyl, C 2 .i 0 -alkynyl, C 3 .i 0 -cycloalkyl, C 4-I0 - cycloalkenyl, Ci.io-alkylamino-, Ci- 10 -dialkylamino-, Ci.io-alkoxyl, Ci.io-thioalkoxyl and ring of said ring system is optionally substituted independently with 1-5 substituents of halo, haloalkyl, CN, NO 2 , NH 2 , OH, oxo, methyl, methoxyl, ethyl, ethoxyl, propyl, propoxyl, isopropyl, isopropoxyl, cyclopropyl, cyclopropylmethoxyl, butyl, butoxyl, isobutoxyl, tert-butoxyl, isobutyl, sec-butyl, tert-butyl, cyclobutyl, pentyl, cyclopentyl, hexyl, cyclohexyl, Ci.io-alkylamino-, Ci.io-dialkylamino-, Ci.io-thioalkoxyl, benzyl or phenyl; and p is O, l or 2.

8. The compound of Claim 7, or a pharmaceutically acceptable salt thereof, wherein

R la is C 1-6 -alkyl, C 1-6 -alkyl-S- C,. 3 -alkyl-, alkyl, C 2 - 4 -alkenyl-S-C 1-3 -alkyl-, C 2-4 -alkenyl-NH-C 1-3 -alkyl- or wherein the alkyl, alkenyl or alkynyl moiety of each is optionally substituted with 1-3 substituents of R 7 ;

R lb is H, halo, haloalkyl, C ]-6 -alkyl, -O-C 1-6 -alkyl, -S-C ]-6 -alkyl, -NH-C^-alkyl, - N-di-Ci- 6 -alkyl, CN, OH or NH 2 ; alternatively, R la and R lb taken together with the carbon atom to which they are attached form a partially or fully saturated 5- or 6-membered ring of carbon atoms optionally including 1-2 heteroatoms selected from O, N, or S, the ring optionally substituted independently with 1-3 substituents of R 7 ; R lc is H;

9. The compound of Claim 1, and pharmaceutically acceptable salts thereof, selected from:

A-1291-WO-PCT - 169 -

N-((lS,2R)-l-(2-cyclobutylideneethyl)-3-(((4'S)-6'-(2,2-dimethylpropyl)-3 1 ,4 t - dihydrospiro[cyclobutane-l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-2- hydroxypropyl)acetamide;

N-((lS)-l-((lR)-2-(((2r,3'R,4S)-6-(2,2-dimethylpropyl)-3'-hydroxy-3,4- dihydrospiro[chromene-2, 1 '-cyclobutan]-4-yl)amino)- 1 -hydroxyethyl)-3 -buten- 1 - yl)acetamide;

N-((lS)-l-((lR)-2-(((4'S)-6'-(2,2-difluoropropyl)-3 t ,4'-dihydrospiro[cyclobutane- 1 ,2'-pyrano[2,3 -b]pyridin]-4'-yl)amino)- 1 -hydroxyethyl)-3 -buten- 1 -yl)-2- (methyloxy)acetamide; N-((lS)-l-((lR)-l-hydroxy-2-(((4 t S)-6'-((2R)-3,3,3-trifluoro-2-methylpropyl)-

3',4'-dihydrospiro[cyclobutane-l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)ethyl)-3-buten-l- yl)acetamide;

N-(( 1 S)- 1 -(( 1 R)-2-(((4'S)-6'-(2,2-dimethylpropyl)-3',4 l -dihydrospiro[cyclobutane- l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-3-buten-l-yl)-2,2- difluoropropanamide;

(2R)-N-((lS)-l-((lR)-l-hydroxy-2-(((4S)-8-(lH-imidazol-l-yl)-6-(2- methylpropyl)-3 ,4-dihydrospiro[chromene-2, 1 '-cyclobutan]-4-yl)amino)ethyl)-3 -buten- 1 - yl)-2-methoxypropanamide;

N-((lS)-l-((lR)-l-hydroxy-2-(((4S)-6-(2-methylpropyl)-8-(l,3-thiazol-2-yl)-3,4- dihydrospiro[chromene-2,r-cyclobutan]-4-yl)amino)ethyl)-3-buten-l-yl)-2- methoxyacetamide;

N-((lS)-l-((lR)-2-(((4S)-6-(2,2-dimethylpropyl)-8-(lH-imidazol-l-yl)-3,4- dihydrospiro[chromene-2, 1 '-cyclobutan]-4-yl)amino)- 1 -hydroxyethyl)-3 -buten- 1 -yl)-2- methoxyacetamide; N-((lS)-l-((lR)-2-(((4S)-6-(2,2-dimethylpropyl)-8-(lH-imidazol-5-yl)-3,4- dihydrospiro[chromene-2,r-cyclobutan]-4-yl)amino)-l-hydroxyethyl)-3-buten-l-yl)-2- methoxyacetamide;

N-((lS)-l-((lR)-2-(((4'S)-6'-(2,2-dimethylpropyl)-8'-(methylamino)-3 1 ,4 1 - dihydrospiro[cyclobutane- 1 ,2'-pyrano[2,3-c]pyridin]-4'-yl)amino)-l -hydroxyethyl)-3- butyn-l-yl)acetamide;

(2R)-N-((lS)-l-((lR)-2-(((4 I S)-6 I -(2,2-dimethylpropyl)-8 t -(methylamino)-3',4 1 - dihydrospiro[cyclobutane- 1 ,2'-pyrano[2,3-c]pyridin]-4'-yl)amino)- 1 - hydroxyethyl)butyl)tetrahydro-2-furancarboxamide;

A-1291-WO-PCT - 170 -

N-((lS)-l-((lR)-2-(((4 I S)-6'-(2,2-dimethylpropyl)-8'-(phenylamino)-3 I ,4 I - dihydrospiro[cyclobutane-l,2'-pyrano[2,3-c]pyridin]-4'-yl)amino)-l-hydrojQ'ethyl)-3- buten- 1 -yl)acetamide;

. N-((lS)-l-((lR)-2-(((4S)-8-(l,3-benzodioxol-5-yl)-6-(2,2-dimethylpropyl)-3,4- dihydrospiro[chromene-2, 1 '-cyclobutan]-4-yl)amino)- 1 -hydroxyethyl)-3 -buten- 1 -yl)-2- methoxyacetamide;

N-^lSH-CClR^-C^'S^S'-Cl^-benzodioxol-S-ylamino)^'^^- dimethylpropyl)-3',4'-dihydrospiro[cyclobutane-l,2'-pyrano[2,3-c]pyridin]-4'-yl)amino)- 1 -hydroxyethyl)-3 -buten- 1 -yl)-2-methoxyacetamide; and N-((lS)-l-((lR)-2-(((4'S)-6'-(2,2-dimethylpropyl)-8'-(l-methyl-lH-imidazol-5- yl)-3',4'-dihydrospiro[cyclobutane-l,2'-pyrano[2,3-c]pyridin]-4'-yl)amino)-l- hydroxyethyl)-3-buten-l-yl)-2-methoxyacetamide.

10. A pharmaceutical composition comprising a compound according to any of claim 1-9 and pharmaceutically acceptable excipient.

11. Use of a compound according to any of claims 1 -9 for the manufacture of a medicament for the treatment of Alzheimer's disease in a subject.

12. Use of a compound according to any of claims 1-9 for the manufacture of a medicament for reducing the levels of beta amyloid peptide in the cerebral spinal fluid or in the brain of a subject.

13. Use of a compound according to any of claims 1 -9 for the manufacture of a medicament for reducing the formation of plaque on the brain of a subject.

14. Use of a compound according to any of claims 1-9 for the manufacture of a medicament for the treatment of 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.

A-1291-WO-PCT - 171 -

15. A method of preparing a compound of Claim 1, the method comprising the step of reacting a compound 20-A

20-A

,wherein A . 1 , A A 2, A A 3, A A 4, R D 2, T R>4, R , X and Z are as defined in claim 1, with a compound

having the structure , wherein R la , R lb , R lc and W are as defined in claim 1 and X is a leaving group selected from a halogen and an HOBt ester, to prepare the compound of claim 1.

Description:

A-1291 -WO-PCT - 1 - Express Mail No. EL732551922US

SUBSTITUTED HYDROXYETHYL AMINE COMPOUNDS AS BETA- SECRET ASE MODULATORS AND METHODS OF USE

This application claims the benefit of U.S. Provisional Application No. 60/931 ,824, filed 25 May 2007 which specification is hereby incorporated here in by reference in its entirety.

FIELD OF THE INVENTION

The invention relates generally to pharmaceutically active compounds, pharmaceutical compositions and methods of use thereof, to treat Beta-Secretase mediated diseases and conditions, including, without limitation, Alzheimer's disease, plaque formation on the brain and related disorders.

BACKGROUND OF THE INVENTION Alzheimer's disease (AD) affects greater than 12 million aging people worldwide. AD accounts for the majority of dementia 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, typically leading to a diminished cognition of self, family and friends. 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 effectively treat AD 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 peptide (A-beta), or A-beta fragments thereof, deposits in the brain (commonly referred to as beta amyloid "plaques" or "plaque deposits") and in cerebral blood vessels (beta amyloid angiopathy). The second change in AD is the formation of intraneuronal tangles, consisting of an aggregate form of the protein tau. Amyloid plaques are thought to be specific for AD, while intraneuronal tangles are also found in other dementia-inducing disorders. Joachim et al., AIz. Dis. Assoc. Dis., 6:7-34 (1992).

A-1291 -WO-PCT - 2 -

Several lines of evidence indicate that progressive cerebral deposition of A-beta plays a seminal role in the pathogenesis of AD and can precede cognitive symptoms by years or even decades. Selkoe, Neuron, 6:487 (1991). Release of A-beta from neuronal cells grown in culture and the presence of A-beta 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 these 2 factors 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 A-beta formation is a causative precursor or factor in the development of AD. More specifically, deposition of A-beta in areas of the brain responsible for cognitive factors is believed to be a major factor in the development of AD. Beta amyloid plaques are primarily composed of amyloid beta peptide (A-beta peptide). A-beta peptide is derived from the proteolytic cleavage of a large transmembrane amyloid precursor protein (APP), and is a peptide ranging in about 39-42 amino acid residues. A-beta 42 (42 amino acids long) is thought to be the major component of these plaque deposits. Citron, Trends in Pharmacological Sciences, 25(2):92-97 (2004).

Several proteases are thought to be involved in the processing or cleavage of APP, resulting in the formation of A-beta peptide. Beta secretase (BACE, also commonly referred to as memapsin) is thought to first cleave APP to generate two fragments: (1) a first N-terminus fragment (beta APP) and (2) a second C-99 fragment, which is subsequently cleaved by gamma secretase to generate the A-beta peptide. APP has also found to be cleaved by alpha-secretase to produce alpha-sAPP, a secreted form of APP that does not result in beta-amyloid plaque formation. This alternate pathway precludes the formation of A-beta peptide. A decription 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 beta-secretase specific cleavage site. BACE is present in two forms, BACE 1 and BACE 2, designated as such depending upon the

A- 1291 -WO-PCT - 3 - specifϊc cleavage site of APP. Beta secretase is described in Sinha et al., Nature, 402:537- 554 (1999) (p510) and PCT application WO 2000/17369. It has been proposed that A- beta peptide accumulates as a result of APP processing by BACE. Moreover, in vivo processing of APP at the beta secretase cleavage site is thought to be a rate-limiting step in A-beta production. Sabbagh, M. et al., AIz. Dis. Rev. 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. BACEl knockout mice have failed to produce A-beta. When crossed with transgenic mice that over express APP, the progeny show reduced amounts of A-beta in brain extracts as compares with control animals (Luo et al., Nature Neuroscience, 4:231 - 232 (2001)). This evidence further supports the concept that inhibition of beta secretase activity and a corresponding reduction of A-beta in the brain should provide a therapeutic method for treating AD and other beta amyloid or plaque related disorders.

Several approaches have been taken to potentially treat AD and plaque-related disorders. One approach has been to attempt to reduce the formation of plaque on the brain, by inhibiting or reducing the activity of BACE. For example, each of the following PCT publications: WO 03/045913, WO 04/043916, WO 03/002122, WO 03/006021, WO 03/002518, WO 04/024081, WO 03/040096, WO 04/050619, WO 04/080376, WO 04/099376, WO 05/004802, WO 04/080459, WO 04/062625, WO 04/042910, WO 05/004803, WO 05/005374, WO 03/106405, WO 03/062209, WO 03/030886, WO 02/002505, WO 01/070671, WO 03/057721, WO 03/006013, WO 03/037325, WO 04/094384, WO 04/094413, WO 03/006423, WO 03/050073, WO 03/029169 and WO 04/000821, describe inhibitors of BACE, useful for treating AD and other beta-secretase mediated disorders.

BRIEF DESCRIPTION OF THE INVENTION The present invention provides a new class of compounds useful for the modulation of beta secretase activity. To that end, the compounds of the invention are useful for the regulation or reduction of the formation of A-beta peptide and, consequently, the regulation and/or reduction of beta amyloid plaque formation on the brain. Accordingly, the compounds are useful for the treatment of Alzheimer's disease and other beta secretase and/or plaque mediated disorders. For example, the compounds are useful for the prophylaxis and/or treatment, acute and/or chronic, of AD and other

A-1291 -WO-PCT - 4 - diseases or conditions involving the deposition or accumulation of beta amyloid peptide, and formation of plaque, on the brain.

The compounds provided by the invention, including stereoisomers, tautomers, solvates, pharmaceutically acceptable salts, derivatives or prodrugs thereof, are generally defined by Formula I

I wherein R la , R lb , R lc , B, W, R 3 , R 4 and R 5 are as described below. In another embodiment, the invention provdes compounds of general Formula II

wherein A 1 , A 2 , A 3 , A 4 , R la , R lb , R lc , R 2 , R 4 , R 5 , W, X and Z are defined herein.

The invention also provides procedures for making compounds of Formulas I and II, and any sub-Formulas thereof, as well as intermediates useful in such procedures. The invention further provides pharmaceutical compositions, which comprise one or more compounds of the invention, methods for the treatment of beta secretase mediated diseases, such as AD, using the compounds and compositions of the invention. For example, and in one embodiment, the invention provides a pharmaceutical composition comprising an effective dosage amount of a compound of Formula I in association with at least one pharmaceutically acceptable excipient.

The foregoing merely summarizes certain aspects of the invention and is not intended, nor should it be construed, as limiting the invention in any way. All patents and other publications recited herein are hereby incorporated by reference in their entirety.

DETAILED DESCRIPTION OF THE INVENTION

A-1291-WO-PCT - 5 -

In one embodiment of the invention, the compounds, including stereoisomers, tautomers, solvates, pharmaceutically acceptable salts, derivatives or prodrugs thereof, are generally defined by

I wherein R la is H, halo, Ci.io-alkyl, C 2 - 8 -alkenyl or C 2 - 8 -alkynyl, wherein 1, 2 or 3 carbon atoms of said CpCioalkyl, C 2 -C 8 -alkenyl or C 2 -C 8 -alkynyl is optionally replaced with a heteroatom selected from O, S, S(O), S(O) 2 and N, and optionally substituted independently with one or more substituents of R 7 ; R lb is H, halo, haloalkyl, C 1-6 -alkyl, -O-C,. 6 -alkyl, -S-C,. 6 -alkyl, -NH-C^-alkyl, -

N-di-Ci. 6 -alkyl, CN, OH or NH 2 , wherein the Ci -6 -alkyl and the Ci -6 -alkyl portion of -O- Ci -6 -alkyl, -S-Ci_ 6 -alkyl, -NH-Ci_ 6 -alkyl and -N-di-C^-alkyl are optionally substituted independently with 1-5 substituents of R 7 ; alternatively, R la and R lb taken together with the carbon atom to which they are attached form a partially or fully saturated 3-, 4-, 5- or 6-membered ring of carbon atoms optionally including 1-2 heteroatoms selected from O, N, or S, the ring optionally substituted independently with 1-3 substituents of R 7 ;

R lc is H, halo, haloalkyl, C,. 6 -alkyl, -O-C^-alkyl, -S-d. 6 -alkyl, -NH-Ci -6 -alkyl, - N-di-Ci. 6 -alkyl, CN, OH or NH 2 ; W is -C(=0)-, - OC(=O) -, -NHC(=O)-, -S(=0) b - or -NHS(=O) b -, wherein b is 1 or 2;

B is R 2 -(CR 2a R 2a ) h -, wherein

R 2 is Ci-C 6 alkyl, C,-C 6 haloalkyl, C 2 -C 8 alkenyl or C 2 -C 8 alkynyl, wherein each of said Ci-C 6 alkyl, C 2 -C 8 alkenyl and C 2 -C 8 alkynyl is optionally substituted independently with one or more substituents of R 7 ; each R 2a , independently, is H, halo, OH, NO 2 , CN, NH 2 , C, -Ci 0 alkyl, C r Cio alkoxyl or haloalkyl; and h is O, 1, 2 or 3; each R 3 , independently, is H, haloalkyl, CN, Ci -6 -alkyl, C 2 . 8 -alkenyl, C 2-8 -alkynyl, C 3 . 8 -cycloalkyl or C4_ 8 -cycloalkenyl, each of the C 2 . 8 -alkenyl, C 2 . 8 -alkynyl, C 3-

A- 1291 -WO-PCT - 6 - g-cycloalkyl and optionally comprising 1 -2 heteroatoms selected from N, O and S and optionally substituted with 1-5 substituents of R 7 ;

R 4 is H, haloalkyl, CN, Ci. 6 -alkyl, Ci -6 -alkenyl, Ci. 6 -alkynyl or Ci. 6 -cycloalkyl, wherein each of the Ci_ 6 -alkyl, Ci -6 -alkenyl, Ci. 6 -alkynyl and Ci -6 -cycloalkyl is optionally substituted with 1-5 substitutions of R 7 ;

R 5 is

wherein X 1 is CR 8 R 8 , C(=O), O, S, S(O) 2 or NR 8 ; each X 2 , independently, is CR 8 R 8 ; each ofY 1 , Y 2 and Y 3 , independently, is CR 8 R 8 , O, S or NR 8 ; Z 2 taken together with the carbon atoms to which it is attached is a partially or fully unsaturated 5-6 membered monocyclic ring, said ring formed of carbon atoms optionally including 1-3 heteroatoms selected from O, N and S; m is 0, 1 or 2; o is 0, 1, 2, 3, 4 or 5; and p is O, 1, 2, 3, 4 or 5 provided that (a) no more than two of Y 1 , Y 2 and Y 3 is O, S or NR 8 and (b) when o is 0, then each of Y 1 and Y 2 is CR 8 R 8 ; each R 7 , independently, is H, halo, haloalkyl, CN, OH, NO 2 , NH 2 , acetyl, oxo, Q- l o-alkyl, C 2 .i 0 -alkenyl, C 2 .i 0 -alkynyl, C 3 .i 0 -cycloalkyl, C 4 -io-cycloalkenyl, C M0 - alkylamino-, Ci.io-dialkylamino-, Ci.io-alkoxyl, Ci.i 0 -thioalkoxyl or a saturated or partially or fully unsaturated 3-8 membered monocyclic or a 6-12 membered bicyclic, said ring system formed of carbon atoms optionally including 1-3 heteroatoms if monocyclic or 1-6 heteroatoms if bicyclic, said heteroatoms selected from O, N, or S,

A-1291 -WO-PCT - 7 - wherein each of the Cuo-alkyl, C 2- io-alkenyl, C 2- io-alkynyl, Cs.io-cycloalkyl, C 4-I0 - cycloalkenyl, Ci. 10 -alkylamino-, Ci.io-dialkylamino-, Ci.io-alkoxyl, C M o-thioalkoxyl and ring of said ring system is optionally substituted independently with 1-5 substituents of halo, haloalkyl, CN, NO 2 , NH 2 , OH, oxo, methyl, methoxyl, ethyl, ethoxyl, propyl, propoxyl, isopropyl, isopropoxyl, cyclopropyl, cyclopropylmethoxyl, butyl, butoxyl, isobutoxyl, tert-butoxyl, isobutyl, sec-butyl, tert-butyl, cyclobutyl, pentyl, cyclopentyl, hexyl, cyclohexyl, Ci.io-alkylamino-, Ci.io-dialkylamino-, Ci.io-thioalkoxyl, benzyl or phenyl; each R 8 , independently, is H, halo, haloalkyl, CN, OH, NO 2 , NH 2 , acetyl, oxo, C 1 . l o-alkyl, C 2 .i 0 -alkenyl, C 2- i 0 -alkynyl, Cs.io-cycloalkyl, C 4 -io-cycloalkenyl, C M0 - alkylamino-, Ci.iQ-dialkylamino-, Q.io-alkoxyl, Ci.io-thioalkoxyl or a saturated or partially or fully unsaturated 3-8 membered monocyclic or a 6-12 membered bicyclic, said ring system formed of carbon atoms optionally including 1 -3 heteroatoms if monocyclic or 1-6 heteroatoms if bicyclic, said heteroatoms selected from O, N, or S, wherein each of the Ci.io-alkyl, C 2 .i 0 -alkenyl, C 2- i 0 -alkynyl, Cs-io-cycloalkyl, C 4-10 - cycloalkenyl, Ci.io-alkylamino-, Ci.io-dialkylamino-, Ci.io-alkoxyl, Cj.io-thioalkoxyl and ring of said ring system is optionally substituted independently with 1 -5 substituents of R 9 ; and

R 9 is halo, haloalkyl, CN, OH, NO 2 , NH 2 , oxo, acetyl, methyl, methoxyl, ethyl, ethoxyl, propyl, propoxyl, isopropyl, isopropoxyl, cyclopropyl, cyclopropylmethoxyl, butyl, butoxyl, isobutoxyl, tert-butoxyl, isobutyl, sec-butyl, tert-butyl, cyclobutyl, pentyl, cyclopentyl, hexyl, cyclohexyl, benzyl, phenyl, Ci_ 10 -alkylamino-, Ci.io-dialkylamino-, Ci. lo -thioalkoxyl or a partially or fully saturated or unsaturated 3-6 membered monocyclic formed of carbon atoms optionally including 1-3 heteroatoms selected from O, N, or S, and optionally substituted independently with 1 -5 substituents of halo, haloalkyl, CN, NO 2 , NH 2 , OH, oxo, acetyl, methyl, methoxyl, ethyl, ethoxyl, propyl, propoxyl, isopropyl, isopropoxyl, cyclopropyl, cyclopropylmethoxyl, butyl, butoxyl, isobutoxyl, tert-butoxyl, isobutyl, sec-butyl, tert-butyl, cyclobutyl, pentyl, cyclopentyl, hexyl, cyclohexyl, benzyl or phenyl. In another embodiment, Formula I includes compounds wherein R la is H, halo,

Ci.io-alkyl, C 2 .g-alkenyl or C 2 . 8 -alkynyl, wherein 1, 2 or 3 carbon atoms of said Q- Ci θ alkyl, C 2 -C 8 -alkenyl or C 2 -C 8 -alkynyl is optionally replaced with a heteroatom selected from O, S, S(O), S(O) 2 and N, and optionally substituted independently with one or more substituents of R 7 , in conjunction with any of the above or below embodiments.

A- 1291 -WO-PCT - 8 -

In another embodiment, Formula I includes compounds wherein R la is H, halo, Cj.io-alkyl, C 2-8 -alkenyl or C 2 -8-alkynyl, wherein 1, 2 or 3 carbon atoms of said Q- Ci O alkyl, C 2 -C 8 -alkenyl or C 2 -C 8 -alkynyl is optionally replaced with a heteroatom selected from O, S, S(O), S(O) 2 and N, and optionally substituted independently with 1-3 substituents of halo, haloalkyl, CN, OH, NO 2 , NH 2 , acetyl, Q.io-alkyl, C 2 .i 0 -alkenyl, C 2- l o-alkynyl, C 3 .i 0 -cycloalkyl, Gno-cycloalkenyl, Ci.io-alkylamino-, Ci.io-dialkylamino-, Ci. l o-alkoxyl and Ci.io-thioalkoxyl, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein R la is Ci -6 -alkyl, C 2 . 6 alkenyl, C 2 . 6 -alkynyl, -NH-C L β-alkyl-, -O-C,. 6 -alkenyl-, -S-C 2 . 6 -alkenyl-, -NH-C 2 . 6 -alkenyl-, -O-Ci_ 6 -alkynyl-, -S-C^alkynyl-, -NH-Ci-e-alkynyl-, -C,. 4 alkyl-O-C M -alkyl-, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein R lb is H, halo, haloalkyl, C^-alkyl, -O-Ci. 6 -alkyl, -S-C^-alkyl, -NH-C, -6 -alkyl, -N-di-d. 6 -alkyl, CN, OH or NH 2 , wherein the Ci -6 -alkyl and the portion of -O-Ci. 6 -alkyl, -S-Ci. 6 - alkyl, -NH-Ci. 6 -alkyl and -N-di-C]. 6 -alkyl are optionally substituted independently with 1-5 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein R lb is H, halo, haloalkyl, d. 6 -alkyl, -O-C 1-6 -alkyl, -S-C 1-6 -alkyl, -NH-C,. 6 -alkyl, -N-di-C,. 6 -alkyl, CN, OH or NH 2 , in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein R la and R lb taken together with the carbon atom to which they are attached form a partially or fully saturated 3-, A-, 5- or 6-membered ring of carbon atoms optionally including 1-2 heteroatoms selected from O, N, or S, the ring optionally substituted independently with 1-3 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein R lc is H, halo, haloalkyl, C^-alkyl, -O-C,. 6 -alkyl, -S-Ci. 6 -alkyl, -NH-C, -6 -alkyl, -N-di-Ci. 6 -alkyl, CN, OH or NH 2 , in conjunction with any of the above or below embodiments. In another embodiment, Formula I includes compounds wherein R lc is H, halo, haloalkyl, Ci. 6 -alkyl, CN, OH or NH 2 , in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein R la is Ci -6 -alkyl, C 2 . 6 alkenyl, C 2-6 -alkynyl, -O-C 1-6 -alkyl-, -S-C 1-6 -alkyl-, -NH-C 1 . 6 -alkyl-, -O-C,. 6 -alkenyl-,

A-1291 -WO-PCT - 9 -

-S-C 2 . 6 -alkenyl-, -NH-C 2 . 6 -alkenyl-, -O-Cβ-alkynyl-, -NH-C L β-alkynyl-, -C M alkyl-O-C M -alkyl-,

R lb is H, halo, haloalkyl, Ci -6 -alkyl, -O-C 1-6 -alkyl, -S-C 1-6 -alkyl, -NH-C 1-6 -alkyl, - N-di-C,. 6 -alkyl, CN, OH or NH 2 ; and R lc is H, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein W is -C(=0)-, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein W is - 0C(=0) -, in conjunction with any of the above or below embodiments. In another embodiment, Formula I includes compounds wherein W is -NHC(=O)-

, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein W is -S(=O) b - wherein b is 1 or 2, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein W is -NHS(=O) b - wherein b is 1 or 2, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein B is R 2 - (CR 2a R 2a ) h - wherein R 2 is Ci-C 6 alkyl, Q-C 6 haloalkyl, C 2 -C 8 alkenyl or C 2 -C 8 alkynyl , each of which is optionally substituted independently with one or more substituents of R 7 , each R 2a , independently, is H, halo, OH, NO 2 , CN, NH 2 , Ci-Ci 0 alkyl, Ci-Ci 0 alkoxyl or haloalkyl, and h is 1, 2 or 3, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein one R 2a is H and the other R 2a is OH, NO 2 , CN, NH 2 , C r C 10 alkyl, C,-C 10 alkoxyl or haloalkyl, in conjunction with any of the above or below embodiments. In another embodiment, Formula I includes compounds wherein each R 2a , independently, is H, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein h is 1 , in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein h is 2, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein B is R 2 -O- (CR 23 R 2 V and each R 2a , independently, is H, CN, Ci-Qo alkyl, Ci-Ci 0 alkoxyl or haloalkyl, in conjunction with any of the above or below embodiments.

A-1291-WO-PCT - 10 -

In another embodiment, Formula I includes compounds wherein B is R 2 -S- (CR 23 R 2 V and each R 2a , independently, is H, CN, Ci-Ci 0 alkyl, C r Ci 0 alkoxyl or haloalkyl, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein B is R 2 -NR 2a - (CR 2a R 2a ) h - and each R 2a , independently, is H, CN, Ci-Ci 0 alkyl, C r Ci 0 alkoxyl or haloalkyl, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein B is R 2 -O- (CH 2 ) h -, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein B is R 2 -S- (CH 2 ) h -, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein B is R 2 -NH- (CH 2 ) h -, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein B is R 2 - (CR 2a R 2a ) h - wherein R 2 is Ci -Cβ alkyl optionally substituted independently with one or more substituents of R 7 , each R 2a , independently, is H, OH, NO 2 , CN, NH 2 , Cj -Ci 0 alkyl, Ci-Cio alkoxyl or haloalkyl, and h is 1, 2 or 3, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein B is R 2 - (CR 2a R 2a ) h - wherein R 2 is C 2 -Cg alkenyl optionally substituted independently with one or more substituents of R 7 , each R 2a , independently, is H, OH, NO 2 , CN, NH 2 , C r Cio alkyl, Ci-Cio alkoxyl or haloalkyl, and h is 1, 2 or 3, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein B is R 2 - (CR 2a R 2a ) h - wherein R 2 is C 2 -C 8 alkynyl optionally substituted independently with one or more substituents of R 7 , each R 2a , independently, is H, OH, NO 2 , CN, NH 2 , Ci-Ci 0 alkyl, Ci-Cio alkoxyl or haloalkyl, and h is 1, 2 or 3, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein each R 3 , independently, is H, haloalkyl, CN, Ci-β-alkyl, C 2 . 8 -alkenyl, C 2 _ 8 -alkynyl, C 3 . 8 -cycloalkyl or C^s-cycloalkenyl, each of the Ci -6 -alkyl, C 2 . 8 -alkenyl, C 2-8 -alkynyl, C 3 . 8 -cycloalkyl and C 4-8 -cycloalkenyl optionally comprising 1-2 heteroatoms selected from N, O and S and optionally substituted with 1-5 substituents of R 7 , in conjunction with any of the above or below embodiments.

A-1291 -WO-PCT - 11 -

In another embodiment, Formula I includes compounds wherein R 3 is H, haloalkyl, CN, Ci.i O -alkyl, C 2 .io-alkenyl or C 2 -io-alkynyl, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein R 3 is H, haloalkyl or Ci-io-alkyl, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein each R 3 , independently, is H, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein R 4 is H, haloalkyl, CN, Ci. 6 -alkyl, Ci -6 -alkenyl, Ci. 6 -alkynyl or wherein each of the Ci- 6 -alkyl, Ci. 6 -alkenyl, Ci_ 6 -alkynyl and C^-cycloalkyl is optionally substituted with 1-5 substitutions of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein R 4 is H, haloalkyl, CN or Ci-β-alkyl, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein R 4 is H, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein R 5 is

wherein X 1 is CR 8 R 8 , C(=O), O, S, S(O) 2 or NR 8 ; each X 2 , independently, is CR 8 R 8 ; each of Y 1 , Y 2 and Y 3 , independently, is CR 8 R 8 , O, S or NR 8 ;

Z 2 taken together with the carbon atoms to which it is attached is a partially or fully unsaturated 5-6 membered monocyclic ring, said ring formed of carbon atoms optionally including 1-3 heteroatoms selected from O, N and S; m is O, 1 or 2;

34

A-1291 -WO-PCT - 12 - o is 0, 1, 2, 3, 4 or 5; and p is 0, 1, 2, 3, 4 or 5 provided that (a) no more than two of Y 1 , Y 2 and Y 3 is O, S or NR 8 and (b) when o is 0, then each of Y 1 and Y 2 is CR 8 R 8 , in conjunction with any of the above or below embodiments.

In the immediately preceeding embodiment, Formula I includes compounds wherein X 1 is CR 8 R 8 , in conjunction with any of the above or below embodiments.

In the immediately preceeding embodiment, Formula I includes compounds wherein X 1 is CH 2 , in conjunction with any of the above or below embodiments. In the preceeding embodiment, Formula I includes compounds wherein X 1 is

C(=O), in conjunction with any of the above or below embodiments.

In the preceeding embodiment, Formula I includes compounds wherein X 1 is O, in conjunction with any of the above or below embodiments.

In the preceeding embodiment, Formula I includes compounds wherein X 1 is S, in conjunction with any of the above or below embodiments.

In the preceeding embodiment, Formula I includes compounds wherein X 1 is S(O) 2 , in conjunction with any of the above or below embodiments.

In the preceeding embodiment, Formula I includes compounds wherein X 1 is NR 8 , in conjunction with any of the above or below embodiments. In the preceeding embodiment, Formula I includes compounds wherein X 2 is

CR 8 R 8 , in conjunction with any of the above or below embodiments.

In the preceeding embodiment, Formula I includes compounds wherein X 2 is CH 2 , in conjunction with any of the above or below embodiments.

In the preceeding embodiment, Formula I includes compounds wherein each of Y 1 , Y 2 and Y 3 , independently, is CR 8 R 8 , in conjunction with any of the above or below embodiments.

Li the preceeding embodiment, Formula I includes compounds wherein each of Y 1 , Y 2 and Y 3 , independently, is CR 8 R 8 , O, S or NR 8 , in conjunction with any of the above or below embodiments. In the preceeding embodiment, Formula I includes compounds wherein each of

Y 1 , Y 2 and Y 3 , independently, is CHR 8 , in conjunction with any of the above or below embodiments.

In the preceeding embodiment, Formula I includes compounds wherein each of

A- 1291 -WO-PCT - 13 -

Y 1 , Y 2 and Y 3 , independently, is CH 2 , in conjunction with any of the above or below embodiments.

In the preceeding embodiment, Formula I includes compounds wherein no more than two of Y 1 , Y 2 and Y 3 , independently, is O and the remaining of Y 1 , Y 2 and Y 3 , independently, is CR 8 R 8 , in conjunction with any of the above or below embodiments. In the preceeding embodiment, Formula I includes compounds wherein one of Y 1 , Y 2 and Y 3 , independently, is O and the other two of Y 1 , Y 2 and Y 3 , independently, is CR 8 R 8 , in conjunction with any of the above or below embodiments.

In the preceeding embodiment, Formula I includes compounds wherein Y 2 is O and Y'and Y 3 , independently, are CR 8 R 8 , in conjunction with any of the above or below embodiments.

In the preceeding embodiment, Formula I includes compounds wherein Y 2 is S and Y'and Y 3 , independently, are CR 8 R 8 , in conjunction with any of the above or below embodiments. In the preceeding embodiment, Formula I includes compounds wherein Y 2 is

-NR 8 - and Y'and Y 3 , independently, are CR 8 R 8 , in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein Z 2 is an optionally substituted phenyl, pyridine, pyrimidine, pyridazine, pyrazine, pyridone, pyrrole, imidazole, pyrazole, triazole, thiophene, thiazole, thiadiazole, isothiazole, furan, oxazole, oxadiazole or isoxazole ring, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein Z 2 is a 5- or 6- membered aromatic ring which is substituted with a chemical moiety which reduces CYP enzyme activity, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein Z 2 is a phenyl, pyridine, pyrimidine, triazine, pyridazine, pyrazine, pyridone, pyrrole, imidazole, pyrazole, triazole, thiophene, thiazole, thiadiazole, isothiazole, furan, oxazole, oxadiazole or isoxazole ring, each of which is substituted with a chemical moiety which reduces CYP enzyme activity, in conjunction with any of the above or below embodiments.

A-1291-WO-PCT - 14 -

In another embodiment, Formula I includes compounds wherein R 5 is

wherein each of A 1 , A 2 , A 3 and A 4 , independently, is CR 8 or N, provided that no more than two of A 1 , A 2 , A 3 and A 4 is N; m, o, R 8 , X 2 , Y 1 , Y 2 and Y 3 are as defined herein; X 1 is CR 8 R 8 , C(=O), O, S, S(O) 2 or NR 8 ; and p is 0, 1, 2 or 3, , in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein R 5 is

wherein o is 1 or 2; p is O, 1, 2 or 3; each of A 1 , A 2 , A 3 and A 4 , independently, is CR 8 or N, provided that no more than two of A 1 , A 2 , A 3 and A 4 is N; X 1 is CHR 8 , C(=O), O or NR 12 ;

A-1291-WO-PCT - 15 -

Y 3 is CR 12 or O; and each R 8 , independently, is H, halo, haloalkyl, CN, OH, NO 2 , NH 2 , acetyl, oxo, Ci.io-alkyl, C 2- io-alkenyl, C 2 -io-alkynyl, C M o-cycloalkyl, Gno-cycloalkenyl, Ci -I0 - alkylamino-, Ci.io-dialkylamino-, Ci.io-alkoxyl, Cj.io-thioalkoxyl or a ring selected from phenyl, pyridyl, pyrimidinyl, triazinyl, thiophenyl, fiiryl, tetrahydrofuranyl, pyrrolyl, pyrazolyl, thieno-pyrazolyl, imidazolyl, triazolyl, tetrazolyl, thiazolyl, thiadiazolyl, oxazolyl, oxadiazolyl, isoxazolyl, isothiazolyl, oxazolinyl, isoxazolinyl, thiazolinyl, pyrrolidinyl, pyrazolinyl, morpholinyl, piperidinyl, piperazinyl, pyranyl, dioxolyl, dioxazinyl, cyclopropyl, cyclobutyl, azetidinyl, cyclopentyl, cyclohexyl and cycloheptyl, wherein each of the Ci -]0 -alkyl, C 2- io-alkenyl, C 2- io-alkynyl, C 4-I0 - cycloalkenyl, Ci.io-alkylamino-, Ci -10 -dialkylamino-, Ci.io-alkoxyl, Ci.io-thioalkoxyl and ring is optionally substituted independently with 1-5 substituents of halo, haloalkyl, CN, NO 2 , NH 2 , OH, oxo, methyl, methoxyl, ethyl, ethoxyl, propyl, propoxyl, isopropyl, isopropoxyl, cyclopropyl, cyclopropylmethoxyl, butyl, butoxyl, isobutoxyl, tert-butoxyl, isobutyl, sec-butyl, tert-butyl, cyclobutyl, pentyl, cyclopently, hexyl, cyclohexyl, Ci -I0 - alkylamino-, Ci.io-dialkylamino-, Ci.io-thioalkoxyl, ben∑yl or phenyl, in conjunction with any of the above or below embodiments.

In another embodiment, Formula I includes compounds wherein R la is C M -alkyl- S(O) 2 -Ci -3 -alkyl-, C,^-alkyl-NH-C 1-3 -alkyl, di-(Ci -3 -alkyl)-N-C, -3 -alkyl, C 2-4 -alkenyl-O- Ci -3 -alkyl-, C 2-4 -alkenyl-S-Ci -3 -alkyl-, or C 2-4 -alkynyl-NH-Ci. 3 -alkyl-, wherein the alkyl, alkenyl or alkynyl moiety of each is optionally substituted with 1-3 substituents of R 7 ;

W is -C(=0)-; B is R 2 -(CR 2a R 2l V, wherein each R 2a , independently, is H or C r C 6 alkyl; h is 1 or 2; and

R 2 is C]-C 4 alkyl, C 2 -C 4 alkenyl or C 2 -C 4 alkynyl, each of which is optionally substituted independently with 1-5 substituents of R 7 ; each R 3 , independently, is H, haloalkyl, CN, C 2-6 -alkenyl or C 2-6 - alkynyl;

R 4 is H, CN or C M0 -alkyl;

A- 1291 -WO-PCT - 16 -

R 5 is

wherein each of A 1 , A 2 , A 3 and A 4 , independently, is CR 8 or N, provided that no more than two of A 1 , A 2 , A 3 and A 4 is N; m, o, R 8 , X 2 , Y 1 , Y 2 and Y 3 are as defined in claim 1;

X 1 is CR 8 R 8 , C(O), O, S, S(O) 2 or NR 8 ; and p is 0, 1, 2 or 3; each R 7 , independently, is H, Cl, F, Br, I, haloalkyl, CN, OH, NO 2 , NH 2 , acetyl, methyl, methoxyl, ethyl, ethoxyl, allyl, propyl, propoxyl, isopropyl, isopropoxyl, cyclopropyl, cyclopropylmethoxyl, butyl, butoxyl, isobutoxyl, tert-butoxyl, isobutyl, sec- butyl, tert-butyl, cyclobutyl, pentyl, cyclopentyl, hexyl, cyclohexyl, Ci-io-alkylamino-, Ci.io-dialkylamino-, Ci.io-thioalkoxyl, benzyl or phenyl; each R 8 , independently, is Cl, F, Br, I, haloalkyl, CN, OH, NO 2 , NH 2 , acetyl, methyl, methoxyl, ethyl, ethoxyl, allyl, propyl, propoxyl, isopropyl, isopropoxyl, cyclopropyl, cyclopropylmethoxyl, butyl, butoxyl, isobutoxyl, tert-butoxyl, isobutyl, sec- butyl, tert-butyl, cyclobutyl, pentyl, neopentyl, cyclopentyl, hexyl, cyclohexyl, Ci -I0 - alkylamino-, Ci-io-dialkylamino-, Ci.jo-thioalkoxyl or a ring system selected from phenyl, pyridyl, pyrimidinyl, triazinyl, thiophenyl, furyl, tetrahydrofuranyl, pyrrolyl, pyrazolyl, thieno-pyrazolyl, imidazolyl, triazolyl, tetrazolyl, thiazolyl, thiadiazolyl, oxazolyl, oxadiazolyl, oxadiazolyl, isoxazolyl, isothiazolyl, oxazolinyl, isoxazolinyl, thiazolinyl, pyrrolidinyl, pyrazolinyl, moφholinyl, piperidinyl, piperazinyl, pyranyl, dioxozinyl, 2,3-

A- 1291 -WO-PCT - 17 - dihydro-l,4-benzoxazinyl, 1,3-benzodioxolyl and azetidinyl, said ring system optionally substituted independently with 1-3 substituents of R 9 ; and

R 9 is H, halo, haloalkyl, CN, OH, NO 2 , NH 2 , acetyl, C 1-10 -alkyl, C 2- io-alkenyl, C 2- l o-alkynyl, C 3-7 -cycloalkyl, C 4-7 -cycloalkenyl, Ci-io-alkylamino-, Ci.io-dialkylamino-, Ci- 10 -alkoxyl, Ci.jo-thioalkoxyl.

In another embodiment, the present invention includes compounds generally defined by Formula II:

π or a stereoisomer, tautomer, solvate or pharmaceutically acceptable salt thereof, wherein each of A 1 and A 2 , independently, is CH or CR 6 ; one of A 3 and A 4 , independently, is CH or CR 3 and the other of A 3 and A 4 , independently, is N, CH or CR 6 ;

R la is H, halo, haloalkyl, C^-alkyl, -O-C 1-6 -alkyl, -S-C 1-6 -alkyl, -NH-d-e-alkyl, - N-di-Ci. 6 -alkyl, CN, OH or NH 2 , wherein the C ]-6 -alkyl portion of -O-C 1-6 -alkyl, -S-C 1-6 - alkyl, -NH-Ci -6 -alkyl and -N-di-Ci_ 6 -alkyl are optionally substituted independently with 1-5 substituents of R 7 ;

R lb is H, halo, haloalkyl, C^-alkyl, -O-C ]-6 -alkyl, -S-C w -alkyl, -NH-Ci-e-alkyl, - N-di-Ci- 6 -alkyl, CN, OH or NH 2 , wherein the C 1-6 -alkyl portion of -O-C 1-6 -alkyl, -S-Q -6 - alkyl, -NH-Q^-alkyl and -N-di-Q^-alkyl are optionally substituted independently with 1-5 substituents of R 7 ; alternatively, R la and R lb taken together with the carbon atom to which they are attached form a partially or fully saturated 3-, 4-, 5- or 6-membered ring of carbon atoms optionally including 1-2 heteroatoms selected from O, N, or S, the ring optionally substituted independently with 1-3 substituents of R 7 ;

R lc is H, halo, haloalkyl, C 1-6 -alkyl, -O-Ci- 5 -alkyl, -S-Q^-alkyl, -NH-Q.e-alkyl, - N-di-C^-alkyl, CN, OH Or NH 2 ;

W is -C(=O)-, -C(=S>, - OC(O) -, -NHC(=O)-, -S(=O) b - or -NHS(=O) b -, wherein b is 1 or 2;

A- 1291 -WO-PCT - 18 -

R 2 is Ci-C 3 alkyl, -OC 1 -C 3 alkyl, C 2 -C 3 alkenyl or C 2 -C 3 alkynyL, wherein each of said Ci-C 3 alkyl, -OCi-C 3 alkyl, C 2 -C 3 alkenyl and C 2 -C 3 alkynyl is optionally substituted independently with 1-3 substituents of R 7 ;

R 3 is -NH-Ci-βalkyl, -NH-partially or fully unsaturated 5- or 6-membered monocyclic ring formed of carbon atoms wherein said ring optionally including 1-3 heteroatoms selected from O, N, or S and optionally substituted with 1-5 substituents of R 7 , -NH-l,3-benzodiox-5-yl optionally substituted with 1-3 substituents of R 7 , 1,3- benzodiox-5-yl optionally substituted with 1-3 substituents of R 7 or partially or fully unsaturated 5- or 6-membered monocyclic ring formed of carbon atoms wherein said ring optionally including 1-3 heteroatoms selected from O, N, or S and optionally substituted with 1-5 substituents of R 7 ;

R 4 is H, halo or C w -alkyl;

R 5 is H, halo, haloalkyl, oxo, C 1-6 -alkyl, -O-Ci-e-alkyl, -S-C^-alkyl, -NH-Ci-βalkyl, -N-di-Ci- 6 -alkyl, CN, OH or NH 2 , wherein the Q-6-alkyl and the C^-alky! portion -NH-Ci-β-alkyl and are optionally substituted independently with 1-5 substituents of R 7 ;

X is CH 2 , CHR 6 , CR 6 R 6 , C(=0), O, NH, NR 6 , or S(O) 0 wherein o is 0, 1 or 2;

Z is a 3-6 membered spirocyclic ring formed of carbon atoms optionally including 1-3 heteroatoms selected from O, N and S and optionally substituted independently with 1-5 substituents of R 7 ; each R 6 , independently, is halo, haloalkyl, CN, OR 7 , NHR 7 , -S-Ci- 6 -alkyl, C 2-6 -alkynyl, C 3- g-cycloalkyl or C 4-8 - cycloalkenyl, wherein the C 2-6 -alkynyl, C 3- g-cycloalkyl, C 4-8 - cycloalkenyl and the Ci- 6 -alkyl portion and are optionally substituted with 1-5 substituents of R 7 ; or R 6 is a fully saturated or partially or fully unsaturated 5- or 6-membered monocyclic or bicyclic ring formed of carbon atoms, said ring optionally including 1-4 heteroatoms selected from O, N, or S and optionally substituted with one or more substituents of R 7 ; and each R 7 , independently, is H, halo, haloalkyl, CN, OH, NO 2 , NH 2 , acetyl, oxo, Q-

10 -alkyl, C 2- io-alkenyl, C 2- i 0 -alkynyl, C 3- i 0 -cycloalkyl, Gno-cycloalkenyl, Ci -I0 - alkylamino-, Cj.io-dialkylamino-, Q.io-alkoxyl, Ci.io-thioalkoxyl or a fully saturated or partially or fully unsaturated 3-8 membered monocyclic or a 6-12 membered bicyclic, said ring system formed of carbon atoms optionally including 1-3 heteroatoms if

A- 1291 -WO-PCT - 19 - monocyclic or 1-6 heteroatoms if bicyclic, said heteroatoms selected from O, N, or S, wherein each of the Ci-io-alkyl, C 2- io-alkenyl, C 2- io-alkynyl, Ca-io-cycloalkyl, C 4-I0 - cycloalkenyl, Ci.io-alkylamino-, Cj.io-dialkylamino-, Ci.i θ -alkoxyl, Ci-io-thioalkoxyl and ring of said ring system is optionally substituted independently with 1-5 substituents of halo, haloalkyl, CN, NO 2 , NH 2 , OH, oxo, methyl, methoxyl, ethyl, ethoxyl, propyl, propoxyl, isopropyl, isopropoxyl, cyclopropyl, cyclopropylmethoxyl, butyl, butoxyl, isobutoxyl, tert-butoxyl, isobutyl, sec-butyl, tert-butyl, cyclobutyl, pentyl, cyclopentyl, hexyl, cyclohexyl, Ci.io-alkylamino-, Ci-io-dialkylaminor, Ci-io-thioalkoxyl, benzyl or phenyl; provided the compound is not

N-((lS)-l-((lR)-2-(((4 t S)-6'-(2,2-dimethylpropyl)-3 t ,4'-dihydrospiro[cyclobutane- l^'-pyranoP^-^pyridinJ^'-yOamino^l-hydroxyethyO-S-methylbuty Oacetamide;

N-((lS)-l-((lR)-2-(((4'S)-6 1 -(2,2-dimethylpropyl)-3',4 l -dihydrospiro[cyclobutane- l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-3-me thyl-3-buten-l- yl)acetamide;

N-((lS)-l-((lR)-2-(((4 I S)-6'-(2,2-dimethylpropyl)-3',4 I -dihydrospiro[cyclobutane- 1 ,2'-pyrano [2,3 -b]pyridin]-4'-yl)amino)- 1 -hydroxyethyl)-3 -pentyn- 1 -yl)acetamide;

N-((lS)-l-((lR)-2-(((4'S)-6'-(2,2-dimethylpropyl)-3 I ,4'-dihydrospiro[cyclobutane- 1 ,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)- 1 -hydroxyethyl)-4-penten- 1 -yl)acetamide; N-((lS>l-((lR)-2-(((ls,3R,4'S)-6'-(2,2-dimethylpropyl)-3- methyl-3 l ,4 1 - dihydrospiro[cyclobutane- 1 ,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)- 1 -hydroxyethyl)-4- penten- 1 -yl)acetamide;

N-((lS,2R)-3-(((4S)-6-ethyl-3,4-dihydrospiro[chromene-2,l '-cyclobutan]-4- yl)amino)-2 -hydroxy- 1 -((methyloxy)methyl)propyl)acetamide; N-((lS,2R)-3-(((4S)-6-ethyl-3,4-dihydrospiro[chromene-2,l'-c yclobutan]-4- yl)amino)-2-hydroxy- 1 -((propyloxy)methyl)propyl)acetamide;

N-((lS,2R)-3-(((4S)-6-ethyl-3,4-dihydrospiro[chromene-2,l '-cyclobutan]-4- yl)amino)-2-hydroxy- 1 -(((phenyhnethyl)oxy)methyl)propyl)acetamide;

N-((lS)-l-((lR)-2-(((4'S)-6'-(2,2-dimethylpropyl)-3',4'-d ihydrospiro[cyclobutane- 1 ,2'-pyrano [2,3 -b]pyridin]-4'-yl)amino)- 1 -hydroxyethyl)-3 -buten- 1 -yl)acetamide;

N-((lS)-l-((lS)-2-(((4 I S)-6'-(2,2-dimethylpropyl)-3 I ,4'-dihydrospiro[cyclobutane- l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-3-bu ten-l-yl)acetamide;

N-(( 1 S)- 1 -(( 1 R)-2-(((4'S)-6 1 -(2,2-dimethylpropyl)-3 t ,4'-dihydrospiro[cyclobutane- l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-4,4, 4-trifluorobutyl)acetamide;

A-1291-WO-PCT - 20 -

N-((lS,3E)-l-((lR)-2-(((4 1 S)-6'-(2,2-dimethylpropyl)-3',4 1 - dihydrospiro[cyclobutane- 1 ,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)- 1 -hydroxyethyl)-3- penten- 1 -yl)acetamide;

N-((lS)-l-((lR)-2-(((4 I S)-6'-(2,2-dimethylpropyl)-3',4 l -dihydrospiro[cyclobutane- 1 ,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l -hydroxyethyl)-2-propen- 1 -yl)acetamide;

N-((l S)-I -(( lR)-2-(((4'S)-6 1 -(2-fluoro-2-methylpropyl>3',4 1 - dihydrospiro[cyclobutane-l,2'-pyrano[2,3-b]pyridin]-4'-yl)am ino)-l-hydroxyethyl)-3- buten-l-yl)acetamide; and

N-((lS)-l-((lR)-2-(((4'S)-6'-(2,2-dimethylpropyl)-3',4 I -dihydrospiro[cyclobutane- 1 ,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)- 1 -hydroxyethyl)-3-hexyn- 1 -yl)acetamide.

Thus, the specific compounds listed above are excluded from the scope of the present invention and from the scope of Formulas I and π described herein.

In another embodiment, the compounds of Formulas I and II include compounds wherein R la is H, halo, Q.io-alkyl, C 2- 8-alkenyl or C 2-8 -alkynyl, wherein 1, 2 or 3 carbon atoms of said Ci-Ci O alkyl, C 2 -Cg-alkenyl or C 2 -Cg-alkynyl is optionally replaced with a heteroatom selected from O, S, S(O), S(O) 2 and N, and optionally substituted independently with one or more substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and II include compounds wherein R la is H, halo, Ci-io-alkyl, C 2-8 -alkenyl or C 2- g-alkynyl, wherein 1, 2 or 3 carbon atoms of said Ci-Ci O alkyl, C 2 -C 8 -alkenyl or C 2 -C 8 -alkynyl is optionally replaced with a heteroatom selected from O, S, S(O), S(O) 2 and N, and optionally substituted independently with 1-3 substituents of halo, haloalkyl, CN, OH, NO 2 , NH 2 , acetyl, Ci -I0 - alkyl, C 2 . 10 -alkenyl, C 2- io-alkynyl, C 3- io-cycloalkyl, Ci.io-alkylamino-, Ci-io-dialkylamino-, Ci.io-alkoxyl and Ci.io-thioalkoxyl, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and II include compounds wherein R la is C 1-6 -alkyl, -NH-C]- 6 -alkyl-, -O-C^-alkenyl-, -S-C 2-6 -alkenyl-, -NH-C 2-6 -alkenyl-, -O-C^-alkynyl-, -S-Ci- 6 alkynyl-, -NH-C 1-6 -alkynyl-, -C M alkyl-O-C M -alkyl-, -Ci. 4 -alkyl-S-C M -alkyl- or -C M - alkyl-NH-C M -alkyl-, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and II include compounds wherein R la is H, halo, haloalkyl, -

A- 1291 -WO-PCT - 21 -

N-di-C 1-6 -alkyl, CN, OH orNH 2 , wherein the Cwalkyl and the C^-alkyl portion of -O- and -N-di-C ]-6 -alkyl are optionally substituted independently with 1-5 substituents of R 7 , in conjunction with any of the above or below embodiments. In another embodiment, the compounds of Formulas I and π include compounds wherein R la is C^-alkyl, -O-Ci -6 -alkyl, -S-C^-alkyl, -NH-C w -alkyl, wherein the C 1-6 - alkyl and the is optionally substituted independently with 1-5 substituents of R 7 , in conjunction with any of the above or below embodiments. In another embodiment, the compounds of Formulas I and π include compounds wherein R lb is H, halo, haloalkyl, C 1-6 -alkyl, -O-C 1-6 -alkyl, -S-C^-alkyl, -NH-C 1-6 -alkyl, - N-di-Ci- 6 -alkyl, CN, OH or NH 2 , wherein the and the C 1-6 -alkyl portion of -O- are optionally substituted independently with 1-5 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and π include compounds wherein R lb is H, halo, haloalkyl, - N-di-C 1-6 -alkyl, CN, OH or NH 2 , wherein the C^-alkyl portion of -O-C 1-6 -alkyl, -S-C 1-6 - alkyl, -NH-Ci -6 -alkyl and -N-di-C 1-6 -alkyl are optionally substituted independently with 1-5 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and II include compounds wherein R lb is H, halo, haloalkyl, d-e-alkyl, -O-C 1-6 -alkyl, -S-C^-alkyl, -NH-C 1-6 -alkyl, - N-di-Ci-β-alkyl, CN, OH or NH 2 , in conjunction with any of the above or below embodiments. In another embodiment, the compounds of Formulas I and π include compounds wherein R lb is H, F, Cl, Br, CF 3 , C 2 F 5 , C^-alkyl, -O-C^-alkyl, -S-C^-alkyl, -NH-C 1-6 - alkyl, wherein the C 1-6 -alkyl and the Q^-alkyl portion of -O-C^-alkyl, -S-C 1-6 -alkyl and - NH-C^-alkyl is optionally substituted independently with 1-3 substituents of R 7 , in conjunction with any of the above or below embodiments. In another embodiment, the compounds of Formulas I and π include compounds wherein each of R la and R lb , independently, is H, methyl, ethyl, propyl, butyl, methoxyl, ethoxyl, propoxyl, butoxyl, each of which is optionally substituted independently with 1-3 substituents of halo, OH, NH 2 and CN, and provided that both of R la and R lb are not H, in conjunction with any of the above or below embodiments.

A-1291-WO-PCT - 22 -

In another embodiment, the compounds of Formulas I and π include compounds wherein R la and R lb taken together with the carbon atom to which they are attached form a partially or fully saturated 3, 4-, 5- or 6-membered ring of carbon atoms optionally including 1-2 heteroatoms selected from O, N, or S, the ring optionally substituted independently with 1-3 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and II include compounds wherein R la and R lb taken together with the carbon atom to which they are attached form a partially or fully saturated 4-, 5- or 6-membered ring of carbon atoms optionally including 1-2 heteroatoms selected from O, N, or S, the ring optionally substituted independently with 1-3 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and π include compounds wherein R la and R lb taken together with the carbon atom to which they are attached form a ring selected from tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, morpholin-2-yl, oxetan-2- yl, oxetan-3-yl, cyclopropyl, cyclobutyl, cyclopenty and cyclohexyl, the ring optionally substituted independently with 1-3 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and π include compounds wherein R lc is H, halo, haloalkyl, - N-di-Ci- 6 -alkyl, CN, OH or NH 2 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and II include compounds wherein R lc is H, halo, haloalkyl, C^-alkyl, CN, OH OrNH 2 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and II include compounds wherein R la is C^-alkyl, C 2-6 alkenyl, C 2-6 -alkvnyl, -O-Ci-e-alkyl-, -S-C^-alkyl-, -NH-C 1- 6 -alkyl-, -S-C 1 . 6 alkynyl-, -NH-C^-alkynyl-, -C M alkyl-O-C M -alkyl-, -C M -alkyl-S-C M -alkyl- or -C M - alkyl-NH-Q- 4 -alkyl-;

R lb is H, halo, haloalkyl, C^-alkyl, -O-C^-alkyl, -S-C^-alkyl, -NH-C^-alkyl, - N-di-C,_ δ -alkyl, CN, OH or NH 2 ; and

R lc is H, in conjunction with any of the above or below embodiments.

A-1291-WO-PCT - 23 -

In another embodiment, the compounds of Formulas I and π include compounds wherein each of R la and R lb , independently, is H, methyl, ethyl, propyl, butyl, methoxyl, ethoxyl, propoxyl, butoxyl, each of which is optionally substituted independently with 1-3 substituents of halo, OH, NH 2 and CN, and provided that both of R la and R lb are not H, and R lc is H, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and π include compounds wherein R la and R lb taken together with the carbon atom to which they are attached form a partially or fully saturated 3, 4-, 5- or 6-membered ring of carbon atoms optionally including 1-2 heteroatoms selected from O, N, or S, the ring optionally substituted independently with 1-3 substituents of R 7 and R lc is H, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and II include compounds wherein R la and R lb taken together with the carbon atom to which they are attached form a partially or fully saturated 4-, 5- or 6-membered ring of carbon atoms optionally including 1-2 heteroatoms selected from O, N, or S, the ring optionally substituted independently with 1-3 substituents of R 7 and R lc is H, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and π include compounds wherein R la and R lb taken together with the carbon atom to which they are attached form a ring selected from tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl, the ring optionally substituted independently with 1-3 substituents of R 7 and R lc is H, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and II include compounds wherein each of R la and R lb , independently, is H, methyl, ethyl, propyl, butyl, methoxyl, ethoxyl, propoxyl, butoxyl, each of which is optionally substituted independently with 1-3 substituents of halo, OH, NH 2 and CN, and provided that both of R la and R lb are not H, and R lc is H, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and II include compounds wherein W is -C(=O)-, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and II include compounds wherein W is - OC(=O) -, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and II include compounds wherein W is -NHC(=O)-, in conjunction with any of the above or below embodiments.

A-1291-WO-PCT - 24 -

In another embodiment, the compounds of Formulas I and π include compounds wherein W is -S(=O) b - wherein b is 1 or 2, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and π include compounds wherein W is -NHS(=O) b - wherein b is 1 or 2, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas I and π include compounds wherein W is -C(=O)-, -C(=S)-, - OC(=O) -, -NHC(O)-, -S(=0) b - or -NHS(=O) b -, wherein b is 1 or 2, in conjunction with any of the above or below embodiments. In another embodiment, the compounds of Formulas I and π include compounds wherein W is -C(=0)-, -C(=S)- or -S(=O) b -, wherein b is 1 or 2, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas II includes compounds wherein each of A 1 , A 2 , A 3 and A 4 , independently, is N, CH or CR 6 , provided that no more than two of A 1 , A 2 , A 3 and A 4 is N, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas II includes compounds wherein each of A 1 and A 2 , independently, is CH and one of A 3 and A 4 , independently, is N while the other of A 3 and A 4 , independently, is CH or CR 6 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas π includes compounds wherein A 1 is CH, A 2 is CR 6 and one of A 3 and A 4 , independently, is N while the other of A 3 and A 4 , independently, is CH or CR 6 , in conjunction with any of the above or below embodiments. In another embodiment, the compounds of Formulas π includes compounds wherein A 1 is CH, A 2 is CR 6 , A 3 is N and A 4 is CH or CR 3 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas π includes compounds wherein A 1 is CH, A 2 is CR 6 , A 3 is CH or CR 3 and A 4 is N, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas π includes compounds wherein A 1 is CH, A 2 is CR 6 and each of A 3 and A 4 , independently, CH, CR 6 or N, provided no more than one of A 3 and A 4 is N, in conjunction with any of the above or below embodiments.

A-1291-WO-PCT - 25 -

In another embodiment, the compounds of Formulas II includes compounds wherein A 1 is CH, A 2 is CR 6 and each of A 3 and A 4 , independently, CH or CR 3 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas II includes compounds wherein A 1 is CH, A 2 is CR 6 , A 3 is CH and A 4 is CR 3 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas II includes compounds wherein one of A 3 and A 4 , independently, is CH or CR 3 and the other of A 3 and A 4 , independently, is N, CH or CR 6 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas II includes compounds wherein one of A 3 and A 4 , independently, is CR 3 and the other of A 3 and A 4 , independently, is N, CH or CR 6 , in conjunction with any of the above or below embodiments. In another embodiment, the compounds of Formulas II includes compounds wherein R 2 is C]-C 3 alkyl, -OC 1 -C 3 alkyl, C 2 -C 3 alkenyl or C 2 -C 3 alkynyl, wherein each of said Ci-C 3 alkyl, -OCi-C 3 alkyl, C 2 -C 3 alkenyl and C 2 -C 3 alkynyl is optionally substituted independently with 1-3 substituents of R 7 , in conjunction with any of the above or below embodiments. In another embodiment, the compounds of Formulas π includes compounds wherein R 2 is Ci-C 3 alkyl, C 2 -C 3 alkenyl or C 2 -C 3 alkynyl, wherein each of said Ci-C 3 alkyl, C 2 -C 3 alkenyl and C 2 -C 3 alkynyl is optionally substituted independently with 1-3 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas π includes compounds wherein R 2 is C r C 3 alkyl or -OCi-C 3 alkyl, each of which is optionally substituted independently with 1-3 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas π includes compounds wherein R 2 is C 2 -C 3 alkenyl, optionally substituted independently with 1-3 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas π includes compounds wherein R 2 is C 2 -C 3 alkynyl, optionally substituted independently with 1-3 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formulas II includes compounds

A- 1291 -WO-PCT - 26 - wherein R 2 is -OC 1 -C 3 alkyl, optionally substituted independently with 1-3 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein R 3 is -NH-Ci-βalkyl, -NH-partially or fully unsaturated 5- or 6-membered monocyclic ring formed of carbon atoms wherein said ring optionally including 1-3 heteroatoms selected from O, N, or S and optionally substituted with 1-5 substituents of R 7 , -NH-l,3-benzodiox-5-yl optionally substituted with 1-3 substituents of R 7 , 1,3- benzodiox-5-yl optionally substituted with 1-3 substituents of R 7 or partially or fully unsaturated 5- or 6-membered monocyclic ring formed of carbon atoms wherein said ring optionally including 1-3 heteroatoms selected from O, N, or S and optionally substituted with 1-5 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula π includes compounds wherein R 3 is -NH-partially or fully unsaturated 5- or 6-membered monocyclic ring formed of carbon atoms wherein said ring optionally including 1-3 heteroatoms selected from O, N, or S and optionally substituted with 1-5 substituents of R 7 or -NH-l,3-benzodiox-5-yl optionally substituted with 1-3 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula π includes compounds wherein R 3 is l,3-benzodiox-5-yl optionally substituted with 1-3 substituents of R 7 or partially or fully unsaturated 5- or 6-membered monocyclic ring formed of carbon atoms wherein said ring optionally including 1-3 heteroatoms selected from O, N, or S and optionally substituted with 1-5 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein R 3 is a partially or fully unsaturated 5- or 6-membered monocyclic ring formed of carbon atoms wherein said ring optionally including 1-3 heteroatoms selected from O, N, or S and optionally substituted with 1-5 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein R 3 is -NH-partially or fully unsaturated 5- or 6-membered monocyclic ring formed of carbon atoms wherein said ring optionally including 1-3 heteroatoms selected from O, N, or S and optionally substituted with 1-5 substituents of R 7 or a partially or fully unsaturated 5- or 6-membered monocyclic ring formed of carbon atoms wherein said ring optionally including 1-3 heteroatoms selected from O, N, or S

A- 1291 -WO-PCT - 27 - and optionally substituted with 1-5 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein R 4 is H, halo or d- 6 -alkyl, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein R 4 is H or C^-alkyl, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein R 4 is H, F or methyl, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein R 4 is H or methyl, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein R 4 is H, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula π includes compounds wherein R 5 is H, halo, haloalkyl, oxo, Ci- 6 -alkyl, -S-Ci_ 6 -alkyl, -NH-C ]-6 alkyl, -N-di-C 1-6 -alkyl, CN, OH or NH 2 , wherein the C^-alkyl and the Q-e-alkyl portion of -O-C 1-6 -alkyl, -NH-Ci- 6 -alkyl and are optionally substituted independently with 1-5 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula π includes compounds wherein R 5 is H, halo, haloalkyl, -O-C 1-6 -alkyl, -S-C ]-6 -alkyl, -NH-C ]-6 -alkyl, CN, OH OrNH 2 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein R 5 is H, F, Cl, Br, CF 3 , C 2 F 5 , CH 2 CF 3 , methly, ethyl, methoxyl, ethoxyl, CN, OH or NH 2 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein each R 6 , independently, is halo, haloalkyl, Ci-6-alkyl, -S-C 1-O - alkyl, -NH-C^-alkyl, -N-di-C 1-6 -alkyl, CN, OR 7 , NHR 7 , C 2-6 -alkenyl, C 2-6 -alkynyl, C 3-8 - cycloalkyl or C^-cycloalkenyl, wherein the Ci-6-alkyl, C 2-6 -alkenyl, C 2-6 -alkynyl, C 3-8 - cycloalkyl, C 4-8 -cycloalkenyl and the Ci_s-alkyl - and are optionally substituted with 1-5 substituents of R 7 ;

A-1291-WO-PCT - 28 - or R 6 is a fully saturated or partially or fully unsaturated 5- or 6-membered monocyclic or bicyclic ring formed of carbon atoms, said ring optionally including 1-4 heteroatoms selected from O, N, or S and optionally substituted with one or more substituents of R 7 , in conjunction with any of the above or below embodiments. In another embodiment, the compounds of Formula π includes compounds wherein each R 6 , independently, is halo, haloalkyl, -S-Ci -6 - alkyl, -NH-C 1-6 -alkyl, -N-di-C 1-6 -alkyl, CN, OR 7 , NHR 7 , C 2 ^-alkenyl, C 2-6 -alkynyl, C 3-8 - cycloalkyl or C 4-g -cycloalkenyl, wherein the C 2 - 6 -alkenyl, C 2 - 6 -alkynyl, C 3-8 - cycloalkyl, C 4-8 -cycloalkenyl and the C^-alkyl portion of -O-Ci- 6 -alkyl, -S-C^-alkyl, - and are optionally substituted with 1-5 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula π includes compounds wherein R 6 is a fully saturated or partially or fully unsaturated 5- or 6-membered monocyclic or bicyclic ring formed of carbon atoms, said ring optionally including 1-4 heteroatoms selected from O, N, or S and optionally substituted with one or more substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula π includes compounds wherein each R 6 , independently, is halo, haloalkyl, -O-Ci- 6 -alkyl, -S-Cj -6 - alkyl, -NH-Ci -6 -alkyl, CN, OR 7 , NHR 7 , C 2-6 -alkenyl, C 2-6 -alkynyl, C 3-8 - cycloalkyl, C 4-8 -cycloalkenyl or a ring selected from phenyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, triazinyl, thiophenyl, furyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, thiazolyl, oxazolyl, isoxazolyl, isothiazolyl, thiadiazolyl, oxadiazolyl, pyrrolidinyl, oxazolinyl, isoxazolinyl, thiazolinyl, pyrazolinyl, morpholinyl, piperidinyl, piperazinyl and pyranyl, wherein the C^-alkyl, C 2 ^-alkenyl, C 2 ^-alkynyl, C 3-8 - cycloalkyl, C 4-8 -cycloalkenyl, ring and the C ]-6 -alkyl portion of -O-C 1-6 -alkyl, -S-Ci -6 - alkyl, -NH-C ]-6 -alkyl and -N-di-Ci-β-alkyl are optionally substituted with 1-5 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein each R 6 , independently, is halo, haloalkyl, Ci- 6 -alkyl, -S-Ci -6 - alkyl, -NH-C 1-6 -alkyl, -N-di-Co-alkyl, CN, OR 7 , NHR 7 , C 2 ^-alkenyl, C 2-6 -alkynyl, C 3-8 - cycloalkyl or C 4 . 8 -cycloalkenyl, wherein the C 2-6 -alkenyl, C -β-alkynyl, C 3-8 - cycloalkyl, C 4-8 -cycloalkenyl and the C w -alkyl portion - and -N-di-Ci-β-alkyl are optionally substituted with 1-5 substituents of R 7 , in conjunction with any of the above or below embodiments.

A- 1291 -WO-PCT - 29 -

In another embodiment, the compounds of Formula π includes compounds wherein X is CH 2 , CHR 6 , CR 6 R 6 , C(=O), O, NH, NR 6 , or S(O) 0 wherein o is O, 1 or 2, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula π includes compounds wherein X is CH 2 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula π includes compounds wherein X is CHR 6 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein X is CR 6 R 6 , in conjunction with any of the above or below embodiments. In another embodiment, the compounds of Formula π includes compounds wherein X is C(=O), in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein X is O, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula π includes compounds wherein X is NH, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula π includes compounds wherein X is NR 6 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein X is S(O) 0 wherein o is 0, 1 or 2, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein X is CH 2 , CHR 6 , O, NH, NR 6 , or S(O) 0 wherein o is 0, 1 or 2, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein X is CH 2 , C(=O), O, NH, NR 6 , or S(O) 0 wherein o is O, 1 or 2, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein X is C(=O), O, NH, NR 6 , or S(O) 0 wherein o is O, 1 or 2, in conjunction with any of the above or below embodiments. In another embodiment, the compounds of Formula II includes compounds wherein X is O, NH, NR 6 , or S(O) 0 wherein o is O 5 1 or 2, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein X is O, NH or NR 6 , in conjunction with any of the above or below embodiments.

A- 1291 -WO-PCT - 30 -

In another embodiment, the compounds of Formula π includes compounds wherein Z is a 3-6 membered spirocyclic ring formed of carbon atoms optionally including 1-3 heteroatoms selected from O, N and S and optionally substituted independently with 1-5 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula π includes compounds wherein Z is a cyclopropyl, cyclobutyl or cyclopentyl ring wherein 0, 1 or 2 carbon atoms of the ring are, independently, replaced with an oxygen atom and the ring optionally substituted independently with 1-5 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein Z is a ring of

wherein R 7 is as defined herein and p is 0, 1, 2, 3 or 4, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds wherein Z is a 3-6 membered spirocyclic ring formed of carbon atoms optionally including 1-3 heteroatoms selected from O, N and S and optionally substituted independently with 1-5 substituents of R 7 , in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II includes compounds and pharmaceutically acceptable salt forms thereof, wherein

A 1 is CH;

A 2 is CR 6 ; each of A 3 and A 4 , independently, is CH, CR 3 or N, provided no more than one of A 3 and A 4 is N;

A- 1291 -WO-PCT - 31 -

R la is Ci-6-alkyl, C 2-6 alkenyl, C 2-6 -alkynyl, d-ealkyl-O-C^-alkyl-, C ]-6 -alkyl-S- C 1-3 -alkyl-, C 1-6 -alkyl-S(O) 2 -Ci -3 -alkyl-, Ci -6 -alkyl-NH-C 1-3 -alkyl, di-(C 1-3 -alkyl)-N-C, -3 - alkyl, C 2-4 -alkenyl-O-Ci. 3 -alkyl-, C 2-4 -alkenyl-S-Ci -3 -alkyl-, C 2 - 4 -alkenyl-NH-C 1-3 -alkyl- or C 2 - 4 -alkynyl-NH-Ci -3 -alkyl-, wherein the alkyl, alkenyl or alkynyl moiety of each is optionally substituted with 1-3 substituents of R 7 ;

R lb is H, halo, haloalkyl, C^-alkyl, -O-C 1-6 -alkyl, -S-C 1-6 -alkyl, -NH-C^-alkyl, - N-di-Ci-6-alkyl, CN, OH OrNH 2 ; alternatively, R la and R lb taken together with the carbon atom to which they are attached form a partially or fully saturated 5- or 6-membered ring of carbon atoms optionally including 1-2 heteroatoms selected from O, N, or S, the ring optionally substituted independently with 1-3 substituents of R 7 ;

R lc is H;

W is -C(=0)-;

R 2 is C]-C 3 alkyl, -OCi-C 3 alkyl, C 2 -alkenyl or C 2 -alkynyl, wherein each of said Ci-C 3 alkyl, C 2 -alkenyl and C 2 -alkynyl is optionally substituted independently with 1-2 substituents of R 7 ;

R 3 is a ring selected from phenyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, triazinyl, thiophenyl, furyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, thiazolyl, oxazolyl, isoxazolyl, isothiazolyl, thiadiazolyl, oxadiazolyl, pyrrolidinyl, oxazolinyl, isoxazolinyl, thiazolinyl, pyrazolinyl, morpholinyl, piperidinyl, piperazinyl and pyranyl, said ring optionally substituted with 1-5 substituents of R 7 ;

R 4 is H or d- 4 -alkyl;

R 5 is H, halo, haloalkyl, Ci -6 -alkyl, -O-C 1-6 -alkyl, -S-C 1-6 -alkyl, -NH-Ci -6 -alkyl, -N-di-C 1-6 -alkyl, CN, OH or NH 2 ; X is CH 2 , CHR 6 , C(O) or O;

Z is a cyclopropyl, cyclobutyl or cyclopentyl ring wherein 0, 1 or 2 carbon atoms of the ring are, independently, replaced with an oxygen atom and the ring optionally substituted independently with 1-5 substituents of R 7 ; and each R 6 , independently, is halo, haloalkyl, -S-Ci^- alkyl, -NH-C^-alkyl, -N-di-C^-alkyl, CN, OR 7 , NHR 7 , C 2 ^-alkenyl, C 2-6 -alkynyl, C 3-8 - cycloalkyl, C 4 _g-cycloalkenyl or a ring selected from phenyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, triazinyl, thiophenyl, furyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, thiazolyl, oxazolyl, isoxazolyl, isothiazolyl, thiadiazolyl, oxadiazolyl, pyrrolidinyl, oxazolinyl, isoxazolinyl, thiazolinyl, pyrazolinyl, morpholinyl, piperidinyl,

A- 1291 -WO-PCT - 32 - piperazinyl and pyranyl, wherein the C 2-6 -alkenyl, C 2-6 -alkynyl, C 3-8 - cycloalkyl, C 4- g-cycloalkenyl, ring and portion -S-Ci- 6 - alkyl, and are optionally substituted with 1-5 substituents ofR 7 .

In another embodiment, the invention provides compounds, and pharmaceutically acceptable salt forms thereof, having a general Formula II-A:

wherein one of A 3 and A 4 , independently, is CR 3 and the other of A 3 and A 4 , independently, is N, CH or CR 6 ;

R la is H, halo, haloalkyl, C 1-6 -alkyl, -O-C^-alkyl, -S-C^-alkyl, -NH-C 1-6 -alkyl, - portion of -O- C 1-6 -alkyl, -S-Ci -6 -alkyl, -NH-Ci-β-alkyl and are optionally substituted independently with 1-5 substituents of R 7 ;

R lb is H, halo, haloalkyl, C^-alkyl, -O-C^-alkyl, -S-C^-alkyl, -NH-C^-alkyl, - portion of -O- C 1-6 -alkyl, -NH-Ci-6-alkyl and -N-di-Ci-6-alkyl are optionally substituted independently with 1-5 substituents of R 7 ; alternatively, R la and R lb taken together with the carbon atom to which they are attached form a partially or fully saturated A-, 5- or 6-membered ring of carbon atoms optionally including 1-2 heteroatoms selected from O, N, or S, the ring optionally substituted independently with 1-3 substituents of R 7 ;

R lc is H, F, Cl, methyl, ethyl, methoxyl, ethyoxyl, CN, OH OrNH 2 ;

R 2 is Ci-C 3 alkyl, -OCi-C 3 alkyl, C 2 -alkenyl or C 2 -alkynyl, wherein each of said C]-C 3 alkyl, -OC 1 -C 3 alkyl, C 2 -alkenyl and C 2 -alkynyl is optionally substituted independently with 1-2 substituents of R 7 ;

R 3 is a ring selected from phenyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, triazinyl, thiophenyl, furyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, thiazolyl,

A- 1291 -WO-PCT - 33 - oxazolyl, isoxazolyl, isothiazolyl, thiadiazolyl, oxadiazolyl, pyrrolidinyl, oxazolinyl, isoxazolinyl, thiazolinyl, pyrazolinyl, morpholinyl, piperidinyl, piperazinyl and pyranyl, said ring optionally substituted with 1-5 substituents of R 7 ;

R 4 is H, halo or C M -alkyl; R 5 is H, F, CF 3 , C M -alkyl, -O-Cwalkyl, -S-C M -alkyl, -NH-C M -alkyl,

CN, OH or NH 2 , wherein the and the C 1-4 -alkyl portion of -O-C M -alkyl, -S-C 1- 4 -alkyl and -N-Ci- 4 -alkyl are optionally substituted independently with 1-3 substituents of halo or OH; each R 6 , independently, is halo, haloalkyl, C^-alkyl, -O-Ci -6 -alkyl, -S-Ci -6 - alkyl, -NH-d-e-alkyl, -N-di-C w -alkyl, CN, OR 7 , NHR 7 , C 2-6 -alkenyl, C 2-6 -alkynyl, C 3-8 - cycloalkyl or C 4-8 -cycloalkenyl, wherein the C 2-6 -alkenyl, C 2 ^-alkynyl, C 3-8 - cycloalkyl, C 4-8 -cycloalkenyl and the portion of -O-Ci- 6 -alkyl, - are optionally substituted with 1-5 substituents of R 7 ; each R 7 , independently, is H, halo, haloalkyl, CN, OH, NO 2 , NH 2 , acetyl, oxo, Ci- l o-alkyl, C 2- io-alkenyl, C 2- io-alkynyl, Q-io-cycloalkyl, C^io-cycloalkenyl, C MO - alkylamino-, Ci.io-dialkylamino-, Ci.io-alkoxyl, Ci-io-thioalkoxyl or a fully saturated or partially or fully unsaturated 3-8 membered monocyclic or a 6-12 membered bicyclic, said ring system formed of carbon atoms optionally including 1-3 heteroatoms if monocyclic or 1-6 heteroatoms if bicyclic, said heteroatoms selected from O, N, or S, wherein each of the Ci.io-alkyl, C 2- io-alkenyl, C 2- io-alkynyl, C 4- io- cycloalkenyl, Ci.io-alkylamino-, Ci-io-dialkylamino-, Ci.io-alkoxyl, Ci.io-thioalkoxyl and ring of said ring system is optionally substituted independently with 1-5 substituents of halo, haloalkyl, CN, NO 2 , NH 2 , OH, oxo, methyl, methoxyl, ethyl, ethoxyl, propyl, propoxyl, isopropyl, isopropoxyl, cyclopropyl, cyclopropylmethoxyl, butyl, butoxyl, isobutoxyl, tert-butoxyl, isobutyl, sec-butyl, tert-butyl, cyclobutyl, pentyl, cyclopentyl, hexyl, cyclohexyl, Ci.io-alkylamino-, Ci-io-dialkylamino-, Ci.io-thioalkoxyl, benzyl or phenyl; and p is O, 1 or 2.

In another embodiment, the compounds of Formula EI-A include compounds wherein p is O or 1, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II-A include compounds wherein p is 1, in conjunction with any of the above or below embodiments. In another embodiment, the compounds of Formula II-A include

A-1291-WO-PCT - 34 - compounds wherein p is 0, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of II- A include compounds wherein R la is Ci-6-alkyl, -O-C 1-6 -alkyl, -S-d-e-alkyl, -NH-C 1-6 -alkyl, wherein the Ci -6 - alkyl and the C 1-6 -alkyl portion of -O-C 1-6 -alkyl, is optionally substituted independently with 1-5 substituents of R 7 ;

R lb is H, halo, haloalkyl, C^-alkyl, -O-C 1-6 -alkyl, -S-C,^-alkyl, -NH-C 1-6 -alkyl, wherein the portion of -O-Ci-6-alkyl, -S-Ci-β-alkyl and -NH- is optionally substituted independently with 1-5 substituents of R 7 ; alternatively, R la and R lb taken together with the carbon atom to which they are attached form a partially or fully saturated A-, 5- or 6-membered ring of carbon atoms optionally including 1 or 2 oxygen atoms and optionally substituted independently with 1- 3 substituents of R 7 ; and

R lc is H, F, Cl, methyl or ethyl, in conjunction with any of the above or below embodiments.

In another embodiment, the compounds of Formula II-A includes compounds wherein each of the specific embodiments for A 3 , A 4 , R la , R lb , R lc , R 2 , R 2a , R 3 , R 4 , R 5 , R 6 , R 7 , m, n and p, respectively, of compounds of Formula π above apply for compounds of Formula II-A.

In another embodiment, the invention provides compounds, and pharmaceutically acceptable salt forms thereof, having a general Formula II-B:

π-B wherein each of the independent embodiments for A 3 , R la , R lb , R lc , R 2 , R 2a , R 3 , R 4 , R 5 , R 6 , R 7 and p, respectively, of compounds of Formulas II and II-A apply for compounds of Formula II-B.

In another embodiment, the invention provides compounds of Formula II-C:

A- 1291 -WO-PCT - 35 -

π-c or a pharmaceutically acceptable salt thereof, wherein each of A 1 and A 2 , independently, is CH or CR 6 ; one of A 3 and A 4 , independently, is CH or CR 3 and the other of A 3 and A 4 , independently, is N, CH or CR 6 ;

R la is H, halo, haloalkyl, - N-di-C^-alkyl, CN, OH or NH 2 , wherein the C ]-6 -alkyl portion of -O-C^-alkyl, -S-C,^- alkyl, -NH-Ci-β-alkyl and are optionally substituted independently with 1-5 substituents of R 7 ;

R lb is H, halo, haloalkyl, C^-alkyl, -O-C^-alkyl, -S-C^-alkyl, -NH-C 1-6 -alkyl, - N-di-C 1-6 -alkyl, CN, OH or NH 2 , wherein the portion of -O-C^-alkyl, -S-C u - alkyl, and are optionally substituted independently with 1-5 substituents of R 7 ; alternatively, R la and R lb taken together with the carbon atom to which they are attached form a partially or fully saturated 3-, 4-, 5- or 6-membered ring of carbon atoms optionally including 1-2 heteroatoms selected from O, N, or S, the ring optionally substituted independently with 1-3 substituents of R 7 ;

R lc is H, halo, haloalkyl, -NH-C 1-6 -alkyl, - N-di-C 1-6 -alkyl, CN, OH or NH 2 ;

W is -C(O)-, -C(=S)-, - OC(=O) -, -NHC(O)-, -S(OV or -NHS(OV, wherein b is 1 or 2;

R 2 is C 1 -C 3 alkyl, -OC r C 3 alkyl, C 2 -C 3 alkenyl or C 2 -C 3 alkynyl, wherein each of said C 1 -C 3 alkyl, -OC r C 3 alkyl, C 2 -C 3 alkenyl and C 2 -C 3 alkynyl is optionally substituted independently with 1-3 substituents of R 7 ;

R 3 is -NH-partially or fully unsaturated 5- or 6-membered monocyclic ring formed of carbon atoms wherein said ring optionally including 1-3 heteroatoms selected from O, N, or S and optionally substituted with 1-5 substituents of R 7 , -NH-l,3-benzodiox-5-yl optionally substituted with 1-3 substituents of R 7 , 1,3-

A- 1291 -WO-PCT - 36 - benzodiox-5-yl optionally substituted with 1-3 substituents of R 7 or partially or fully unsaturated 5- or 6-membered monocyclic ring formed of carbon atoms wherein said ring optionally including 1-3 heteroatoms selected from O, N, or S and optionally substituted with 1-5 substituents of R 7 ; R 4 is H, halo or C^-alkyl;

R 5 is H, halo, haloalkyl, oxo, C^-alkyl, -O-C^-alkyl, -S-C 1-6 -alkyl, -NH-C 1-6 alkyl, -N-di-Ci -6 -alkyl, CN, OH or NH 2 , wherein the C 1-6 -alkyl and the portion of -0-Ci.β-alkyl, -S-Ci-β-alkyl, and -N-di-Ci_6-alkyl are optionally substituted independently with 1-5 substituents of R 7 ; X is CH 2 , CHR 6 , CR 6 R 6 , C(=O), O, NH, NR 6 , or S(O) 0 wherein o is O, 1 or 2;

Z is a 3-6 membered spirocyclic ring formed of carbon atoms optionally including 1-3 heteroatoms selected from O, N and S and optionally substituted independently with 1-5 substituents of R 7 ; each R 6 , independently, is halo, haloalkyl, C 1-6 -alkyl, CN, OR 7 , NHR 7 , -S-C^-alkyl, -N-di-Ci -6 -alkyl, C 2-6 -alkenyl, C 2-6 -alkynyl, C 3 . 8 -cycloalkyl or C 4-8 - cycloalkenyl, wherein the Ci- 5 -alkyl, C 2 -6-alkenyl, C 2 . 6 -alkynyl, Cs-g-cycloalkyl, C 4-8 - cycloalkenyl and the portion and are optionally substituted with 1-5 substituents of R 7 ; or R 6 is a fully saturated or partially or fully unsaturated 5- or 6-membered monocyclic or bicyclic ring formed of carbon atoms, said ring optionally including 1-4 heteroatoms selected from O, N, or S and optionally substituted with one or more substituents of R 7 ; and each R 7 , independently, is H, halo, haloalkyl, CN, OH, NO 2 , NH 2 , acetyl, oxo, Q- 10 -alkyl, C 2- io-alkenyl, C 2- i 0 -alkynyl, Cs-io-cycloalkyl, C 4- io-cycloalkenyl, C MO - alkylamino-, Cj.io-dialkylamino-, Cj.io-alkoxyl, Ci-io-thioalkoxyl or a fully saturated or partially or fully unsaturated 3-8 membered monocyclic or a 6-12 membered bicyclic, said ring system formed of carbon atoms optionally including 1-3 heteroatoms if monocyclic or 1-6 heteroatoms if bicyclic, said heteroatoms selected from O, N, or S, wherein each of the Ci-io-alkyl, C 2- i 0 -alkenyl, C 2- io-alkynyl, Cs^o-cycloalkyl, C 4- io- cycloalkenyl, Ci -10 -alkylamino-, Ci-io-dialkylamino-, Ci.io-alkoxyl, Q.io-thioalkoxyl and ring of said ring system is optionally substituted independently with 1-5 substituents of halo, haloalkyl, CN, NO 2 , NH 2 , OH, oxo, methyl, methoxyl, ethyl, ethoxyl, propyl, propoxyl, isopropyl, isopropoxyl, cyclopropyl, cyclopropylmethoxyl, butyl, butoxyl, isobutoxyl, tert-butoxyl, isobutyl, sec-butyl, tert-butyl, cyclobutyl, pentyl, cyclopentyl,

A-1291-WO-PCT - 37 - hexyl, cyclohexyl, Ci.io-alkylamino-, Ci-urdialkylamino-, Ci.io-thioalkoxyl, benzyl or phenyl; provided the compound is not

N-((lS)-l-((lR)-2-(((4 I S)-6 t -(2,2-dimethylpropyl)-3 t ,4 I -dihydrospiro[cyclobutane- 1 ,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l -hydroxyethyl)-3-methylbutyl)acetamide;

N-((lS)-l-((lR)-2-(((4 I S)-6 1 -(2,2-dime%lpropyl)-3',4'-dihydrospiro[cyclobutane- l,2 l -pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-3-methyl -3-buten-l- yl)acetamide;

N-((lS)-l-((lR)-2-(((4'S)-6 t -(2,2-dimethylpropyl)-3',4 l -dihydrospiro[cyclobutane- l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-3-pe ntyn-l-yl)acetamide;

N-(( 1 S)- 1 -(( lR)-2-(((4'S)-6'-(2,2-dimethylpropyl)-3 ',4'-dihydrospiro[cyclobutane- l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-4-pe nten-l-yl)acetamide;

N-((lS)-l-((lR)-2-(((ls,3R,4'S)-6'-(2,2-dimethylpropyl)-3 -methyl-3',4'- dihydrospiro[cyclobutane-l,2'-pyrano[2,3-b]pyridin]-4'-yl)am ino)-l-hydroxyethyl)-4- penten- 1 -yl)acetamide;

N-((lS,2R)-3-(((4S)-6-ethyl-3,4-dihydrospiro[chromene-2,l '-cyclobutan]-4- yl)amino)-2-hydroxy- 1 -((methyloxy)methyl)propyl)acetamide;

N-((lS,2R)-3-(((4S)-6-ethyl-3,4-dihydrospiro[chromene-2,l '-cyclobutan]-4- yl)amino)-2-hydroxy- 1 -((propyloxy)methy l)propyl)acetamide; N-(( 1 S,2R)-3 -(((4S)-6-ethyl-3 ,4-dihydrospiro[chromene-2, 1 '-cyclobutan]-4- yl)amino)-2-hydroxy- 1 -(((phenyhnethyl)oxy)methyl)propyl)acetamide;

N-((lS)-l-((lR)-2-(((4 l S)-6 1 -(2,2-dimethylpropyl)-3',4'-dihydrospiro[cyclobutane- l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-3-bu ten-l-yl)acetamide;

N-((lS)-l-((lS)-2-(((4 I S)-6'-(2,2-dimethylpropyl)-3',4 I -dihydrospiro[cyclobutane- l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-3-bu ten-l-yl)acetamide;

N-((lS)-l-((lR)-2-(((4 l S)-6'-(2,2-dimethylpropyl)-3',4 I -dihydrospiro[cyclobutane- l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-4,4, 4-trifluorobutyl)acetamide;

N-((lS,3E)-l-((lR)-2-(((4'S)-6'-(2,2-dimethylpropyl)-3',4 '- dihydrospiro[cyclobutane-l,2'-pyrano[2,3-b]pyridin]-4'-yl)am ino)-l-hydroxyethyl)-3- penten- l-yl)acetamide;

N-((lS)-l-((lR)-2-(((4'S)-6 t -(2,2-dimethylpropyl)-3',4'-dihydrospiro[cyclobutane- 1 ,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)- 1 -hydroxyethyl)-2-propen- 1 -yl)acetaraide;

A-1291-WO-PCT - 38 -

N-(( 1 S)- 1 -(( 1 R>2-(((4'S)-6'-(2-fluoro-2-methylpropyl)-3',4 I - dihydrospiro[cyclobutane- 1 ,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)- 1 -hydroxyethyl)-3- buten-l-yl)acetamide; and

N-((lS)-l-((lR)-2-(((4 I S)-6 1 -(2,2-dimethylpropyl)-3 I ,4'-dihydrospiro[cyclobutane- 1 ,2'-pyrano [2,3 -b]pyridin]-4'-yl)amino)- 1 -hydroxyethyl)-3 -hexyn- 1 -yl)acetamide.

In another embodiment, the invention provides compounds, and pharmaceutically acceptable salt forms thereof, wherein each of the independent embodiments for Formulas π and π-A apply for compounds of Formula II-C.

In another embodiment, the invention provides compounds, and pharmaceutically acceptable salt forms thereof, having a general Formula II-D:

π-D wherein each of A 3 , R la , R lb , R lc , R 2 , R 2a , R 3 , R 4 , R 5 , R 6 , R 7 and p are defined herein with respect to Formula II, II-A and II-B above. In another embodiment, the invention provides the compound of Formula π, or a pharmaceutically acceptable salt thereof, selected from

N-((lS,2R)-l-(2-cyclobutylideneethyl)-3-(((4 1 S)-6'-(2,2-dimethylpropyl)-3',4 t - dihydrospiro[cyclobutane-l,2'-pyrano[2,3-b]pyridin]-4'-yl)am ino)-2- hydroxypropyl)acetamide; N-((lS)-l-((lR)-2-(((2r,3'R,4S)-6-(2,2-dimethylpropyl)-3'-hy droxy-3,4- dihydrospiro[chromene-2, 1 '-cyclobutan]-4-yl)amino)- 1 -hydroxyethyl)-3 -buten- 1 - yl)acetamide;

N-((lS)-l-((lR)-2-(((4'S)-6 I -(2,2-difluoropropyl)-3',4'-dihydrospiro[cyclobutane- l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-3-bu ten-l-yl)-2- (methyloxy)acetamide;

N-(( 1 S)- 1 -(( 1 R)- 1 -hydroxy-2-(((4'S)-6'-((2R)-3 ,3 ,3 -trifluoro-2-methylpropyl)- 3 l ,4'-dihydrospiro[cyclobutane-l,2'-pyrano[2,3-b]pyridin]-4'-y l)ammo)ethyl)-3-buten-l- yl)acetamide;

A-1291-WO-PCT - 39 -

N-((lS)-l-((lR)-2-(((4 1 S)-6'-(2,2-dimethylpropyl)-3 I ,4 I -dihydrospiro[cyclobutane- l,2'-pyrano[2,3-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)-3-bu ten-l-yl)-2,2- difluoropropanamide;

(2R)-N-(( 1 S)- 1 -(( 1 R)- 1 -hydroxy-2-(((4S)-8-( 1 H-ϊmidazol- 1 -yl)-6-(2- methylpropyl)-3 ,4-dihydrospiro[chromene-2, 1 '-cyclobutan]-4-yl)amino)ethyl)-3 -buten- 1 - yl)-2-methoxypropanamide;

N-(( 1 S)- 1 -(( 1 R)- 1 -hydroxy-2-(((4S)-6-(2-methylpropyl)-8-( 1 ,3-thiazol-2-yl)-3 5 4- dihydrospiro[chromene-2,r-cyclobutan]-4-yl)amino)ethyl)-3-bu ten-l-yl)-2- methoxyacetamide; N-((lS)-l-((lR)-2-(((4S)-6-(2,2-dimethylpropyl)-8-(lH-imidaz ol-l-yl)-3,4- dihydrospiro[chromene-2, 1 '-cyclobutan]-4-yl)amino)-l -hydroxyethyl)-3 -buten- 1 -yl)-2- methoxyacetamide;

N-((lS)-l-((lR)-2-(((4S)-6-(2,2-dimethylpropyl)-8-(lH-imi dazol-5-yl)-3,4- dihydrospiro[chromene-2, 1 '-cyclobutan]-4-yl)amino)- 1 -hydroxyethyl)-3 -buten- 1 -yl)-2- methoxyacetamide;

N-(( 1 S)- 1 -(( 1 R)-2-(((4 I S)-6 l -(2,2-dimethylpropyl)-8'-(methylamino)-3 ',4'- dihydrospirofcyclobutane-l^'-pyranoP^-cJpyridinl^'-yOaminoyi -hydroxyethyl^S- butyn-1 -yl)acetamide;

(2R)-N-((lS)-l-((lR)-2-(((4 1 S)-6'-(2,2-dimethylpropyl)-8'-(methylamino)-3 l ,4 1 - dihydrospiro[cyclobutane- 1 ,2'-pyrano[2,3-c]pyridin]-4'-yl)amino)- 1 - hydroxyethyl)butyl)tetrahydro-2-fiirancarboxamide;

N-(( 1 S)- 1 -(( lR)-2-(((4'S)-6 1 -(2,2-dimethylpropyl)-8'-(phenylamino)-3 ',4'- dihydrospiro[cyclobutane-l,2'-pyrano[2,3-c]pyridin]-4'-yl)am ino)-l-hydroxyethyl)-3- buten-1 -yl)acetamide; N-((lS)-l-((lR)-2-(((4S)-8-(l,3-benzodioxol-5-yl)-6-(2,2-dim ethylpropyl>3,4- dihydrospiro[chromene-2, 1 '-cyclobutan]-4-yl)amino)- 1 -hydroxyethyl)-3 -buten- 1 -yl)-2- methoxyacetamide;

N-αiS^l-CClR^-C^'S^S'-Cl^-benzodioxol-S-ylamino^ό 1 ^^- dimethylpropy^-S'^'-dihydrospirotcyclobutane-l^'-pyranoPjS-c Jpyridinj^'-y^amino)- l-hydroxyethyl)-3 -buten- l-yl)-2-methoxyacetamide; and

N-((lS)-l-((lR)-2-(((4 1 S)-6'-(2,2-dimethylpropyl)-8 1 -(l-methyl-lH-imidazol-5- yl)-3',4'-dihydrospiro[cyclobutane-l,2'-pyrano[2,3-c]pyridin ]-4'-yl)amino)-l- hydroxyethyl)-3-buten-l-yl)-2-methoxyacetamide.

A-1291-WO-PCT - 40 -

In another embodiment, the invention provides each of the Examplary compounds, and stereoisomers, tautomers, solvates, pharmaceutically acceptable salts, derivatives or prodrugs thereof, and related intermediates, described herein.

DEFINITIONS

The following definitions should assist in understanding the invention described herein.

The term "comprising" is meant to be open ended, i.e., all encompassing and non- limiting. It may be used herein synonymously with "having." Comprising is intended to include each and every indicated component while not excluding any other components or elements.

The term "C α- palkyl", when used either alone or within other terms such as "haloalkyl" and "alkylamino", embraces linear or branched radicals having α to β number of carbon atoms (such as C 1 -Ci 0 ; Ci-C 6; or C 1 -C 4 ). Unless otherwise specified, one or more carbon atoms of the "alkyl" radical may be substituted, such as with a cycloalkyl moiety. Examples of "alkyl" radicals include methyl, cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, ethyl, cyclopropylethyl, cyclobutylethyl, cyclopentylethyl, n-propyl, isopropyl, n-butyl, cyclopropylbutyl, isobutyl, .fee-butyl, tert- butyl, pentyl, isoamyl, hexyl and the like. The term "C α _ β alkenyl", when used alone or in combination, embraces linear or branched radicals having at least one carbon-carbon double bond in a moiety having a number of carbon atoms in the range from α and β. Included within alkenyl radicals are "lower alkenyl" radicals having two to about six carbon atoms and, for example, those radicals having two to about four carbon atoms. Examples of alkenyl radicals include, without limitation, ethenyl, propenyl, allyl, propenyl, butenyl and 4-methylbutenyl. The terms "alkenyl" and "lower alkenyl", embrace radicals having "cis" and "trans" orientations, or alternatively, "E" and "Z" orientations, as appreciated by those of ordinary skill in the art.

The term "C a-P alkynyl", when used alone or in combination, denotes linear or branched radicals having at least one carbon-carbon triple bond in a moiety having a number of carbon atoms in the range from α and β. Examples of alkynyl radicals include "lower alkynyl" radicals having two to about six carbon atoms and, for example,

A-1291-WO-PCT - 41 - lower alkynyl radicals having two to about four carbon atoms. Examples of such radicals include, without limitation, ethynyl, propynyl (propargyl), butynyl, and the like.

The term "C a-p -alkyl", "C α-β -alkenyl" and "C α-β -alkynyl", when used with other terms such as " wherein 1, 2 or 3 carbon atoms of said C α-p -alkyl, C α-p -alkenyl or C 2α-β - alkynyl is optionally replaced with a heteroatom selected from O, S, S(O), S(O) 2 and N" embraces linear or branched radicals wherein one or more of the carbon atoms may be replaced with a heteroatom. Examples of such "alkyl" radicals include -O-methyl, -O- ethyl, -CH 2 -O-CH 3 , -CH 2 CH 2 -O-CH 3 , -NH-CH 2 , -CH 2 CH 2 -N(CH 3 )-CH 3 , -S-(CH 2 ) 3 CH 2 , -CH 2 CH 2 -S-CH 3 and the like. Accordingly, such radicals also include radicals encompassed by -OR 7 where R 7 may be defined as a C α . p -alkyl. Examples of such "alkenyl" radicals include -NH-CH 2 CH=CH 2 , -S-CH 2 CH 2 CH=CHCH 3 and the like. Simlar examples exist for such "alkynyl" radicals, as appreciated by those skilled in the art.

The term "C α-P alkoxyl" when used alone or in combination, embraces linear or branched oxygen-containing alkyl radicals each having α to β number of carbon atoms (such as C 1 -C] 0 ). The terms "alkoxy" and "alkoxyl", when used alone or in combination, embraces linear or branched oxygen-containing radicals each having alkyl and substituted alkyl portions of one or more carbon atoms. Examples of such radicals include methoxy, ethoxy, propoxy, butoxy and tert-butoxy. Alkoxy radicals may be further substituted with one or more halo atoms, such as fluoro, chloro or bromo, to provide "haloalkoxy" radicals or with other substitution. Examples of such radicals include fluoromethoxy, chloromethoxy, trifluoromethoxy, trifluoroethoxy, fluoroethoxy, fluoropropoxy and cyclopropylmethoxy.

The term "aryl", when used alone or in combination, means a carbocyclic aromatic moiety containing one, two or even three rings wherein such rings may be attached together in a fused manner. Every ring of an "aryl" multi-ring system need not be aromatic, and the ring(s) fused to the aromatic ring may be partially or fully unsaturated and include one or more heteroatoms selected from nitrogen, oxygen and sulfur. Thus, the term "aryl" embraces aromatic radicals such as phenyl, naphthyl, indenyl, tetrahydronaphthyl, dihydrobenzafuranyl, anthracenyl, indanyl, benzodioxazinyl, and the like. The "aryl" group may be substituted, such as with 1 to 5 substituents including lower alkyl, hydroxyl, halo, haloalkyl, nitro, cyano, alkoxy and lower

A- 1291 -WO-PCT - 42 - alkylamino, and the like. Phenyl substituted with -0-CH 2 -O- or -0-CH 2 -CH 2 -O- forms an aryl benzodioxolyl substituent.

The term "carbocyclic", also referred to herein as "cycloalkyl", when used alone or in combination, means a partially or fully saturated ring moiety containing one ("monocyclic"), two ("bicyclic") or even three ("tricyclic") rings wherein such rings may be attached together in a fused manner and formed from carbon atoms. Examples of saturated carbocyclic radicals include saturated 3 to 6-membered monocyclic groups such as cyclopropane, cyclobutane, cyclopentane and cyclohexane.

The terms "ring" and "ring system" refer to a ring comprising the delineated number of atoms, the atoms being carbon or, where indicated, a heteroatom such as nitrogen, oxygen or sulfur. Where the number of atoms is not delineated, such as a "monocyclic ring system" or a "bicyclic ring system", the numbers of atoms are 3-8 for a monocyclic and 6-12 for a bicyclic ring. The ring itself, as well as any substitutents thereon, may be attached at any atom that allows a stable compound to be formed. The term "nonaromatic" ring or ring system refers to the fact that at least one, but not necessarily all, rings in a bicyclic or tricyclic ring system is nonaromatic.

The term "cycloalkenyl", when used alone or in combination, means a partially or fully saturated cycloalkyl containing one, two or even three rings in a structure having at least one carbon-carbon double bond in the structure. Examples of cycloalkenyl groups include C 3 -C 6 rings, such as compounds including, without limitation, cyclopropene, cyclobutene, cyclopentene and cyclohexene. The term also includes carbocyclic groups having two or more carbon-carbon double bonds such as "cycloalkyldienyl" compounds. Examples of cycloalkyldienyl groups include, without limitation, cyclopentadiene and cycloheptadiene. The term "halo", when used alone or in combination, means halogens such as fluorine, chlorine, bromine or iodine atoms.

The term "haloalkyl", when used alone or in combination, embraces radicals wherein any one or more of the alkyl carbon atoms is substituted with halo as defined above. For example, this term includes monohaloalkyl, dihaloalkyl and polyhaloalkyl radicals such as a perhaloalkyl. A monohaloalkyl radical, for example, may have either an iodo, bromo, chloro or fluoro atom within the radical. Dihalo and polyhaloalkyl radicals may have two or more of the same halo atoms or a combination of different halo radicals. Examples of haloalkyl radicals include fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, pentafluoroethyl,

A-1291-WO-PCT - 43 - heptafluoropropyl, difluorochloromethyl, dichlorofluoromethyl, difluoroethyl, difluoropropyl, dichloroethyl and dichloropropyl. "Perfluoroalkyl", as used herein, refers to alkyl radicals having all hydrogen atoms replaced with fluoro atoms. Examples include trifluoromethyl and pentafluoroethyl. The term "heteroaryl", as used herein, either alone or in combination, means a fully unsaturated (aromatic) ring moiety formed from carbon atoms and having one or more heteroatoms selected from nitrogen, oxygen and sulfur. The ring moiety or ring system may contain one ("monocyclic"), two ("bicyclic") or even three ("tricyclic") rings wherein such rings are attached together in a fused manner. Every ring of a "heteroaryl" ring system need not be aromatic, and the ring(s) fused thereto (to the heteroaromatic ring) may be partially or fully saturated and optionally include one or more heteroatoms selected from nitrogen, oxygen and sulfur. The term "heteroaryl" does not include rings having ring members of -O-O-, -O-S- or -S-S-.

Examples of unsaturated heteroaryl radicals, include unsaturated 5- to 6- membered heteromonocyclyl groups containing 1 to 4 nitrogen atoms, including for example, pyrrolyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazolyl [e.g., 4H-l,2,4-triazolyl, IH-1, 2,3 -triazolyl, 2H-1,2,3- triazolyl] and tetrazole; unsaturated 7- to 10- membered heterobicyclyl groups containing 1 to 4 nitrogen atoms, including for example, quinolinyl, isoquinolinyl, quinazolinyl, isoquinazolinyl, aza-quinazolinyl, and the like; unsaturated 5- to 6-membered heteromonocyclic group containing an oxygen atom, for example, pyranyl, 2-furyl, 3- furyl, benzofuryl, etc.; unsaturated 5 to 6-membered heteromonocyclic group containing a sulfur atom, for example, 2-thienyl, 3-thienyl, benzothienyl, etc.; unsaturated 5- to 6- membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, for example, oxazolyl, isoxazolyl, oxadiazolyl [e.g., 1,2,4-oxadiazolyl, 1,3,4- oxadiazolyl, 1,2,5-oxadiazolyl]; unsaturated 5 to 6-membered heteromonocyclic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms, for example, thiazolyl, isothiazolyl, thiadiazolyl [e.g., 1,2,4-thiadiazolyl, 1,3,4-thiadiazolyl, 1,2,5-thiadiazolyl]. The term "heterocyclic", when used alone or in combination, means a partially or fully saturated ring moiety containing one, two or even three rings wherein such rings may be attached together in a fused manner, formed from carbon atoms and including one or more heteroatoms selected from N, O or S. Examples of saturated heterocyclic radicals include saturated 3 to 6-membered heteromonocyclic groups containing 1 to 4 nitrogen atoms [e.g. pyrrolidinyl, imidazolidinyl, piperidinyl, pyrrolinyl, piperazinyl]; saturated 3

A- 1291 -WO-PCT - 44 - to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms [e.g. morpholinyl]; saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms [e.g., thiazolidinyl]. Examples of partially saturated heterocyclyl radicals include dihydrothienyl, dihydropyranyl, dihydrofuryl and dihydrothiazolyl.

The term "heterocycle" also embraces radicals where heterocyclic radicals are fused/condensed with aryl radicals: unsaturated condensed heterocyclic group containing 1 to 5 nitrogen atoms, for example, indolyl, isoindolyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, indazolyl, benzotriazolyl, tetrazolopyridazinyl [e.g., tetrazolo [1,5- b]pyridazinyl]; unsaturated condensed heterocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms [e.g. benzoxazolyl, benzoxadiazolyl]; unsaturated condensed heterocyclic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms [e.g., benzothiazolyl, benzothiadiazolyl]; and saturated, partially unsaturated and unsaturated condensed heterocyclic group containing 1 to 2 oxygen or sulfur atoms [e.g. benzofuryl, benzothienyl, 2,3-dihydro-benzo[l,4]dioxinyl and dihydrobenzofuryl]. Examples of heterocyclic radicals include five to ten membered fused or unfused radicals.

Examples of partially saturated and fully saturated heterocyclyls include, without limitation, pyrrolidinyl, imidazolidinyl, piperidinyl, pyrrolinyl, pyrazolidinyl, piperazinyl, morpholinyl, tetrahydropyranyl, thiazolidinyl, dihydrothienyl, 2,3-dihydro- benzo[l,4]dioxanyl, indolinyl, isoindolinyl, dihydrobenzothienyl, dihydrobenzofuryl, isochromanyl, chromanyl, 1,2-dihydroquinolyl, 1,2,3,4-tetrahydro-isoquinolyl, 1,2,3,4- tetrahydro-quinolyl, 2,3,4,4a,9,9a-hexahydro-lH-3-aza-fluorenyl, 5,6,7-trihydro- 1,2,4- triazolo[3,4-a]isoquinolyl, 3,4-dihydro-2H-benzo[l,4]oxazinyl, benzo[l,4]dioxanyl, 2,3- dihydro-lH-lλ'-benzo[d]isothiazol-6-yl, dihydropyranyl, dihydrofuryl and dihydrothiazolyl, and the like.

The phrase "a saturated or partially or fully unsaturated 3-8 membered monocyclic or a 6-12 membered bicyclic, said ring system formed of carbon atoms optionally including 1-3 heteroatoms if monocyclic or 1-6 heteroatoms if bicyclic, said heteroatoms selected from O, N, or S" as used herein is intended to encompass all monocyclic and bicyclic rings as small as three atoms to as large as 12 atoms in size, including both carbocyclic rings and heterocyclic, aromatic and non-aromatic rings. The non-aromatic rings may be partially or fully saturated in nature.

The term "alkylamino" includes "N-alkylamino" where amino radicals are independently substituted with one alkyl radical. Preferred alkylamino radicals are "lower

A-1291-WO-PCT - 45 - alkylamino" radicals having one to six carbon atoms. Even more preferred are lower alkylamino radicals having one to three carbon atoms. Examples of such lower alkylamino radicals include N-methylamino, and N-ethylamino, N-propylamino, N- isopropylamino and the like. The term "dialkylamino" includes "N, N-dialkylamino" where amino radicals are independently substituted with two alkyl radicals. Preferred alkylamino radicals are "lower alkylamino" radicals having one to six carbon atoms. Even more preferred are lower alkylamino radicals having one to three carbon atoms. Examples of such lower alkylamino radicals include N,N-dimethylamino, N,N-diethylamino, and the like. The term "carbonyl", whether used alone or with other terms, such as

"aminocarbonyl", denotes -(C=O)-. "Carbonyl" is also used herein synonymously with the term "oxo".

The term "aminocarbonyl" denotes an amide group of the formula -C(=O)NH 2 .

The term "alkylthio" or "thioalkoxy" embraces radicals containing a linear or branched alkyl radical, of one to ten carbon atoms, attached to a divalent sulfur atom. An example of "alkylthio" or "thioalkoxy" is methylthio,(CH 3 S-).

The term "Formula I" includes any sub formulas, such as Formulas I-A, π, II- A, πi-A, m-B or rv.

The term "pharmaceutically-acceptable" when used with reference to a compound of Formulas I-IV is intended to refer to a form of the compound that is safe for administration. For example, a salt form, a solvate, a hydrate, a prodrug or derivative form of a compound of Formulas I - IV, which has been approved for mammalian use, via oral ingestion or other routes of administration, by a governing body or regulatory agency, such as the Food and Drug Administration (FDA) of the United States, is pharmaceutically acceptable.

Included in the compounds of Formulas I-IV are the pharmaceutically acceptable salt forms of the free-base compounds. The term "pharmaceutically-acceptable salts" embraces salts commonly used to form alkali metal salts and to form addition salts of free acids or free bases. As appreciated by those of ordinary skill in the art, salts may be formed from ionic associations, charge-charge interactions, covalent bonding, complexation, coordination, etc. The nature of the salt is not critical, provided that it is pharmaceutically acceptable.

Suitable pharmaceutically acceptable acid addition salts of compounds of Formulas I-FV may be prepared from an inorganic acid or from an organic acid. Examples

A-1291-WO-PCT - 46 - of such inorganic acids are hydrochloric, hydrobromic, hydroiodic, hydrofluoric, nitric, carbonic, sulfuric and phosphoric acid. Appropriate organic acids may be selected from aliphatic, cycloaliphatic, aromatic, arylaliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids, examples of which include, without limitation, formic, acetic, adipic, butyric, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, mesylic, 4-hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic, ethanesulfonic, ethanedisulfonic, benzenesulfonic, pantothenic, 2-hydroxyethanesulfonic, toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, camphoric, camphorsulfonic, digluconic, cyclopentanepropionic, dodecylsulfonic, glucoheptanoic, glycerophosphonic, heptanoic, hexanoic, 2-hydroxy-ethanesulfonic, nicotinic, 2-naphthalenesulfonic, oxalic, palmoic, pectinic, persulfuric, 2-phenylpropionic, picric, pivalic propionic, succinic, thiocyanic, undecanoic, stearic, algenic, β-hydroxybutyric, salicylic, galactaric and galacturonic acid. Suitable pharmaceutically-acceptable base addition salts of compounds of Formulas I - FV include metallic salts, such as salts made from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc, or salts made from organic bases including, without limitation, primary, secondary and tertiary amines, substituted amines including cyclic amines, such as caffeine, arginine, diethylamine, N-ethyl piperidine, histidine, glucamine, isopropylamine, lysine, morpholine, N-ethyl morpholine, piperazine, piperidine, triethylamine, disopropylethylamine and trimethylamine. All of these salts may be prepared by conventional means from the corresponding compound of the invention by reacting, for example, the appropriate acid or base with the compound of Formulas I-IV.

Also, the basic nitrogen-containing groups can be quaternized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides, and others. Water or oil-soluble or dispersible products are thereby obtained. Additional examples of such salts can be found in Berge et al., J. Pharm. Sci.,

66:1 (1977). Conventional methods may be used to form the salts. For example, a phosphate salt of a compound of the invention may be made by combining the desired compound free base in a desired solvent, or combination of solvents, with phosphoric acid in a desired stoichiometric amount, at a desired temperature, typically under heat

A-1291-WO-PCT - 47 -

(depending upon the boiling point of the solvent). The salt can be precipitated upon cooling (slow or fast) and may crystallize (i.e., if crystalline in nature), as appreciated by those of ordinary skill in the art. Further, hemi-, mono-, di, tri- and poly-salt forms of the compounds of the present invention are also contemplated herein. Similarly, hemi-, mono-, di, tri- and poly-hydrated forms of the compounds, salts and derivatives thereof, are also contemplated herein.

The term "derivative" is intended to encompass any salt of a compound of this invention, any ester of a compound of this invention, or any other compound, which upon administration to a patient is capable of providing (directly or indirectly) a compound of this invention, or a metabolite or residue thereof, characterized by the ability to the ability to modulate an enzyme.

The term "pharmaceutically-acceptable derivative" as used herein, denotes a derivative which is pharmaceutically acceptable.

The term "prodrug", as used herein, denotes a compound which upon administration to a subject or patient is capable of providing (directly or indirectly) a compound of this invention. Examples of prodrugs would include esterified or hydroxylated compounds where the ester or hydroxyl groups would cleave in vivo, such as in the gut, to produce a compound according to Formula I-TV. A "pharmaceutically- acceptable prodrug" as used herein, denotes a prodrug which is pharmaceutically acceptable. Pharmaceutically acceptable modifications to the compounds of Formula I-FV are readily appreciated by those of ordinary skill in the art.

The compound(s) of Formulas I-TV may be used to treat a subject by administering the compound(s) as a pharmaceutical composition. To this end, the compound(s) can be combined with one or more excipients, including without limitation, carriers, diluents or adjuvants to form a suitable composition, which is described in more detail herein.

The term "excipient", as used herein, denotes any pharmaceutically acceptable additive, carrier, adjuvant, or other suitable ingredient, other than the active pharmaceutical ingredient (API), which is typically included for formulation and/or administration purposes. "Diluent" and "adjuvant" are defined hereinafter.

The terms "treat", "treating," "treatment," and "therapy" as used herein refer to therapy, including without limitation, curative therapy, prophylactic therapy, and preventative therapy. Prophylactic treatment generally constitutes either preventing the

A- 1291 -WO-PCT - 48 - onset of disorders altogether or delaying the onset of a pre-clinically evident stage of disorders in individuals.

The phrase "effective dosage amount" is intended to quantify the amount of each agent, which will achieve the goal of improvement in disorder severity and the frequency of incidence over treatment of each agent by itself, while avoiding adverse side effects typically associated with alternative therapies. Accordingly, this term is not limited to a single dose, but may comprise multiple dosages required to bring about a therapeutic or prophylactic response in the subject. For example, "effective dosage amount" is not limited to a single capsule or tablet, but may include more than one capsule or tablet, which is the dose prescribed by a qualified physician or medical care giver to the subject. The term "leaving group" (also denoted as "LG") generally refers to groups that are displaceable by a nucleophile. Such leaving groups are known in the art. Examples of leaving groups include, but are not limited to, halides (e.g., I, Br, F, Cl), sulfonates (e.g., mesylate, tosylate), sulfides (e.g., SCH 3 ), N-hydroxsuccinimide, N-hydroxybenzotriazole, and the like. Nucleophiles are species that are capable of attacking a molecule at the point of attachment of the leaving group causing displacement of the leaving group. Nucleophiles are known in the art. Examples of nucleophilic groups include, but are not limited to, amines, thiols, alcohols, Grignard reagents, anionic species (e.g., alkoxides, amides, carbanions) and the like.

GENERAL SYNTHETIC PROCEDURES

The present invention further comprises procedures for the preparation of compounds of Formulas I and π. The compounds of Formulas I-II can be synthesized according to the procedures described in the following Schemes 1-4, wherein the substituents are as defined for Formulas I and π above, except where further noted. The synthetic methods described below are merely exemplary, and the compounds of the invention may also be synthesized by alternate routes utilizing alternative synthetic strategies, as appreciated by persons of ordinary skill in the art.

The following list of abbreviations used throughout the specification represent the following and should assist in understanding the invention: ACN, MeCN - acetonitrile

Aq., aq. - aqueous

Ar - argon (gas)

BOP - benzotriazol-1-yl-oxy

A- 1291 -WO-PCT - 49 - hexafluorophosphate

Cs 2 CO 3 cesium carbonate

CHCl 3 chloroform

CH 2 Cl 2 , DCM dichloromethane, methylene chloride CuI copper iodide

DCC dicyclohexylcarbodiimide

DIC 1 ,3-diisopropylcarbodiimide

DIEA, DIPEA diisopropylethylamine

DME dimethoxyethane DMF dirnethylfbrmamide

DMAP 4-dimethylaminopyridine

DMS dimethylsulfide

DMSO dimethylsulfoxide

EDC, EDCI 1 -(3 -dimethylaminopropyl)-3 -ethylcarbodiimide Et 2 O diethyl ether

EtOAc ethyl acetate

FBS fetal bovine serum

G, gm gram h, hr hour H 2 hydrogen

H 2 O water

HATU O-(7-azabenzotriazol-l-yl)-N,N,N',N'- tetramethyluroniumhexafluorophosphate

HBr hydrobromic acid HCl hydrochloric acid

HOBt 1-hydroxybenzotriazole hydrate

HOAc acetic acid

HPLC high pressure liquid chromatography

IPA, IpOH isopropyl alcohol K 2 CO 3 potassium carbonate

KI potassium iodide

LG leaving group

LiOH lithium hydroxide

MgSO 4 magnesium sulfate

A-1291-WO-PCT - 50 -

MS mass spectrum

MeOH methanol

N 2 nitrogen NaCNBH 3 sodium cyanoborohydride Na 2 CO 3 sodium carbonate

NaHCO 3 sodium bicarbonate

NaH sodium hydride

NaI sodium iodide

NaBH 4 sodium borohydride NaOH sodium hydroxide

Na 2 SO 4 sodium sulfate

NH 4 Cl ammonium chloride

NH 4 OH ammonium hydroxide

P(M)U) 3 tri(tert-butyl)phosphine PBS phospate buffered saline

Pd/C palladium on carbon

Pd(PPh 3 ) 4 palladium(0)triphenylphosphine tetrakis

Pd(dppf)Cl 2 palladium(l,l- bisdiphenylphosphinoferrocene)

II chloride

Pd(PhCN) 2 Cl 2 palladium di-cyanophenyl dichloride Pd(OAc) 2 palladium acetate Pd 2 (dba) 3 tris(dibenzylideneacetone) dipalladium PyBop benzotriazol- 1 -yl-oxy-tripyrrolidino-phosphonium hexafluorophosphate

RT, rt room temperature RBF, rbf round bottom flask TLC, tic thin layer chromatography TBTU O-benzotriazol-l-yl-N,N,N',N'-tetramethyluronium tetrafluoroborate

TEA, Et 3 N triethylamine TFA trifluoroacetic acid

A- 1291 -WO-PCT - 51 -

THF tetrahydrofuran

UV ultraviolet light

While the synthetic strategy for preparing the compounds of Formulas I - π may vary, as appreciated by persons skilled in the art, one strategy for devising a method of making compounds of these formulas is by retro-synthetic disconnection. For example,

II

π-A

As shown in Formulas I-Q and II-A above, each squiggly line represents a possible point of bond-construction, whose order is generally dependent upon the particular compound being synthesized. Such bond construction methods are generally described in synthetic Schemes 1 - 5c below.

Scheme 1 - hydrolysis

-. activation

A- 1291 -WO-PCT - 52 -

Scheme 1 describes a few methods for preparing C(R la )(R' b )(R lc )-W acids, useful for preparing compounds of Formulas I-II (see scheme 2) wherein W is -C(O)- or - S(O) 2 - and each of R la , R 1 b and R lc , independently, is Ci., 0 -alkyl, C 2- i 0 -alkenyl, C 2-I0 - alkynyl, R'-Ci.io-alkyl-, R 1 -C 2- io-alkenyl- or R 1 -C 2- ] 0 -alkynyl- ("L" in scheme 1 corresponds to the Ci-io-alkyl, C 2 -io-alkenyl or C 2 -io-alkynyl of A defined in A-W above or OfC(R 13 XR 1 b )(R lc ) defined herein). Desired C(R^)(R 1 b )(R lc )-W groups and A-W groups may be commercially available and purchased, or may be made by known, conventional methods. As shown, esters 1 can be hydrolyzed to their corresponding acids 2 using known bases, such as NaOH or LiOH. Acids 2 can then be coupled to an amine (not shown) to prepare compounds of Formula I-II. Similarly, sulfonic acids 1' can be converted to an activated sulfonate 2' by reaction with oxalyl chloride, for example, to prepare the corresponding sulfonyl chloride 2'. The sulfonyl chloride 2' can be reacted with an amine to prepare compounds of Formula I-II.

Scheme 2

A-1291-WO-PCT - 53-

Desired C(R la )(R lb )(R lc )-W groups of Formula I and A-W groups of Formula π, which may be substituted with various substitutions including one or more R 7 groups, can be coupled to the core hydroxyl-propyl backbone structure, generally designated in Scheme 2 as "Pr" group, by various coupling methods as described in Scheme 2. In each of the 4 sub-schemes, X refers generally to a "LG" or a "leaving group" such as a halide (bromine, chlorine or iodine), alkylsulfonate and other known groups (also see definitions herein) which generally forms an electrophilic species (E + ) and m is an integer from 0-1. The NH 2 group (primary amine) is a nucleophilic species (Nu " ), as is secondary amines, hydroxides, alkoxides, an anionic carbon species and the like, which should be sufficiently strong to the attack the E + species and displace the leaving group X thereby effecting a coupling of A-W to the Pr backbone. Examples of suitable electrophilic carbonyl species include, without limitation, acid halides, mixed anhydrides, aldehydes, carbamoyl-chlorides, sulfonyl chlorides, acids activated by coupling with activating reagents such as TBTU, HBTU, HATU, HOBT, BOP, PyBOP and carbodiimides (DCC, EDC and the like), and other electrophilic species including halides, isocyanates, daizonium ions and the like.

The coupled adduct of C(R la )(R lb )(R lc )-W and Pr or A-W and Pr, shown as products in sub-schemes 1-4, can be brought about using various conventional methods. For example, an amide or a sulfonamide linkage, as shown in sub-schemes 2 and 4, can be made utilizing an amine on the Pr intermediate and an activated electrophilic species, on the A-W group such as the acid chloride or sulfonyl chloride as shown. The reaction proceeds generally in the presence of a suitable solvent and/or base. Suitable solvents include, without limitation, generally non-nucleophilic, anhydrous solvents such as toluene, CH 2 Cl 2 , THF, DMF, DMF, N,N-dimethylacetamide and the like, including solvent combinations thereof. The solvent may range in polarity, as appreciated by those skilled in the art. Suitable bases include, for example, tertiary amine bases such as DIEA, TEA, carbonate bases such as Na 2 CO 3 , K 2 CO 3 , Cs 2 CO 3 , hydrides such as NaH, KH, borohydrides, cyanoborohydrides and the like, alkoxides such as NaOCH 3 , and the like. The base itself may also serve as a solvent. The reaction may optionally be run neat, i.e., without any base and/or solvent. These coupling reactions are generally fast and conversion occurs typically in ambient conditions. However, depending upon the particular substrate, such reactions may require heat, as appreciated by those skilled in the art.

A-1291-WO-PCT - 54-

Similarly, carbamates as illustrated in sub-scheme 1 and ureas as illustrated in sub-scheme 3 may be made as shown, wherein X has the same definition as above, using the same coupling methods described above for sub-schemes 2 and 4. While the above .methods are so described, they are not exhaustive, and other methods for linking. C(R la )(R lb )(R lc )-W groups and A-W groups and desired Pr groups together may be utilized as appreciated by those skilled in the art.

The coupling methods described in sub-schemes 1-4 of scheme 2 are also applicable for coupling desired C(R la )(R lb )(R lc )-W and A-W intermediates to desired Pr intermediates not containing desired R 5 groups, although sub-schemes 1-4 as illustrated do contain R 5 groups.

Scheme 3a

10 11

Amine intermediate 9 (one embodiment of an R ring of the compounds of Formula I) can be prepared according to the method generally described in Scheme 3 a. As shown, spiro-substituted- (ring Z) or gem-dialky-substituted (not shown) oxo-R 5 ring intermediates 6 (shown where R 5 of Formula I is a spiro fused chroman or aza-chroman ring, as in Formulas IL, II- A, IH-A and IQ-B) can be converted directly to the amino- intermediate 9 using known reductive amination methods, such as in the presence of

A-1291-WO-PCT - 55- sodium cyanoborohydride and ammonium acetate. Alternatively, the carbonyl may be reduced to the corresponding alcohol using conventional reducing reagents or catalytic hydrogenation conditions, and then converted to form the corresponding azido- intermediate 8 using known reagents, such as DPPA, in the presence of a suitable base as shown. Intermediate 8 may be reduced with a suitable reducing agent or by known methods, including triphenylphosphine, trimethylphosphine or lithium aluminum hydride (LAH), to produce the desired amino adduct 9.

Yet another method of forming the amine adduct 9, can be via an imine formation to form compound 10. The imine double bond of compound 10 may then be successively reduced and deprotected to yield the primary amine product 9. Such steps may be conducted using known, conventional methods, as appreciated by those skilled in the art.

Scheme 3b

9

Alternatively, amine intermediates 9 may be prepared by the method shown in scheme 3b above. Desirably substituted compounds 9a may first be treated with a strong base, such as LDA or n-butyl lithium, to form an anion that may then be added to a sufinylimine intermediate 9b (nucleophilic group X may be protected or unprotected as appreciated by one of ordinary skill in the art) to form the corresponding coupled adduct 9c. Open intermediate 9c (X may first be deprotected as necessary) may subsequently be treated with a strong base, such as NaH (wherein, e.g., X is a nucleophile such as OH or NHR 6 ) or TBAF (wherein, e.g., X is OSiR 3 ) to form intermediate 11. Intermediate 11 may then be deprotected to provide spiro amine compound 9.

A-1291-WO-PCT - 56-

Scheme 3c

LG = Cl, Br, I 1 OTf, etc.

9f

Amine intermediate 9 can also be prepared from other amine 9 precursors such as 9d containing an appropriate leaving group (LG, e.g., Cl, Br, I, OTf, etc) as shown in scheme 3c above. Using this method, compound 9d, with the amino group used as is, mono-protected (compound 9e), or doubly protected (compound 9f having PGl and PG2 protecting groups as shown), can be coupled with the requisite neucleophilic reagents with a catalyst such as a Pd-catalyst selected from appropriate sources. The said neucleophilic reagents can be selected from, but not limited to, commercial or pre-formed

A- 1291 -WO-PCT - 57- boronic reagents, stannane reagents, Zinc- or Magnesium-derived metallic reagents. After deprotection if necessary, amine intermediate 9 can be obtained.

Scheme 4

= protecting group 12 14'

12 14

amination

3. 2. deprotections

13'

14' PG = protecting group

13 14 1

PG = protecting group

Scheme 4a describes, generally, multiple different methods for constructing the bond between the amino-propyl backbone starting material (also referred to herein as "Pr") or intermediate 12' (sub-scheme 1) or 12 (sub-scheme 2) and an R 5 ring intermediate 9, thereby synthesizing a desired intermediate 14' or a final compound 14 of Formulas I-H. One method to make this bond is to react an epoxide intermediate 12 or 12' (Note: the epoxide 12 or 12' may be purchased commercially or made via known, published methods such as from the olefin precursor), with an amino-R 5 intermediate 9, as shown. The reaction may proceed in the presence of a polar solvent, such as an alcohol or dioxanes, and may require additional reagents, as appreciated by those skilled in the

A-1291-WO-PCT - 58- art. Additionally, the reaction may require heat for a period of time. Note that while the scheme described the addition of heat, this is by way of example only, and not every reaction would necessarily require heat, as appreciated by those of ordinary skill in the art. The protecting group may be removed using an acid, such as HCl, such that the bonded adduct 14' is recovered as an HCl salt.

Alternatively, desired intermediates 14' may be synthesized starting with an amine-protected aldehyde intermediate 13' (sub-scheme 3) or 13 (sub-scheme 4) and condensing the aldehyde with a primary or secondary amine 9 to form an imine (not shown, generally formed in-situ and not isolated). The imine can then be reduced using a known reducing agent, such as a hydride or borohydride, the reduced intermediate may be deprotected to provide an intermediate 14' having an amine useful to prepare compounds 14 of Formulas I-π.

Scheme 5a

Scheme 5a describes, generally, one method for constructing gem-dialkyl or spiro

(not shown) compounds 9 in schemes 3a and 3b above, by first preparing the corresponding bromo-intermediate 19 as schematically illustrated above. As shown, a methoxy pyridine compound 15 can be reacted with a bromine source, such bromine in HOAc, in the presence of an acid to form the corresponding dibrominated intermediate 16. Compound 16 can be treated with a suitably strong base, such as a lithium base (e.g. BuLi) in the presence of a suitable non-protic, anhydrous colvent, such as ether, to form the lithiated species, which may then be treated with a suitable aldehyde, such as the allylic aldehyde shown above, to afford the corresponding alcohol adduct 18. Intermediate 18 may then be treated with a suitable acid, such as HBr, to protonate and condense the compound effecting ring closure to afford the cyclized adduct 19.

A- 1291 -WO-PCT - 59-

Scheme 5b

Bromine source solvent/aq.

Scheme 5b describes, generally, another method for constructing gem-dialkyl (not shown) or spiro (shown) compounds 9 in schemes 3a and 3b above, by first preparing the corresponding bromo-keto-intermediate 25 as schematically illustrated above. As shown, a methoxy picolinic acid 20 can be reacted with an aqueous bromine source, such bromine, in the presence of a suitable solvent, such as DCM/water, to form the corresponding brominated intermediate 21. The acid group of compound 21 can be converted to the corresponding Weinreb amide under known conditions, such as using EDC-HCl in the presence of HOBt, a base such as TEA, and a suitable solvent such as DCM. Weinreb amide 22 may be treated with a desired Grignard reagent, such as vinylmagnesium bromide as shown above, in the presence of a suitable solvent, such as THF, to form the allylic ketone species 23. Alternatively, the Weinreb amide species may be bypassed by treating compounds 22 directly with the Grignard reagents, such the one shown above, to afford compounds 23. Compound 23 can undergo a Grubb's metathesis, such as by utilizing exo-methylene cyclobutane as shown above, to form intermediate 24, which may then be cyclized to ring closure using a suitable acidic environment, such as in EtOH/HCl, to provide the desired compounds 25. An additional method to prepare mono-substituted aza-chroman compounds, but not gem-dialkyl or spiro aza-chroman compounds 25 is described in Sarges et at, J. Med Chem., 1990, 33, 1859-1865, which disclosure is hereby incorporated herein by reference in its entirety. The keto-intermediate 25 can then be converted to the corresponding primay amino species using the chemistry taught herein.

A- 1291 -WO-PCT - 60-

Scheme 5c _, H ςO ° MePO(OMe) 2 , Me0H ' reflUX 1 A)Me ™F / PhMe / 0° C

21 26 27

28 24 25

Scheme 5 c describes, generally, yet another method for constructing gem-dialkyl (not shown) or spiro (shown) intermediates 24 in scheme 5b above. Compound 24 in turn may be converted to the desired bromo-keto-intermediate 25 as schematically illustrated above. As shown, brominated intermediate 21 from scheme 5b can be reacted with an acid, such as sulfuric acid as shown, in a protic solvent, such as MeOH, to form the corresponding methyl ester intermediate 26. The ester group of compound 26 can be converted to the corresponding phosphonate ester under Horner-Emmons type conditions, which are known in the art, such as using a phosphonate species in the presence of a strong base, such as LiHMDS as shown, and a suitable solvent such as THF and toluene. The resulting phosphonate adduct 27 may be deprotonated with a strong base, such as with IiOMe in the presence of an alcoholic solvent such as MeOH and/or I-PrOH, and the lithium enolate can then be reacted with cyclobutanone to afford the adduct compound 24 in high yield. Intermediate 24 may then be cyclized to ring closure using suitable conditions, such as those shown above in scheme 5c, to provide the desired compounds 25. Additional description of useful methods which may be used to prepare compounds similar to compound 25 are described in general in Harada et at, JP patent application no. 08099982A , Yasuda et al, J. Org. Chem. 2004, 69, pg 1958, Yazbeck et al, Org. Process Res. Dev. 2006, JO, pg 655, and in Keneko et al, Chem. Pharm. Bull., 2004, 52, pg 675, which disclosures are hereby incorporated herein by reference in its entirety. The keto- intermediate 25 can then be converted to the corresponding primay amino species using the chemistry taught herein.

It should be appreciated that schemes 5a, 5b and 5c illustrate exemplary methods for preparing the right-side spiro pieces of compounds of Formulas I and II. Reaction yields for each step in schemes 5a and 5b range from about 50% to 90+%. Accordingly, these methods may provide a more efficient process for preparing desired intermediates

A-1291-WO-PCT - 61-

25. Further, utilizing these methods may afford other spirocyclic rings of differeing sizes and heteroatoms, encompassed in the compounds of the present invention.

To enhance the understanding and appreciation of the present invention, the following specific examples (starting reagents, intermediates and compounds of Formulas I-H) are set forth. The following analytical methods were used to purify and/or characterize the compounds, and intermediates, described in the examples below.

Chromatography: Unless otherwise indicated, crude product-containing residues were purified by passing the crude material orconcentrate through an ISCO brand silica gel column (pre-packed or individually packed with SiC^) and eluting the product off the column with a solvent gradient as indicated. For example a description of (330 g SiO 2 , 0- 40% EtOAc/Hexane) means the product was obtained by elution from the column packed with 330gms of silica, with a solvent gradient of 0% to 40% EtOAc in Hexanes.

Preparative HPLC Method:

Unless otherwise 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, C 18, 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 1 H NMR spectra were run on a Bruker series 300

MHz instrument or a Bruker series 400 MHz instrument 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.

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 instrument or an Agilent 1100 series LC/MSD system. Compounds having

A-1291-WO-PCT - 62- 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.

The compounds disclosed and described herein have been named using either (1) the naming convention provided with Chem-Draw Ultra 8.0 software, available in Chem Office, or (2) by the ISIS database software (Advanced Chemistry Design Labs or ACD software). In some instances, compounds were named with the term "spirocarbocycle" inserted where appropriate. For example, where the chroman is substituted with 2,2- spirocyclobutyl, "2,2-spirocyclobutyl" have been added to the Chem-Draw nomenclature in the appropriate place.

Examples

The Examples, described herein below, represent various exemplary starting materials, intermediates and compounds of Formulas I-II, which should assist in a better understanding and appreciation of the scope of the present invention and of the various methods which may be used to synthesize compounds of Formulas I and II. It should be appreciated that the general methods above and specific examples below are illustrative only, for the purpose of assistance and of understanding the present invention, and should not be construed as limiting the scope of the present invention in any manner.

tert-Butyl ClS^S^l-^ert-butyldimethylsilylo^-S-cyclobutylidene-l- hydroxypentan-3-ylcarbamate A solution of tert-butyl (2S,3S)-2-(tert-butyldimethylsilyloxy)-l-hydroxyhex-5-en-3- ylcarbamate (1.0 g, 2.9 mmol) and Grubbs Catalyst 2 nd generation (0.12 g, 0.14 mmol) in DCE (6mL) was treated with methylenecyclobutane (0.79 g, 12 mmol) and heated to 80 0 C in a sealed vial overnight. The reaction mixture was diluted with EtOAc, washed with saturated NaHCO 3 solution, and brine. The organics were dried over MgSO 4 and concentrated under reduced pressure. Purification of the crude residue by column

A-1291-WO-PCT - 63- chromatography gave tert-butyl (2S,3S)-2-(tert-butyldimethylsilyloxy)-5- cyclobutylidene-l-hydroxypentan-3-ylcarbamate (0.900 g, 81% yield) as a 2:1 mixture with starting alkene.

Example 2

(4S,5S)-tert-Butyl 4-(but-2-ynyl)-5-(hydroxymethyl)-2^dimethyloxazolidine-3- carboxylate

Step 1: (4S. 5S)-tert-butyl 4-(but-2-vnylV5-((tert-butyldimethylsilyloxy ' )niethylV2,2- dimethyloxazolidine-3 -carboxylate

A solution of (4S,5S)-tert-butyl 5-((tert-butyldimethylsilyloxy)methyl)-2,2-dimethyl-4- (prop-2-ynyl)oxazolidine-3-carboxylate (0.60 g, 1.6 mmol) in THF (5 mL) was cooled to - 78°C and butyllithium (0.75 mL, 1.9 mmol) was added dropwise. After 5 min, the mixture was re-cooled to -78°C and iodomethane (0.15 mL, 2.3 mmol) was added dropwise. The mixture was stirred at 0 0 C for 2 h and brought to RT and stirred for 15 h. The reaction mixture was quenched with sat NH 4 Cl, filtered, concentrated, and chromatographed on silica gel eluting with hexanes to afford a light yellow oil as (4S,5S)- tert-butyl 4-(but-2-ynyl)-5-((tert-butyldimethylsilyloxy)methyl)-2,2-di methyloxazolidine- 3-carboxylate (0.463g, 74% yield). MS m/z: 298 (M+l). Step 2: (4S.5SVtert-butyl 4-(but-2-vnyl)-5-(hvdroxymethyl)-2.2dimethyloxazolidine-3- carboxylate

The title compound was obtained using the procedure analogous to that described in WO2007061670, which specification is hereby incorporated by reference herein in its entirety. MS m/z: 184 (M+l).

Example 3

N-((lS)-l-((lR)-2-(((4'S)-6'-(2^-Dimethylpropyl)-3',4'-di hydrospiro[cyclobutane- l^'-pyrano[2β-b]pyridin]-4'-yl)amino)-l-hydroxyethyl)butyl) acetainide

A- 1291 -WO-PCT - 64-

To a 50 mL RBF containing N-((l S)-l-((lR)-2-(((4S)-6-(2,2-dimethylpropyl)-3,4- dihydrospirofchromene^ j r-cyclobutanJ^-yOaminoVl-hydroxyethyO-S-buten-l- yl)acetamide (164 mg, 395 μmol) was added EtOAc (20 mL ) and the mixture was allowed to stir at 23 0 C for 2 min. At this time, EtOH was added because the alkene was not completely soluble in EtOAc. Pd/C (75 mg) was added to the reaction, then hydrogen gas was bubbled through the reaction mixture for 10 min. The reaction was allowed to stir under a balloon of hydrogen gas for Ih and then filtered through a plug of silica gel under celite washing with 15% MeOH (2.0 M in ammonia) in EtOAc. The solvent was removed to give the title compound as a white solid. MS m/z: 418.2 (M+H).

Example 4

N-((lS)-l-((lR)-2-(((4'S)-6'-(2^-Dimethylpropyl)-8 t -(methylamino)-3',4'- dihydrospiroIcycIobutane-l^'-pyranotZ^-cJpyridinJ^'-yOamino^ l-hydroxyethyl)- 3-buten-l-yl)acetamide.

To a flame-dried microwave vial under Ar gas was added N-((lS)-l-((lR)-2-(((4'S)-8'- chloro-6'-(2,2-dimethylpropyl)-3',4'-dihydrospiro[cyclobutan e-l,2'-pyrano[2,3-c]pyridin]- 4'-yl)amino)-l-hydroxyethyl)-3-buten-l-yl)acetamide (120 mg, 267 μmol), Pd 2 dba 3 (49 mg, 53 μmol), and DavePhos (46 mg, 117 μmol). The vial was purged with N 2 5x, then THF (3 mL), methanamine (533 μl, 1067 μmol), and LiHMDS (2400 μl, 2400 μmol) - were added. The vial was sealed and heated in a microwave at 110 0 C for 10 min. The reaction mixture was directly purified by reverse phase HPLC to give the title compound as an amorphous white solid. MS m/z: 445.3(M+1).

(4S)-2^σ-Spirocyclobutan-6-neopentyI-3,4-dihydro-2H-pyra no[3^-b]pyridin-4-amine

Step 1 : 5-(methoxymethoxy)-2-neopentylpyridine-N-oxide

5-(Methoxymethoxy)-2-neopentylpyridine (11.0 g, 52.6 mmol) was dissolved in CH 2 Cl 2 (200 mL) to which mCPBA (18.1 g, 105 mmol) was added, and the mixture was stirred

A- 1291 -WO-PCT - 65- under N 2 for about 4 h. The mixture was quenched with IM NaOH (200 mL) and stirring was continued vigorously for 10 min. The mixture was extracted with CH 2 Cl 2 (2 x 200 mL), the combined organic layers were washed with saturated NaCl, dried (Na 2 SO 1 O, and evaporated to give 5-(methoxymethoxy)-2-neopentylpyridine-N-oxide (11.8 g, 99.7% yield) as a brown oil which was used without purification in the next step. Step 2: 3-(methoxymethoxyV6-neopentylpicolinonitrile

5-(Methoxymethoxy)-2-neopentylpyridine-N-oxide (11.5 g, 51 mmol) was dissolved in CH 2 Cl 2 (50 mL) to which benzoyl chloride (12 ml, 102 mmol) and (trimethylsilyl)formonitrile (14 ml, 102 mmol) were added. The mixture was stirred under N 2 4 h, quenched with saturated NaHCO 3 (150 mL), and extracted with CH 2 Cl 2 (3 x 100 mL). The combined organic layers were washed with saturated NaHCO 3 (2 x 50 mL), dried (MgSO 4 ), and evaporated to give the crude product as a brown oil, which was purified by ISCO (330 g SiO 2 , 0-40% EtOAc/Hexane) to give 3-(methoxymethoxy)-6- neopentylpicolinonitrile (8.8 g, 74% yield) as a clear oil. Step 3: l-(3-(methoxymethoxy)-6-neopentylpyridin-2-yl)ethanone

3-(Methoxymethoxy)-6-neopentylpicolinonitrile (8.3 g, 35 mmol) was dissolved in THF (125 mL). The solution was cooled to 0 0 C and methylmagnesium chloride (24 ml, 71 mmol) (3.0 M in Et 2 O) was added. The reaction mixture stirred for 2 h at it under N 2 then quenched with saturated NH 4 Cl (50 mL) and extracted with EtOAc (3 x 50 mL). The organic layer was dried over MgSO 4 and evaporated to give the crude product as a yellow oil. Purification of the crude residue by ISCO (40 g SiO 2 , 0-40% EtOAc/Hexane) gave 1- (3-(methoxymethoxy)-6-neopentylpyridin-2-yl)ethanone (3.8 g, 43% yield) as a clear, light orange oil. Step 4: l-(3-hvdroxy-6-neopentylpyridin-2-vDethanone A solution of l-(3-(methoxymethoxy)-6-neopentylpyridin-2-yl)ethanone (3.75 g, 15 mmol) in (2:1:1) 5 M HCl : /-PrOH : THF (100 mL) was stirred 16 h at rt. The mixture was concentrated to remove the THF and /-PrOH. The resulting solution consisting of the product in aqueous HCl was quenched by slow addition to a solution of saturated aqueous NaHCO 3 (500 mL) containing excess solid NaHCO 3 (28 g). The aqueous layer was extracted with CH 2 Cl 2 (3 x 100 mL), the organic layers combined and washed with saturated aqueous NaCl (100 mL), dried (MgSO 4 ), and concentrated to give the crude product as a brown oil. The product was purified by ISCO (0-10% EtOAc/Hexanes) to give l-(3-hydroxy-6-neopentylpyridin-2-yl)ethanone (1.98 g, 64% yield) as a clear, colorless oil.

A-1291-WO-PCT - 66-

Step 5: 2.2-spirocvclobutan-6-neopentyl-2.3-dihydropyranor3.2-b1pyri din-4-one A mixture of l-(3-hydroxy-6-neopentylpyridin-2-yl)ethanone (1.90 g, 9167 μmol), pyrrolidine (2296 μl, 27501 μmol), and cyclobutanone (2570 mg, 36667 μmol) in CH 3 CN (20 mL) was heated in a 65 0 C oil bath for 3 h. The mixture was cooled to rt, then diluted with EtOAc (25 mL), washed with H 2 O, saturated aqueous NH 4 Cl, saturated aqueous NaCl, dried (MgSO 4 ), and concentrated. Purification of the resulting crude materially ISCO (40 g SiO 2 , 10-20% EtOAc/Hexanes) gave 2,2-spirocyclobutan-6-neopentyl-2,3- dihydropyrano[3,2-b]pyridin-4-one (710 mg, 29.9% yield) as a yellow solid. Step 6: (4R)-2.2-spirocyclobutan-6-neopentyl-3 ^-dihydro-σH-pyranoP ,2-b]pyridin-4-ol To a vial containing 2,2-spirocyclobutan-6-neopentyl-2,3-dihydropyrano[3,2-b]pyri din-4- one (710 mg, 2738 μmol) was added sodium formate (1862 mg, 27377 μmol) and tetrabutylammonium bromide (26.5 mg, 82.1 μmol). Toluene (5 mL) and H 2 O (2.5 mL) were added and the solution purged 3 x with N 2 , then with an Ar balloon for 15 min.

[( 1 J?,2i?)-2-Amino- 1 ,2-diphenyl-N-(p-tolylsulfonyl)ethylamido]chloro(η 6 -^- cymene)ruthenium(π) (53.4 mg, 82.1 μmol) was added and the biphasic reaction was stirred at rt under Ar for 24 h. H 2 O (10 mL) was added and the reaction was extracted with CH 2 Cl 2 (3 x 20 mL). The combined organic layers were dried (Na 2 SO 4 ), and concentrated to give a brown oil, which was purified by ISCO (40 g SiO 2 , 5-40% EtOAc/Hexane) gives (4R)-2,2-spirocyclobutan-6-neopentyl-3,4-dihydro-2H-pyrano[3 ,2- b]pyridin-4-ol (460 mg, 64.3% yield) as a clear oil.

Step 7: (4S " )-4-azido-2,2-spirocvclobutan-6-neopentyl-3.4-dihvdro-2H-pyr ano[3.2- b]pyridine

To a solution of (4R)-2,2-spirocyclobutan-6-neopentyl-3,4-dihydro-2H-pyrano[3 ,2- b]pyridin-4-ol (460 mg, 1760 μmol) in toluene (4 mL) is added diphenylphosphoryl azide (531 μl, 2464 μmol) then l,8-diazabicyclo(5.4.0)-7-undecene (368 μl, 2464 μmol). The reaction mixture was stirred under N 2 at rt 23 h. The clear, light yellow solution first turned into a brown cloudy/opaque solution after 30 min. To speed up the reaction rate, the mixture was heated to 40 0 C and stirred an additional 5 h. Water (20 mL) was added and the reaction mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were dried (Na 2 SO 4 ), and concentrated to give the crude product as a brown oil, which was purified by ISCO (40 g SiO 2 , 0-20% EtOAc/Hexane) gives (4S)-4-azido-2,2- spirocyclobutan-6-neopentyl-3,4-dihydro-2H-pyrano[3,2-b]pyri dine (250 mg, 49.6% yield) as a white solid.

A-1291-WO-PCT - 67-

Step 8: (4S)-2.2-spirocvclobutan-6-neopentyl-3.4-dihydro-2H-pyranor3 ,2-blpyridin-4- amine

A solution of (4S)-4-azido-2,2-spirocyclobutan-6-neopentyl-3,4-dihydro-2H- pyrano[3,2- b]pyridine (250 mg, 873 μmol) in methanol (10 mL) was purged with N 2 (3 x), then palladium (260 mg, 244 μmol) (10 wt% on carbon) was added. The reaction was purged with H 2 (3 x), then stirred at rt under H 2 1.5 h. The suspension was filtered through a pad of Celite, MeOH wash (4 x 5 mL), and the solution concentrated to give the crude product (245 mg) as a white oily solid, which was purified by ISCO (12 g SiO 2 , 0-10% MeOH/CH 2 Cl 2 ) to give the title compound as a white solid.

(4S)-6-(l,l-Difluoroethoxy)-2^-spirocyclobutan-3,4-dihydr o-2H-pyrano[2β- b] pyridin-4-amine. Step 1 : (4S)-/gr/-Butyl-6-acetyl-2.2-spirocvclobutan-3.4-dihvdro-2H- pyranor23- b1pyridin-4-yl(allyl)carbamate.

To a solution of (4S)-ter/-butyl 6-bromo-2,2-spirocyclobutan-3,4-dihydro-2H-pyrano[2,3- b]pyridin-4-yl(allyl)carbamate (8.1 g, 20 mmol) in Et 2 O (100 mL) at -78 0 C was added fert-butyllithium (23 ml, 40 mmol) over 3 minutes. The reaction was allowed to stir 10 min. at -78 0 C, then acetaldehyde (4.5 ml, 79 mmol) was added, the reaction was then warmed to rt over 30 min, and quenched with NH 4 CI (200 mL). The reaction was extracted with EtOAc (3 x 100 mL), the combined organic layers were washed with saturated NaCl (100 mL), dried (Na 2 4 ), and concentrated to give crude product as a dark yellow/orange oil. The crude was carried on into the next step without purification. To a solution of the crude product from above in CH 2 Cl 2 (50 mL) at 0 0 C, was added sodium bicarbonate (6.64 g, 79.0 mmol) and Dess-Martin periodinane (10.5 g, 24.7 mmol) simultaneously. The ice bath was removed and the reaction was stirred for 2 h at rt, then quenched with saturated Na 2 SO 3 (300 mL), extracted with CH 2 Cl 2 (3 x 200 mL), and concentrated. The crude material was purified by ISCO (10-50% EtOAc/Hexane) to give the title compound (4.10 g, 55.7% yield over 2 steps) as a clear, light yellow oil.

A-1291-WO-PCT - 68-

Step 2: (4S)-l-(4-amino-2,2-spirocyclobutan-3,4-dihvdro-2H-pyranor2. 3-b1pyridin-6- vDethanone

To a 150 mL rbf with (4S)-tert-butyl 6-acetyl-2,2-spirocycldbutan-3,4-dihydro-2H- pyrano[2,3-b]pyridin-4-yl(allyl)carbamate (2.05 g, 5504 μmol) and CH 2 Cl 2 (50 mL) was added 2,2,2-trifluoroacetic acid (5089 μl, 66048 μmol). The reaction was allowed to stir at RT for 5 h, then diluted with CH 2 Cl 2 (50 mL). The mixture was washed with saturated NaHCO 3 (2 x 100 mL) and the organic layer degassed with Argon for 10 minutes. The degassed solution was treated with l,3-dimethylpyrimidine-2,4,6(lH,3H,5H)-trione (2.21 g, 14154 μmol) and tetrakistriphenylphosphine palladium(O) (254 mg, 220 μmol) and stirred at it for 24 hours. The reaction mixture was washed with NaOH (IN, 2 X 50 mL), and HCl (IN, 2 X 50 mL). The acidic aqueous layer was then basified to pH 14 with NaOH (5N, 25 mL) and extracted with DCM (3 X 50 mL). The combined organic layers were dried (Na 2 SO 4 ) and concentrated to give (4S)-l-(4-amino-2,2-spirocyclobutan-3,4- dihydro-2H-pyrano[2,3-b]pyridin-6-yl)ethanone (810 mg, 63.4%) as a light yellow oil. Step 3 : (4S)-6-( 1 , 1 -difluoroethoxy)-2,2-spirocyclobutyl-3.4-dihvdro-2H-pyranor2 ,3- blpyridin-4-amine

To a 20 mL polyethylene vial was added (4S)-l-(4-amino-2,2-spirocyclobutan-3,4- dihydro-2H-pyrano[2,3-b]pyridin-6-yl)ethanone (410 mg, 1765 μmol) and HF/pyridine (1 mL). Xenon difluoride (359 mg, 2118 μmol) was added to the mixture followed by CH 2 Cl 2 (1 mL). The reaction was stirred at rt 24 h, then quenched by slowly adding it to saturated NaHCO 3 (100 mL) with solid NaHCO 3 (5 g). The mixture was extracted with CH 2 Cl 2 (3 x 50 mL). The combined organic layers were collected, dried (Na 2 SO 4 ) and concentrated to give the crude product as a brown oil. The crude was purified by ISCO (2xl2g SiO 2 stacker, 0-8% MeOH/CH 2 Cl 2 ) to give (4S)-6-(l,l-difluoroethoxy)-2,2- spirocyclobutyl-3,4-dihydro-2H-pyrano[2,3-b]pyridin-4-amine as an orange oil.

A-1291-WO-PCT - 69-

(lR^S,4'S)-2-Hydroxy-3',4'dihydrospiro[cyclobutane-l^ l -pyrano[2 r 3-b]pyridin-4'- amine and (lS^R,4'S)-2-hydroxy-3',4'dihydrospiro[cyclobutane-l^'-pyran o[23- b] py ridin-4'-amine

Step 1: (cisVl-Allyl^-rbenzyloxy^cvclobutanol and (trans)- l-allyl-2- (benzyloxy)cvclobutanol

To a RBF under argon was added 2-(benzyloxy)cyclobutanone (8.0 g, 45 mmol) and THF (100 mL). The reaction was cooled to 0 0 C and allylmagnesium bromide (113 ml, 113 mmol) (1.0 M in Et 2 O) was added over 20 min. The clear colorless solution turned into a tan solution with a white suspension. The ice bath was removed and the reaction warmed to rt and stirred 7 h. The reaction was quenched by slow addition to saturated aqueous NH 4 Cl (500 mL). The reaction was diluted with EtOAc (300 mL) and extracted. The aqueous layer was extracted with EtOAc (2 x 250 mL), the combined organic layers washed with saturated NaCl (250 mL), dried (Na 2 SO 4 ), and concentrated to give a clear, light yellow oil, which was purified by ISCO (33O g SiO 2 , 10-40% EtOAc/Hexane) gives the less polar isomer (cis)-l-allyl-2-(benzyloxy)cyclobutanol (4.0 g, 40% yield) followed by the more polar isomer (trans)- l-allyl-2-(benzyloxy)cyclobutanol (2.4 g, 24% yield) as a clear, colorless oils.

Step 2: ((1.2-cis)-l-Allyl-2-(benzγloxy)cvclobutoxy)(tert-butyl)dim ethylsilane (Cis)-l-Allyl-2-(benzyloxy)cyclobutanol (4.00 g, 18.3 mmol) was dissolved in CH 2 Cl 2 (100 mL) to which tert-butyldimethylsilyl trifluoromethanesulfonate (5.05 ml, 22.0 mmol) and N-ethyl-N-isopropylpropan-2-amine (3.99 ml, 22.9 mmol) were added. The reaction mixture was stirred at rt for 5 h. The reaction was quenched with 10% NaCU 3 (300 mL), and the aqueous layer was extracted with CH 2 Cl 2 (2 x 100 mL). The combined organic layers were washed with saturated NaCl (50 mL), dried (Na 2 SO 4 ), and concentrated to give a yellow oil, which was purified by ISCO (40 g SiO 2 , 100% Hexane to give ((cis)-l-allyl-2-(benzyloxy)cyclobutoxy)(tert-butyl)dimethyl silane (5.38 g, 88.3% yield) as a clear, colorless oil.

Step 3: 2-((1.2-cis)-2-(ωenzyloxy)-l-(tert-buWldimethylsilyloxy) cvclobutvDacetaldehyde ((cis)-l-Allyl-2-(benzyloxy)cyclobutoxy)(tert-butyl)dimethyl silane (5.37 g, 16 mmol) was dissolved in /-butanol (30 mL, 319 mmol) and H 2 O (30 mL) followed by the addition of 4-methylmorpholine n-oxide (3.4 g, 29 mmol) in one portion. After the reactants dissolved, osmium tetroxide (5.1 mL, 0.40 mmol) was added and the reaction mixture was stirred at RT for 17 h. The reaction mixture was worked up by the addition of 6g of sodium sulfite and allowed to stir for 1 h. The reaction mixture was extracted with ether

A-1291-WO-PCT - 70- and the organic phase was concentrated and used directly in the next step.The crude was dissolved in 1:1 f-BuOH/H 2 O (60 mL) and sodium periodate (6.2 g, 29 mmol) was added. The mixture was stirred for 3h. Then H 2 O (100 mL) was added and the mixture was extracted with Et 2 O (3 x 100 mL). The combined organic layers were dried (MgSO 4 ) and concentrated. The crude was purified by ISCO (12O g SiO 2 , 5-20% EtOAc/Hexane to give 2-((cis)-2-(benzyloxy)-l-(tert-buryldimethylsilyloxy)cyclobu tyl) acetaldehyde (4.0 g, 74% yield) as a clear, colorless oil.

Step 4: 2-(d.2-cis)-2-(Benzyloxγ)-l-(tert-butyldimethylsilyloxy)cvc lobutylVl-(2-fluoro- 5-neopentylpyridin-3-yl)ethanol To a flame-dried 250 mL rbf with 2,2,6,6-tetramethylpiperidine (3.41 ml, 20.1 mmol) was added THF (40 mL) and the solution is cooled to -78 0 C. w-Butyllithium (10.8 ml, 1.60 M, 17.2 mmol) was added dropwise and the reaction was warmed to 0 °C and stirred 5 min. The reaction was recooled to -78 0 C and 2-fluoro-5-neopentylpyridine (2.40 g, 14.3 mmol) in THF (10 mL) was added and the reaction stirred 45 min at -78 0 C. Then 2- ((cis)-2-(benzyloxy)-l-(tert-butyldimethylsilyloxy)cyclobuty l) acetaldehyde (4.00 g, 12.0 mmol) in THF (10 mL) was added dropwise. The reaction mixture was stirred 15 min at - 78 0 C, then quenched by addition of saturated NH 4 Cl (50 mL), warmed to rt, diluted with H 2 O (50 mL) and extracted with Et 2 O (3 x 100 mL). The combined organic layers were washed with saturated NaCl (100 mL), dried (Na 2 SO 4 ), and concentrated to give a crude product, which was purified by ISCO (12O g SiO 2 , 0-20% EtOAc/Hexane) to give the title compound (5.18 g, 86.3%) as a 1:1 mixture of diastereomers, as a clear, light yellow oil. Step 5: (l,2-cisV2-(BenzyloxyVl-(2-(2-fluoro-5-neopentylpyridin-3-yl )-2- hydroxyethvDcvclobutanol To a flame-dried 100 mL rbf with 2-((l,2-cis)-2-(benzyloxy)-l-(tert- butyldimethylsilyloxy)cyclobutyl)- 1 -(2-fluoro-5 -neopentylpyridin-3 -yl)ethanol (5.18 g, 10 mmol) was added THF (10 mL) followed by TBAF (12 ml, 1.0 M in THF, 12 mmol). The reaction was stirred at rt for 30 min, then diluted with H 2 O (100 mL) and extracted with Et 2 O (3 x 50 mL). The combined organic layers were washed with saturated NaCl (100 mL), dried (Na 2 SO 4 ) and concentrated to give a crude materiall, which was purified by ISCO (120 g SiO 2 , 0-20% EtOAc/Hexane) to give (l,2-cis)-2-(benzyloxy)-l-(2-(2- fluoro-5-neopentylpyridin-3-yl)-2-hydroxyethyl)cyclobutanol (3.25 g, 81%) as a 1 :1 mixture of diastereomers, a clear, light yellow oil.

Step 6: (1.2-cisV2-Benzyloxy — 6'neopentyl-3'.4'dihydrospirorcvclobutane-1.2'- pvranor2.3-b1pvridin-4'-one

A-1291-WO-PCT - 71-

To a flame-dried 100 mL rbf with (l,2-cis)-2-(benzyloxy)-l-(2-(2-fluoro-5- neopentylpyridin-3-yl)-2-hydroxyethyl)cyclobutanol (3.06 g, 7.9 mmol) was added THF (500 mL) followed by NaH (1.6 g, 39 mmol, 60% in mineral oil). The reaction was heated in a 60 0 C oil bath under N 2 for 4 h, then cooled to rt and quenched with saturated NH 4 Cl (200 mL). The aqueous layer was extracted with EtOAc (3 x 100 mL), and the combined organic layers dried (MgSO 4 ) and concentrated to give the crude alcohol, which was used in the next step without purification. The crude material was dissolved in CH 2 Cl 2 (100 mL) and Dess-Martin periodinane (3.3 g, 7.9 mmol) and sodium bicarbonate (0.66 g, 7.9 mmol) were added at the same time. The reaction was stirred for 2 h at rt. The reaction was quenched with saturated aqueous Na 2 3 (100 mL), extracted, then extracted with additional CH 2 Cl 2 (2 x 100 mL). The combined organic layers were washed with saturated NaCl (100 mL), dried (Na 2 SO 4 ), and concentrated to give the crude product as a yellow oil. Purification of the oil by ISCO (12O g SiO 2 , 0-80% EtOAc/Hexane) gave the title compound (2.67 g, 93%) as a clear, light yellow oil. Step 7: (1R.2S. 4'RV2-Benzyloxy-3'.4'dihvdrospirolcvclobutane-1.2'-Dyranor2. 3- b " |pyridin-4'-ol

To a stirred solution of (s)-2-methyl-cbs-oxazaborolidine (0.70 ml, 0.70 mmol) in THF (10 mL) at 0 0 C was added borane-methyl sulfide complex (1.2 ml, 12 mmol) followed by a solution of (l^-cis^-benzyloxy-ό'neopentyl-S'^'dihydrospirofcyclobutane -l^ 1 - pyrano[2,3-b]pyridin-4'-one (2.57 g, 7.0 mmol) in THF (20 mL) dropwise via syringe pump over about 2.8 h. The reaction was stirred an additional 30 min, then was quenched by dropwise addition (1 drop/10 sec) of 5 M HCl (25 mL) at 0 0 C. After 15 mL HCl was added, bubbling had ceased and the addition rate was increased as the ice bath was removed. The reaction was stirred an additional 2 h at rt. The reaction was recooled to 0 0 C and neutralized with 5 M NaOH (27 mL). The mixture was then extracted with

EtOAc (2 x 150 mL), washed with saturated aqueous NaCl (200 mL), dried (MgSO 4 ), and concentrated to give a yellow oil. Purification of the oil by ISCO (12O g SiO 2 , 20% EtOAc/Hexane) gave a mixture of (1R,2S, 4'R)-2-benzyloxy-6'-neopentyl- 3',4'dihydrospiro[cyclobutane-l,2'-pyrano[2,3-b]pyridin-4'-o l (1.3 g, 50%) and (1S,2R, 4'R)-2-benzyloxy-6'-neopentyl-3 l ,4'dihydrospiro[cyclobutane-l,2'-pyrano[2,3-b]pyridin- 4'-ol (1.3 g, 50%) as a white foam.

Step 9: πR.2S.4'SV2-Benzyloxy-6'-neopentyl-3'.4'dihvdrospirorcvclob utane-1.2'- pyranor2,3-blpyridin-4'-azide and (lS,2R,4'SV2-benzyloxy-6'neopentyl- 3'.4'dihvdrospirorcyclobutane- 1.2'-Pvranor2.3-blpyridin-4'-azide

A- 1291 -WO-PCT - 72-

To a solution of (1,2-cis, 4'R)-2-benzyloxy-6'-neopentyl-3',4'dihydrospiro[cyclobutane- l,2'-pyrano[2,3-b]pyridin-4'-ol (2.6 g, 7.1 mmol) (1:1 mixture of 1,2-spirocyclobutyl diastereomers) in toluene (14 mL) was added diphenylphosphoryl azide (2.1 ml, 9.9 mmol) then l,8-diazabicyclo(5.4.0)-7-undecene (1.5 ml, 9.9 mmol). The reaction was stirred under N 2 at rt for 18 h. The clear, light yellow solution turned into a yellow cloudy/opaque solution after 10 min. Water (100 mL) was added and the reaction mixture extracted with EtOAc (3 x 200 mL). The combined organic layers were washed with saturated NaCl (150 mL), dried (MgSO 4 ), and concentrated to give the crude product as a brown oil. To a solution of the brown oil from above in 10: 1 THFZH 2 O (40 mL) at 0 0 C is added NaOH (2.85 ml, 14.3 mmol). After 5 min, trimethylphosphine (2.52 ml, 28.5 mmol) was added dropwise over 4 min. The ice bath was allowed to melt as the reaction warmed to rt and stirred a total of 18 h. The mixture was recooled to 0 °C and 5 N HCl (50 mL) was added. The resulting mixture was extracted with CH 2 Cl 2 (3 x 100 mL), the combined organic layers were washed with 2.5 N HCl (2 x 50 mL). The combined aqueous layers were cooled to 0 °C and basified to pH 14 with 5 N NaOH (200 mL). The aqueous layer was extracted with CH 2 Cl 2 (3 x 100 mL) the combined organic layers dried (Na 2 SO 4 ), and concentrated to give 2.9 g crude product as a viscous yellow oil. Purification of the oil by ISCO (120 SiO 2 , 0-10% MeOH/CH 2 Cl 2 gradient elution) gave a 1:1 mixture of the title compounds (1.870 g, 71.6% yield) as a yellow oil.

Step 10: (lR.2S.4'SV2-Hvdroxy-6'-neopentyl-3'.4'dihvdrospirorcvclobut ane-1.2'- pyranor2,3-b1pyridin-4'-amine and (lS.2R.4'S)-2-hvdroxy-6'neopentyl-

3',4'dihvdrospirorcvclobutane-l,2'-pyranor2,3-b1pyridin-4 '-amine

To a solution of (l,2-cis,4'S)-2-benzyloxy-6'neopentyl-3',4'dihydrospiro[cycl obutane- l,2'-pyrano[2,3-b]pyridin-4'-azide (1.320 g, 3.6 mmol) in MeOH (50 mL) under Ar is added Pd Black (76.7 mg, 720 μmol). H 2 gas was bubbled though the suspension for 15 min. The reaction was then stirred at rt under an atmosphere OfH 2 (balloon) for 48 h. After 48 h, the H 2 atmosphere was replaced with N 2 , and palldium hydroxide (506 mg, 720 μmol) was added, the reaction was sparged with H 2 and stirred for 24 h at rt. The H 2 atmosphere was replaced with N 2 , and the suspension was filtered through a plug of

Celite, washed with MeOH (3 x 50 mL), and the combined filtrates were concentrated in vacuo to give the crude product. Purification of the crude material by ISCO (12O g SiO 2 , 0-30% MeOH/CH 2 Cl 2 gradient elution) gave a mixture of (lR,2S,4'S)-2-hydroxy-6'- neopentyl-3',4'dihydrospiro[cyclobutane-l,2'-pyrano[2,3-b]py ridin-4'-amine (415 mg,

A-1291-WO-PCT - 73-

41.7% yield) and (lS,2R,4'S)-2-hydroxy-6'neopentyl-3',4'dihydrospiro[cyclobut ane-l,2'- pyrano[2,3-b]pyridin-4'-amine as a yellow solid.

Example 8

(lR^R,4'S)-2-Hydroxy-6'-neopentyl-3',4'dihydrospiro[cyclo butane-l^'-pyrano[23- b]pyridin-4'-amine and (lS^S,4'S)-2-hydroxy-6'neopentyl- 3',4'dihydrospiro[cyclobutane-l^'-pyrano[2β-b]pyridin-4'-am ine

The title compounds were prepared by a method analogous to that described in Example 7 above, starting with (trans)- l-allyl-2-(benzyloxy)cyclobutanol as the starting material.

(S)-3-(4-Amino-8-fluoro-2^-spirocyclobutyl-6-yl)-2^-dimet hylpropanenitrile Step 1: l-(5-Bromo-3-fluoro-2-hvdroxyphenyl)ethanone

4-Bromo-2-fluorophenyl acetate (126 g, 540 mmol) in 1,2-dichlorobenzene (53 mL) was added dropwise to aluminum(III) chloride (72 g, 540 mmol) in 1,2-dichlorobenzene (64 mL) with vigorous stirring to give a red solution. The solution was heated to 120 0 C for 60 hours, cooled, diluted with DCM, and added to IN HCl at 0 0 C. The layers were separated and the aqueous layer was extracted with DCM. The combined organic layers were washed with IN HCl, water, brine, and dried over sodium sulfate and concentrated. The crude material was taken up in hexanes and added to aqueous IN NaOH at 0 0 C. The solid was collected and washed with hexanes. The aqueous filtrate and solid were acidified with concentrated HCl and extracted with ethyl acetate. The combined organic

A- 1291 -WO-PCT - 74- layers were washed with water, brine, dried over sodium sulfate, and concentrated. The crude solid was recrystallized from MeOH to afford the title compound (46 g, 37% yield). Step 2: ό-Bromo-S-fluoro^^-spirocvclobutvM-one l-(5-Bromo-3-fluoro-2-hydroxyphenyl)ethanone (15.00 g, 64 mmol), pyrrolidine (8 ml, 97 mmol), DIPEA (11 ml, 64 mmol), and cyclobutanone (9 ml, 129 mmol) were heated at 65 0 C for 12 hours. After cooling, the reaction was diluted with EtOAc and washed with IN HCl. The aqueous layer was extracted with EtOAc. The organic layers were combined, washed with water, brine, dried over sodium sulfate and concentrated. The residue was chromatographed on a silica gel column (10:1 Hexanes/Ether) to afford the title compound (9.73 g, 53% yield) as an orange solid. MS m/z: 285.0(100%, M). Step 3: (R " )-6-Bromo-8-fluoro-2.2-spirocvclobutyl-4-ol

(s)-2-Methyl-cbs-oxazaborolidine, IM in toluene (3.41 ml, 3.41 mmol) was added to a solution of borane-dimethyl sulfide (4.86 ml, 51.2 mmol) in 74 mL of toluene at 0 0 C. After stirring 20 minutes, 6-bromo-8-fluoro-2,2-spirocyclobutyl-4-one (9.73 g, 34.1 mmol) was added via syringe pump in 106 mL of toluene over 1.5 hour at -5° C. After stirring an additional 30 minutes at -5 0 C the reaction was quenched by the addition of methanol and then IN HCl. The mixture was extracted with ethyl acetate and the combined organic layers were washed 2x with 50% saturated ammonium chloride, brine, and dried over sodium sulfate. Concentration of the filterd organic layer afforded the title compound as yellow oil.

Step 4: (S " )-4-Azido-6-bromo-8-fluoro-2.2-spirocyclobutyl

Diphenyl azidophosphate (4.90 ml, 22.7 mmol) was added to a solution of (R)-6-bromo- 8-fluoro-2,2-spirocyclobutyl-4-ol (4.35 g, 15.2 mmol) and DBU (3.43 ml, 22.7 mmol) in toluene (28 mL). The reaction was allowed to stir 48 hours and was filtered through a pad of silica gel and washed with EtOAc. Concentration of the EtOAc afforded the title compound.

Step 5: (S)-6-Bromo-8-fluoro-2.2-spirocvclobutyl-4-amine

Raney nickel (2800), slurry, in water (0.4 g, 6 mmol) was added to (S)-4-azido-6-bromo- 8-fluoro-2,2-spirocyclobutyl. (4.73 g, 15 mmol) dissolved in i-PrOH (150 mL). Hydrazine, monohydrate (5 ml, 76 mmol) was added and the reaction mixture was stirred 30 minutes before being filtered through a pad of Celite washing with ethanol. The EtOH solvent was concentrated, and the resulting crude material was purified by silica gel chromatography (20:1 DCM/MeOH (2M NH 3 ) to afford the title product. MS m/z: 269.0 (100%, M- 17).

A-1291-WO-PCT - 75-

Step 6: (SVtert-Butyl β-bromo-S-fluoro^^-spirocvclobutvM-ylcarbamate (S)-6-Bromo-8-fluoro-2,2-spirocyclobutyl-4-amine (3.00 g, 10 mmol), TEA (2.2 ml, 16 mmol), and BOC-anhydride (3.0 g, 14 mmol) were stirred in DCM (30 mL) for 12 hrs and concentrated. The crude material was taken up in EtOAc and washed with saturated ammonium chloride, water, brine, dried over sodium sulfate and concentrated. The crude material was purified by recrystallization from methanol and water to afford the title product as a white solid.

Step 7: (SVtert-Butyl allvKό-bromo-S-fluoro^^-spirocvclobutvM-yl ' lcarbamate (S)-tert-Butyl 6-bromo-8-fluoro-2,2-spirocyclobutyl-4-ylcarbamate (5.7 g, 15 mmol) was dissolved in DMF (70 mL) and cooled to 0° C. NaH (0.71 g, 18 mmol) was added carefully to the mixture and the solution was allowed to stir for 40 minutes. Allyl bromide (1.4 ml, 16 mmol) was added and the reaction mixture was stirred 45 minutes and then diluted with saturated aqueous ammonium chloride. Water was added and the solution was extracted with ether. The combined organic layers were washed with water, brine, dried over magnesium sulfate, filtered and concentrated to afford the title compound. MS m/z: 370.1 (100%, M-55).

Step 8: (SVtert-Butyl allyl(8-fluoro-6-(hvdroxymethyl)-2,2-spirocvclobutyl-4- vDcarbamate (S)-tert-Butyl allyl(6-bromo-8-fluoro-2,2-spirocyclobutyl-4-yl)carbamate (6.30 g, 15 mmol) was dissolved in diethyl ether (75 mL) and cooled to -78 0 C. tert-butyllithium (1.7 M) (19 ml, 33 mmol) was added dropwise to give a dark orange solution. After 20 minutes, DMF (13 ml, 163 mmol) was added and the solution was stirred for 45 minutes before being quenched by the addition of saturated ammonium chloride and water. The aqueous solution was extracted with EtOAc and the combined organic layers were washed with water, brine, dried over sodium sulfate and concentrated. The crude material was dissolved in 80 mL of MeOH, cooled to 0 0 C and NaBH 4 (0.84 g, 22 mmol) was added to the cooled mixture. After stirring 40 minutes the reaction mixture and was quenched by addition of saturated ammonium chloride and water. The aqueous solution was extracted with EtOAc and the combined organic layers were washed with water, brine, dried over sodium sulfate and concentrated. The crude residue was purified by column chromatography (4: 1 Hex/EtOAc) to give the title product. Step 9: (SVtert-Butyl allyl(6-(2-cvano-2-methylpropylV8-fluoro-2,2-spirocyclobutyl -4- vDcarbamate

A- 1291 -WO-PCT - 76-

Dibromotriphenylphosphorane (4.28 g, 10.1 mmol) was added to a solution of (S)-tert- butyl allyl (S-fluoro-o-thydroxymethyl^^-spirocyclobutyM-yOcarbamate (3.48 g, 9.22 mmol) and N-ethyl-N-isopropylpropan-2-amine (1.61 ml, 9.22 mmol) in DCM (80 mL) at 0 0 C. After stirring 45 minutes at 0 0 C and 30 minutes at ambient temperature, the reaction was concentrated and taken up in THF (40 mL). In a seperate flask, diisopropylamine (8.21 ml, 58.1 mmol) was added to THF (90 mL) and the solution was cooled to -78 0 C. «-Butyllithium (22.1 ml, 55.3 mmol) was added and the solution was stirred 20 minutes at 0 0 C. Isobutyronitrile (4.96 ml, 55.3 mmol) was added and the yellow solution was stirred 40 minutes at 0 0 C before the intermediate benzyl bromide described above in THF (40 mL) was added dropwise via addition funnel. The reaction was stirred at 0 0 C and after 1 hour was complete. The reaction was quenched with saturated ammonium chloride and was extracted with EtOAc and the combined organic layers were washed with water, brine, dried over sodium sulfate and concentrated. The crude material was purified by silica gel chromatography (1.5:1 Hex/EtOAc)to afford the title product. MS m/z: 373.3(100%, M-55).

Step 10: (S)-3-(4-(AllylaminoV8-fluoro-2,2-spirocvclobutyl-6-yl)-2,2- dimethylpropanenitrile

(S)-tert-Butyl allyl (6-(2-cyano-2-methylpropyl)-8-fluoro-2,2-spirocyclobutyl-4- yl)carbamate (2.90 g, 6.8 mmol) and TFA (25 ml, 324 mmol) were stirred in DCM (50 mL) for 3 hours and concentrated. The crude product was taken up in DCM and 1 N NaOH and separated. The aqueous layer was extracted with DCM and the combined organic layers were washed with brine and dried over sodium sulfate. Concentration afforded the title product which was used without further purification. MS m/z: 329.3 (100%, M+l). Step 11: (S)-3-(4-Amino-8-fluoro-2,2-spirocyclobutyl-6-ylV2.2-dimethy lpropanenitrile (S)-3-(4-(Allylamino)-8-fiuoro-2,2-spirocyclobutyl-6-yl)-2,2 -dimethylpropanenitrile was dissolved in degassed (N 2 ) DCM (40 mL) and 1,3-dimethylbarbituric acid (3.2 g, 20 mmol) was added. After two minutes, Pd(PPh 3 ) 4 (0.78 g, 0.68 mmol) was added and the reaction was stirred at ambient temperature for 12 hours. The reaction was diluted with DCM and 10% aqueous sodium carbonate and the layers were separated. The aqueous layer was extracted with DCM and the combined organic layers were washed with brine, dried over sodium sulfate and concentrated. The crude product was purified by silica gel chromatography (20: 1 DCM/MeOH (2M NH 3 )) to afford the title product. MS m/z: 289.2 (37%, M+l); 272.2 (100%, M-16).

A-1291-WO-PCT - 77-

(S)-8-Fluoro-2£-spirocyclobutyl-6-neopentylchroman-4-yl- amiiie

To zinc(II) chloride (0.5 M THF) (50.3 ml, 25.1 mmol) was added neopentylmagnesium chloride (1.0 M ether) (39.6 ml, 39.6 mmol) in a sealed tube and stirred 20 minutes. 1,1'- bis(diphenylphosphino)ferrocene-palladium dichloride (0.557 g, 0.762 mmol) was added followed by (S)-6-bromo-8-fluoro-2,2-spirocyclobutyl-4-amine (2.18 g, 7.62 mmol) in THF (4 mL). The tube was sealed and heated to 7O 0 C for 12 h. The reaction was cooled and dilute with EtOAc and aqueous solution of a 9:1 saturated ammonium chloride/ammonium hydroxide solution (PH = 9) and separated. The aqueous layer was extracted with EtOAc, and the combined organic layers were washed with a 9:1 saturated ammonium chloride/ammonium hydroxide solution (PH = 9), water, brine, dried over sodium sulfate, and concentrated. Purification by silica gel chromatography (40:1 DCM/MeOH 2MNH 3 ) afforded the title product. MS m/z: 261.2 (100%, M-16).

(S)-3-(4-Amino-2^-spirocyclobutyl-6-yl)-2,2-dimethylpropanen itrile

The title compound was prepared according to steps 1-11 of the procedure described in Example 9 above.

Example 12

A- 1291 -WO-PCT - 78-

(S)-8-Fluoro-2^-tetrahydrospirofuranyl-6-neopentylchroman -4-amine

Step 1: ό-Bromo-S-fluoro-σ^-tetrahvdrospirofuranylchroman^-one l-(5-Bromo-3-fluoro-2-hydroxyphenyl)ethanone (0.200 g, 0.858 mmol), dihydrofuran- 3(2H)-one (0.222 g, 2.57 mmol), and pyrrolidine (0.142 ml, 1.72 mmol) were dissolved in MeCN (0.5 mL) and heated in the microwave for 20 minutes fixed at 60 0 C. After cooling, the reaction was diluted with EtOAc and washed with IN HCl. The aqueous layer was extracted with EtOAc. The organic layers were combined, washed with water, brine, dried over sodium sulfate, and concentrated. The crude product was purified by silica gel chromatography (1 :4 EtOAc/hexanes) to afford the titled products. MS m/z: 301.0 (100%, M).

Step 2: (R)-6-Bromo-8-fluoro-2.2-tetrahvdrospirofuranylchroman-4-ol (s)-2-Methyl-cbs-oxazaborolidine (0.767 ml, 0.767 mmol) was added to a solution of borane-methyl sulfide complex (1.09 ml, 11.5 mmol) in 16 mL of toluene at 0 0 C. After stirring 20 minutes, 6-bromo-8-fluoro-2,2-tetrahydrospirofuranylchroman-4-one (2.31 g, 7.67 mmol) was added via syringe pump in 23 mL of toluene over 1.5 hour at -5° C. After stirring an additional 30 minutes at -5 0 C the reaction was quenched by the addition of MeOH and then IN HCl. The mixture was extracted with ethyl acetate and the combined organic layers were washed twice with 50% saturated ammonium chloride, brine, and dried over sodium sulfate. Concentration of the filtered organic layer afforded the titled product as a yellow oil.

Step 3: (S)-4-Azido-6-bromo-8-fluoro-2.2-tetrahvdrospirofuranylchrom an Diphenylphosphoryl azide (1.93 ml, 8.96 mmol) was added to a solution of (R)-6-bromo- 8-fluoro-2,2-tetrahydrospirofuranylchroman-4-ol (1.81 g, 5.97 mmol) and DBU (1.35 ml, 8.96 mmol) in toluene (10 mL). The reaction was allowed to stir 48 hours and was filtered through a pad of silica gel with ethyl acetate. Concentration of the filtered organic layer afforded the titled products which were used without further purification. Step 4. (SV6-Bromo-8-fluoro-2.2-tetrahvdrospirofuranylchroman-4-yl-a mine Raney nickel (2800, as a slurry in water) (0.19 g, 3.3 mmol) was added to (S)-4-azido-6- bromo-8-fluoro-2,2-tetrahydrospirofuranylchroman. (1.2 g, 3.7 mmol) dissolved in i-

A- 1291 -WO-PCT - 79-

PrOH (50 mL). Hydrazine hydrate (1.1 ml, 18 mmol) was added and the reaction was stirred 30 minutes and then filtered through a pad of celite with ethanol, concentrated, and purified by silica gel chromatography (20: 1 DCM/MeOH-NH3) to afford the titled products. MS m/z: 302.1 (5%, M); 285.1 (100%, M-17). Step 5 : (SV8-Fluoro-2.2-tetrahvdrospirofuranyl-6-neopentylchroman-4- amine To zinc chloride, 0.5M solution in THF (31 ml, 15 mmol) was added 2,2- dimethylpropylmagnesium chloride, 1.0 M solution in diethyl ether (25 ml, 25 mmol) in a sealed tube and stirred 20 minutes. l,r-Bis(diphenylphosphino)ferrocene-palladium dichloride (0.2 g, 0.3 mmol) was added to the mixture followed by (S)-6-bromo-8-fluoro- 2,2-tetrahydrospirofuranylchroman-4-amine (0.932 g, 3 mmol) in THF (8 mL). The tube was sealed and heated to 70 0 C for 12 h. The reaction was cooled and diluted with DCM and an aq. solution of a 9:1 saturated ammonium chloride/ammonium hydroxide and the layers were separated. The aqueous layer was extracted with DCM, and the combined organic layers were washed with a 9:1 saturated ammonium chloride/ammonium hydroxide solution, water, brine, dried over sodium sulfate, and concentrated. Purification of the crude concentrate by silica gel chromatography (20:1 DCM/MeOH-NH3) afforded the title compounds.

(S)-7,7-Spirocyclobutyl-3-neopentyl-5,6,7,8-tetrahydroqui nolin-5-amine

Step 1: 3-Amino-5.5-spirocvclobutylcvclohex-2-enone

5,5- Spirocyclobutylcyclohexane-l,3-dione (15.2 g, 99.9 mmol), HOAc (5.15 ml, 89.9 mmol), and ammonium acetate (15.4 g, 200 mmol) were refluxed in benzene (250 mL) with a Dean-Stark trap for 5 hrs. After cooling, the product was filtered from the reaction mixture and the solid was taken up in EtOAc and washed with saturated sodium bicarbonate, brine, dried over sodium sulfate, and concentrated to afford the title product as a yellow solid. Step 2: 4.4-Dimethvlpentanal

A-1291-WO-PCT - 80-

DIBAL-H (1 M Hexanes)(80 ml, 80 mmol) was added to 4,4-dimethylpentanenitrile (5.9 g, 53 mmol) in DCM (200 mL) at 0 0 C. After stirring for 1.5 hrs the reaction was quenched with concentrated HCl and diluted with DCM. The aqueous layer was extracted with DCM and the combined organic layers were washed with concentrated HCl, water, brine, and dried over sodium sulfate. The organics were filtered and concentrated to afford the title compound which was used without further purification. Step 3: (ZV2-(Ethoxymethylene)-4.4-dimethylpentanal

4,4-Dimethylpentanal (4.20 g, 36.8 mmol, Step 2) in 36 mL of THF was added over 4 hours by syringe pump to a solution of sodium methoxide, 25 wt. % in methanol (15.1 ml, 66.2 mmol), 15 mL of methanol, and methyl formate (166 ml, 2685 mmol). Following the addition, the reaction was diluted with benzene (50 mL) and DMF (40 mL). The solvents were distilled off at 90 0 C leaving the DMF and residual benzene. Bromoethane (11.0 ml, 147 mmol) was added and the solution was heated at 40° C for 48 hrs and then cooled. The solution was diluted with water, and the reaction was extracted with ether. The combined organic layers were washed with water, brine, and dried over magnesium sulfate, filtered and concentrated toafford the title compound. Step 4: 7-Spirocvclobutyl-3-neopentyl-7.8-dihvdroquinolin-5(6FD-one (Z)-2-(Ethoxymethylene)-4,4-dimethylpentanal (5.22 g, 31 mmol) and 3-amino-5,5- spirocyclobutylcyclohex-2-enone (4.6 g, 31 mmol) were heated to 140 0 C in propionic acid (30 ml, 399 mmol)for 12 hours. After cooling, the reaction was diluted with ethyl acetate and washed 2x with IN NaOH. The aqueous layer was extracted with ethyl acetate and the combined organic layers were washed with brine, dried over sodium sulfate, and concentrated. Purification of the crude by silica gel chromatography (3:1 Hexanes/EtOAc) afforded the titled product (3.1 g, 39% yield). MS m/z: 258.2 (100%, M+l).

Step 5: (RV7.7-Spirocvclobutyl-3-neopentyl-5.6.7.8-tetτahvdroquinol in-5-ol. (s)-2-Methyl-cbs-oxazaborolidine, 1 M in toluene (1.2 ml, 1.2 mmol) was added to a solution of borane-dimethyl sulfide (1.7 ml, 18 mmol) in 26 mL of toluene at 0 0 C. After stirring 20 minutes, 7,7-spirocyclobutyl-3-neopentyl-7,8-dihydroquinolin-5(6H)-on e (3.10 g, 12 mmol) was added to the mixture via syringe pump in 37 mL of toluene over 1.5 hour at -5 0 C. After stirring an additional 30 minutes at -5 0 C the reaction was quenched by the addition of methanol and then IN HCl. The mixture was extracted with ethyl acetate and the combined organic layers were washed 2 times with 50% saturated

A-1291-WO-PCT - 81- ammonium chloride, brine, and dried over sodium sulfate, filtered and concentrated to afford the titled product.

Step 6: (S)-5-Azido-7.7-spirocvclobutyl-3-neopentyl-5.6.7.,8-tetrahy droquinoline Dipheηylphosphoryl azide (3.5 ml, 16 mmol) was added to a solution of (R)-7,7- spirocyclobutyl-3-neopentyl-5,6,7,8-tetrahydroquinolin-5-ol (2.8 g, 11 mmol) and DBU (2.4 ml, 16 mmol). The reaction was allowed to stir 24 hours and was filtered through a pad of silica gel washing and eluting with ethyl acetate. Concentration of the EtOAc afforded the titled product which was used without further purification. Step 7: (S)-7.7-Spirocvclobutyl-3-neopentyl-5.6.7,8-tetrahvdroquinol in-5-amine A solution of (S)-5-azido-7,7-spirocyclobutyl-3-neopentyl-5,6,7,8-tetrahyd roquinoline (1.21 g, 4 mmol) in ethanol (30 mL) was purged with nitrogen gas. Palladium, 5% on calcium carbonate, poisoned with lead (0.5 ml, 4 mmol) was added and the flask was flushed with a balloon of hydrogen gas. The reaction was allowed to stir under an atmosphere of hydrogen gas for 4 hours. After purging the reaction with nitrogen gas, the . reaction was filtered through a pad of celite with ethanol. The reaction was concentrated and purified by silica gel column (20:1 DCM/ MeOH 2N NH 3 ) to afford the titled product. MS m/z: 259.3(100%, M+l).

(S)-8,8-Difluoro-7,7-spirocyclobutyl-3-neopentyl-5,6,7,8- tetrahydroquinoIin-5-amine

Step 1: (S)-tert-butyl 7,7-spirocvclobutyl-3-neopentyl-5,6,7,8-tetrahydroquinolin-5 - ylcarbamate

(S)-7,7-Spirocyclobutyl-3-neopenryl-5,6,7,8-tetrahydroqui nolin-5-amine (3.47 g, 13.4 mmol), TEA (2.81 ml, 20.1 mmol), and di-tert-butyl dicarbonate (2.93 g, 13.4 mmol) were stirred in DCM (60 mL) for 12 hrs and concentrated. The crude material was taken up in ethyl acetate and washed with saturated ammonium chloride, water, brine, dried over sodium sulfate, and concentrated. The crude material was purified by column chromotography (10:1 to 1:1 Hexanes/EtOAc) to afford the titled product.

A- 1291 -WO-PCT - 82-

Step 2: N-oxide of (SVtert-butyl 7,7-spirocyclobutyl-3-neopentyl-5.6,7,8- tetrahydroquinolin-5 -ylcafbamate w-Chloroperbenzoic acid (0.662 g, 2.30 mmol) was added to a solution of (S)-tert-butyl 7,7-spirocyclobutyl-3-neopentyl-5,6,7,8-tetrahydroquinolin-5 -ylcarbamate (0.688 g, 1.92 mmol) in DCM (20 mL) and the solution was stirred 12 hrs before being diluted with aqueous saturated sodium bicarbonate and aqueous sodium thiosulfate. The mixture was stirred vigorously for 1.5 hrs then separated. The aqueous layer was extracted with DCM and the combined organic layers were washed with aqueous sodium thiosulfate, 10% sodium carbonate, water, brine, and dried over sodium sulfate. The organic layer was filterd and concentrated to afford the title product.

Step 3: (SVtert-Butyl 8-hydroxy-7J-spirocvclobutyl-3-neopentyl-5.6,7,8- tetrahydroquinolin-5 -ylcarbamate

The N-oxide from above (0.719 g, 1.92 mmol, Step 2) was dissolved in DCM (9 mL). Trifluoroacetic anhydride (1.33 ml, 9.60 mmol) was added and the reaction was refluxed for 2 hours and concentrated. The crude product was dissolved in THF (4.5 mL) and aqueous saturated sodium bicarbonate was added by pipette in dropwise fashion until no further bubbling was observed. The reaction was diluted with ethyl acetate and water and the layers were separated. The aqueous layer was extracted with ethyl acetate and the combined organic layers were washed with water, brine, and dried over sodium sulfate, filtered and concentrated. The crude product was purified by silica gel chromatography (25:1 DCM/MeOH) to afford the titled product. MS m/r. 375.3(100%, M+l). Step 4: (S)-tert-Butyl 7.7-spirocvclobutyl-3-neopentyl-8-oxo-5.6.7.8-tetrahvdroquin olin- 5-ylcarbamate (S)-tert-Butyl 8-hydroxy-7,7-spirocyclobutyl-3-neopentyl-5,6,7,8-tetrahydro quinolin-5- ylcarbamate (0.285 g, 0.761 mmol) and Dess-Martin Periodinane (0.968 g, 2.28 mmol) were stirred 12 hrs in DCM (7 mL). The reaction was diluted with ether and aqueous sodium thiosulfate and saturated aqueous sodium bicarbonate and stirred vigorously. The layers were separated and the aqueous layers were extracted with ether and the combined organic layers were washed with saturated aqueous sodium bicarbonate and water and concentrated. The residue was taken up in ethyl acetate and washed with brine, dried over sodium sulfate, and concentrated. Purification by silica gel chromatography (1:1 Hexanes/EtOAc) afforded the titled product. MS m/r. 373.3 (100%, M+l). Step 5: (SVtert-Butyl 8.8-difluoro-7.7-spirocvclobutyl-3-neopentyl-5,6J.8- tetrahvdroαuinolin-5-ylcarbamate

A-1291-WO-PCT - 83-

(S)-tert-Butyl 7,7-spirocyclobutyl-3-neopentyl-8-oxo-5,6,7,8-tetrahydroquin olin-5- ylcarbamate (0.183 g, 0.491 mmol) was dissolved in DCM (1.5 mL) and cooled to -78 0 C. DAST (0.130 ml, 0.983 mmol) was added and the reaction mixture was allowed to warm to RT over 12 hrs and stirred for four additional days. The reaction was diluted with DCM. and aqueous 10% sodium carbonate and separated. The aqueous layer was extracted with DCM and the combined organic layers were washed with water, brine, and dried over sodium sulfate. Concentration of the filtered organic solvent afforded the titled product. MS m/z: 395.2 (100%, M+l). Step 6: (S " )-8.8-Difluoro-7.7-spirocvclobutyl-3-neopentyl-5.6.7.8-tetra hydroquinolin-5- amine

(S)-tert-Butyl 8,8-difluoro-7,7-spirocyclobutyl-3-neopentyl-5,6,7,8-tetrahy droquinolin-5- ylcarbamate (0.194 g, 0.49 mmol) was stirred in a 2:1 solution of DCM and TFA (4.5 mL) for 3 hours and concentrated. The crude product was taken up in chloroform and IN aq. NaOH and the layers were separated. The aqueous layer was extracted with chloroform and the combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated. The crude product was purified by silica gel chromatography (30:1 DCMZMeOH-NH 3 ) to afford the titled product. MS m/z: 295.2 (100%, M+l).

(S)-2^-SpirocyclobutyI-6-(3 r 3 r 3-trifluoropropyl)-3,4-dihydro-2H-pyrano[2^- b] py ridin-4-amine

Step 1 : ( 1 s.5 s)-9-(3.3.3 -Trifluoropropyl)-9-bora-bicvclo [3.3.1 lnonane 3,3,3-Trifluoroprop-l-ene (2.9 g, 30 mmol) was condensed with a cold finger and the liquid was allowed to drop into a 500 mL RBF containing 9-BBN, 0.5M in THF (58 ml, 29 mmol) cooled in a dry ice/iPrOH bath. After the addition was complete the solution was allowed to stir under N 2 and slowly warm to RT.

A- 1291 -WO-PCT - 84-

Step 2: (S)-tert-butyl 2.2-spirocvclobutyl-6-(3.3.3-trifluoropropyl ' )-3.4-dihvdro-2H- pyranor2,3-b1pyridin-4-ylcarbamate

To a 500 mL RBF containing (ls,5s)-9-(3,3,3-trifluoropropyl)-9-bora- bicyclo[3.3.1]nonane (6.3 g, 29 mmol, step 1) in THF (58 mL) was added, toluene:EtOH (110 mL, 10:1), (S)-tert-butyl 6-bromo-2,2-spirocyclobutyl-3 ,4- dihydro-2H-pyrano[2,3-b]pyridin-4-ylcarbamate (5.12 g, 14 mmol), sodium carbonate, 10% aqueous (28.0 ml, 26 mmol), and palladium tetrakis (790 mg, 0.68 mmol, Strem). The solution was stirred at 8O 0 C. After 16 hours, LC-MS shows reaction to be ~20% complete and not progressing. The reaction was concentrated to half volume and the layers seperated and the aqueous layer extracted with EtOAc (2 X 30 mL). The combined organic layers were concentrated in vacuo and adsorbed onto a plug of silica gel and chromatographed through a Redi-Sep® pre-packed silica gel column (120 g), eluting with 0% to 20% EtOAc in hexane, to provide (S)-tert-butyl 2,2-spirocyclobutyl-6-(3,3,3-trifluoropropyl)-3,4-dihydro-2H - pyrano[2,3-b]pyridin-4-ylcarbamate (1.01 g, 19% yield, 56% brsm) as a yellow oil that began to solidify upon standing under vacuum overnight. Step 3; (S ' )-2.2-spirocyclobutyl-6-(3.3.3-trifluoropropyl')-3,4-dihvdro -2H-pyranor2.3- b]pyridin-4-amine To a 250 mL round bottomed flask containing (S)-tert-buryl 2,2-spirocyclobutyl-6-(3,3,3- trifluoropropyl)-3,4-dihydro-2H-pyrano[2,3-b]pyridin-4-ylcar bamate (1.00 g, 2588 μmol) in EtOAc (40 mL) was added HCl (4M in dioxane, 5 mL). The solution was stirred at RT. After 4 hours, another 5 mL of HCl was added to the mixture. After a further 16 hours, the reaction was poured into sat'd NaHCO3. The aqueous layer was extracted with EtOAc and the combined organic layers washed with brine, dried over MgSO 4 , and concentrated in vacuo to give (S)-2,2-spirocyclobutyl-6-(3,3,3-trifluoropropyl)-3,4- dihydro-2H-pyrano[2,3-b]pyridin-4-amine (700 mg, 94.5% yield), as a yellow oil. MS m/z: 287.2 (M+l).

Example 16

A- 1291 -WO-PCT - 85-

(S)-tert-Butyl 6-allyl-2^-spirocyclobutyl-3,4-dihydro-2H-pyrano[2^-b]pyridi n-4- ylcarbamate

Step 1: (S)-tert-Butyl 6-allyl-2,2-spirocvclobutyl -S^-dihvdro^H-pyranoβ.S-bipyridin- 4-ylcarbamate A solution of tris(dibenzylideneacetone)dipalladium (o) (0.186 g, 0.203 mmol), tri-t- butylphosphonium tetrafluoroborate (0.354 g, 1.22 mmol), cesium fluoride (3.09 g, 20.3 mmol), and (S)-tert-butyl 6-bromo-2,2-spirocyclobutyl -3,4-dihydro-2H-pyrano[2,3- b]pyridin-4-ylcarbamate (1.500 g, 4.06 mmol) in dioxane(12mL) was thoroughly degassed then treated with allyltributylstannane (6.23 ml, 20.3 mmol). The reaction mixture was heated to 100 0 C and allowed to stir 4 hours. The reaction mixture was then diluted with EtOAc, washed with saturated KF solution, water, and brine. The organic layer was dried over MgSθ 4 and concentrated under reduced pressure. Purification of the crude residue by column chromatography gave (S)-tert-butyl 6-allyl-2,2-spirocyclobutyl - 3,4-dihydro-2H-pyrano[2,3-b]pyridin-4-ylcarbamate (0.820 g, 61.1% yield) as a light yellow solid.

Step 2: (SVtert-Butyl 6-(cvclopropylmethyl)- 2,2-spirocyclobutyl -3,4-dihydro-2H- pyrano| " 2.3-b1pyridin-4-ylcarbamate

A solution of (S)-tert-butyl 6-allyl-2,2-spirocyclobutyl -3,4-dihydro-2H-pyrano[2,3- b]pyridin-4-ylcarbamate (0.710 g, 2 mmol) in DCM(1.5mL ) was cooled to -10°C and treated with diethylzinc IM in hexanes (21 ml, 21 mmol), followed by dropwise addition of chloroiodomethane (3 ml, 43 mmol). After 1 hour an additional portion of diethylzinc and chloroiodomethane was added. After an additional hour at RT the reaction mixture was quenched with concentrated NH 4 Cl solution and diluted with ether. The organic layer was washed with 2N NaOH, brine, dried over Na 2 SO 4 and concentrated under reduced pressure. The crude residue was retreatted with the reaction and work-up conditions three times. Purification of the crude residue by column chromatography gave (S)-tert-butyl 6- (cyclopropylmethyl)- 2,2-spirocyclol>utyl -3,4-dihydro-2H-pyrano[2,3-b]pyridin-4- ylcarbamate (0.300 g, 41% yield) as a clear oily solid with minor impurites. Step 3: (S)-6-(Cvclopropylmethyl)- 2.2-spirocyclobutyl -3,4-dihydro-2H-pyranor2.3- blpyridin-4-amine

A solution of (S)-tert-Butyl 6-(cyclopropylmethyl)- 2,2-spirocyclobutyl -3,4-dihydro-2H- pyrano[2,3-b]pyridin-4-ylcarbamate (0.300 g, 0.87 mmol) and DIEA (0.30 ml, 1.7 mmol) in DCM(2mL) was treated with trimethylsilyltriflate (0.19 ml, 1.0 mmol) and was allowed to stir at RT overnight. An additional 4 equivalents of DIEA then 4 equivalents

A- 1291 -WO-PCT - 86- of trimethylsilyltriflate were then added. After an additional 2 hours of stirring the reaction mixture was quenched with 4N HCl in dioxane and allowed to stir for 1 hr. The reaction mixture was diluted with EtOAc, washed with 2N NaOH, water, and brine. The organics were dried over MgSθ 4 and concentrated under reduced pressure. Purification of the crude residue by column chromatography gave (S)-6-(cyclopropylmethyl)- 2,2- spirocycloburyl -3,4-dihydro-2H-pyrano[2,3-b]pyridin-4-amine as a 2:1 mixture an impurity.

Example 17

tert-Butyl (S)-6-(3-hydroxy-2-methylpropyl)- 2,2-spirocyclobutyl -3,4-dihydro-2H- pyrano[2,3-b]pyridin-4-ylcarbamate

Step 1: tert-butyl (S)-6-(3-hvdroxy-2-methylpropyl)- 2,2-spirocvclobutyl -3,4-dihvdro- 2H-pyranor2.3-b1pyridin-4-ylcarbamate A solution of tert-butyldimethyl(2-methylallyloxy)silane (9 g, 47 mmol) and 9-BBN 0.5M in THF (95 ml, 47 mmol) was degassed and allowed to stir at RT for 3 hours. A separate solution of palladium(II) acetate (0.2 g, 0.9 mmol) and S-Phos (1 g, 3 mmol) in THF(5mL) and benzene(5mL) was thoroughly degassed and allowed to stir at RT for one hour. The palladium solution was then added to the borane solution, followed by potassium phosphate (8 g, 38 mmol) and a solution of (S)-tert-butyl 6-bromo-2,2- spirocyclobutyl -3,4-dihydro-2H-pyrano[2,3-b]pyridin-4-ylcarbamate ( (3.5 g, 9 mmol) in DMF(15mL). The reaction mixture was degassed and heated to 100 0 C 4 hours. Cesium fluoride (14 g, 95 mmol) was added followed by ImL water and the reaction mxiture was heated to 100 0 C overnight. The reaction mixture was decanted to a clean flask and concentrated. The crude residue was treated with TBAF IM in THF (19 ml, 19 mmol) and was allowed to stir at RT for 2 hours. The reaction mixture was diluted with EtOAc, washed with saturated NH4C1 solution, water, and brine. The organics were dried over MgSO 4 and concentrated under reduced pressure. Purification of the crude residue by column chromatography gave tert-butyl (S)-6-(3-hydroxy-2-methylpropyl)- 2,2- spirocyclobutyl -3,4-dihydro-2H-pyrano[2,3-b]pyridin-4-ylcarbamate (1.444 g, 42% yield) as a yellow solid.

A-1291-WO-PCT . - 87-

Step 2: tert-Butyl (SV6-(2-methyl-3-oxopropyl)- 2.2-spirocvclobutyl -3,4-dihvdro-2H- pyranor2.3-blpyridin-4-ylcarbamate

A solution of tert-butyl (S)-6-(3-hyaroxy-2-methylpropyl)- 2,2-spirocyclobutyl -3,4- dihydro-2H-pyranot2,3-b]pyridin-4-ylcaτbamate (2.290 g, 6.32 mmol) in.wet DCM (2OmL) was treated with Dess-Martin periodinane(3.22 g, 7.58 mmol) and allowed to stir for 2 hours. The reaction mixture was diluted with 2OmL ether, treated with ImL saturated NaHCθ 3 solution followed by sodium thiosulfate (4.99 g, 31.6 mmol) and was allowed to stir at RT for 1 hr. The reaction mixture was then diluted with EtOAc, washed with saturated NaHCC> 3 solution, water, and brine. The organic layer was dried over MgSO 4 and concentrated under reduced pressure. Purification of the crude residue by column chromatography gave tert-butyl (S)-6-(2-methyl-3-oxopropyl)- 2,2- spirocyclobutyl -3,4-dihydro-2H-pyrano[2,3-b]pyridin-4-ylcarbamate (1.43 g, 62.8% yield) as a sticky white solid. Step 3: tert-Butyl (SV6-(3.3-difluoro-2-methylpropylV 2.2-spirocvclobutyl -3.4-dihydro- 2H-pyranor2.3-blpyridin-4-ylcarbamate

A solution of tert-butyl (S)-6-(2-methyl-3-oxopropyl)- 2,2-spirocyclobutyl -3,4-dihydro- 2H-pyrano[2,3-b]pyridin-4-ylcarbamate (1.180 g, 3.27 mmol) in DCM(30mL) was cooled to -78°C and treated with deoxofluor (0.905 ml, 4.91 mmol). The solution was allowed to warm to RT and triethylamine trihydrofluoride (0.0534 ml, 0.327 mmol) was added. After 1.5 hours, so an additional equivalent of deoxofluor was added, and the reaction mixture was allowed to stir at RT for an additional hour. The reaction mixture was concentrated under reduced pressure, and the crude residue was purified by column chromatography yielding tert-butyl (S)-6-(3,3-difluoro-2-methylpropyl)- 2,2-spirocyclobutyl -3,4-dihydro- 2H-pyrano[2,3-b]pyridin-4-ylcarbamate (0.940 g, 75.1% yield) as a white solid. Step 4: (4S)-6-(3.3-difluoro-2-methylpropylV 2.2-spirocvclobutyl -3.4-dihvdro-2H- pyranor2,3-blpyridin-4-amine

A solution of tert-butyl (S)-6-(3,3-difluoro-2-methylpropyl)- 2,2-spirocyclobutyl -3,4- dihydro-2H-pyrano[2,3-b]pyridin-4-ylcarbamate (1.015 g, 2.65 mmol) in DCM(IOmL) was treated with TFA (10.2 ml, 133 mmol) and allowed to stir overnight at RT. The reaction mixture was then concentrated under reduced pressure. The crude residue was dissolved in EtOAc 5 washed with IN NaOH and brine. The organics were dried over MgSO 4 and concentrated under reduced pressure yielding (4S)-6-(3,3-difluoro-2- methylpropyl)- 2,2-spirocyclobutyl -3,4-dihydro-2H-pyrano[2,3-b]pyridin-4-amine (0.720 g, 96.1% yield). The product was a mixture of diastereomers that was separted by chiral

A-1291-WO-PCT - 88-

HPLC and the resulting single isomers were used for preparing compounds of Formulas I and π.

Example 18

(lS^S,4'S)-3-Hydroxy-3',4'-dihydrospiro[cyclobutane-l^'-pyra no[23-c]pyridin-4'- amine

Step 1: 3-(Methoxymethoxy)pyridine

Pyridin-3-ol (25 g, 260 mmol) was added to a stirring mixture of NaH (11 g of a 60 wt% dispersion with mineral oil, 260 mmol) and DMF (350 mL) at 0 0 C. After 30 min, the reaction mixture was allowed to warm to RT, stirred for 90 min, and then chloromethoxymethane (20 mL, 260 mmol) was added. After 18 h, the reaction mixture was partitioned between ethyl acetate and saturated aqueous NaHCO 3 . The layers were separated, the organic material was washed with saturated aqueous NaHCθ 3 , brine, dried (Na 2 SO 4 ), filtered, and the filtrate was concentrated. The residue was dissolved with CH 2 Cl 2 , the solution was filtered through a plug of silica gel (sequential elution; 9:1 — * 1:1 hexane-ethyl acetate), and the second filtrate was concentrated to give 1O g (27%) of 3-(methoxymethoxy)pyridine as a clear yellow oil. Step 2: l-(3-(Methoxymethoxy)pyridin-4-yl)ethanol A solution of 3-(methoxymethoxy) pyridine (9.8 g, 70 mmol) and THF (40 mL) was added to a stirring mixture of tert-butyllithium (91 mL of a 1.7 M solution with pentane, 160 mmol) and THF (100 mL) at -78 0 C. After 1 h, acetaldehyde (9.9 mL, 180 mmol) was added, and the reaction mixture was stirred for 3 h and then warmed to RT. After 21 h, the reaction mixture was partitioned between ethyl acetate and saturated aqueous NaHCO 3 , the layers were separated, the organic material was washed with saturated aqueous NaHCO 3 , brine, dried (Na 2 SO 4 ), filtered, and the filtrate was concentrated. The residue was purified by flash chromatography on silica gel (1:1 hexane-ethyl acetate) to afford 4.6 g (36%) of l-(3-(methoxymethoxy)pyridin-4-yl)ethanol as a colorless solid. Step 3: l-(3-(methoxymethoxy ' )pyridin-4-vDethanone Dess-Martin periodinane (18 g, 43 mmol) was added to a stirring mixture of of l-(3- (methoxymethoxy) pyridin-4-yl)ethanol (4.6 g, 25 mmol), NaHCO 3 (6.3 g, 75 mmol), and CHCl 3 (75 mL) at RT. After 24 h, 1.0 M aqueous Na 2 S 2 O 3 was added, the reaction

A-1291-WO-PCT - 89- mixture was stirred for 90 min, partitioned between ethyl acetate and 1.0 M aqueous Na 2 S 2 Os, the layers were separated, the organic material was washed with 1.0 M aqueous Na 2 S 2 Oa, water, brine, dried (Na 2 SO 4 ), filtered, and the filtrate was concentrated. The residue was purified by flash chromatography on silica gel (gradient elution; 2:1 -→ 1:1 hexane-ethyl acetate) to give 3.9 g (86%) of l-(3-(methoxymethoxy)pyridin-4- yl)ethanone as a clear yellow-orange oil.

Step 4: (lS.SSVS-tert-Butyldimethylsiloxy-S'^'-dihvdrospirorcvclobut ane-l^'- pyranor2,3-c1pyridin-4'-one; and (lS,3R)-3-tert-Butyldimethylsiloxy-3'.4'- dihvdrospirorcvclobutane- 1 ,2'-pyranor2.3-clpyridin-4'-one 3-(tert-Butyldimethylsilyloxy) cyclobutanone (15 g, 77 mmol), l-(3-Hydroxypyridin-4- yl)ethanone (10.5000 g, 77 mmol) and pyrrolidine (19 ml, 230 mmol) were dissolved in 500 ml CH 3 CN and stirred at 65 0 C for 2h. TLC analysis revealed the disappearance of the SM and the formation of a single new spot. The mixture was evaporated (100 ml residue) and partitioned between water and EtOAc. The phases were separated and the aqueous was extracted 3x with EtOAc. The combined organic extracts were dried over MgSO 4 and evaporated and the mixture was purified via glass col. chromatography. The title compounds (8.500 g, 35% yield) were obtained as a yellow solid (1:1 mixture of stereo isomers) Step S^lSJS^'RVS-tert-Butyldimethylsiloxy-S'^'-dihvdrospirorcvclo butane-Lσ'- pyranor2.3-clpyridin-4'-oU (lS.3R.4'RV3-tert-Butyldimethylsiloxy-3'.4'- dihvdrospirofcvclobutane- 1 ,2'-pyranor2.3-c1pyridin-4'-ol

(lS,3S)-3-tert-Butyldimethylsiloxy-3',4'-dihydrospiro[cyclob utane-l,2'-pyrano[2,3- c]pyridin-4'-one and (1 S,3R)-3-tert-Butyldimethylsiloxy-3',4'-dihydrospiro[cyclobut ane- l,2'-pyrano[2,3-c]pyridin-4'-one (1:1 mixture of stereo isomers) (8.5000 g, 26.61 mmol) were dissolved in 150 ml toluene and 50 ml water was added. Ar gas was bubbled through the mixture for 15 min. Tetrabutylammonium bromide (0.2573 g, 0.7982 mmol), sodium formate (18.09 g, 266.1 mmol) and TPAP (0.5190 g, 0.7982 mmol) were added and the mixture was stirred for 14h under an Ar atmosphere. The mixture was partitioned between EtOAc and water and the phases were separated. The aqueous was extracted 3 times with EtOAc, dried over MgSO 4 and evaporated. Glass col. chrom (10-50% EtOAc in hex.) provided the title compounds (6.630 g, 77.51% yield) as yellow oil. (1:1 mixture of stereo isomers). MS m/z: 322.2 (M+l).

A- 1291 -WO-PCT - 90-

pyranor2.3-c1pyridin-4'-a2ide: (lS.3R.4'SV3-tert-Butyldimethylsiloxy-3'.4'- dihvdrospirorcyclobutane-1.2'-pyranor2.3-clpyridin-4'-azide

(lS,3S,4'R)-3-tert-Butyldimethylsiloxy-3',4'-dihydrospiro[cy clobutane-l,2'-pyrano[2,3- c]pyridin-4'-ol, (lS,3R,4'R)-3-tert-Butyldimethylsiloxy-3',4'-dihydrospiro[cy clobutane- l,2'-pyrano[2,3-c]pyridin-4'-ol (1:1 mixture of stereo isomers) (6.6300 g, 20.62 mmol), diphenyl azidophosphate (6.667 ml, 30.93 mmol) and DBU (4.626 ml, 30.93 mmol) were dissolved in 40 ml CH 2 Cl 2 and stirred over the weekend. Monitoring revealed that the starting materials were almost consumed and product formed, but still a large portion of the phosphonate ester remained. 40 ml of Water was added and the mixture was extracted 3 times with Et 2 O 5 dried over MgSO 4 and evaporated. The crude product was used w/o purification in the next step. MS m/z: 347.2 (M+l).

Step 7: (1 S.3S.4'S)-3-tert-Butyldimethylsiloxy-3'.4'-dihydrospirorcvcl obutane-1.2'- pyranor2,3-c1pyridin-4'-amine; (lS.3R,4'SV3-tert-Butyldimethylsiloxy-3'.4'- dihvdrospiro[cvclobutane-1.2'-pyranor2,3-c1pyridin-4'-amine

The crude (lS,3S,4'S)-3-tert-Butyldimethylsiloxy-3',4'-dihydrospiro[cy clobutane-l,2'- pyrano[2,3-c]pyridin-4'-azide and ( 1 S,3R,4'S)-3-tert-Butyldimethylsiloxy-3',4'- dihydrospirotcyclobutane-l^'-pyranop^-cjpyridin^'-azide (1:1 mixture of stereo isomers) from the previous reaction (6.9 g, 20 mmol) was dissolved in 200 ml THF and lithium aluminum hydride, 2M in THF (30 ml, 60 mmol) was added at 0 0 C. The mixture was stirred for 60 min and hydrolyzed with Na 2 SO 4 -IOH 2 O until gas evolution had ceased. The mixture was filtered and evaporated and purified. (2-10% MeOH in CH 2 Cl 2 ) glass col. chromatography provided the title compounds) as a yellow oil as a mixture of diastereomers. The diastereomers were separated by SFC. (lS,3S,4'S)-3-tert- Butyldimethylsiloxy-3',4'-dihydrospiro[cyclobutane-l,2'-pyra no[2,3-c]pyridin-4'-amine (1.200 g, 19% yield) and (lS,3R,4'S)-3-tert-Butyldimethylsiloxy-3',4'- dihydrospiro[cyclobutane-l,2'-pyrano[2,3-c]pyridin-4'-amine were obtained. MS m/z: 321.2 (M+l).

Example 19

A- 1291 -WO-PCT - 91-

(lS^S^'SVo'-CZ^-Dimethylpropyl^-fluoro-S'^'-dihydrospiroI cyclobutane-l^'- pyrano[23-b]pyridin-4'-amine

Step 1: αS.,3R.4'SVtert-butyl 6'-(2.2-DimethylpropylV3-hvdroxy-3'.4'- dihvdrospirorcvclobutane-1.2'-pyranor2.3-b]pyridin-4'-carbam ate

(lS,3R,4'S)-6'-(2,2-Dimethylpropyl)-3-hydroxy-3',4'-dihydros piro[cyclobutane-l,2'- pyrano[2,3-b]pyridin-4'-amine; (1 S,3 S,4 l S)-6'-(2,2-Dimethylpropyl)-3-hydroxy-3',4 I - dihydrospiro[cyclobutane-l,2'-pyrano[2,3-b]pyridin-4'-amine (mixture of stereoisomers) (7.5000 g, 21.472 mmol) was dissolved in 400 ml THFTH 2 O (1:1) and carbonic acid monosodium salt (9.0189 g, 107.36 mmol) and di-tert-butylpyrocarbonate (9.3723 g, 42.944 mmol) were added. The mixture was stirred over night and 300 ml H 2 O was added and the product was extracted out with EtOAc (3x 800 ml). The combined organic phases were dried over MgSO 4 and evaporated and purified by chiral purification (SFC). MS m/z: 377.3 (M+l). Alternatively. πS.3S.4'SVtert-butyl 6'-(2.2-DimethylpropylV3-fluoro-3'.4'- dihvdrospiro[cvclobutane-1.2'-pyranor2.3-blpyridin-4'-carbam ate can be made by: (lS,3R,4 l S)-6'-(2,2-Dimethylpropyl)-3-hydroxy-3',4'-dihydrospiro[cycl obutane-l,2 t - pyrano[2,3-b]pyridin-4'-(BOC)-amine (1.050 g, 2.789 mmol) was dissolved in 13 ml CH 2 Cl 2 and DAST (0.4790 ml, 3.626 mmol) was added. The reaction was heated to 45 0 C for 4h and cooled to RT. The mixture was poured into 15 ml of sat. NaHCO 3 and extracted 3x with EtOAc (3x 150 ml). The combined organic extracts were dried over MgSO4 and evaporated. The crude product was used without purification in the next step. MS m/z: 379.2 (M+l). Step 2: (lS.3S.4'S)-6'-f2.2-Dimethylpropyn-3-fluoro-3'.4'-dihvdrospi rolcvclobutane-1.2'- pyranor2,3-b " |pyridin-4'-amine

The crude product from step 1 was dissolved in 5 ml MeOH and treated with 20 ml 4M HCl in dioxane. The mixture was stirred for 4h at RT. and evaporated. The messy mixture was purified on HPLC. The combined HPLC fractions were basified (10%

A- 1291 -WO-PCT - 92-

Na2CO3, aq.) and extracted with EtOAc (3x 250 ml). The title compound (0.180 g) was obtained as a yellow solid, MS m/z: 279.2 (M+l).

(4'S)-6'-(2-Fluoro-2-methylpropyl)-3',4'-dihydrospiro[cyc lobutane-l^'-pyrano[2β- c]pyridin-4'amine

Step 1: l-(5-(methoxymethoxy)pyridin-2-yl ' )-2-methylpropan-2-ol 5-(Methoxymethoxy)-2-methylpyridine (22.1500 g, 144.6 mmol) was dissolved in 1500 ml THF and cooled to -78 0 C. tert-Butyllithium (97.82 ml, 166.3 mmol) was added and the mixture was stirred for 15 min. Acetone, (42.52 ml, 578.4 mmol) was added and stirring of the mixture was continued for 15 min. The reaction was hydrolyzed with 300 ml H 2 O and extracted with 4L EtOAc (2x). The combined organic extracts were dried over MgSO 4 and evaporated. Glas col. chrom (20-100% EtOAc provided the 2 products: l-(5-(methoxymethoxy) pyridin-2-yl)-2-methylpropan-2-ol (7.5000 g, 24.55% yield) and 2-(5-(methoxymethoxy)-2-methylpyridin-4-yl)propan-2-ol (15.00 g, 49.10% yield). MS m/z: 212.0 (M+l).

Step 2: 2-(2-fluoro-2-methylpropyl)-5-(methoxymethoxy)pyridine l-(5-(methoxymethoxy)pyridin-2-yl)-2-methylpropan-2-ol (7.600 g, 36.0 mmol) was dissolved in 200 ml CH 2 Cl 2 and cooled to -78 0 C. DAST (9.51 ml, 72.0 mmol) was added drop wise to the solution and stirring was continued for 30 min The mixture was allowed to warm up to 0 0 C and was hydrolyzed with NaHCO 3 (200 ml). Stirring was continued in the cold until gas evolution had ceased and the phases were separated. The aqueous was extracted 2x with EtOAc and the combined organic layers were dried over MgSO 4 and evaporated. Glass col. Chromatography of the crude material provided 2-(2-fluoro-2-methylpropyl)-5-(methoxymethoxy)pyridine (5.80 g, 75.6% yield) as a pale yellow oil.

Step 3 : 1 -(2-(2-fluoro-2-methylpropylV5-(methoxymethoxy')pyridin-4-v ethanol. 2,2,6,6-Tetramethylpiperidine (8.26 ml, 49.0 mmol) was dissolved with 270 ml THF and 1-butyllithium (14.1 ml, 35.4 mmol) was added at -78 0 C. The mixture was stirred for 10

A-1291-WO-PCT - 93- min in an ice bath and cooled back to r78 0 C. A solution of 2-(2-fluoro-2-methylpropyl)-, 5-(methoxymethoxy)pyridine (5.8000 g, 27.2 mmol) in 20 ml THF was added dropwϊse and the reaction was stirred for 20 min. Acetylaldehyde (7.65 ml, 136 mmol) was added to the dark red solution and the color disappeared. Stirring was continued for 20 min and the mixture was hydrolyzed with 50 ml of water. The mixture was warmed to RT and extracted 3x with CH 2 Cl 2 (300 ml each). The combined organic extracts were dried over MgS0 4 and evaporated and purified via glass col. chrom. (30-80% EtOAc in hex.) to provide l-(2-(2-fluoro-2-methylpropyl)-5-(methoxymethoxy)pyridin-4-y l)ethanol a white solid. MS m/z: 258.2 (M+l).

(5)-6-(2^-Difluoropropyl)-2^-(2'^'-difluorocyclobutyl)-3, 4-dihydro-2H- pyrano[2β-b]pyridin-4-amine Step 1: l-(6-Fluoropyridin-3-yl)propan-2-one

Anhydrous, de-gassed THF (20 mL) was added to 2-(dicyclohexylphosphiήo)-2'- methylbiphenyl (1.2 g, 3.2 mmol) and tris(dibenzylideneacetone)dipalladium (0) chloroform adduct (1.4 g, 1.4 mmol), and the resulting solution was warmed to 45 0 C and sparged with N 2 . After 20 min, the reaction mixture was allowed to cool to RT, then added to a stirring, degassed mixture of 5-bromo-2-fluoropyridine (4.8 g, 27 mmol), finely ground potassium phosphate tribasic (14 g, 68 mmol), and acetone (100 mL, 1400 mmol) at RT, and the resulting mixture was sparged with N 2 . After 20 min, the reaction mixture was heated at reflux for 24 h, the reaction mixture was allowed to cool to room temperature, filtered through a 0.45 μm Teflon filter, and the filtrate was concentrated. The residue was purified by flash chromatography on silica gel (gradient elution; 4:1 → 2:1 hexane-ethyl acetate) to give 2.3 g (55%) of l-(6-fluoropyridin-3-yl)propan-2-one as a brown oil. Step 2: 5-(2,2-Difluoropropyl)-2-fluoropyridine

A- 1291 -WO-PCT - 94-

Diethylaminosulfurtrifluoride (9.8 mL, 75 mmol) was added to a stirring solution of l-(6- fluoropyridin-3-yl)ρropan-2-one (2.3 g, 15 mmol), ethanol (0.18 mL, 3.0 mmol), and CH 2 Cl 2 (60 mL) at RT. After 24 h, the reaction mixture was added to a rapidly stirring solution of aqueous 10% Na 2 CO 3 . After 1 h, ethyl acetate was added, the layers were separated, the organic layer was washed sequentially with aqueous 10% Na 2 CO 3 and brine, dried (Na 2 SO 4 ), filtered, and the filtrate was concentrated. The residue was purified by flash chromatography on silica gel (9: 1 hexane-ethyl acetate) to afford 1.5 g (57%) of 5-(2,2-difluoropropyl)-2-fluoropyridine as a yellow-orange oil. SteD 3: (4S)-2-α.3-Bis(tert-butyldimethylsiloxy > )cvclobutyl ' )-l-f5-(2.2-difluoropropylV2- fluoropyridin-3-yl)-ethyl-((/?)-tert-butylsulfϊnyl)amine

Butyllithium (4.1 mL of a 2.5 M solution with toluene, 10 mmol) was added to a stirring solution of 2,2,6,6-tetramethylpiperidine (2.0 mL, 12 mmol) and THF (43 mL) at -78 0 C. After 5 min, the reaction mixture was raised above the cooling bath for 10 min, re-cooled to -78 0 C, and then a solution of 5-(2,2-difluoropropyl)-2-fluoropyridine (1.5 g, 8.6 mmol) and THF (8.6 mL) was added. After 30 min, a solution of 2-(l,3-bis(tert- butyldimethylsiloxy)cyclobutyl)acetaldehyde (R)-tert-butylsulfϊnylimine (4.4 g, 9.4 mmol) and THF (9.4 mL) was added. After 20 min, saturated aqueous NaHCO 3 was added, the reaction mixture was allowed to warm to RT, partitioned between saturated aqueous NaHCO 3 and ethyl acetate, the layers were separated, the organic layer was washed with saturated aqueous NaHCO 3 , brine, dried (NaSO 4 ), filtered, and the filtrate was concentrated. The residue was purified by flash chromatography on silica gel (gradient elution; 4:1 → 3:1 → 2:1 hexane-ethyl acetate) to afford 2.9 g (53%) of (45)-2- (l,3-bis(tert-butyldimethylsiloxy)cyclobutyl)-l-(5-(2,2-difl uoropropyl)-2-fluoropyridin-3- yl)-ethyl-((i?)-tert-butylsulfinyl)amine as a yellow solid. SteD 4: (S)-6-r2.2-difluoropropyl ' )-2.2-fCR)-2'-hvdroxy > )cvclobutyl-3.4-dihvdro-2H- pyranor2.3-blpyridin-4-amine and (y)-6-(2,2-difluoropropylV2.2-((.S r )-2'- hvdroxy)cvclobutyl-3.4-dihvdro-2H-pyrano[2.3-blpyridin-4-ami ne Hydrogen fluoride (24 mL of a 70 wt% solution with pyridine, 1300 mmol) was added to (S)-2-(l,3-bis(tert-butyldimethylsiloxy)cyclobutyl)-l-(5-(2, 2-difluoropropyl)-2- fluoropyridin-3-yl)-ethyl-((/?)-tert-butylsulfinyl) amine (1.7 g, 2.7 mmol) in Teflon™ reaction vessel, and the reaction mixture was heated at 80 0 C. After 48 h, the reaction mixture was added to aqueous 10% Na 2 CO 3 , the mixture was stirred vigorously for 2 h, ethyl acetate was added, the layers were separated, the organic material was washed with aqueous 10% Na 2 CO 3 , brine, dried (Na 2 SO 4 ), filtered, and the filtrate was concentrated to

A-1291-WO-PCT - 95- afford 0.64 g (84%) of a mixture of (S)-6-(2,2-difluoropropyl)-2,2-((φ-2'- hydroxy)cyclobutyl-3,4-dihydro-2H-pyrano[2,3-b]pyridin-4-ami ne and (iS)-6-(2,2- difluoropropyl)-2,2-((S)-2'-hydroxy)cyclobutyl-3,4-dihydro-2 H-pyrano[2,3-b]pyridin-4- amine as a yellow solid. Step 5: (SVtert-butyl 6-(2,2-difluoropropyl')-2.2-((-R)-2^-hvdroxy ' )cvclobutyl -3.4-dihvdro- 2H-pyranor2.3-blpyridin-4-ylcarbamate and (SVtert-butyl 6-(2.2-difluoropropylV2.2-((5 r )- 2'-11VdFOXV-)CVcIObUtVl -3,4-dihydro-2H-pyranor2.3-blpyridin-4-ylcarbamate Di-tert-butyl dicarbonate (0.64 g, 2.9 mmol) was added to a stirring solution of the (5)-6- (2,2-difluoropropyl)-2,2-((R)-2'-hydroxy)cyclobutyl-3,4-dihy dro-2H-pyrano[2,3- b]pyridin-4-amine and (5)-6-(2,2-difluoropropyl)-2,2-((S)-2'-hydroxy)cyclobutyl-3, 4- dihydro-2H-pyrano[2,3-b]pyridin-4-amine mixture (0.64 g, 2.3 mmol), CH 2 Cl 2 (23 mL), and diisopropyethylamine (2.0 mL, 11 mmol) at RT. After 24 h, aqueous 10% Na 2 CC> 3 was added, the mixture was stirred vigorously for 1 h, EtOAc was added, the layers were separated, the organic layer was washed with aqueous 10% Na 2 CO 3 , brine, dried (Na 2 SO 4 ), filtered, and the filtrate was concentrated. The residue was purified by flash chromatography on silica gel (19:1 CH 2 Cl 2 -methanol) to afford 0.32 g (37%) of a mixture of (5)-tert-butyl 6-(2,2-difluoropropyl)-2,2-((R)-2'-hydroxy)cyclobutyl -3,4-dihydro-2H- pyrano[2,3-b]pyridin-4-ylcarbamate and (SJ-tert-butyl 6-(2,2-difluoropropyl)-2,2-((5)-2'- hydroxy)cyclobutyl -3,4-dihydro-2H-pyrano[2,3-b]pyridin-4-ylcarbamate as a yellow- brown solid.

Step 6: (SHert-butyl 6-(2,2-difluoropropyl)-2,2-cyclobutan-2'-one-3.4-dihvdro-2H- pyranor2,3-b1pyridin-4-ylcarbamate

Dess-Martin periodinane (0.49 g, 1.2 mmol) was added to a mixture of (5)-tert-butyl 6-

(2,2-difluoropropyl)-2,2-((i?)-2'-hydroxy)cyclobutyl -3,4-dihydro-2H-pyrano[2,3- b]pyridin-4-ylcarbamate and (5)-tert-butyl 6-(2,2-difluoropropyl)-2,2-((5)-2'- hydroxycyclobutyl) -3,4-dihydro-2H-pyrano[2,3-b]pyridin-4-ylcarbamate (0.32 g, 0.83 mmol), CH 2 Cl 2 (8.3 mL), and NaHCO 3 (0.21 g, 2.5 mmol) at RT. After 2 h, the reaction mixture was purified by flash chromatography on silica gel (1 : 1 hexane-ethyl acetate) to afford 0.24 g (75%) of (5)-tert-butyl 6-(2,2-difluoropropyl)-2,2-cyclobutan-2'-one-3,4- dihydro-2H-pyrano[2,3-b]pyridin-4-ylcarbamate as a colorless solid.

Step 7: (.SVtert-butyl 6-(2.2-difluoropropylV2.2-(2'.2'-difluorocyclobutyl) -3.4-dihvdro-

2H-pyrano[2.3-blpyridin-4-ylcarbamate

Diethylaminosulfur trifluoride (0.41 mL, 3.1 mmol) was added to a stirring solution of

(5)-tert-butyl 6-(2,2-difluoropropyl)-2,2-cyclobutan-2'-one-3,4-dihydro-2H- pyrano[2,3-

A-1291-WO-PCT - 96- b]pyτidin-4-ylcarbamate (0.24 g, 0.63 mmol), CH 2 Cl 2 (6.3 mL), and ethanol (7.3 DL, 0.13 mmol) at RT. After 24 h, the reaction mixture was added to a stirring solution of aqueous 10% Na 2 3 , the mixture was stirred for 1 h, partitioned between EtOAc and aqueous 10% Na 2 COa, the layers were separated, the organic layer was washed with aqueous 10% Na 2 3 , brine, dried (Na 2 SO 4 ), filtered, and the filtrate was concentrated. The residue was purified by flash chromatography on silica gel (2:1 hexane-ethyl acetate) to give 0.15 g (59%) of of (5)-tert-butyl 6-(2,2-difluoropropyl)-2,2-(2',2'- difluorocyclobutyl) -3,4-dihydro-2H-pyrano[2,3-b]pyridin-4-ylcarbamate as a colorless solid. Step 8: (4S)-6-(2,2-difluoropropylV2.2-(2'.2'-difluorocvclobutylV3.4 -dihvdro-2H- pyranor2,3-blpyridin-4-amine

Hydrogen chloride (0.93 mL of a 4.0 M solution with 1,4-dioxane, 3.7 mmol) was added to a stirring solution of (5)-tert-bury 1 6-(2,2-difluoropropyl)-2,2-(2',2'-difluorocyclobutyl) -3,4-dihydro-2H-pyrano[2,3-b]pyridin-4-ylcarbamate (0.15 g, 0.37 mmol) and CH 2 Cl 2 (3.7 mL) at RT. After 24 h, the reaction mixture was concentrated, the residue was partitioned between aqueous 10% Na 2 CO 3 and ethyl acetate, the layers were separated, the organic material was washed with aqueous 10% Na 2 CO 3 , brine, dried (Na 2 SO 4 ), filtered, and the filtrate was concentrated to afford (5)-6-(2,2-difluoropropyl)-2,2-(2',2'- difluorocyclobutyl)-3,4-dihydro-2H-pyrano[2,3-b]pyridin-4-am ine as a yellow solid.

(4S)-2^-SpirocycIobutyl-6-(l y 33^-tetrafluoro-2-methylpropyl)-3,4-dihydro-2H- pyrano(2,3-b)pyridiiie-4-amine Step 1: tert-Butyl allyl((SV2.2-spirocvclobutyl-6-(3.3.3-trifluoro-l-hvdroxy-2- methylpropyl)-3,4-dihvdro-2H-pyrano(2,3-b)pyridine-4-yl)carb amate To a cooled (-78 0 C) solution of (S)-tert-butyl allyl(6-bromo-2,2-spirocyclobutyl-3,4- dihydro-2H-pyrano(2,3-b)pyridine-4-yl)carbamate (8.70 g, 21 mmol) in diethylether was added tert-butyllithium (25 ml, 43 mmol) dropwise. After stirred for 15 min, the fresh distilled 3,3,3-trifluoro-2-methylpropanal (5.8 ml, 53 mmol) was added, and the reaction

A- 1291 -WO-PCT - 97- was stirred for 30 min, and then quenched with saturated NH4C1. The resulted mixture was allowed to warm to RT and extracted with EtOAc (3x). The organic layers were combined, dried over Na2SO4, filtered and concentrated. The residue was purified on silica gel column to afford the title compound as a mixture of isomers (4.5 g, 46% yield) as light yellow oil. MS m/z: 457 (M+l).

Step 2: tert-Butyl allyl((S ' )-2,2-spirocvclobutyl-6-(1.3.3,3-tetrafluoro-2-methylpropylV

3.4-dihvdro-2H-pyrano(2.3-b)pyridine-4-yl ' )carbamate

To a cooled (-78 0 C) solution of tert-butyl allyl((S)-2,2-spirocyclobutyl-6-(3,3,3-trifluoro-

1 -hydroxy-2-methylpropyl)-3 ,4-dihydro-2H-pyrano(2,3-b)pyridine-4-yl)cafbamate ( 1.24 g, 2.7 mmol) in toluene was added (diethylamino)sulfur trifluoride (0.54 ml, 4.1 mmol) via a syringe. The reaction was stirred for 50 min, then quenched with saturated NH 4 Cl (10 ml) and warmed to RT. The layers were separated. The aqueous layer was extracted with EtOAc (2 x 20 ml). The organic layers were combined, dried over Na 2 SO 4 , filtered and concentrated. The crude residue was purified on a silica gel column (10-15% EtOAc/hexane) to afford the title compound as a mixture of isomers as colorless oil. MS m/z: 459 (M+l).

Step 3: (4S)-N-Allyl-2.2-spirocvclobutyl-6-(1.3.3.3-tetrafluoro-2-me thylpropyl ' )-3.4- dihvdro-2H-pyrano(2.3-b)pyridine-4-amine To a solution of tert-butyl allyl((S)-2,2-spirocyclobutyl-6-(l,3,3,3-tetrafluoro-2- methylpropyl)-3,4-dihydro-2H-pyrano(2,3-b)pyridine-4-yl)carb amate (430 mg, 938 μmol) in MeOH was added hydrogen chloride 4.0m in 1,4-dioxane (2.0 ml, 8000 μmol). The reaction was stirred for 2 days (over the weekend) at RT, then concentrated and neutralized with 10% Na 2 CO 3 and extracted with DCM (3x). The organic layers were combined, dried over Na 2 SO 4 and filtered. The filtrate was concentrated and dried in vacuum to afford the title compound as a mixture of isomers as a light yellow oil. MS+ m/z: 359 (M+l).

Step 4: (4SV2.2-Spirocvclobutyl-6-(l J,3.3-tetrafluoro-2-methylpropyl)-3.4-dihvdro-2H- pyrano(2.3-b')pyridine-4-amine The crude product from step 3 above was dissolved in CH 2 Cl 2 (10 ml) to which 1,3- dimethylpyrimidine-2,4,6(lH,3H,5H)-trione (439 mg, 2814 μmol) was added. The mixture was purged with N 2 gas for 10 min and tetrakis(triphenylphosphine)palladium (0) (54 mg, 47 μmol) was added. The reaction was heated at 40 0 C for 3 h, then cooled and diluted with DCM and washed with 10% Na 2 CO 3 (2x). The aqueous layer was back extracted with EtOAc (2x). The organic layers were combined, dried over Na 2 SO 4 ,

A-1291-WO-PCT - 98- fϊltered and concentrated. The residue was dried in vacuum to afford the title compound as mixture of isomers as a yellow oil. MS mJz: 319 (M+l).

Example 23

(S)-6-tert-Butoxy-2^-spirocyclobutyl-3,4-dihydro-2H-pyrano[2 r 3-b]pyridin-4-amine

Step 1 : (SV4-Azido-2.2-spirocvclobutyl-3.4-dihvdro-2H-pyranor2.3-b1p yridin-6-ol A 25-mL RBF was charged with (S)-4-azido-6-bromo-2,2-spirocyclobutyl-3,4-dihydro- 2H-pyrano[2,3-b]pyridine (0.100 g, 0.34 mmol), and the solid was dissolved in THF (3.0 mL). The solution was cooled to -78 0 C, and a solution of butyllithium (2.50 M, 0.20 ml, 0.51 mmol) in hexane was added, followed immediately by triisopropyl borate (0.079 ml, 0.34 mmol. After 45 minutes, the reaction solution was warmed to 0 0 C. After 30 minutes, a mixture of aqueous 30% hydrogen peroxide (0.35 ml, 3.4 mmol), and sodium hydroxide (2.50 M, 0.81 ml, 2.0 mmol) was added, and the mixture was warmed to ambient temperature. After 30 minutes the mixture was concentrated, the crude residue was taken up in half-saturated NH 4 Cl (10 mL) and extracted with DCM (3 x 20 mL). The organic layers were combined, dried over sodium sulfate and concentrated. The crude material was purified through silica gel (25 mL) using 50% EtOAc-hexane to afford the title compound. MS m/z 233 (M+l). Step 2: (S)-4-Azido-6-tert-butoxy-2,2-spirocvclobutyl-3,4-dihydro-2H -pyranol2.3- blpyridine

In a microwave vessel, the (S)-4-azido-2,2-spirocyclobutyl-3,4-dihydro-2H-pyrano[2,3- b]pyridin-6-ol (0.024 g, 0.10 mmol) was suspended in DCM (2 mL), and trifluoromethanesulfonic acid (0.011 ml, 0.12 mmol) was added to the mixture, The reaction mixture was cooled to -78 0 C. 2-Methylprop-l-ene (0.97 ml, 10 mmol) was separately liquefied at -78 0 C and added to the reaction mixture via a pipet. The reaction vessel was sealed and allowed to warm to RT while stirring overnight. The reaction was diluted with DCM (60 mL), and the organic phase was extracted with dilute sodium carbonate (2 x 6 mL), then with dilute brine (6 mL). The organic phase was dried over sodium sulfate, filtered and concentrated, and the crude residue was purified through

A-1291-WO-PCT - 99- silica gel (20 mL) using 38% EtOAc-hexane to afford the title compound (15 mg, 0.052 mmol, 50%) as a white solid. MS m/z 289 (M+l).

Step 3: (S)-6-tert-Butoxy-2.2-spirocvclobutyl-3,4-dihvdro-2H-pyranor 23-blpyridin-4- amine In a 25-mL RBF, the (S)-4-azido-6-tert-butoxy-2,2-spirocyclobutyl-3,4-dihydro-2H - pyrano[2,3-b]pyridine (0.036 g, 0.12 mmol) and Pd/C (10%, 0.0036 g) were taken up in EtOAc (2.5 mL), and the mixture was stirred at ambient temperature under an atmosphere of hydrogen. After 16 h, the mixture was filtered through Celite, and the filtrate was concentrated. Purification of the concentrate through silica gel (20 mL), which had been deactivated with triethylamine (2 mL) using 0.5% MeOH-DCM, afforded the title compound. MS m/z 263 (M+l).

6-(2'^'-Dimethylpropyl)-2^-spirocycIobutyl-3-fluro-3,4-dihyd ro-2H-pyrano[2^- b] py ridin-4-amine

Step 1: (±) 6-Bromo-2,2-spirocvclobutyl-3-fluro-3.4-dihvdro-2H-pyranor2. 3-b1pyridin-4- one

A mixture of 6-bromo-2,2-spirocycloburyl-3,4-dihydro-2H-pyrano[2,3-b]pyri din-4-one (4.9 g, 18 mmol) and SLECTFLUOR (7.1 g, 20 mmol) in 40 ml of anhydrous MeOH was heated at 110-130 0 C in a presure bottle for 16 h. The mixture was cooled down and the solids were filtered off. The filtrate was concentrated to give an oil which was purified by silica gel chromatography using EtOAc-Hexanes (0-12%) to give the tile compound as a clear oil which solidified upon drying. Step 2: (±) 6-Bromo-2.2-spirocvclobutyl-3-fluro-3.4-dihvdro-2H-pyranor2, 3-b1pyridin-4- yl-(S)-tert-butylsulfinylimine

A solution of (±) 6-Bromo-2,2-spirocyclobutyl-3-fluro-3,4-dihydro-2H-pyrano[2, 3- b]pyridin-4-one (3.0 g, 10 mmol) and (S)-2-methylpropane-2-sulfinamide (2.5 g, 21 mmol) in 5 ml of THF was treated with tetraethoxytitanium (8.7 ml, 42 mmol) at rt for 18h. The reaction mixture was diluted with 100 ml of EtOAc and the resulting solution

A- 1291 -WO-PCT - 100-

- was added dropwise to 150 ml of sat. aq. NaHCθ 3 . White precϊpiates formed, and the mixture was stirred at rt vigorously for 1 h. The EtOAc layer was carefully decanted; the rest of mixture was filtered through a celite pad with Na 2 SO 4 . The celite pad was washed with 150 ml of EtOAc. The filtrates were combined and the EtOAc layer was separated. All organic layers were combined, dried (Na 2 SO 4 ) and concentrated to give an oil that was purified by Isco (0-30% EtOAc in hexanes) to give the title compound as a yellow foam.

Step 3: (4R " )-6-Bromo-2,2-spirocvclobutyl-3-fluro-3.4-dihvdro-2H-pyranor 2.3-b1pyridin- 4-amine A solution of (±) 6-Bromo-2,2-spirocyclobutyl-3-fluro-3,4-dihydro-2H-pyrano[2, 3- b]pyridin-4-yl-(S)-tert-butylsulfinylimine from Step 2 (2.55 g, 6.6 mmol) in 20 ml of THF:H 2 O (98:2) at -50 0 C was treated with sodium borohydride (0.74 g, 20 mmol) and the resulting mixture was stirred and warmed up to rt over 2 h, and then stirred overnight. The solvents were then removed, The crude esidue was triturated with DCM, washed with sat. aq. NaHCO 3 (2x75 ml), dried over Na 2 SO 4 and concentrated to give the title compound as an oil.

Step 4: (4R)-tert Butyl 6-Bromo-2,2-spirocvclobutyl-3-fluro-3,4-dihydro-2H-pyranor2, 3- b1pyridin-4-carbamate A solution of (4R)-6-Bromo-2,2-spirocyclobutyl-3-fluro-3,4-dihydro-2H-pyra no[2,3- b]pyridin-4-amine (1.23 g, 4.3 mmol) in 15 ml of dry DCM was treated with Boc anhydride (4.3 ml, 4.3 mmol) at rt overnight. The reaction solvent was removed and the resulting crude residue was purified by ISCO (0-20% EtOAc on hexanes) to give the titled compound. Step 5: (4R)-6-(2'2'-dimethylpropyl)-2.2-spirocvclobutyl-3-fluro-3.4 -dihydro-2H- pyrano [2,3 -blpyridin-4-amine

To a 50 mL RBF was added (4R)-tert butyl 6-bromo-2,2-spirocyclobutyl-3-fluro-3,4- dihydro-2H-pyrano[2,3-b]pyridin-4-carbamate (580 mg, 1498 μmol), dioxane (10 mL). The solution was degassed with N 2 for 10 minutes, and Pd catalyst (53 mg, 75 μmol) was added to the solution. A solution of neopentylzinc(H) iodide, in THF (8.0 ml, 4000 μmol) was then added and the reaction mixture was stirred at RT under N 2 for 16 hours. The reaction mixture was quenched with water (20 mL) and acidified to pH 2 with IN HCl. The mixture was extracted with EtOAc (2 X 40 mL). The combined organic layers were washed with brine and concentrated in vacuo to give a dark brown oil, which was then treated with MeOH (30 mL) and HCl (4M in dioxane, 10 mL) and stirred overnight. The

A-1291-WO-PCT - 101- material was concentrated in vacuo and taken up in DCM (5% MeOH was added to improve solubility), and extracted with IN HCl (2 X 20 mL). The combined acidic aqueous layers were washed with DCM (20 mL), neutralized with sat'd NaHCO 3 and extracted with DCM (3 X 20 mL). The combined organic layers were concentrated in vacuo to give the title compound.

(4S)-6-(2'^'-Dimethyl-3' r 3' r 3'-trifluoro-propyl)-2^-spirocycIobutyl-3,4-dihydro-2H- pyrano [2,3-b] py ridin-4-amine

Step 1: l.Ll-Trifluoro-2-(4-methoxyphenyl)propan-2-ol

A solution of l-bromo-4-methoxybenzene (19 ml, 150 mmol) in 300 ml of dry THF at - 78 0 C was treated with n-butyllithium (96 ml, 154 mmol) (1.6 M or 15% in hexanes, Strem). The resulting mixture was stirred for an additional 30 min. At -78 °C, 1,1,1- trifluoropropan-2-one (16 ml, 180 mmol) was added and the mixture was stirred -78 °C and slowly warmed up overnight. The reaction mixture was treated with 100 ml of 5N HCl and concentrated. The layers were separated. The bottom layer (organic) was diluted with 50 ml of DCM, washed with 1x100 ml OfH 2 O, and dried (MgSO 4 ) and concentrated to give an oil which was purified by silica gel chromatography eluting with 0-5% EtOAc in hexanes to provide the title compound.

Step 2: l-(2-chloro-lJ.l-trifluoropropan-2-yl)-4-methoxybenzene A solution of l,l,l-trifluoro-2-(4-methoxyphenyl)propan-2-ol (22.5 g, 102 mmol) in 150 ml of anhydrous toluene was treated sequentially with pyridine (8.26 ml, 102 mmol) and dropwise sulfuryl dichloride (11.2 ml, 153 mmol) at 0 0 C. The resulting mixture was then heated at 55 0 C for 18 h. The reaction was diluted with 100 ml of EtOAc, washed with 100 ml OfH 2 O and 100 ml of IN HCl, dried over Na 2 SO 4 , and concentrated to give an oil which was filtered through a silca gel pad eluting with 0-5% EtOAc in hexanes to afford the title compound. Step 3: l-Methoxv-4-π.l.l-trifluoro-2-methylpropan-2-vDbenzene

A- 1291 -WO-PCT - 102-

A solution of l-(2-chloro-l,l,l-trifluoropropan-2-yl)-4-methoxybenzene (18.0 g, 75 mmol) in 400 ml of hexanes was treated with trimethylaluminum (151 ml, 302 mmol) (2.0 M in heptane) at rt and the resulting mixture was heated at 95 °C overnight. The reaction mixture was cooled in an ice-water bath, conc.HCl was added to the reaction mixture dropwise. Fumes were generated. After about 10 ml of cone. HCl was added, more rapid addition of acid was possible due to the near complete quenching of the unreacted AlMe3. After addition of 100 ml of cone. HCl, 250 ml OfH 2 O was added and the mixture was stirred vigorously for 1 h. The layers were separated. The org layer was dried (Na 2 SO 4 ) and concentrated to give the title compound as an oil, which solidified upon drying on the vac line.

Step 4: 3,3J-Trifluoro-2.2-dimethylpropanoic acid

To a biphasic mixture of l-methoxy-4-(l,l,l-trifluoro-2-methylpropan-2-yl)benzene (10.0 g, 45.8 mmol) and sodium periodate (137 g, 642 mmol) in a mixture solvent of CCUrCH 3 CNtH 2 O (2:2:3, 550 ml) was added slowly ruthenium(iπ) chloride hydrate (0.517 g, 2.29 mmol) (black powder) while the reaction flask was kept in an ice-water bath. The mixture turned yellow rapidly then red after 5 minutes, ans was stirred for 0.5 h. The ice-water bath was remioved and the reaction mixture was stirred vigorously, via a mechanical stirrer, overnight. The solids were filtered off, washed with 100 ml of DCM, 100 ml OfH 2 O. The filtrate (pH ~1) was basified with 10 N NaOH to pH > 11, and the layers were separated, and the aq. layer was extracted with 130 ml of DCM. The aq. layer was then acidified carefully with cone. HCl to pH ~ 1, extracted with 3x150 ml of DCM. The org layers were combined, dried (Na 2 SO 4 ), and concentrated to give the title compound as an oil, which became a semi-solid upon drying on the vac line. Step 5: Naphthalen-2-ylmethyl 3.3.3-trifluoro-2,2-dimethylpropanoate A mixture of 3,3,3-trifluoro-2,2-dimethylpropanoic acid (2.96 g, 19.0 mmol), 2-

(bromomethyl)naphthalene (4.19 g, 19.0 mmol), and potassium carbonate (7.86 g, 56.9 mmol) in 50 ml of anhydrous DMF was stirred at 30 0 C overnight. Diluted with 150 ml ofEtOAc, washed with 150 ml OfH 2 O, 2x100 ml of IN NaOH, 2x100 ml of IN HCl, dired (Na 2 SO 4 ) and concentrated to give an oil that was purified by ISCO using EtOAc in hexanes (5%) as eluents to provide the the title compound as an oil.

Step 6: (4S)-6-f2'.2'-dimethyl-3\3',3'-trifluoro-propyl)-2,2-spirocv clobutyl-3.4-dihvdro-

2H-pyranor2.3-blpyridin-4-amine

The title compound was obtained by a method analogous to the method examplified

WO2007061670.

A-1291 -WO-PCT - 103-

Example 26

(4S)-6-Neopenyl-[(2-2-spirocydobutyl)-3'-cis-cyano)]-23-d ihydropyrano[2β-b- ] pyridin-4-amine Step 1: 3-Cvanocvclobutanone

To a stirred mixture of 3-methylenecyclobutanecarbonitrile (5.0 g, 54 mmol) and ruthenium trichloride hydrate (0.086 ml, 1.2 mmol) in DCM/MeCN/H 2 O (215/215/315 ml) was added sodium meta periodate (12 ml, 225 mmol) in several portions (30 min.). The reaction mixture was slowly warmed to RT and stirred in 3h. The precipitated solid was filtered off. The filtrate was extracted with DCM (3x); dried over MgSO 4 , concentrated and filtered through a short plug of silical gel, concentrated, to give the title compound as a light brown oil, which solidified upon standing at rt. Step 2: 3-(2-(5-Bromo-2-rnethoxypyridin-3-yl)-2-oxoethylidene) cvclobutanecarbonitrile A mixture of lithium (Z)-l-(5-bromo-2-methoxypyridin-3-yl)-2- (dimethoxvphosphoryl)ethenolate (2.0 g, 5.8 mmol) and 3-oxocyclobutanecarbonitrile (1.1 g, 12 mmol) in p-dioxane (6 ml) was heated at 120 0 C by Microwave in Ih. The mixture was cooled, taken up in H 2 O, extracted with EtOAc (3x), dried over MgSO 4 , concentrated to provide the title compound. MS (m+1): 307.0. Step 3: 6-Bromo-r(2,2-spirocvclobutyl)-3'-cvano)l-2.3-dihydropyrano[ 2.3-b-lpyridine-4- one

A mixture of 3-(2-(5-bromo-2-methoxypyridin-3-yl)-2-oxoethylidene) cyclobutanecarbonitrile (3.4 g, 11 mmol), sodium iodide (1.8 ml, 44 mmol), and chlorotrimethyl silane (5.6 ml, 44 mmol) in MeCN (40 ml) was stirred at rt for 24h, concentrated, taken up in H 2 O, extracted with DCM (3x), washed with saturated NH 4 Cl, brine, dried over MgSO 4 , concentrated and purified by ISCO (20% EtOAc/Hexanes) to give the title compound as a yellow solid.

Step 4: (4R)-6-Bromo-r(2.2-spirocvclobutyl)-3'-cvano)l-23-dihvdropyr anor2.3-b- ]pyridine-4-ol To a stirred solution of (S)-2-methyl-CBS-oxazaborolidine (IM in toluene; 1.0 ml, 1.0 mmol) in toluene (5 ml) was added a solution of borane-methyl sulfide complex (0.5 ml, 5 mmol) in toluene (20 ml) and a solution of 6-bromo-[(2,2-spirocyclobutyl)-3'-cyano)]-

A-1291-WO-PCT - 104-

2,3-dihydropyrano[2,3-b-]pyridine-4-one (1.40 g, 5 mmol) in toluene (20 ml) in 30 min at 0 °C. The reaction mixture was stirred for another 15min. then slowly quenched with 10% aq. HCl, extracted with EtOAc (3x), washed with NaHCO 3 , brine, dried over MgSO 4 , concentrated to give the title compound as a light yellow solid. MS (m+1): 296.1 Step 5: (4R")-6-Bromo-4-tert-butyldimethylsilyloxo-[(2.2-spirocvclob utylV3'-cvano)l-2,3- dihydropyranor2.3-b-lpyridine

To a stirred mixture of (4R)-6-bromo-[(2,2-spirocycloburyl)-3'-cyano)]-2,3- dihydropyrano[2,3-b-]pyridine-4-ol (5.7 g, 19 mmol) and lH-imidazole (22 ml, 193 mmol) in DMF (70 ml) was added tert-butylchlorodimethylsilane (15 g, 97 mmol). The reaction mixture was stirred at rt in 24h, added water, extracted with ether (3x), dried over MgSO 4 , concentrated and purified by ISCO (15% EtOAc/Hexanes) to give the title compound. MS (m+1): 410.4.

Step 6: (4R)-6-(2'2-dimethylpropyl ' )-4-tert-butyldimethylsilyloxo-r(2.2-spirocvclobutyl ' )- 3'-cvano)l-2,3-dihvdropyranor2,3-b-1pyridine To a stirred solution of neopentylmagnesium chloride (1M,15 ml, 15 mmol) at 0 °C was added dropwise a solution of zinc(II) chloride (8 ml, 8 mmol). The mixture was gradually warmed to rt in 30 min. PdCl 2 (dppf) 2 (0.2 g, 0.2 mmol) and a solution of (4R)-6-bromo- 4-tert-butyldimethylsilyloxo-[(2,2-spirocyclobutyl)-3'-cyano )]-2,3-dihydropyrano[2,3-b- Jpyridine (1.56 g, 4 mmol) in THF (20 ml) were successively added to the mixture. The reaction mixture was stirred at 40 °C overnight, then cooled, quenched with saturated NH 4 Cl, extracted with EtOAc, dried over MgSO 4 , concentrated to provide the title compound. MS (m+1): 401.6.

Step 7: (4R)-6-Neopenyl-r(2,2-spirocyclobutyl)-3 '-cis-cvano)1-2,3-dihydropyranor2,3-b- dihvdropyranor2,3-b-1pyridine-4-ol

To a stirred solution of (4R)-6-neopentyl-4-tert-butyldimethylsilyloxo-[(2,2- spirocyclobutyl)-3'-cyano)]-2,3-dihydropyrano[2,3-b-]pyridin e (1.5 g, 4 mmol) in THF (10 ml) was added tetrabutylammonium fluoride, 1.0M in THF (7 ml, 7 mmol). The reaction mixture was stirred in 2h, quenched with H 2 O, extracted with EtOAc, dried over MgSO 4 , concentrated and purified by ISCO (40% EtOAc/Hexanes with 12Og column) to separate the cis- and trans- isomers of the title compound. MS (m+1): 287.4. Step 8: (4S)-4-Azido-6-neopenyl-[(2.2-spirocvclobutyl)-3 '-cis-cvanoV|-2.,3- dihvdropyranoβj-b-lpvridine

A-1291-WO-PCT - 105-

To a stirred solution of (4R)-6-neopenyl-[(2,2-spirocyclobutyl)-3'-cis-cyano)]-2,3- dihydropyrano[2,3-b-]pyridine-4-ol (1.05 g, 3.67 mmol) in toluene (30 ml) was added DPPA (1.03 ml, 4.77 mmol) dropwise. After stirring for 15min., DBU (0.713 ml, 4.77 mmol) was slowly added, and the reaction mixture was stirred at RT for 16h. H 2 O was added and the mixture was extracted with EtOAc (3x), washed with brine, dried over MgSO 4 , concentrated to give the title compound as a brown oil. MS (m+1): 312.4. Step 9: (4S)-6-Neopenyl-IY 2.2-spirocvclobutylV3 '-cis-cvano)1-2,3-dihydropyranor2.3-b- lpyridin-4-amine A mixture of (4S)-4-azido-6-neopenyl-[(2,2-spirocyclobutyl)-3'-cis-cyano) ]-2,3- dihydropyrano[2,3-b-]pyridine (3 g, 10 mmol) and triphenylphosphine (3 g, 10 mmol) in THF (20 ml) was stirred at RT in 2h, 3 ml OfH 2 O was added and heated at 80 0 C in 4h. 40 ml of 10%aq. HCl was added and the mixture was heated for 10 min. at 80 0 C, then cooled and extracted with toluene, (discarded). The acidic aqueous layer was neutralized with solid Na 2 CO 3 , extracted with DCM (3x), dried over MgSO 4 , purified by ISCO (3% MeOH/DCM) to give the title compound as a yellow foam. MS (m+ 1 ): 286.4.

Example 27

(S)-6-Bromo-N 8 -ethyl-2^,-spirocyclobutyl-3,4-dihydro-2H-chromene-4,8-diain ine Step 1: l-(3-amino-5-bromo-2-hydroxyphenyl)ethanone

A mixture of l-(5-bromo-2-hydroxy-3-nitrophenyl)ethanone (25 g, 96 mmol), iron (27 g, 481 mmol), and NH 4 Cl (5.1 g, 96 mmol) in EtOH/H 2 O (5:1, 300 ml) was heated at reflux in 2h, the mixture was cooled filtered the solid, the filtrate was concentrated, taken up in H 2 O, extracted with DCM (3x), dried over MgSO 4 , concentrated and purified by ISCO (10% EtOAc/Hexanes) to give the title compound as a yellow solid. MS (m+2): 232.1. Step 2: 8-Amino-6-bromo-2.2-spirocvclobutyl-2.3-dihvdrochromen-4-one A mixture of l-(3-amino-5-bromo-2-hydroxyphenyl)ethanone (4.5 g, 20 mmol), cyclobutanone (3 ml, 39 mmol), and pyrrolidine (5 ml, 59 mmol) in p-dioxane (80 ml) was heated at 65 'C for 24h. The mixture was cooled, taken up in dilute acid HCl stirred, extracted with EtOAc (3x), dried over MgSO 4 , concentrated and purified by ISCO (0- 20% in 30 min.) to give the title compound as an orange solid. MS (m+2): 284.1.

A- 1291 -WO-PCT - 106-

Step 3 : (4R)-8-Amino-6-bromo-2.2-spirocvclobutyl-2.3-dihvdrochronien -4-ol To a stirred solution of (s)-2-methyl-CBS-oxazaborolidine, IM in toluene (1 ml, 1 mmol) in toluene (2 ml) was added a solution of borane-methyl sulfide complex (5 ml, 11 mmol) in toluene (20 ml) followede by addition of a solution of 8-amino-6-bromo-2,2- spirocyclobutyl-2,3-dihydrochromen-4-one (3 g, 11 mmol) in toluene (40 ml) dropwise. After the reaction was complete as monitored by TLC, it was quenched with 10% aq. HCl (40 ml), stirred for 15min., extracted with EtOAc (3x), washed with brine, dried over MgSO 4 , filtered and concentrated to give the title compound as a purple foam. MS (m+1): 285.2. Step 4: (4R)-8-Amino-6-bromo-4-tertbutyldimethylsilyloxo-2.2-spirocv clobutyl-2,3- dihvdrochromene

A mixture of 8-amino-6-bromo-2,2-spirocyclobutyl-2,3-dihydrochromen-4-ol (3.2 g, 11 mmol) and imidazole (1 ml, 12 mmol) in DCM (30 ml) was added tert- butylchlorodimethylsilane (2 g, 12 mmol). The mixture was stirred for 3h, then H 2 O was added and the layers were separated, dried over MgSO 4 , concentrated and the crude was purified by ISCO (5% EtOAc/Hexanes) to give the title compound as a colorless oil. MS (m+1): 399.4.

Step 5 : (4RV8-ethylamino-6-bromo-4-tertbutyldimethylsilyloxo-2.2-spi rocvclobutyl-2,3- dihydrochromene A mixture of (4R)-8-amino-6-bromo-4-tertbutyldimethylsilyloxo-2,2-spirocy clobutyl-2,3- dihydrochromene (2 g, 5 mmol), acetaldehyde (0.3 ml, 5 mmol), and trimethyl orthoformate (4 ml, 40 mmol) in DCE (20 ml) was stirred at RT in 30 min. Sodium triacetoxyborohydride (5 g, 25 mmol) was added and stirred in 3h, quenched with diluted aq. HCl, extracted with DCM (3x), dried over MgSO 4 , concentrated and purified by ISCO (5% EtOAc/Hexanes) to give the title compound as a light yellow oil. MS (m+1): 428.4. Step 6: (4R)-6-Bromo-8-(ethylamino')-2.2-spirocvclobutyl-3.4-dihvdro -2H-chromen-4-ol A mixture of (4R)-8-ethylamino-6-bromo-4-tertbutyldimethylsilyloxo-2,2- spirocyclobutyl-2,3-dihydrochromene (0.900 g, 2.2 mmol) in THF (15 ml) was added tetrabutylammonium fluoride (2.6 ml, 2.6 mmol). The reaction mixture was stirred at RT in 2h. H 2 O was added and the mixture was extracted with EtOAc (3x), dried over MgSO 4 , concentrated to give the title compound as a brown oil. MS (m+1): 304.4. Step 7: (S)-6-Bromo-N 8 -ethyl-2.2,-spirocvclobutyl-3,4-dihvdro-2H-chromene-4,8- diamine

A- 1291 -WO-PCT - 107-

The title compound was obtained, as a white foam, by a method analogous to that described in Steps 8-9 of Example 26 above. MS (m+1): 312.2.

Example 28

(S)-2^-Spirocyclobutyl-6-trifluoromethyl-3,4-dihydro-2H-p yraiio[2,3 — c]pyridine-4- amine

Step 1: 5-MethoxymethoxyV2-(trifluoromethyl ' )pyridine

To a stirred suspension of NaH (3.1 ml, 74 mmol) in DMF (100 ml) was added 6- (trifluoromethyl) pyridin-3-ol (10 g, 61 mmol) in several portions. The mixture was stirred for 30 min., then chlorodimethyl ether (5.0 ml, 64 mmol) was added dropwise and stirring was continued for 3h. The reaction was cooled to O 0 C and quenched slowly by addition of H 2 O. The solution was extracted with ether (3x), the organic layers were dried over MgSθ 4 , filtered and concentrated to give the title compound as a yellow oil. MS (m+1): 208.2.

Step 2: l-(5-(Methoxymethoxy)-2-(trifluoromethyl)pyridine-4-yl)ethan ol To a stirred solution of piperidine, 2,2,6,6-tetramethyl- (9.2 ml, 54 mmol) in THF (400 ml) at -78 °C was added butyllithium (~2.5 m in toluene; 18 ml, 45 mmol) dropwise. The reaction was then stirred at 0 °C for 5 min at -78 °C, and a solution of 5- (methoxymethoxy)-2-(trifluoromethyl)pyridine (7.5 g, 36 mmol) in THF (100 ml) was added dropwise. The mixture was stirred for an additional 10 min, then acetaldehyde (20 ml, 362 mmol) was added and stirring continued for 15 min. the reaction was slowly quenched with H 2 O, warmed to RT, extracted with EtOAc (3x), combine organic layers were dried over MgSO 4 , filtered, concentrated and the crude product was purified by ISCO (20% EtOAc/Hexanes) to give the title compound as a light yellow solid. MS (m+1): 252.2.

Step 3: l-(5-(methoxymethoxy)-2-(trifluoromethyl)pyridin-4-yl ' )ethanone To a stirred mixture of l-(5-(methoxymethoxy)-2-(trifluoromethyl) pyridin-4-yl)ethanol (3.2 g, 13 mmol) and NaHCO 3 (3.2 g, 38 mmol) in DCM (100 ml) was added Dess Martin Periodinane (5.9 g, 14 mmol). The reaction mixture was stirred at rt in 16h, the solid was filtered, the filtrate was concentrated, then taken up and stirred in ether/EtOAc.

A-1291-WO-PCT - 108-

The precipitated white solids were filtered and discarded. The filtrate was concentrated to give the title compound as a light yellow oil. MS (m+1): 250.2. Step 4: l-(5-hvdroxy-2-(trifluoromethyl ' )pyridh-4-yl ' )ethanone

A mixture of l-(5-(methoxymethoxy)-2-(trifluoromethyl)pyridin-4-yl)ethano ne (3.2 g, 13 mmol) and 5N HCl (40 ml) in i-PrOH/THF (1: 1, 40 ml) was stirred at 45 °C overnight. The mixture was cooled, concentrated , taken up in H 2 O, neutralized with saturated NaHCO 3 , and extracted with DCM (3x). The organic layers were combined, dried over Na 2 SO 4 , and concentrated to give the title compound as a tan solid. MS (m+1): 206.2 Step 5: (S)-2.2-Spirocvclobutyl-6-trifluoromethyl-3,4-dihvdro-2H-pyr anor2,3 — c]pyridine-4-amine

The title compound was obtained by a method analogous to that described in Steps 4-7 of Example 18 above. MS (m+1): 259.2.

Example 29

(4S)-[(2^-SpirocydobutyI-3'(trans)-hydroxyI)]-6-neopentyl -3,4-dihydro-2H- chromen-4-amine and (4S)-[(2^-Spirocyclobutyl-3'(cis)-hydroxyl)]-6-neopentyl-3,4 - dihydro-2H-chromen-4-amine

Step 1: 2,2-Dichloro-3-oxocyclobutyl pivalate To a stirred mixture of vinyl pivalate (30 g, 234 mmol) and zinc (31 g, 468 mmol) in ether (300 ml) was added a solution of 2,2,2-trichloroacetyl chloride (55 g, 304 mmol) in ether (300 ml) dropwise (2-3h) in a water bath. (Note: fast addition causes the reaction temp.to elevate) while maintaining the reaction temperature between 15-30 °C. After the reaction was done (stained with KMnO 4 solution), it was filtered through Celite. The filtrate was washed with cold water, brine, dried over MgSO 4 and concentrated to give the title compound as an orange solid. Step 2: 3-Oxocvclobutyl pivalate

To a stirred suspension of zinc dust (103 g, 1568 mmol) in HOAc (200 ml) was added a solution of 2,2-dichloro-3-oxocyclobutyl pivalate (75 g, 314 mmol) in HOAc (400 ml) dropwise in an ice bath. The reaction mixture was stirred for Ih, filtered the solid through celite and washed with DCM. The DCM layer was washed with H 2 O, NaHCO 3 , brine,

A-1291 -WO-PCT - 109- dried over MgSO 4 , filterd and concentrated. The crude material was purified by ISCO (10% EtOAc/Hexanes) to give the title compound as a light yellow oil. Step 3: 3-Hvdroxylcvclobutyl pivalate

To a stirred solution of 3-oxocyclobutyl pivalate (15.1 g, 88.7 mmol) in ethanol (100 ml) at 0 °C was added sodium borohydride (4.69 ml, 133 mmol) in several portions. The reaction was stirred for 30 min, slowly quenched with 10% aqueous HCl and concentrated to remove ethanol. The solution was taken up with more 10% HCl, extracted with DCM (3x), washed with brine, dried over MgSO 4 and concentrated to give the title compound as a light yellow oil. Step 4: 3-(tert-Butyldimethylsilyloxy)cvclobutyl pivalate

To a stirred mixture of 3-hydroxycyclobutyl pivalate (16.60 g, 96.4 mmol) and diea (25.2 ml, 145 mmol) in DCM (100 ml) at 0 °C was added tert-butyldimethylsilyl triflate (31.0 ml, 135 mmol) dropwise. The reaction was stirred for 2h, then quenched with H 2 O. The layers were separated, and the organic layer was washed with saturated NaHCθ 3 , brine, dried over MgSO 4 and concentrated to give the title compound as a light brown oil. Step 5: 3-(tert-butyldimethylsilyloxy)cvclobutanol

To a stirred solution of 3-(tert-butyldimethylsilyloxy)cyclobutyl pivalate (4.32 g, 15 mmol) in THF (20 ml) at 0 °C was added diisobutylaluminum hydride, 1.0m solution In hexanes (48 ml, 48 mmol) dropwise. The reaction was stirred in Ih, then slowly quenched with Rochelle's salt. The quenched mixture was stirred and layers were separated. The organic layer was dried over MgSO 4 and concentrated to give the title compound as a colorless oil.

Step 6: 3-(tert-butyldimethylsilyloxy ' )cvclobutanone A mixture of 3-(tert-butyldimethylsilyloxy) cyclobutanol (2.59 g, 13 mmol), sodium bicarbonate (3 ml, 38 mmol), and Des-Martin periodonane (7 g, 15 mmol) in DCM (40 ml) was stirred at RT in 4h, the solid was filtered; the filtrated was purified by ISCO (5% EtOAc/Hexanes) to give the title compound as a colorless oil. Step 7: (4S)-[(2.2-Spirocvclobutyl-3ϊtrans)-hvdroxyl)l-6-neopentyl- 3.4-dihydro-2H- chromen-4-amine and (4S)-|Y2.2-Spirocvc1obutyl-3 '(cis)-hydroxyl)l-6-neopentyl-3,4- dihydro-2H-chromen-4-amine

The title compounds were obtained, by a method analogous to that described in Steps 4-7 of Example 18 above, after separation of the cis- and trans-isomers by reverse phase HPLC. MS (m+1): 261.2.

A- 1291 -WO-PCT - 110-

Example 30

l-(3-(Methoxymethoxy)pyridin-4-yl)ethanone

Step 1: 3-(methoxymethoxy * )pyridine Pyridin-3-ol (25 g, 260 mmol) was added to a stirring mixture of NaH (11 g of a 60 wt% dispersion with mineral oil, 260 mmol) and DMF (350 mL) at 0 0 C. After 30 min, the reaction mixture was allowed to warm to RT, stirred for 90 min, and then chloromethoxymethane (20 mL, 260 mmol) was added. After 18 h, the reaction mixture was partitioned between ethyl acetate and saturated aqueous NaHCO 3 , the layers were separated, the organic layer was washed with saturated aqueous NaHCOs, brine, dried (Na 2 SO 4 ), filtered, and the filtrate was concentrated. The residue was dissolved with CH 2 Cl 2 , the solution was filtered through a plug of silica gel (sequential elution; 9:1 → 1 : 1 hexane-ethyl acetate), and the second filtrate was concentrated to give 1O g (27%) of 3-(methoxymethoxy)pyridine as a clear yellow oil. Step 2: l-(3-(methoxymethoxy ' )pyridin-4-yl ' )ethanol

A solution of 3-(methoxymethoxy)pyridine (9.8 g, 70 mmol) and THF (40 mL) was added to a stirring mixture of tert-butyllithium (91 mL of a 1.7 M solution with pentane, 160 mmol) and THF (100 mL) at -78 0 C. After 1 h, acetaldehyde (9.9 mL, 180 mmol) was added, and the reaction mixture was stirred for 3 h and then warmed to RT. After 21 h, the reaction mixture was partitioned between ethyl acetate and saturated aqueous NaHCθ3, the layers were separated, the organic material was washed with saturated aqueous NaHCO 3 , brine, dried (Na 2 SO 4 ), filtered, and the filtrate was concentrated. The residue was purified by flash chromatography on silica gel (1:1 hexane-ethyl acetate) to afford 4.6 g (36%) of l-(3-(methoxymethoxy)pyridin-4-yl)ethanol as a colorless solid. Step 3: l-(3-(methoxymethoxy)pyridin-4-yl)ethanone

Dess-Martin periodinane (18 g, 43 mmol) was added to a stirring mixture of of l-(3- (methoxymethoxy)pyridin-4-yl)ethanol (4.6 g, 25 mmol), NaHCO 3 (6.3 g, 75 mmol), and CHCl 3 (75 mL) at RT. After 24 h, 1.0 M aqueous Na 2 S 2 O 3 was added, the reaction mixture was stirred for 90 min, partitioned between ethyl acetate and 1.0 M aqueous Na 2 S 2 O 3 , the layers were separated, the organic layer was washed with 1.0 M aqueous Na 2 S 2 O 3 , water, brine, dried (Na 2 SO 4 ), filtered, and the filtrate was concentrated. The

A-1291-WO-PCT - I ll- residue was purified by flash chromatography on silica gel (gradient elution; 2: 1 — > 1 : 1 hexane-ethyl acetate) to give 3.9 g (86%) of l-(3-(methoxymethoxy)pyridin-4- yl)ethanone as a clear yellow-orange oil.

Example 31

2-Isopropoxyacetic acid

Step 1 : tert-Butyl 2-isopropoxyacetate

In a 250-mL flask, 35% aqueous sodium hydroxide (29.3 g, 256 mmol), propan-2-ol (0.785 ml, 10.3 mmol) tetrabutylammonium chloride (0.214 g, 0.769 mmol), and tert- butyl 2-bromoacetate (1.00 g, 5.13 mmol) were taken up in benzene (25 mL). The mixture was stirred at ambient temperature overnight. The reaction was concentrated to remove most of the benzene. The aqueous residue was diluted further with water (100 mL) and the aqueous phase was extracted with 75% ether-hexane (3 x 33 mL). The organics were combined, washed with water (10 mL) then with saturated brine (10 mL).

The organics were dried over magnesium sulfate, filtered, and concentrated to afford the title compound (119 mg, 0.683 mmol, 13%).

Step 2: 2-Isopropoxyacetic acid

In a 25-mL RBF, the tert-butyl 2-isopropoxyacetate (0.120 g, 0.69 mmol) was dissolved in DCM (1.5 mL). The solution was cooled to 0 0 C. TFA (0.53 ml, 6.9 mmol) was added, and the solution was stirred at 0 0 C for 30 min. Then the ice bath was removed and the mixture was stirred at ambient temperature overnight. The mixture was then concentrated to afford the title compound.

Example 32

Lithium 2-methoxybutanoate

Step 1.: Methyl 2-methoxybutanoate

A 250-mL RBF was charged with DMSO (15 mL). n-Butyllithium (2.50 M, 8.45 ml, 21.1 mmol) was added to the mixture as a solution in hexane. A solution of 2-

A- 1291 -WO-PCT - 112- hydroxybutanoic acid (1.00 g, 9.61 mmol) was dissolved in DMSO (15 mL). The solution was transferred via cannula into the dimsyl anion reaction mixture. The resulting mixture was stirred at ambient temperature for 2.5 h. Iodomethane (1.44 ml, 23.1 mmol) was added and the reaction mixture was stirred overnight. The mixture was diluted with water (125 mL) and.the aqueous layer was extracted with ether (50 mL). The organic layer was washed with water (2 x 5 mL), then with saturated brine (5 mL), then was dried over magnesium sulfate, filtered, and concentrated to afford the title compound. Step 2: Lithium 2-methoxybutanoate

In a 15-mL RBF was dissolved methyl 2-methoxybutanoate (0.027 g, 0.20 mmol) in methanol (0.75 mL). A solution of lithium hydroxide monohydrate (0.0086 g, 0.20 mmol) in water (0.75 mL) was added, and the mixture was stirred overnight. The mixture was concentrated to afford the title compound.

The following examples in Table I were prepared by methods and steps analogous to those described in Examples 4-5 above. Provided also is the mass spectral data and BACE enzyme and cell-based assay data (IC 5 o's in uM) for each example, where available.

Table !

A- 1291 -WO-PCT -113-

A-1291-WO-PCT -114-

A-1291-WO-PCT -115-

A-1291-WO-PCT -116-

A-1291-WO-PCT -117-

A- 1291 -WO-PCT -118-

A-1291-WO-PCT -119-

A- 1291 -WO-PCT -120-

A-1291-WO-PCT -121-

A-1291-WO-PCT - 122 -

The following compounds in Tables 2 and 3 are additional representative examples of Formulas I-II, as provided by the present invention.

A-1291-WO-PCT -123-

Table 3

A-1291-WO-PCT - 124 -

Synthesis of (S)-8-chloro-2^-spirocyclobutyI-6-neopentyl-3,4-dihydro-2H- py rano [2,3-c] pyridin-4-amine

Step 1: 2-bromo-5-(methoxymethoxy)pyridine

To a solution of 6-bromopyridin-3-ol (25 g, 144 mmol) in DMF (300 mL) at 0°C under N 2 is added portionwise NaH (5.7 g, 144 mmol) over 5 min. The reaction was stirred 1 h, then chloro(methoxy)methane (12 g, 144 mmol) was added and the reaction stirred an additional 1 h at O 0 C. Saturated sodium bicarbonate (500 mL) was added slowly and the suspension stirred 30 min and warmed to it. The solution was extracted with EtOAc (3 x 400 mL), the combined organic layers washed with H 2 O (500 mL), saturated NaCl (500 mL), dried (Na 2 SO 4 ), and concentrated in vacuo to give the title compound as a brown oil. Step 2: 5-(methoxymethoxy)-2-neopentylpyridine

To a solution of 2-bromo-5-(methoxymethoxy)pyridine (30.5 g, 140 mmol) in THF (5 mL) at 0 0 C under N 2 is added dichloro-((bis-diphenylphosphino)ferrocenyl)- palladium(II)(4.88 g, 5.5 mmol) followed by dropwise addition of neopentylmagnesium chloride (155 mL 5 155 mmol) over 2 min. After addition, the cooling bath was removed and the reaction stirred 3 h at rt. The reaction was cooled to 0 0 C and saturated NH 4 Cl

A- 1291 -WO-PCT - 125 -

(500 mL) was added, and the aqueous layers extracted with EtOAc (3 x 200 mL). The combined organic layers were washed with saturated NaCl, dried (Na 2 SO 4 ) and concentrated to give a red oil. Purification by vacuum filtration through a silica plug (9 x 7 cm, dry load, 10-20%% EtOAc/Hexanes) gives 5-(methoxymethoxy)-2- neopentylpyridine as a light yellow oil.

Step 3: l-(5-(methoxymethoxy)-2-neopentylpyridin-4-yl)ethanol

To a solution of 5-(methoxymethoxy)-2-neopentylpyridine (16.5 g, 79 mmol) and in THF (200 mL) -78°C is added terf-butyllithium (46 ml, 79 mmol) (1.7 M in pentane ) over 2 min via cannula. The reaction was stirred at -78 0 C 30 min, and acetaldehyde (11 ml, 197 mmol) was added. The reaction was stirred at -78°C 10 min, then the reaction was warmed to rt and stirred 3 h. The reaction was quenched by addition of saturated aqueous NH 4 Cl (400 mL), extracted with EtOAc (3 x 200 mL), the combined organic layers washed with saturated NaCl (10 mL), dried (Na 2 SO 4 ), and concentrated to give a orange oil, which was purified by chromatography on an ISCO (330 g SiO 2 , 10%-50% EtOAc/Hexane) gives l-(5-(methoxymethoxy)-2-neopentylpyridin-4-yl)ethanol as a clear, light yellow oil.

Step 4: l-(5-(methoxymethoxy)-2-neopentylpyridin-4-yl)ethanone To a solution of l-(5-(methoxymethoxy)-2-neopentylpyridin-4-yl)ethanol (24.4 g, 96.3 mmol) and sodium bicarbonate (32.4 g, 385 mmol) in CHCl 3 (500 mL) at 0 0 C was added Dess-Martin Periodinane (53.1 g, 125 mmol). The reaction was stirred 5 h, quenched with saturated aqueous Na 2 SO 3 (300 mL), extracted with CH 2 Cl 2 (3 x 250 mL), the combined organic layers washed with saturated NaCl (300 mL), dried (Na 2 SO 4 ), and concentrated to give a yellow oil. Purification by ISCO (330 g SiO 2 , 20% EtOAc/Hexane) gives l-(5-(methoxymethoxy)-2-neopentylpyridin-4-yl)ethanone as a clear, colorless oil.

Step 5: l-(5-hydroxy-2-neopentylpyridin-4-yl)ethanone

A solution of l-(5-(methoxymethoxy)-2-neopentylpyridin-4-yl)ethanone (21.6 g, 86 mmol) in (2: 1 : 1) 5 M HCl : i-PrOH : THF (800 mL) was stirred 4 h at rt. The mixture was concentrated to remove the THF and /-PrOH. The resulting solution consisting of the product in aqueous HCl was quenched by slow addition to a solution of saturated aqueous NaHCO 3 (500 mL) containing excess solid NaHCO 3 (50 g). The aqueous layer was extracted with CH 2 Cl 2 (3 x 250 mL), the organic layers combined and washed with saturated aqueous NaCl (250 mL), dried (MgSO 4 ), and concentrated to give l-(5- hydroxy-2-neopentylpyridin-4-yl)ethanone as a brown oil.

A-1291-WO-PCT - 126 -

Step 6: 2,2-spirocvclobutyl-6-neopentyr-2.3-dihvdropyranor2.3-clpyri din-4-one A mixture of l-(5-hydroxy-2-neopentylpyridin-4-yl)ethanone (14.8 g, 71 mmol) (76894- 11), Hϋnig's base (12 ml, 71 mmol) pyrrolidine (8.9 ml, 107 mmol), and cyclobutanone (13 ml, 179 mmol) in toluene (300 mL) with a Dean-Stark trap was heated in a 140 0 C oil bath for 2 h. The mixture was cooled to rt, then diluted with EtOAc (25 mL), washed with H 2 O, saturated aqueous NH 4 Cl, saturated aqueous NaCl, dried (MgSO 4 ), and concentrated. Purification by ISCO (12O g SiO 2 , 10-20% EtOAc/Hexane) gives the title compound as a yellow solid. Step 7: 2,2-spirocvclobutyl-6-neopentyl-7-oxo-2,3-dihydropyrano [2,3-c]pyridin-4-one 2,2-spirocyclobutyl-6-neopentyl-2,3-dihydropyrano[2,3-c]pyri din-4-one (5.00 g, 19 mmol) was dissolved in 100 ml CHCl 3 and cooled to 0 0 C, mCPBA (10.0 g, 58 mmol) was added portionwise and the reaction was stirred under N 2 and allowed to warm slowly to rt; stirring was continued for 17 h. The mixture was then cooled to 0 0 C, IM NaOH (100 mL) was added, and stirring was continued vigorously for 10 min. The mixture was extracted with CH 2 Cl 2 (3 x 100 mL), the combined organic layers washed with saturated sodium chloride (100 mL), dried (Na 2 SO 4 ) and evaporated to give the title compound as a white solid.

Step 8: S-chloro^.σ-spirocvclobutyl-ό-neopentyl^^-dihvdro pyranor2.3-clpyridin-4-one 2,2-spirocyclobutyl-6-neopentyl-7-oxo-2,3-dihydropyrano[2,3- c]pyridin-4-one. (5.3 g, 19 mmol) was taken up in phosphoryl trichloride (20 mL, 218 mmol) and the mixture was heated to 8O 0 C for 2 h under N 2 . The reaction mixture was quenched by slow addition to vigorously stirred cold 10% aqueous NaCO 3 (300 mL), extracted with EtOAc (3 x 200 mL), the combined organic layers were washed with saturated NaCl (200 mL), dried (Na 2 SO 4 ), and concentrated to give a brown oil. Purification by ISCO (120 g SiO 2 , 10% EtOAc/Hexane) gives the title compound as a light yellow solid.

Step 9: (R)-8-chloro-2.2-spirocvclobutyl-6-neopentyl-3.4-dihvdro-2H- pyranor2.3- c]pyridin-4-ol

To a stirred solution of (s)-2-methyl-cbs-oxazaborolidine (1.7 ml, 1.7 mmol) in THF (20 mL) at O 0 C is added borane-methyl sulfide complex (14 ml, 28 mmol) followed by a solution of 8-chloro-2,2-spirocyclobutyl-6-neopentyl-2,3-dihydropyrano[2 ,3-c]pyridin-4- one (4.90 g, 17 mmol) in THF (40 mL) dropwise via syringe pump over 2.8 h. The reaction was stirred an additional 30 min, then was quenched by dropwise addition (1 drop/10 sec) of 5 M HCl (25 mL) at 0 0 C, after 15 mL HCl was added, bubbling had ceased and the addition rate was increased as the ice bath was removed. The reaction was

A-1291-WO-PCT - 127 - stirred an additional 2 h at rt The reaction was recooled to 0 0 C and neutralized with 5-M NaOH (27 mL). The mixture was then extracted with EtOAc (2 x 150 mL), washed with saturated aqueous NaCl (200 mL), dried (MgSθ 4 ), and concentrated in vacuo. Purification by ISCO (12O g SiO 2 , 20% EtOAc/Hexane) gives the title compound as a white foam.

Step 10: (S)-4-a2ido-8-chloro-2.2-spirocvclobutyl-6-neopentyl-3.4-dih ydro-2H- pyranor2.3-c1pyridine

To a solution of (R)-8-chloro-2,2-spirocyclobutyl-6-neopentyl-3,4-dihydro-2H- pyrano[2,3-c]pyridin-4-ol (2.33 g, 7.9 mmol) in toluene (43 mL) is added diphenylphosphoryl azide (2.4 ml, 11 mmol) then l,8-diazabicyclo(5.4.0)-7-undecene (1.6 ml, 11 mmol). The reaction was stirred under N 2 at rt 4 days. The clear, light yellow solution first turned into a yellow cloudy/opaque solution after 10 min. Water (100 mL) was added and the reaction mixture extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with saturated NaCl (150 mL), dried (MgSO 4 ), and concentrated to give the title compound as a brown oil which was used in the next step without purification.

Step 11: (S)-8-chloro-2,2-spirocvclobutyl-6-neopentyl-3.4-dihvdro-2H- pyranor2,3- cipyridin-4-amine To a solution of (S^-azido-δ-chloro^^-spirocyclobutyl-ό-neopentyl-S^-dihydr o^H- pyrano[2,3-c]pyridine (2.21 g, 6.9 mmol) in 10:1 THFZH 2 O (44 mL) at 0 0 C is added

NaOH (3.0 ml, 15 mmol) (5 N). After 5 min, trimethylphosphine (2.4 ml, 28 mmol) was added dropwϊse over 4 min. The reaction went from brown to pink to purple as N 2 evolution occurred. The ice bath was allowed to melt as the reaction warmed to rt and stirred a total of 15 h. The mixture was re-cooled to 0°C and 5 N HCl (25 mL) was added. The resulting mixture was extracted with EtOAc (3 x 50 mL), the combined organic layers were washed with 2.5 N HCl (2 x 25 mL). The combined aqueous layers were cooled to 0 0 C and basified to pH 14 with 5 N NaOH (100 mL). The aqueous layer was extracted with EtOAc (3 x 50 mL) the combined organic layers dried (Na 2 SO 4 ), and concentrated to give the crude product as a viscous yellow oil. The combined organic layers were combined with the crude product from above and concentrated to give a crude yellow oil. Purification of the crude oil by flash chromatography (5 x 15 cm SiO 2 , 0-10% MeOH/CH 2 Cl 2 gradient elution) gave the title compound as a yellow oil.

A-1291-WO-PCT - 128-

Synthesis of (4S)-6-(2-fluoro-2-methylpropyl)-8-(lH-imidazol-l-yl)-2,2- spirocyclobutylchroman-4-amine Step 1 : rR)-(6-Bromo-2.2-spirocvclobutyl-3.4-dihvdro-2H-chromen-4-yl oxy)(tert- butvDdimethylsilane

To a round bottomed flask was added (R)-6-bromo-2,2-spiτocyclobutyl-3,4-dihydro-2H- chromen-4-ol (37.7 g, 140 mmol), CH 2 Cl 2 (600 mL), DIPEA (39.0 ml, 210 mmol) and an ice bath. After cooling for 20 minutes, TBS triflate (32.0 ml, 139 mmol) was added. After stirring for 45 minutes, the ice bath was removed and an additional 6 mL of TBS triflate were added. After stirring for a further 30 minutes, the reaction was washed with water (100 mL), HCl (IM, 200 mL), and sat'd NaHCO 3 (100 mL). The organic layer was concentrated in vacuo. The crude orange oil was taken up in hexane (100 mL) and eluted through a plug of silica gel (600 mL Frit) using 2%EtOAc/hexane to give (R)-(6-bromo- 2,2-spirocyclobutyl-3,4-dihydro-2H-chromen-4-yloxy)(tert-but yl)dimethylsilane (51.05 g, 95.1% yield), as a colorless oil. The material was carried forward without further characterization. MS m/z: 253.1 (M-H-OTBS).

Step 2: (R)-I -(4-(Tert-butyldimethylsilyloxy)-2.2-spirocvclobutyl-3.4-dih ydro-2H- chromen-6-yl)propan-2-one To a RBF was added MePhos (1.7 g, 4.7 mmol), dichloro[l,F- bis(diphenylphosphino)ferrocene]palladium (TT) (1.4 g, 2.0 mmol), and THF (29 mL). The solution was degassed with N 2 for 20 minutes. The solution was heated to 45°C for 20 minutes, then allowed to cool to RT. To a separated 500 mL RBF was added (R)-(6- bromo-2,2-spirocyclobutyl-3,4-dihydro-2H-chromen-4-yloxy)(te rt-butyl)dimethylsilane (15.0 g, 39 mmol), potassium phosphate (21.77 g, 103 mmol) (freshly ground) and degassed acetone, 99% (140 ml, 1904 mmol). The solution of catalyst and ligand were cannulated into the acetone solution and the entire reaction further degassed with N 2 for 20 minutes. The solution was then stirred at reflux. After 24 hours, the reaction was allowed to cool to RT and filtered through a cartridge of celite. The filtrate was concentrated in vacuo and adsorbed onto a plug of silica gel and chromatographed

A-1291-WO-PCT - 129 - through a Redi-Sep® pre-packed silica gel column (120 g), eluting with 0% to 5% EtOAc in hexane, to provide (R)-l-(4-(tert-butyldimethylsilyloxy)-2,2-spirocycloburyl-3, 4- dihydro-2H-chromen-6-yl)propan-2-one as a light yellow oil. MS m/z: 229.1 (M-H- OTBS). Step 3: (R)-l-(4-(Tert-butyldimethylsilyloxy)-2.2-spirocvclobutyl-3, 4-dihvdro-2H- chromen-6-yl)-2-methylpropan-2-ol

To a RBF was added (R)-l-(4-(tert-butyldimethylsilyloxy)-2,2-spirocyclobutyl-3, 4- dihydro-2H-chromen-6-yl)propan-2-one (4.97 g, 13.8 mmol) and THF (60 mL, anhydrous). The solution was cooled to 0°C and treated with methylmagnesium chloride (5.50 ml, 16.5 mmol). The solution was allowed to warm to RT over 1 hour, then stirred at RT for 2 hours. The reaction was then partitioned between EtOAc:water and the resulting aqueous layer extracted with EtOAc (50 mL). The combined organic layers were washed with brine, dried over MgSO , and concentrated in vacuo to give (R)- 1 -(4-

(tert-butyldimethylsilyloxy)-2,2-spirocyclobutyl-3,4-dihy dro-2H-chromen-6-yl)-2- methylpropan-2-ol as a yellow oil that solidified. MS m/z: 245.2 (M-H-OTBS).

Step 4: (R)-l-(8-Bromo-4-(tert-butyldimethylsilyloxy ' )-2.2-spirocvclo-butyl-3.4-dihydro-

2H-chromen-6-yl)-2-methylpropan-2-ol

To a RBF was added (R)-l-(4-(tert-butyldimethylsilyloxy)-2,2-spirocyclobutyl-3, 4- dihydro-2H-chromen-6-yl)-2-methylpropan-2-ol (5.04 g, 13.4 mmol), sodium bromide (2.75 g, 26.8 mmol), acetone:water (1:1, 160 mL), and Oxone (8.23 g, 13.4 mmol). The reaction warmed about 10 0 C. After 6 hours, the reaction was filtered and the filtrate extracted with EtOAc (2 X 50 mL). The combined organic layers were concentrated in vacuo and adsorbed onto a plug of silica gel and chromatographed through a Redi-Sep® pre-packed silica gel column (80 g), eluting with 0% to 10% EtOAc in hexane, to provide (R)- 1 -(8-bromo-4-(tert-butyldimethylsilyloxy)-2,2-spirocyclo-buty l-3 ,4-dihydro-2H- chromen-6-yl)-2-methylpropan-2-ol as a light yellow oil. MS m/z: 325.0 (M-H-OTBS).

Step 5 : 1 -((4RV4-(Tert-butyldimethylsilyloxyV8-( lH-imidazol- 1 -ylVσ.σ-spirocvclobutyl- chroman-6-yl)-2-methylpropan-2-ol

A glass microwave reaction vessel was charged with (R)-l-(8-bromo-4-(tert- butyldimethylsilyloxy)-2,2-spirocyclobutyl-3,4-dihydro-2H-ch romen-6-yl)-2- methylpropan-2-ol (1.30 g, 2.9 mmol), DMF:Water (9:1, 20 mL), cesium carbonate (4.6 g, 14 mmol), copper(I) iodide (0.54 g, 2.9 mmol), lH-imidazole (0.97 g, 14 mmol), and N,N'-dimethylethane-l,2-diamine (0.31 ml, 2.9 mmol). The reaction mixture was stirred and heated in a Biotage Initiator at 180 0 C for 40 minutes. The reaction was set up in

A-1291-WO-PCT - 130 - triplicate. The vials were all poured into 150 mL of water. The aqueous solution was extracted with EtOAc (4 X 40 mL). The combined organic layers were filtered through a celite cartridge to remove an emulsion. The filtrate was washed with water, brine, and concentrated in vacuo to give a brown oil. The oil was adsorbed onto a plug of silica gel and chromatographed through a Redi-Sep® pre-packed silica gel column (40 g), eluting with 0% to 5% MeOH in CH2C12, to provide l-((4R)-4-(tert-butyldimethylsilyloxy)-8- (lH-imidazol-l-yl)-2,2-spirocyclobutyl-chroman-6-yl)-2-methy lpropan-2-ol as a brown solid. MS m/z: 443.3 (M+l). Step 6: l-((4R ' )-4-fTert-butyldimethylsilyloxy ' )-6-(2-fluoro-2-methylpropylV2.2- spirocyclobutyl-3 ,4-dihvdro-2H-chromen-8-yl ' )- 1 H-imidazole

To a RBF was added l-((4R)-4-(tert-butyldimethylsilyloxy)-8-(l H-imidazol- l-yl)-2,2- spirocyclobutyl-chroman-6-yl)-2-methylpropan-2-ol (2.94 g, 6.64 mmol), CH 2 Cl 2 (80 mL), and a dry ice/iPrOH bath. After stirring for 15 minutes, DAST (1.75 ml, 13.3 mmol) was added. After 30 minutes, TLC shows complete conversion. The cooling bath was removed and the material poured into sat'd NaHCθ 3 (25 mL). The aqueous layer was extracted with CH 2 Cl 2 (5 mL) and the combined organic layers were concentrated in vacuo. The crude product was adsorbed onto a plug of silica gel and chromatographed through a Redi-Sep® pre-packed silica gel column (12 g), eluting with 0% to 20% EtOAc in CH2C12, to provide l-((4R)-4-(tert-butyldimethylsilyloxy)-6-(2-fluoro-2- methylpropyl)-2,2-spirocyclobutyl-3,4-dihydro-2H-chromen-8-y l)-l H-imidazole as a brown oil, that solidified upon standing. MS m/z: 445.3 (M+l). Step 7: (4RV6-r2-Fluoro-2-methylDropylV8-( 1 H-imidazol- 1 -ylV2.2-spiro- cvclobutylchroman-4-ol To a RBF was added l-((4R)-4-(tert-butyldimethylsilyloxy)-6-(2-fluoro-2-methylp ropyl)- 2,2-spiro-cyclobutyl-3,4-dihydro-2H-chromen-8-yl)-lH-imidazo le (1.6 g, 3.6 mmol), THF (40 mL), and an ice bath. After stirring for 15 minutes, TBAF (4.0 ml, 4.0 mmol) was added. After 3 hours, LC-MS shows complete conversion. The reaction was poured on to a plug of silica gel and eluted with 50% EtOAc in CH2C12, to provide (4R)-6-(2- fluoro-2-methylpropyl)-8-(l H-imidazol- l-yl)-2,2-spiro-cyclobutylchroman-4-ol as a light yellow solid. This material was carried forward as is. MS m/z: 331.2 (M+l).

Step 8: 1 -((4S)-4-Azido-6-(2-fluoro-2-methylpropyl)-2,2-spirocyclobut yl-3 ,4-dihydro- 2H-chromen-8-yl)- 1 H-imidazole

To RBF was added (4R)-6-(2-fluoro-2-methylpropyl)-8-(l H-imidazol- l-yl)-2,2-spiro- cyclobutylchroman-4-ol (1.18 g, 3.57 mmol), toluene (4 mL), and an ice bath. After

A-1291-WO-PCT - 131 - stirring for 15 minutes, DPPA (1.08 ml, 5.00 mmol) was added, then DBU (0.748 ml, 5.00 mmol). After 24 hours, LC-MS shows complete conversion. The reaction was diluted with water (100 mL) and extracted with EtOAc (3 X 40 mL). The combined organic layers were concentrated in vacuo and adsorbed onto a plug of silica gel and chromatographed through a Redi-Sep® pre-packed silica gel column (12 g), eluting with 0% to 40% EtOAc in hexane, to provide l-((4S)-4-azido-6-(2-fluoro-2-methylpropyl)- 2,2-spirocyclobutyl-3,4-dihydro-2H-chromen-8-yl)-lH-imidazol e as a light golden oil. MS m/z: 356.4 (M+l). Step 9: (4S)-6-(2-Fluoro-2-methylpropylV8-( lH-imidazol-1 -vD-2,2- spirocvclobutylchroman^-amine

To RBF was added l-((4S>4-azido-6-(2-fluoro-2-methylpropyl)-2,2-spirocyclo butyl-3,4- dihydro-2H-chromen-8-yl)-lH-imidazole (1.11 g, 3.1 mmol), THF (100 mL), and a dry ice/iPrOH bath. After stirring for 15 minutes, LAH (12 ml, 12 mmol) was added over 30 minutes. LC-MS showed no progress. The solution was warmed to O 0 C via an ice bath. After a further 30 min, the reaction is -75% complete (via LC-MS). After another hour, the reaction was cautiously quenched with sat'd Rochelle's salt and the organic layer separated. The aqueous layer was extracted with EtOAc (2 X 3O mL). The combined organic layers were washed with brine, concentrated in vacuo, and adsorbed onto a plug of silica gel and chromatographed through a Redi-Sep® pre-packed silica gel column (12 g), eluting with 0% to 5% MeOH in CH2C12, to provide (4S)-6-(2-fluoro-2- methylpropyl)-8-(lH-imidazol-l-yl)-2,2-spirocyclobutylchroma n-4-amine as a light yellow oil. MS m/z: 330.4 (M+l).

Example 201 (also example 221 in Table 4)

N-((2R r 3S)-l-(2^-spirocyclobutane-8-(methylamino)-6-neopentyl-3,4-d ihydro-2H- pyranoP^-clpyridine-^ylamino^-hydroxyhex-S-yn-S-ytyacetaniid e

A- 1291 -WO-PCT - 132 -

Step l: (SVN-((2S.3S ' )-l,2-bis(tert-butyldimethylsilyloxy)-6-(triniethylsilyl ' )hex-5-vn-3- yl)-2-methylpropane-2-sulfϊnamide

Trimethyl(prop-l-ynyl)silane (4 mL, 28 mmol) and N,N,N',N'-tetramethylethanediamine (5 mL, 36 mmol) were dissolved in dry diethyl ether (50 mL) and cooled to - 78 0 C. Tert- butyllithium, 1.7 M solution in pentane (17 mL, 28 mmol) was added dropwise and the heterogenous solution was allowed to warm to 0 0 C for 15 min to give a clear yellow solution, which was recooled to - 78 C. Azeotropically (S,E)-N-((S)-2,3-bis(tert- butyldimethylsilyloxy)propylidene)-2-methylpropane-2-sulfina mide (3.0 g, 7 mmol) in dry THF (20 mL) was added dropwise and the solution was stirred for 1 h until the starting material was consumed. The reaction mixture as quenched with NH 4 Cl and extracted with EtOAc. The combined organics were washed with water, brine, dried over Na 2 SO 4 , filtered, and concentrated to afford a yellow oil as (S)-N-((2S,3S)-l,2-bis(tert- butyldimethylsilyloxy)-6-(trimethylsilyl)hex-5-yn-3-yl)-2-me thylpropane-2-sulfinamide.

Step 2: Tert-butyl (2S,3SV2-(tert-butyldimethylsilyloxy)-l-hvdroxy-6- (trimethylsilyl)hex-5 -vn-3 -ylcarbamate

To a solution of (S)-N-((2S,3S)-l,2-bis(tert-butyldimethylsilyloxy)-6-(trimet hylsilyl)hex- 5-yn-3-yl)-2-methylpropane-2-sulfϊnamide (1.8 g, 3.4 mmol) in dry MeOH (70 mL) at - 20 C was added a solution of 10 % HCl (200 mL, acetyl chloride-MeOH) and stirred at this temperature for 1 H. The mixture was quenched with 10 % Na 2 3 and extracted with DCM (10X). The organic extracts were combined, dried OVCrNa 2 SO 4 , filtered, and concentrated. The yellow oil obtained was dissolved in DCM (30 mL) followed by the addition of TEA (1.4 ml, 10 mmol) and di-tert-butyldicarbonate (1.5 g, 6.7 mmol). The reaction mixture was stirred at rt for 3 h, concentrated, and chromatographed on silica gel using 0-3:1 EtO Ac/hexanes toa fford a light yellow oil as tert-butyl (2S,3S>2-(tert- butyldimethylsilyloxy)-l-hydroxy-6-(trimethylsilyl)hex-5-yn- 3-ylcarbamate. MS m/z: 316.2 (M+l).

A-1291-WO-PCT - 133 -

Step 3: Tert-butyl (2S.3SV2-(tert-butyldimethylsilyloxy')-l-oxo-6-(triniethylsi lyl)hex-5- vn-3 -ylcarbamate

To a solution of tert-butyl (2S,3S)-2-(tert-butyldimethylsilyloxy)-l-hydroxy-6- (trimethylsilyl)hex-5-yn-3 -ylcarbamate (1.83 g, 4.4 mmol) in DCM (20 mL) were added sodium bicarbonate (1.5 g, 18 mmol) and Dess-MartinPeriodinane (2.4 g, 5.7 mmol) and the reaction was stirred at rt for 1 h. The mixture was quenched with sat NaHCC>3 followed by the addition of sodium thiosulfate (3.5 g, 22 mmol) and stirred for an additional 1 h. The reaction was extracted with DCM and the combined organics were washed with brine, dried over Na 2 SO 4 , filtered, and concentrated to afford a light yellow oil as tert-butyl (2S,3S)-2-(tert-butyldimethylsilyloxy)-l-oxo-6-(trimethylsil yl)hex-5-yn- 3-ylcarbamate. MS m/z: 316.2 (M+l).

Step 4: Tert-butyl (2R3S)-2-(tert-butyldimethylsilyloxyVl-(TS>8-chloro-2.2- spirocvclobutane-6-neopentyl-3.4-dihvdro-2H-pyranor2.3-clpyr idin-4-ylamino)-6- (trimethylsilyl)hex-5-vn-3-ylcarbamate

To a solution of (S)-8-chloro-2,2-spirocyclobutane-6-neopentyl-3,4-dihydro-2H - pyrano[2,3-c]pyridine-4-amine (1.4 g, 4.7 mmol) and tert-butyl (2S,3S)-2-(tert- butyldimethylsilyloxy)-l-oxo-6-(trimethylsilyl)hex-5-yn-3-yl carbamate (2.4 g, 5.7 mmol) in DCE (20 mL) was added trimethyl orthoformate (5.2 ml, 47 mmol) dropwise and the resulting mixture was stirred at rt for 1 h. At this point, sodium triacetoxyborohydride (4.0 g, 19 mmol) was added and stirred for 2 h more. The mixture was quenched with 10% Na 2 CO 3 and extracted with DCM. The combined organics were washed with brine, dried over Na 2 SO 4 , filtered, concentrated, and chromatographed on silica gel using 0-3:1 EtOAc/hexanes to afford a yellow oil as tert-butyl (2R,3 S)-2-(tert-butyldimethylsilyloxy)- l-((S)-8-chloro-2,2-spirocyclobutane-6-neopentyl-3,4-dihydro -2H-pyrano[2,3-c]pyridin- 4-ylamino)-6-(trimethylsilyl)hex-5-yn-3-ylcarbamate. MS m/z: 692.3 (M+l).

A-1291-WO-PCT - 134 -

Step 5: QRJS^-ammo-l-^SVS-chloro-σ^-spirocyclobutane-ό-neopentyl^ -dihvdro- 2H-pyranor2.3-clpyridin-4-ylamino)-6-(trimethylsilyl ' )hex-5-vn-2-ol To a solution of (2R,3S)-2-(tert-butyldimethylsilyloxy)-l-((S)-8-chloro-2,2- spirocyclobutane-6-neopentyl-3,4-dihydro-2H-pyrano[2,3-c]pyr idine-4-ylamino)-6- (trimethylsilyl)hex-5-yn-3-ylcarbamate (1.5 g, 2.2 mmol) in MeOH (40 mL) was added 4.0 M HCl in dioxane (40 mL) and the resulting mixture was stirred at 50 C for 1.5 H. The mixture was concentrated and the residue was washed with 1 N NaOH and extracted with DCM. The organic extracts were combined, washed with brine, dried OVCrNa 2 SO 4 , filtered, concentrated, and chromatographed on silica gel using 0-5% MeOH/DCM and 1- 3% 2M NH 3 MeOH/DCM to afford a light yellow foam as (2R,3S)-3-amino-l-((S)-8- chloro-2,2-spirocyclobutane-6-neopentyl-3,4-dihydro-2H-pyran o[2,3-c]pyridin-4- ylamino)-6-(trimethylsilyl)hex-5-yn-2-ol. MS m/z: 478.1 (M+l).

Step 6: N-( (2R.3 SV 1 -((SV S-chloro^-spirocvclobutane-ό-neopentyl^ ,4-dihvdro-2H- pyranor2,3-c]pyridin-4-ylamino-2-hvdroxy-6-(trimethylsilvπh ex-5-vn-3-vπacetamide To a solution of (2R,3S)-3-amino-l-((S)-8-chloro-2,2-spirocyclobutane-6-neope ntyl-3,4- dihydro-2H-pyrano[2,3-c]pyridin-4-ylamino)-6-(trimethylsilyl )hex-5-yn-2-ol (0.24 g, 0.50 mmol) in DMF (2 mL) was added Hunig'sBase (0.088 ml, 0.50 mmol) followed by the addition of l-(lH-imidazol-l-yl)ethanone (0.058 g, 0.53 mmol) and stirred at rt for 17 h. The mixture was diluted in DCM and washed with sat NH 4 CI. The aqueous phase was extracted (3x) with DCM and the combined organics were dried over Na 2 SO 4 , filtered, concentrated, and chromatographed on silica gel using 0-5% MeOH/DCM to afford N- ((2R,3S)-l-((S)-8-chloro-2,2-spirocyclobutane-6-neopentyl-3, 4-dihydro-2H-pyrano[2,3- c]pyridin-4-ylamino-2-hydroxy-6-(trimethylsilyl)hex-5-yn-3-y l)acetamide as a light oil. MS m/z: 520.2 (M+l).

A-1291-WO-PCT - 135 -

Step 7: N-((2R,3S)-l-(2.2-spirocvclobutane-8-(methylamino)-6-neopent yl-3.4-dihvdro- 2H-pyrano [2,3 -clpyridine-4-ylamino-2-hvdroxyhex-5 -vn-3 -vDacetamide A mixture of tris(dibenzylideneacetone)dipalladium(0)(0.044 g, 0.048 mmol), N- ((2R,3S)-l-(8-chloro-2,2-spirocyclobutane-6-neopentyl-3,4-di hydro-2H-pyrano[2,3- c]pyridine-4-ylamino)-2-hydroxy-6-(trimethylsilyl)hex-5-yn-3 -yl)acetamide (0.25 g, 0.48 mmol), lithium bis(trimethylsilyl)amide (4.3 mL, 4.3 mmol), methanamine (0.96 mL, 1.9 mmol), and DavePhos (0.042 g, 0.11 mmol) in a sealed tube was purged with N 2 for 30 min and the mixture was heated at 90 0 C for 20 h. The mixture was brought to rt, quenched with water and extracted with DCM. The combined organics were dried over Na 2 SO 4 , filtered, concentrated, and chromatographed on silica gel using 0-5%

MeOH/DCM and 0-5% MeOH/DCM to afford a yellow solid as N-((2R,3 S)-I -(2,2- spirocyclobutane-8-(methylamino)-6-neopentyl-3,4-dihydro-2H- pyrano[2,3-c]pyridine-4- ylamino-2-hydroxyhex-5-yn-3-yl)acetamide. MS m/z: 443.2 (M+l).

Example 202

N-((2R^S)-l-(2^-spirocyclobutane-8-(methylamino)-6-neopentyl -3,4-dihydro-2H- pyrano[2 r 5-c]pyridine-4-ylamino-2-hydroxyhex-5-yn-3-yl)-2-inethoxyace tainide.

A mixture of tris(dibenzylideneacetone)dipalladium(0)(0.047 g, 0.051 mmol), N- ((2R,3 S)- 1 -(8-chloro-2,2-spirocyclobutane-6-neopentyl-3 ,4-dihydro-2H-pyrano[2,3- c]pyridine-4-ylamino)-2-hydroxy-6-(trimethylsilyl)hex-5-yn-3 -yl)2-methoxyacetamide (0.28 g, 0.51 mmol), lithium bis(trimethylsilyl)amide (4.6 mL, 4.6 mmol), methanamine (1.0 mL, 2.0 mmol), and DavePhos (0.044 g, 0.11 mmol) in a sealed tube was purged with N 2 for 30 min and the mixture was heated at 110 0 C for 10 min in microwave. The mixture was brought to rt, quenched with water and extracted with DCM. The combined organics were dried over Na 2 SO 4 , filtered, concnetrated, and chromatographed on silica gel using 0-5% MeOH/DCM and 0-3% 2 M NH 3 MeOHZDCM to afford a yellow solid as N-((2R,3 S> 1 -(2,2-spirocyclobutane-8-(methylamino)-6-neopentyl-3 ,4-dihydro-2H-

A- 1291 -WO-PCT - 136 - pyrano^S-cJpyridme^-ylammo^-hydroxyhex-S-yn-S-yl^-methoxyace tamide. MS m/z: 473.2 (M+l).

Example 203

Pd 2 (CIbB) 3 (0 1 equiv)

(5)-te/i'-Butyl 2^-spirocyclobutyl-6-neopentyl-8-(thiazol-2-yIaniino)-3,4-di hydro-2H- pyranol∑^-cjpyridin-^ylcarbamate

A 25-mL Schlenk flask was charged with (S)-tert-butyl 8-chloro-2,2-spirocyclobutyl-6- neopentyl-3,4-dihydro-2H-pyrano[2,3-c]pyridin-4-ylcarbamate (266 mg, 674 μmol), thiazol-2 -amine (202 mg, 202 μmol), sodium carbonate (100 mg, 943 μmol), tris(dibenzylideneacetone)dipalladium (62 mg, 67 μmol), and 4,5- bis(diphenylphosphino)-9,9-dimethyl-9H-xanthene (117 mg, 202 μmol). The flask was evacuated and refilled with N 2 (g). This process was repeated twice. Ar (g)-degassed toluene (3.4 mL) was added, and the resulting solution was stirred for 10 min. Water (12 μl, 67 μmol) was added, and the reaction vessel was sealed and heated in a 100 0 C oil bath for 24 h. The mixture was then cooled to RT, diluted with THF (5 mL), filtered through celite, and concentrated under reduced pressure. The residue was purified by chromatography on a 40-g Redi Sep silica gel column with 5-50% EtOAc/hexanes to give the product as a clear oil. MS m/z = 459.3 [M+H] + . Calc'd for C 24 H 35 N 4 O 3 S: 459.2.

(^-Z^-Spirocyclobutyl-ό-neopentyl-λ'S-phenyl-Sjφ^ihydr o-lH-pyranoIl^- c] py ridine-4,8-diamine

A-1291-WO-PCT - 137 -

A dry 2-mL microwave vial was charged with (^-δ-chloro^^-spirocyclobutyl-ό- neopentyl-3,4-dihydro-2H-pyrano[2,3-c]pyridin-4-amine (186 mg, 631 μmol), DavePhos (109 mg, 278 μmol), and tris(dibenzylideneacetone)dipalladium(0) (116 mg, 126 μmol). The vessel was sealed and purged with N 2 (g). THF (3 mL) was added, followed by aniline (345 μl, 3785 μmol). Lithium bis(trimethylsilyl)amide (3785 μl of 1.0 M solution in THF, 3785 μmol) was added dropwise over 25 seconds to give a dark purple solution. This mixture was heated in a Biotage Initiator microwave reactor at 110 0 C for 10 min. The reaction mixture was diluted with 2N HCl solution (aq., 20 mL) and washed with DCM (1 x 20 mL, 2 x 10 mL). The DCM layers were combined and extracted with water (10 mL). The aqueous layers were combined and treated with IN NaOH solution (to pH 13) and extracted with ethyl acetate (3 x 30 mL). The combined organic extracts were dried over MgSO 4 , filtered, and evaporated. The residue was purified by chromatography using a 4Og Redi-Sep column eluting with DCM (2 min), then a gradient of 8% MeOH/0.8%NH 4 θH/DCM over 15 min, then 8% MeOH/0.8%NH 4 θH/DCM for 2 min to give desired product ( 179.5 mg, 81 %) as a tan foam. MS mJz = 352.2 [M+H] + . Calc'd for C 22 H 30 N 3 O: 352.2.

Example 205 (also example 216 in Table 4)

N-((lS)-l-((lR)-2-(((4S)-6-(2^-dimethylpropyl)-8-(lH-imid azol-l-yl)-3,4- dihydrospirolchromene-l.l'-cyclobutanJ^y^amino^l-hydroxyethy ^-S-buten-l-yl)- 2-methoxyacetamide

To a microwave vial is added N-((lS)-l-((lR)-2-(((4S)-8-bromo-6-(2,2-dimethylpropyl)- 3 ,4-dihydrospiro[chromene-2, 1 '-cyclobutan]-4-yl)amino)- 1 -hydroxyethyl)-3-buten- 1 -yl)- 2-methoxyacetamide (100 mg, 191 μmol), cesium carbonate (311 mg, 955 μmol), IH- imidazole (65 mg, 955 μmol), and copper(I) iodide (36 mg, 191 μmol). The vial was purged with N 2 5 x, then 10:1 DMFZH 2 O (2 mL) and Nl,N2-dimethylethane-l,2-diamine (21 μl, 191 μmol) were added. The vial was sealed and heated in a microwave at 185 0 C for 30 min. The crude product was dissolved in DMF, filtered through an Acrodisc filter and purified by reverse phase HPLC to afford the title compound as a white amorphous solid. MS m/z = 511 A [M+H] + .

A-1291-WO-PCT - 138 -

Example 206 (also example 239 in table 4)

N-((lS)-l-((lR)-2-(((4'S)-6'-(2^-dimethylpropyl)-8'-(l-methy l-lH-imidazol-2-yl)- 3',4'-dihydrospiro[cyclobutane-l^'-pyrano[23-c]pyridin]-4 l -yl)ainino)-l- hydroxyethyI)-3-buten-l-yl)-2-methoxyacetamide

A solution of N-CClS^l-CClR^-C^'S^S'-chloro-o'^^-dimethylpropyO-S'^ 1 - dihydrospiro[cyclobutane-l,2'-pyrano[2,3-c]pyridin]-4'-yl)am ino)-l-hydroxyethyl)-3- buten-l-yl)-2-methoxyacetamide (225 mg, 469 μmol) and tetrakis(triphenylphosphine)palladium ( 108 mg, 93.7 μmol) in THF (3.0 mL) is added to a vial with l-methyl-2-(tributylstannyl)-lH-imidazole (217 mg, 586 μmol). The reaction was heated in the microwave at 160 0 C for 20 min. The residue was diluted with H 2 O (10 mL) and extracted with CH 2 Cl 2 (3 x 10 ml). The organic layers were dried (MgSO 4 ), concentrated and purified by reverse phase HPLC to give the title compound as a white amorphous solid. MS m/z = 526.3 [M+H] + .

Example 207

Step 1:

To a 2.0 L round bottom flask containing vinylmagnesium bromide (92 ml, 92 mmol) was added THF (40 mL) and the mixture was allowed to stir at - 78 0 C for 5 min. At this time, TMEDA (23 ml, 154 mmol) was added via syringe followed by (S, Z)-N-((S)-2,3- bis(tert-butyldimethylsilyloxy)propylidene)-2-methylpropane- 2-sulfinamide (13.0 g, 31 mmol) in THF. The reaction was allowed to stir at -78 0 C for 4 h and then allowed to slowly warm to RT overnight. The reaction was quenched by the addition of ammonium chloride (sat, 100 ml). The aqueous layer was extracted with EtOAc, washed with brine, dried sodium sulfate, filtered and concentrated to give a crude oil that was purified by

A-1291-WO-PCT - 139 - silica gel column to give (S)-N-((2S,3S)-l,2-bis(tert-butyldimethylsilyloxy)pent-4-en- 3- yl)-2-methylpropane-2-sulfinamide (5.45 g, 39% yield, higher Rf) and (S)-N-((2S,3R)- l,2-bis(tert-butyldimethylsilyloxy)pent-4-en-3-yl)-2-methylp ropane-2-sulfinamide (7.43 g, 54% yield, lower Rf). Step 2:

To a 1.0 L RBF containing (S)-N-((2S,3S)-l,2-bis(tert-butyldimethylsilyloxy)pent-4-en- 3-yl)-2-methylpropane-2-sulfinamide (5.45 g, 12 mmol) was added EtOH (75 mL ) and

- the mixture was allowed to stir at O 0 C for 10 min. HCl (4 N in dioxane) (15 ml, 61 mmol) was added and the reaction was allowed to stir for 5 h and then quenched by the addition of TEA (17 ml, 121 mmol) followed by DCM (15 ml) and BoC 2 O (5.3 g, 24 mmol). The reaction was allowed to stir overnight and then extracted with EtOAc. The crude oil was purified on a 330 g Isco chromatography column (20 to 35% EtOAc in hexanes) to give tert-butyl (2S,3S)-2-(tert-butyldimethylsilyloxy)-l-hydroxypent-4-en-3- ylcarbamate (3.80 g, 95% yield). Step 3:

To a 500 mL RBF containing tert-butyl (2S,3S)-l,2-bis(tert-butyldimethylsilyloxy)pent- 4-en-3 -ylcarbamate (4.0 g, 9.0 mmol) was added water (10 mL ) and tert-butanol (20 ml), and the mixture was allowed to stir at 23 0 C for 2 min. At this time, NMO (3.2 g, 27 mmol) and osmium tetraoxide (2.8 ml, 9.0 mmol) (2.5 % in butanol, 2 ml) was added and the reaction was allowed to stir overnight. At this time, sodium sulfite (11 g, 90 mmol) in water (75 ml) was added after the reaction was chilled to 0 0 C. EtOAc (100ml) was added and the quenched reaction was allowed to stir for 1 h. The aqueous layer was extracted with EtOAc (3 x 75 ml). The combined organics were washed with HCl (0.1

A-1291-WO-PCT - 140 -

M, 3 x 100 ml), sodium bicarbonate (1 x 150 ml) and brine. The organic layer was dried with magnesium sulfate, filtered and concentrated to give 4.05 g of a foamy off-white solid, rf = 0.45 in 40 % EtOAc in hexanes, not UV active, stains faint green when dilute and white when concentrated. Step 4:

To a 500 mL RBF containing tert-butyl (2R,3S,4S)-4,5-bis(tert-butyldimethylsilyloxy> l,2-dihydroxypentan-3-ylcarbamate (4.05 g, 8.4 mmol) was added water (10 mL ) and tert-butanol (30 ml) the mixture was allowed to stir at 23 0 C for 5 min. At this time, sodium periodiate (5.4 g, 25 mmol) was added in one portion. After 5 min, a white solid appeared and more water (20 ml) and butanol (30 ml) were added to the reaction. After 10 min, thin layer chromatography (TLC) indicated that all the diol starting material was consumed. The mixture was transferred to a separatory funnel with EtOAc (200 ml) and water (150 ml). An emulsion formed that was broken by adding hexane ( 100 ml). The organic layer was washed with water (3 x 100 ml) and brine. The organic layer was dried with magnesium sulfate, filtered and concentrated. Step 5:

To a 500 mL RBF containing crude tert-butyl (2R,3S)-3,4-bis(tert- butyldimethylsilyloxy)-l-oxobutan-2-ylcarbamate (3.85 g, 8.60 mmol) was added methanol (50 mL ) and the mixture was allowed to stir at 0 0 C for 10 min. At this time, sodium borohydride (0.325 g, 8.60 mmol) was added and the reaction was allowed to stir for 1 h and then allowed to warm to 23 0 C for 1 and then quenched with ammonium chloride (sat. 100 ml) and EtOAc (100 ml) and stirred for 1 h. The aqueous layer was

A-1291-WO-PCT - 141 - extracted with EtOAc (3 x 50 ml). The combined organics were washed with water (I x 150 ml) and brine. The solution was dried with sodium sulfate, filtered through a plug of silica gel and concentrated to give 4.20 g that was puriifed on a 120 g Isco chromatography column (10 to 25% EtOAc in hexanes, collect all) to give 3.80 g of a colorless oil. MS m/z 218.1 (M+l).

The following examples in Table 4 were prepared by methods and steps analogous to those described in Examples 4, 5, 201, 202, 205 and 206 herein.

Table 4

A-1291-WO-PCT -142-

A- 1291 -WO-PCT -143-

A-1291 -WO-PCT -144-

A-1291-WO-PCT -145-

A-1291 -WO-PCT - 146 -

The present invention also provides methods for making compounds of Formulas I-π. In another embodiment of the invention, there is provided a method of making a compound of Formula I or π, the method comprising the step of reacting a compound 20

20-A ,wherein A 1 , A 2 , A 3 , A 4 , R 2 , R 4 , R 5 , X and Z are as

defined herein, with a compound having the structure , wherein R la ,

R lb , R lc and W are as defined herein and X is a leaving group, to make a compound of Formulas I or II.

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 of the invention, 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.

The protecting groups may already be present in precursors and should protect the functional groups concerned against unwanted secondary reactions, such as acylations, etherifϊcations, esterifϊcations, oxidations, solvolysis, and similar reactions. It is a characteristic of protecting groups that they readily lend themselves, i.e. without

A-1291-WO-PCT - 147 - undesired secondary reactions, to removal, typically accomplished by solvolysis, reduction, photolysis or other methods of removal such as by enzyme activity, under conditions analogous to physiological conditions. It should also be appreciated that the protecting groups should not be present in the end-products. The specialist knows, or can easily establish, which protecting groups are suitable with the reactions described herein. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the inhibitor 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).

Salts, including pharmaceutically acceptable salts, of a compound of the invention having a salt-forming group may be prepared in a conventional manner or manner known to persons skilled in the art. For example, acid addition salts of compounds of the invention may be obtained by treatment with an acid or with a suitable anion exchange reagent. A salt with two acid molecules (for example a dihalogenide) may also be converted into a salt with one acid molecule per compound (for example a monohalogenide); this may be done by heating to a melt, or for example by heating as a solid under a high vacuum at elevated temperature, for example from 50 0 C to 170 0 C, one molecule of the acid being expelled per molecule of the compound.

Acid salts can usually be converted to free-base compounds, e.g. by treating the salt with suitable basic agents, for example with alkali metal carbonates, alkali metal hydrogen carbonates, or alkali metal hydroxides, typically potassium carbonate or sodium hydroxide. Exemplary salt forms and their preparation are described herein in the Definition section of the application.

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 or diluents. 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

A- 1291 -WO-PCT - 148 - starting materials and other reagents used. Solvents should be able to partially or wholly solubilize the reactants in the absence or presence of catalysts, condensbg 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 dependant 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 compounds of the invention include, without limitation, water; esters, including lower alkyl-lower alkanoates, e.g., EtOAc; ethers including aliphatic ethers, e.g., Et 2 O and ethylene glycol dimethylether or cyclic ethers, e.g., THF; liquid aromatic hydrocarbons, including benzene, toluene and xylene; alcohols, including MeOH, EtOH, 1-propanol, IPOH, n- and t-butanol; nitriles including CH 3 CN; halogenated hydrocarbons, including CH 2 Cl 2 , CHCI 3 and CCL»; acid amides including DMF; sulfoxides, including DMSO; bases, including heterocyclic nitrogen bases, e.g. pyridine; carboxylic acids, including lower alkanecarboxylic acids, e.g., AcOH; inorganic acids including HCl, HBr, HF, H 2 SO 4 and the like; carboxylic acid anhydrides, including lower alkane acid anhydrides, e.g., acetic anhydride; cyclic, linear, or branched hydrocarbons, including cyclohexane, hexane, pentane, isopentane and the like, and mixtures of these solvents, such as purely organic solvent combinations, or water-containing solvent combinations e.g., 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 (liquid and gas phase, and the like), extraction, distillation, trituration, reverse phase HPLC and the like. 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 invention 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

A-1291-WO-PCT - 149 - forming starting materials under the reaction conditions are all "intermediates" included in the invention. Further, structures produced by using starting materials in the form of a reactive derivative or salt, or produced by a compound obtainable by means of the process according to the invention and structures resulting from processing the compounds of the invention in situ are also within the scope of the invention.

New starting materials and/or intermediates, as well as processes for the preparation thereof, are likewise the subject of this invention. In select embodiments, such starting materials are used and reaction conditions so selected as to obtain the desired compound(s). Starting materials of the invention, 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.

Compounds of the present invention can possess, in general, one or more asymmetric carbon atoms and are thus capable of existing in the form of optical isomers as well as in the form of racemic or non-racemic mixtures thereof. The optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, e.g., by formation of diastereoisomeric salts, by treatment with an optically active acid or base. Examples of appropriate acids are tartaric, diacetyltartaric, dibenzoyltartaric, ditoluoyltartaric, and camphorsulfonic acid and then separation of the mixture of diastereoisomers by crystallization followed by liberation of the optically active bases from these salts. A different process for separation of optical isomers involves the use of a chiral chromatography column optimally chosen to maximize the separation of the enantiomers. Still another available method involves synthesis of covalent diastereoisomeric molecules by reacting compounds of the invention with an optically pure acid in an activated form or an optically pure isocyanate. The synthesized diastereoisomers can be separated by conventional means such as chromatography, distillation, crystallization or sublimation, and then hydrolyzed to deliver the enantiomerically pure compound. The optically active compounds of the invention can likewise be obtained by using optically active starting materials. These isomers may be in the form of a free acid, a free base, an ester or a salt. All such isomeric forms of such compounds are expressly included in the present invention.

A-1291-WO-PCT - 150 -

The compounds of this invention may also be represented in multiple tautomeric forms. The compounds may also occur in cis- or trans- or E- or Z- double bond isomeric forms. The invention expressly includes all tautomeric forms of the compounds described herein. All crystal forms of the compounds described herein are expressly included in the present invention.

Substituents on ring moieties (e.g., phenyl, thienyl, etc.) may be attached to specific atoms, whereby they are intended to be fixed to that atom, or they may be drawn unattached to a specific atom, whereby they are intended to be attached at any available atom that is not already substituted by an atom other than H (hydrogen). For example, the R 12 substituent is drawn unattached to any specific atom of ring Z 2 , and therefore each of the n number of R 12 substituents may be attached to any atom of Z 2 .

The compounds of the invention may be modified by appending appropriate functionalities to enhance selective biological properties. Such modifications are known in the art and include those which increase biological penetration into a given biological compartment (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion. By way of example, a compound of the invention may be modified to incorporate a hydrophobic group or "greasy" moiety in an attempt to enhance the passage of the compound through a hydrophobic membrane, such as a cell wall.

Although the pharmacological properties of the compounds of the invention (Formulas I-II) vary with structural change, in general, activity possessed by compounds of Formulas I and π may be demonstrated both in vitro as well as in vivo. Particularly, the pharmacological properties of the compounds of this invention may be confirmed by a number of pharmacological in vitro assays. The following exemplified pharmacological assays have been carried out with the compounds according to the invention. Compounds of the invention were found to modulate BACE activity.

BIOLOGICAL EVALUATION The compounds of the invention may be modified by appending appropriate functionalities to enhance selective biological properties. Surprisingly, the compounds of the present invention exhibit improved pharmacokinetics and pharmacodynamics, which relate, directly and indirectly, to the ability of the compound to be effective for its intended use. For example, the compounds have been surprisingly found to possess

A-1291-WO-PCT - 151 - improved clearance and efflux properties, which readily lend themselves to projecting in- vivo PK and PD properties, which in turn assist in projection of therapeutic target coverage for the compounds and projected efficacious dosages via in-vivo absorption, distribution, metabolism and excretion properties. Increased.biological penetration into a given biological compartment (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection and alter clearance, metabolism and/or rate of excretion are important factors for discovering which compound may be a useful drug and which may not.

Although the pharmacological properties of the compounds of the invention (Formulas I-II) vary with structural change, in general, activity possessed by compounds of Formulas I-II may be demonstrated both in vitro as well as in vivo. The following exemplified pharmacological assays have been carried out with the compounds according to the invention, to assess and characterize the compound's ability to modulate BACE activity and to regulate the cleavage of amyloid beta precursor protein, thereby reducing or inhibiting the production of amyloid beta.

In Vitro Enzymatic BACE FRET (fluorescence resonance energy transfer) Assay (Enzyme Assay data in Table 1)

The assay buffer used in this screen is 0.05 M acetate, pH 4.2, 10% DMSO final, 100 uM genapol (which is a nonionic detergent, below its Critical Micelle Concentration). The Beta Secretase enzyme (0.2nM) is pre-incubated for one hour with inhibitors, typically in about IuL of DMSO according to a serial dilution, are added thereto. The assay is effectively started by the addition of FRET substrate (5OnM) and the combination is incubated for one hour. The FRET assay is terminated with by addition of Tris buffer, which raises the pH to neutrality, and the fluorescence is determined. The FRET substrate is a peptide with commercially available fluorophore and quencher, on opposite sides of the BACE cleavage site. Proteolytic cleavage of the FRET substrate releases quenching of fluorescence (excitation 488 nm and emission 425 run).

Of the compounds tested, the in-vitro BACE FRET enzyme assay data for each of Examples 33-156 is provided in Table 1 and Examples 208-246 are provided in Table 4. The vast majority of those Examples exhibited activities with IC 5O values of 5 μM or less in the in-vitro BACE FRET enzyme assay, a majority of those same Examples exhibited activities with IC 50 values of 1 μM or less in the in-vitro BACE FRET enzyme assay, and

A-1291-WO-PCT - 152 - a majority of those same Examples exhibited activities with IC 50 values of 100 nM or less in the in-vitro BACE FRET enzyme assay.

In Vitro BACE cell-based assay: The cell-based assay measures inhibition or reduction of Aβ40 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 4OK cells/well in 96 well plates (Costar). The cells were cultivated for 24 hours at 37 0 C and 5% CO 2 in DMEM supplemented with 10% FBS. The test compounds were then added to cells in 10-point dose response concentrations with the starting concentration being either 100 μM or 10 μM. The compounds were diluted from stock solutions in DMSO and the final DMSO concentration of the test compounds on cells was 0.1%. After 24 h of incubation with the test compounds the supernatant conditioned media was collected and the Aβ 40 levels were determined using a sandwich ELISA. The IC 50 of the compound was calculated from the percent of control or percent inhibition of Aβ 40 as a function of the concentration of the test compound.

The sandwich ELISA to detect Aβ 40 was performed in 96 well microtiter plates, which were pre-treated with goat anti-rabbit IgG (Pierce). The capture and detecting antibody pair that were used to detect Aβ 40 from cell supernatants were affinity purified pAMO (Biosource) and biotinylated 6E10 (Signet Labs Inc.), respectively. The optimal concentration for the pAb40 antibody was 3 μg/ml in Superblock/TBS (Pierce) that was supplemented with 0.05%Tween 20 (Sigma). Optimal concentration for the detection antibody 6E10-biotinylated was 0.5 μg/ml in Superblock/TBS (Pierce) that had been supplemented with 2% normal goat serum and 2 % normal mouse serum. Cellular supernatants were incubated with the capture antibody for 3 h at 4 0 C, followed by 3 wash steps in TBS-tween (0.05%). The detecting antibody incubation was for 2 h at 4 0 C, again followed by the wash steps as described previously. The final readout of the ELISA is Time-Resolved Fluorescence (counts per minute) using Delfϊa reagents Streptavidin-Europium and Enhancement solutions (Perkin Elmer) and the Victor 2 multilabel counter (Perkin Elmer).

Of the compounds tested, in-the cell-based assay, data for each of Examples 33- 156 is provided in Table 1 and Examples 208-246 are provided in Table 4. The majority of those Examples exhibited activities with IC 5 Q values of 5 μM or less in the cell-based

A-1291-WO-PCT - 153 - assay, a majority of those same Examples exhibited activities with IC 50 values of 1 μM or less in the cell-based assay, and many of those same Examples exhibited activities with IC 50 values of 100 nM or less in the cell-based assay. In vivo Inhibition of Beta-Secretase Several animal models, including mouse, rat, dog, and monkey, may be used to screen for inhibition of beta-secretase activity in vivo following administration of a test compound sample. Animals used in this invention can be wild type, transgenic, or gene knockout animals. For example, the Tg2576 mouse model, prepared and conducted as described in Hsiao et al., 1996, Science 274, 99-102, and other non-transgenic or gene knockout animals are useful to analyze in vivo inhibition of Amyloid beta peptide (Abeta) production in the presence of inhibitory 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 A-beta levels and drug or test compound concentrations (Dovey et al., 2001, Journal of Neiirochemistry, 76,173- 181) 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., 2005, Journal of Pharmacology and Experimental Therapeutics, 313, 902-908), 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 Abeta peptide by specific sandwich ELISA assays based on ECL (Electrochemiluminescence) technology. For example, rat Abeta40 is measured using biotinylated-4G8 (Signet) as a capture antibody and Fab40 (an in-house antibody specific to the C-terminal of Abeta40) 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 20Og male Sprague Dawley rats, Abeta peptide levels are measured for reduction by X%

A-1291-WO-PCT - 154 - and Y% in cerebrospinal fluid and brain, respectively, when compared to the levels measured in the vehicle-treated or contol mice.

Actual vehicles used: Oral: 2% HPMC, 1% Tween80, pH2.2

IV: 5%EtOH, 45%Propylene glycol in 5% Dextrose The compounds of the invention can be shown to possess in-vivo activity by reducing the production or levels of Abeta peptide in the CSF and/or brain of a rat, when orally dosed at about 30mg/kg. Componds of Examples 44, 47, 87-91 and 107 exhibited a 5% or greater reduction of Abeta peptide in the CSF of the rat. Examples 44, 47 and 87- 91 exhibited a 10% or greater reduction of Abeta peptide in the CSF of the rat. Example 47 exhibited a 50% or greater reduction of Abeta peptide in the CSF of the rat. Example 47 exhibited about a 25% reduction of Abeta peptide in the brain of the rat.

CYP Inhibition Assay:

The CYP enxymes in the body are known to function in the metabolic pathway of a given compound. More specifically, CYP enzymes are responsible for metabolic breakdown of compounds. Thus, modulating the activity of one or more of the various CYP enzymes may influence potential metabolism of an administered compound. Particularly, if the compounds of the present invention were to inhibit the CYP enzyme, they may, thereby, reduce the rate of potential in-vivo metabolism of the compound, thus possibly prolonging the bioavailability of that compound. The compounds of the invention were run in the following CYP assays to determine their potential to inhibit specific CYP enzymes.

CYP3A

Pooled human liver microsomes (0.1 mg/mL) are incubated at about 37°C in a phosphate buffer (pH 7.4) with the selective 3A substrate midazolam at a concentration of about 2.5 μM in the presence and absence of a test compound (at about 3 μM cone). The reaction is started with the addition of NADPH (1 mM final concentration). Incubations are stopped after 10 minutes with the addition of organic solvent and 1- hydroxymidazolam metabolite formation is measured by an HPLC MS detection method. The ability of the test compound to inhibit the activity of CYP3A is determined (either % inhibition or an IC 50 can be measured in uM)by the ratio of the amount of metabolite in the presence of the test compound to the amount of metabolite in the absence of test compound. Data for various compounds of the invention in this assay is provided in Table 3.

A-1291-WO-PCT - 155 -

CYP2D6

Pooled human liver microsomes (0.25 mg/mL) are incubated at about 37 0 C in a phosphate buffer (pH 7.4) with the selective 2D6 substrate bufuralol at a concentration of about 5 μM in the presence and absence of a test compound (at about 3 μM cone). The reaction is started with the addition of NADPH (1 mM final concentration). Incubations are stopped after 10 minutes with the addition of organic solvent and 1-hydroxybufuralol metabolite formation is measured by an HPLC MS detection method. The ability of the test compound to inhibit the activity of CYP2D6 is determined (either % inhibition or an IC 5O can be measured in uM) by the ratio of the amount of metabolite in the presence of test compound to the amount of metabolite in the absence of test compound. Data for various compounds of the invention in this assay is provided in Table 3.

Microsomal Stability Assay

The purpose of this assay is to determine to what extent a compound may survive metabolic forces and to help ascertain the degree, time and extent of metabolism of a given compound. Such data is useful for projecting a given compound's ability to remain the plasma and potentially reach a desired target.

Assay: Pooled human or rat liver microsomes (0.25 mg/mL) are incubated at about 37°C in a phosphate buffer (pH 7.4) with a test compound (at a concentration of about 1 μM). The reaction is started with the addition of NADPH (1 mM final concentration). Incubations are stopped after 0 or 30 minutes with the addition of organic solvent. Quenched samples are analyzed for unchanged test compound by reverse phase HPLC with tandem mass spectrometric detection. % Turnover is determined by the ratio of the amount (peak area) of unchanged test compound remaining in incubated samples to the amount of unchanged test compound in non-incubated samples (0 minutes). Intrinsic clearance is estimated assuming first order elimination of compound from the incubation over the 30 minute incubation. Data for various compounds of the invention in this assay (human and rat) is provided in Table 2.

INDICATIONS

Accordingly, compounds of the invention are useful for, but not limited to, the prevention or treatment of beta-secretase related diseases, including Alzheimer's disease. The compounds of the invention have the ability to modulate the activity of beta secretase enzyme, thereby regulating the production of amyloid beta (Abeta peptide) and reducing

A-1291-WO-PCT - 156 - the formation and deposition of Abeta peptide and/or plaque on the brain. In one embodiment of the invention, there is provided a method of treating a disorder related to a beta-secretase enzyme in a subject, the method comprising administering to the subject an effective dosage amount of a compound of Formulas I - II. In another embodiment, there is provided a method of reducing production of amyloid beta, and of reducing plaque formation. In another embodiment, there is provided a method for the treatment, prevention or amelioration of a disease or disorder characterized by the elevated beta- amyloid deposits or beta-amyloid levels in a subject, the method comprising administering to the subject a therapeutically effective amount of a compound according to any of Formulas I, II, II- A, II-B, II-C and II-D. In yet another embodiment, the invention provides a method of treating Alzheimer's disease, cognitive impairment including mild, moderate and/or severe, Down's Syndrome, cognitive decline, senile dementia, cerebral amyloid angiopathy or a neurodegenerative disorder.

Accordingly, the compounds of the invention would be useful in therapy as CNS agents in treating neurological disorders and related conditions.

Besides being useful for human treatment, these compounds are 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 by the invention.

FORMTJLATIONS AND METHOD OF USE

Treatment of diseases and disorders herein is intended to also include therapeutic administration of a compound of the invention, or a pharmaceutical salt thereof, or a pharmaceutical composition of either to a subject (i.e., an animal, preferably a mammal, most preferably a human) which may be in need of preventative treatment, such as, for example, for pain, inflammation and the like. Treatment also encompasses prophylactic administration of a compound of the invention, or a pharmaceutical salt thereof, or a pharmaceutical composition of either to a subject (i.e., an animal, preferably a mammal, most preferably a human). Generally, the subject is initially diagnosed by a licensed physician and/or authorized medical practitioner, and a regimen for prophylactic and/or therapeutic treatment via administration of the compound(s) or compositions of the invention is suggested, recommended or prescribed.

The amount of compound(s) which is/are administered and the dosage regimen for treating neurological disorders and beta-secretase mediated diseases with the

A-1291-WO-PCT - 157 - compounds and/or compositions of this invention 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. Thus, the dosage regimen may vary widely, but can be determined routinely using standard methods. A daily dose of about 0.01 to 500 mg/kg, advantageously between about 0.01 and about 50 mg/kg, more advantageously about 0.01 and about 30 mg/kg, and even more advantageously between about 0.1 and about 10 mg/kg body weight may be appropriate, and should be useful for all methods of use disclosed herein. The daily dose can be administered in one to four doses per day. While it may be possible to administer a compound of the invention alone, in the methods described, the compound administered normally will be present as an active ingredient in a pharmaceutical composition. Thus, in another embodiment of the invention, there is provided a pharmaceutical composition comprising a compound of this invention in combination with a pharmaceutically acceptable carrier, which includes diluents, excipients, adjuvants and the like (collectively referred to herein as "carrier" materials) as described herein, and, if desired, other active ingredients. A pharmaceutical composition of the invention may comprise an effective amount of a compound of the invention or an effective dosage amount of a compound of the invention. An effective dosage amount of a compound of the invention includes an amount less than, equal to or greater than an effective amount of the compound. For example, a pharmaceutical composition in which two or more unit dosages, such as in tablets, capsules and the like, are required to administer an effective amount of the compound, or alternatively, a multi- dose pharmaceutical composition, such as powders, liquids and the like, in which an effective amount of the compound is administered by administering a portion of the composition.

The compound(s) of the present invention may be administered by any suitable route, preferably 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 of the present invention 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 infusion techniques, in dosage unit formulations containing conventional pharmaceutically acceptable carriers, adjuvants, and vehicles.

A-1291-WO-PCT - 158 -

1 For oral administration, the pharmaceutical composition may be in the form of, 1 for example, a tablet, capsule, suspension or liquid. The pharmaceutical composition is preferably 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, advantageously from about 1 to 500 mg, and typically from about 5 to 150 mg. A suitable daily dose for a human or other mammal may vary widely depending on the condition of the patient and other factors, but, once again, can be determined using routine methods and practices. For therapeutic purposes, the active compounds of this invention are ordinarily combined with one or more adjuvants or "excipients" appropriate to the indicated route of administration. If orally administered on a per dose basis, the compounds 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 adjuvants 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 carriers including saline, dextrose, or water, or with cyclodextrin (ie. Captisol), cosolvent solubilization (ie. propylene glycol) or micellar solubilization (ie. Tween 80).

A-1291-WO-PCT - 159 -

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 preferably 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 adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers, buffers etc. Tablets and pills can additionally be prepared with enteric coatings. Such compositions may also comprise adjuvants, such as wetting, sweetening, flavoring, and perfuming agents.

Accordingly, in yet another embodiment of the present invention, there is provided a method of manufacturing a medicament, the method comprising combining an amount of a compound according to Formulas I or II with a pharmaceutically acceptable carrier to manufacture the medicament. In yet another embodiment, the invention provides a method of manufacturing a medicament for the treatment of Alzheimer's disease, the method comprising combining an amount of a compound according to Formulas I or II with a pharmaceutically acceptable carrier to manufacture the medicament.

COMBINATIONS

While the compounds of the invention can be dosed or administered as the sole active pharmaceutical agent, they can also be used in combination with one or more compounds of the invention or in conjunction with other agents. When administered as a combination, the therapeutic agents can be formulated as separate compositions that are

A-1291-WO-PCT - 160 -

' 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 of the present invention 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 coadministration 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 of the present invention may be in conjunction with additional therapies known to those skilled in the art in the prevention or treatment of beta-secretase, gamma-secretase and/or other reagents known in influence the formation and/or deposition of amyloid beta, otherwise responsible for the formation of plaque on the brain. If formulated as a- fixed dose, such combination products employ the compounds of this invention within the accepted dosage ranges. Compounds of Formulas I and EI may also be administered sequentially with known anti-inflammatory agents when a combination formulation is inappropriate. The invention is not limited in the sequence of administration; compounds of the invention may be administered either prior to, simultaneous with or after administration of the known anti-inflammatory agent.

The foregoing description is merely illustrative of the invention and is not intended to limit the invention to the disclosed 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 patents and other publications recited herein are hereby incorporated by reference in their entireties.