Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
AMINOCYCLOBUTANES AS MONOACYLGLYCEROL LIPASE MODULATORS
Document Type and Number:
WIPO Patent Application WO/2021/191391
Kind Code:
A1
Abstract:
Aminocyclobutane compounds of Formula (I), and pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers thereof, pharmaceutical compositions containing them, and methods of using them including methods for treating disease states, disorders, and conditions associated with MGL modulation, such as those associated with pain, psychiatric disorders, neurological disorders (including, but not limited to major depressive disorder, treatment resistant depression, anxious depression, autism spectrum disorders, Asperger syndrome, bipolar disorder), cancers and eye conditions: wherein X, R1, R2a, R2b, R3 and R4 are defined herein.

Inventors:
AMERIKS MICHAEL K (US)
GARCIA-REYNAGA PABLO (US)
Application Number:
PCT/EP2021/057838
Publication Date:
September 30, 2021
Filing Date:
March 25, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
JANSSEN PHARMACEUTICA NV (BE)
International Classes:
C07D263/52; A61K31/423; A61P25/00; A61P27/02; A61P35/00
Foreign References:
US20170283406A12017-10-05
Other References:
MATSUDA ET AL., NATURE, vol. 346, 1990, pages 561 - 4
MUNRO ET AL., NATURE, vol. 365, 1993, pages 61 - 5
HERKENAM ET AL., PROC. NAT. ACAD. SCI., vol. 87, no. 5, 1990, pages 1932 - 1936
DI MARZO ET AL., CURR OPIN LIPIDOL, vol. 18, 2007, pages 129 - 140
PACHER ET AL., AMER J PHYSIOL, vol. 294, 2008, pages HI 133 - H1134
BENITO ET AL., BRIT J PHARMACOL, vol. 153, 2008, pages 277 - 285
CAVUOTO ET AL., BIOCHEM BIOPHYS RES COMMUN, vol. 364, 2007, pages 105 - 110
HAJRASOULIHA ET AL., EUR J PHARMACOL, vol. 579, 2008, pages 246 - 252
DEVANE ET AL., SCIENCE, vol. 258, 1992, pages 1946 - 9
MECHOULAM ET AL., BIOCHEM PHARMACOL, vol. 50, 1995, pages 83 - 90
SUGIURA ET AL., BIOCHEM BIOPHYS RES COMMUN, vol. 215, 1995, pages 89 - 97
BUCZYNSKIPARSONS, BRIT J PHARMACOL, vol. 160, no. 3, 2010, pages 423 - 42
SUGIURA ET AL., PROSTAGLANDINS LEUKOT ESSENT FATTY ACIDS, vol. 66, no. 2-3, 2002, pages 173 - 92
SUGUIRA ET AL., PROG LIPID RES, vol. 45, no. 5, 2006, pages 405 - 46
AHN ET AL., CHEM REV., vol. 108, no. 5, 2008, pages 1687 - 707
LONG ET AL., NAT CHEM BIOL., vol. 5, January 2009 (2009-01-01), pages 37 - 44
SCHLOSBURG ET AL., NAT NEUROSCI, vol. 13, no. 9, September 2010 (2010-09-01), pages 1113 - 9
LONG ET AL., CHEM BIOL., vol. 16, no. 7, 31 July 2009 (2009-07-31), pages 744 - 53
PIOMELLI, NAT REV NEUROSCI, vol. 4, 2003, pages 873 - 884
DINH ET AL., PROC NATL ACAD SCI USA., vol. 99, no. 16, 6 August 2002 (2002-08-06), pages 10819 - 24
STRAIKER ET AL., MOL PHARMACOL., vol. 76, no. 6, December 2009 (2009-12-01), pages 1220 - 7
WALTER ET AL., J NEUROSCI, vol. 24, no. 37, 15 September 2004 (2004-09-15), pages 8068 - 74
LONG ET AL., NAT CHEM BIOL, vol. 5, no. 1, January 2009 (2009-01-01), pages 37 - 44
MULVIHILL ET AL., LIFE SCI, vol. 92, no. 8-9, 19 March 2013 (2013-03-19), pages 492 - 505
BEDSE ET AL., BIOL PSYCHIATRY, vol. 82, no. 7, 1 October 2017 (2017-10-01), pages 488 - 499
BERNAL-CHICO ET AL., GLIA, vol. 3, no. 1, 6 January 2015 (2015-01-06), pages 163 - 76
PATEL ET AL., NEUROSCI BIOBEHAV REV, vol. 6, 7 May 2017 (2017-05-07), pages 56 - 66
BETSE ET AL., TRANSL PSYCHIATRY, vol. 8, no. 1, 26 April 2018 (2018-04-26), pages 92
NOMURA ET AL., SCIENCE, vol. 334, no. 6057, 11 November 2011 (2011-11-11), pages 809 - 13
KATZ ET AL., J NEUROTRAUMA, vol. 32, no. 5, 1 March 2015 (2015-03-01), pages 297 - 306
ZHANG ET AL., J CEREB BLOOD FLOW METAB, vol. 35, no. 4, 31 March 2015 (2015-03-31), pages 443 - 453
PIRO ET AL., CELL REP, vol. 1, no. 6, 28 June 2012 (2012-06-28), pages 617 - 23
WENZEL ET AL., LIFE SCI, vol. 207, 15 August 2018 (2018-08-15), pages 314 - 322
CHEN ET AL., CELL REP, vol. 2, no. 5, 29 November 2012 (2012-11-29), pages 1329 - 39
PASQUARELLI ET AL., NEUROCHEM INT, vol. 0, 11 November 2017 (2017-11-11), pages 14 - 24
PASQUARELLI ET AL., NEUROPHARMACOLOGY, vol. 124, 15 September 2017 (2017-09-15), pages 157 - 169
HERNADEZ-TORRES ET AL., ANGEW CHEM INT ED ENGL, vol. 53, no. 50, 8 December 2014 (2014-12-08), pages 13765 - 70
COVEY ET AL., NEUROPSYCHOPHARMACOLOGY, vol. 43, 2018, pages 2056 - 2063
TERRONE ET AL., EPILEPSIA, vol. 9, no. 1, 5 January 2018 (2018-01-05), pages 79 - 91
VON RUDEN ET AL., NEUROBIOL DIS, vol. 77, May 2015 (2015-05-01), pages 238 - 45
TUO ET AL., J MED CHEM., vol. 60, no. 1, 12 January 2017 (2017-01-12), pages 4 - 46
HILL ET AL., PHARMACOPSYCHIATRY, vol. 41, no. 2, March 2008 (2008-03-01), pages 48 - 53
HILL ET AL., PSYCHONEUROENDOCRINOLOGY, vol. 34, no. 8, September 2009 (2009-09-01), pages 1257 - 1262
HAUER ET AL., REV NEUROSCI, vol. 23, no. 5-6, 2012, pages 681 - 90
HILL ET AL., PSYCHONEUROENDOCRINOLOGY, vol. 38, no. 12, 2013, pages 2952 - 2961
YI ET AL., PROGRESS IN NEURO-PSYCHOPHARMACOLOGY AND BIOLOGICAL PSYCHIATRY, vol. 67, no. 3, 2016, pages 92 - 97
CHRISTENSEN ET AL., THE LANCET, vol. 370, 2007, pages 1706 - 1713
FOLKES ET AL., J CLIN INVEST, vol. 130, no. 4, 2020, pages 1728 - 1742
JUNG ET AL., NATURE COMMUNICATIONS, vol. 3, no. 10, 2012, pages 80
WANG ET AL., MOL PSYCHIATRY, vol. 8, 23 August 2018 (2018-08-23), pages 1798 - 1806
DI MARZO ET AL., ANNU REV MED, vol. 57, 2006, pages 553 - 74
LIGRESTI ET AL., CURR OPIN CHEM BIOL., vol. 13, no. 3, June 2009 (2009-06-01), pages 321 - 31
GUINDON ET AL., BR J PHARMACOL, vol. 163, no. 7, August 2011 (2011-08-01), pages 1464 - 78
KINSEY ET AL., J PHARMACOL EXP THER, vol. 330, no. 3, September 2009 (2009-09-01), pages 902 - 10
WILKERSON ET AL., J PHARMACOL EXP THER, vol. 357, April 2016 (2016-04-01), pages 145 - 56
ALHOUAYEK ET AL., FASEB J, vol. 25, no. 8, August 2011 (2011-08-01), pages 2711 - 21
CURRY ET AL., JPHARMACOL EXP THER, vol. 366, July 2018 (2018-07-01), pages 169 - 18
NITHIPATIKOM ET AL., CANCER RES., vol. 64, no. 24, 15 December 2004 (2004-12-15), pages 8826 - 30
NITHIPATIKOM ET AL., BIOCHEM BIOPHYS RES COMMUN, vol. 332, no. 4, 15 July 2005 (2005-07-15), pages 1028 - 33
NITHIPATIKOM ET AL., PROSTAGLANDINS OTHER LIPIDMEDIAT, vol. 4, no. 1-2, 9 February 2011 (2011-02-09), pages 34 - 43
STICHT ET AL., BR J PHARMACOL, vol. 5, no. 8, 16 April 2012 (2012-04-16), pages 2425 - 35
RAMESH ET AL., JPHARMACOL EXP THER, vol. 39, no. 1, 3 October 2011 (2011-10-03), pages 173 - 85
MILLER ET AL., PHARMACEUTICALS, vol. 11, 2018, pages 50
S.M. BERGE ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19, XP002675560, DOI: 10.1002/jps.2600660104
"Handbook of Pharmaceutical Salts, Properties, Selection, and Use", 2002, WILEY-VCH
ANGEW.CHEM.INT.ED., vol. 47, 2008, pages 6802 - 6806
WILSON SJLOVENBERG TWBARBIER AJ, ANAL BIOCHEM, vol. 318, no. 2, 15 July 2003 (2003-07-15), pages 270 - 5
Attorney, Agent or Firm:
WARNER, James Alexander et al. (GB)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A compound of Formula (I): wherein

X is CH2 or O;

R1 is H;

R2a and R2b are each independently selected from H and Ci-4alkyl;

R3 is selected from:

(i) phenyl, benzyl, or monocyclic heteroaryl, each optionally substituted with one, two, or three substituents selected from: halo, Ci-6alkyl, Ci-6haloalkyl, Ci-6alkyl-OH, OCi-6alkyl, OCi-6haloalkyl, SCi-6alkyl, SFs, Si(CH3)3, NRaRb, C3-6cycloalkyl, OC3-6cycloalkyl, phenyl, O-phenyl, and O-pyridyl, wherein each cycloalkyl, phenyl, or pyridyl is optionally substituted with one or two Ci-4alkyl, Ci-4haloalkyl, or halo groups; or two adjacent ring substituents on the phenyl, benzyl, or monocyclic heteroaryl, taken together with the atoms to which they are attached form a fused monocyclic C5-6cycloalkyl or heterocycloalkyl ring, each ring optionally substituted with one or two Ci-4alkyl, Ci- 4haloalkyl, or halo groups; wherein Ra and Rb are each independently H or Ci-4alkyl;

(ii) a bicyclic heteroaryl optionally substituted with Ci-4alkyl or halo; and

(iii) C3-6alkyl or C3-6cycloalkyl optionally substituted with Ci-4alkyl, Ci-4haloalkyl, or halo;

R4 is Ci-6alkyl; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.

2. A compound of Formula (I): wherein

X is CH 2 or O;

R1 is H;

R2a and R2b are each H;

R3 is selected from: Ci-6alkyl (optionally C3-6alkyl); C3-6cycloalkyl; benzyl; phenyl; phenyl substituted with one or two members each independently selected from: halo, Ci-6alkyl, OCi-6alkyl, C3-6cycloalkyl, and C3-6cycloalkyl substituted with CH3 or CF3; 2, 3 -dihydro- 1H- inden-5-yl; bicyclo[4.2.0]octa-l(6),2,4-trien-3-yl; and 5,6,7,8-tetrahydronaphthalen-2-yl; and

R4 is Ci-6alkyl; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.

3. The compound as claimed in claim 1 or claim 2, wherein X is CFh.

4. The compound as claimed in claim 1 or claim 2, wherein X is O.

5. The compound as claimed in any one of claims 1 to 4, wherein R3 is tert-butyl.

6 The compound as claimed in any one of claims 1 to 4, wherein R3 is

7. The compound as claimed in any one of claims 1 to 4, wherein R3 is

8 The compound as claimed in any one of claims 1 to 4, wherein R3 is benzyl, phenyl, or phenyl substituted with one or two members each independently selected from: F, CFb, CFhCFb, CH(CH3)2, C(CF13)3, OCH3, cyclopropyl, cyclopropyl substituted with CH3 or CF3, and cyclobutyl.

10. The compound as claimed in any one of claims 1 to 4, wherein R3 is 3,5-dimethylphenyl, 3- ethyl-5-methylphenyl, 4-ethyl-3-methylphenyl, 3-isopropylphenyl, or 3-tert-butylphenyl. 11. The compound as claimed in claim 1 or claim 2, wherein X is O, and R2a and R2b are each

H.

12. The compound as claimed in claim 1 or claim 2, wherein X is CFh, and R2a and R2b are each H.

13. The compound as claimed in claim 1 or claim 2, wherein X is O and R3 is phenyl substituted with one or two members each independently selected from: F, Ci-6alkyl, OCFb, cyclopropyl, cyclopropyl substituted with CFb or CF3, and cyclobutyl.

14. The compound as claimed in any one of claims 1 to 13, wherein R4 is CFF or CH2CH3.

15. A compound selected from:

(2s,4S)-N-Methyl-6-oxo-N-((ls,3S)-3-phenylcyclobutyl)-7-oxa-5-azaspiro[3.4]octane-2- carboxamide;

(2s,4S)-N-Methyl-6-oxo-N-((lr,3R)-3-phenylcyclobutyl)-7-oxa-5-azaspiro[3.4]octane-2- carboxamide; (2s,4S)-N-((ls,3S)-3-(3-Cyclobutylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(4-(tert-Butyl)phenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((lr,3R)-3-(4-(tert-Butyl)phenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(3-(tert-Butyl)phenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2r,4S)-N-((ls,3S)-3-(3-(tert-Butyl)phenyl)cyclobutyl)-N-methyl-6-oxo-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(3,5-Dimethylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((lr,3R)-3-(3,5-Dimethylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(3-Isopropylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(3-Methoxyphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((lr,3R)-3-(3-Methoxyphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(4-Isopropylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(3-(tert-Butyl)-4-fluorophenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-Methyl-6-oxo-N-(( 1 s, 3 S)-3 -(3 -( 1 -

(trifluoromethyl)cyclopropyl)phenyl)cyclobutyl)-7-oxa-5-azaspiro[3.4]octane-2- carboxamide;

(2s,4S)-N-Methyl-6-oxo-N-(( 1 r, 3R)-3 -(3 -( 1 -

(trifluoromethyl)cyclopropyl)phenyl)cyclobutyl)-7-oxa-5-azaspiro[3.4]octane-2- carboxamide; (2s,4S)-N-Methyl-6-oxo-N-((ls,3S)-3-(o-tolyl)cyclobutyl)-7-oxa-5-azaspiro[3.4]octane-2- carboxamide;

(2s,4S)-N-Methyl-6-oxo-N-((lr,3R)-3-(o-tolyl)cyclobutyl)-7-oxa-5-azaspiro[3.4]octane-2- carboxamide;

(2s,4S)-N-Methyl-6-oxo-N-((ls,3S)-3-(m-tolyl)cyclobutyl)-7-oxa-5-azaspiro[3.4]octane-2- carboxamide;

(2s,4S)-N-Methyl-6-oxo-N-((lr,3R)-3-(m-tolyl)cyclobutyl)-7-oxa-5-azaspiro[3.4]octane-2- carboxamide;

(2s,4S)-N-((ls,3S)-3-(3-Ethylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(2,3-Dimethylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((lr,3R)-3-(2,3-Dimethylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(4-Cyclopropylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(3,4-Dimethylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(3-Fluoro-4-isopropylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(4-Fluoro-3-isopropylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(2,4-Dimethylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(4-Ethylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(4-Ethyl-3-methylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(4-Cyclopropyl-3-methylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide; (2s,4S)-N-((ls,3S)-3-(3-Cyclopropyl-4-methylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-Methyl-6-oxo-N-((ls,3S)-3-(5,6,7,8-tetrahydronaphthalen-2-yl)cyclobutyl)-7-oxa-

5-azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((lr,3S)-3-Benzylcyclobutyl)-N-methyl-6-oxo-7-oxa-5-azaspiro[3.4]octane-2- carboxamide;

(2s,4S)-N-((ls,3R)-3-Benzylcyclobutyl)-N-methyl-6-oxo-7-oxa-5-azaspiro[3.4]octane-2- carboxamide;

(2s,4S)-N-((ls,3S)-3-(tert-Butyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5-azaspiro[3.4]octane-

2-carboxamide;

(2s,4S)-N-((lr,3R)-3-(tert-Butyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5-azaspiro[3.4]octane-

2-carboxamide;

(2s,4S)-N-((ls,3S)-3-Cyclohexylcyclobutyl)-N-methyl-6-oxo-7-oxa-5-azaspiro[3.4]octane-2- carboxamide;

(2s,4S)-N-((lr,3R)-3-Cyclohexylcyclobutyl)-N-methyl-6-oxo-7-oxa-5-azaspiro[3.4]octane-2- carboxamide;

(2s,4S)-N-Methyl-N-((ls,3S)-3-(4-(l-methylcyclopropyl)phenyl)cyclobutyl)-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(4-(Sec-butyl)phenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-Methyl-N-((ls,3S)-3-(3-(l-methylcyclopropyl)phenyl)cyclobutyl)-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(3-Ethyl-5-methylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(3-Cyclopropyl-5-methylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(3-Isopropyl-5-methylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(3-Chloro-5-methylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide; (2s,4S)-N-((ls,3S)-3-(3-Cyclopropylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(2,3-Dihydro-lH-inden-5-yl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(Bicyclo[4.2.0]octa-l(6),2,4-trien-3-yl)cyclobutyl)-N-methyl-6-oxo-7- oxa-5-azaspiro[3 4]octane-2-carboxamide; and

(2s,4S)-N-((ls,3S)-3-(3,5-dimethylphenyl)cyclobutyl)-N-ethyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide; and pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers thereof.

16. A compound selected from:

(2r,4S)-N-((ls,3S)-3-(3-(/er/-Butyl)phenyl)cyclobutyl)-N-methyl-6-oxo-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(3,5-Dimethylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(3-Isopropylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(4-Isopropylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3 4]octane-2-carboxamide; and

(2s,4S)-N-((ls,3S)-3-(4-Ethylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide; and pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers thereof.

17. A pharmaceutical composition comprising:

(A) a therapeutically effective amount of at least one compound of any one of claims 1, 2, 15, or 16; and

(B) at least one pharmaceutically acceptable excipient.

18. The pharmaceutical composition of claim 17, wherein the compound is a compound as claimed in claim 15.

19. A method of treating a subject suffering from or diagnosed with a disease, disorder, or condition mediated by MGL receptor activity, comprising administering to a subject in need of such treatment a therapeutically effective amount of at least one compound as claimed in any one of claims 1, 2, 15, or 16.

20. The method of claim 19, wherein the MGL receptor mediated disease, disorder, or condition is selected from: pain, psychiatric conditions, neurological conditions, cancers, and eye conditions.

21. The method of claim 19, wherein the MGL receptor mediated disease, disorder or condition is selected from: major depressive disorder, treatment resistant depression, anxious depression, autism spectrum disorders, Asperger syndrome, and bipolar disorder.

22. The method of claim 19, wherein the MGL receptor mediated disease, disorder or condition is inflammatory pain.

Description:
AMINOCYCLOBUTANES AS MONOACYLGLYCEROL LIPASE MODULATORS

FIELD OF THE INVENTION

The present invention is related to certain aminocyclobutane chemical entities having MGL modulating properties, pharmaceutical compositions comprising these chemical entities, chemical processes for preparing these chemical entities and their use in the treatment of diseases, disorders or conditions associated with MGL receptor activity in subjects, in particular humans.

BACKGROUND OF THE INVENTION

Cannabis Sativa and analogs of A 9 -tetrahydrocannabinol have been used since the days of folk medicine for therapeutic purposes. The endocannabinoid system consists of two G-protein coupled receptors, cannabinoid receptor type 1 (CB1) (Matsuda et al ., Nature , 1990, 346, 561-4) and cannabinoid receptor type 2 (CB2) (Munro et al. , Nature , 1993, 365, 61-5). CB1 receptor is one of the most abundant G-protein coupled receptor expressed in the brain (Herkenam et al., Proc. Nat. Acad. Sci., 1990, 87 (5), 1932-1936). CB1 is also expressed peripherally in the liver, gastrointestinal tract, pancreas, adipose tissue, and skeletal muscles (Di Marzo etal. , Curr Opin Lipidol , 2007, 18, 129-140). CB2 is predominantly expressed in immune cells such as monocytes (Pacher et al., Amer J Physiol, 2008, 294, HI 133-H1134) and under certain conditions (inflammation) in the brain ((Benito et al. , Brit J Pharmacol, 2008, 153, 277-285) and in skeletal (Cavuoto et al, Biochem Biophys Res Commun, 2007, 364, 105-110) and cardiac muscles (Hajrasouliha etal, Eur J Pharmacol, 2008, 579, 246-252).

In 1992, N-arachidonoylethanolamine (AEA or anandamide) was found to be an endogenous ligand for cannabinoid receptors (Devane et al, Science, 1992, 258, 1946-9). Subsequently, 2-arachidonoylglycerol (2-AG) was also identified as an additional endogenous ligand for the cannabinoid receptors (Mechoulam et al, Biochem Pharmacol, 1995, 50, 83-90; Sugiura et al, Biochem Biophys Res Commun, 1995, 215, 89-97). Concentrations of 2-AG were reported to be at least 100 times higher than these of anandamide in the rat brain (Buczynski and Parsons, Brit J Pharmacol, 2010, 160 (3), 423-42). Therefore 2-AG may play more essential physiological roles than anandamide in the brain endocannabinoid system (Sugiura et al.

Prostaglandins Leukot Essent Fatty Acids., 2002, Feb-Mar, 66(2-3): 173-92). The endocannabinoid 2-AG is a full agonist for CB1 and CB2 receptors, while anandamide is a partial agonist for both receptors (Suguira et al, Prog Lipid Res , 2006, 45(5):405-46). Unlike many classical neurotransmitters, endocannabinoids signal through a retrograde mechanism. They are synthesized on demand in postsynaptic neurons and then rapidly degraded following binding to presynaptic cannabinoid receptors (Ahn etal, Chem Rev. 2008, 108(5): 1687-707). Monoacylglycerol lipase (MGLL, also known as MAG lipase and MGL) is the serine hydrolase responsible for the degradation of 2-AG into arachidonic acid and glycerol in the central nervous system (Mechoulam et al., Biochem Pharmacol , 1995, 50, 83-90; Sugiura et al., Biochem Biophys Res Commun , 1995, 215, 89-97; Long et al, Nat Chem Biol. 2009 Jan;5(l):37-44; ), Schlosburg etal, Nat Neurosci., 2010, Sep; 13(9): 1113-9) and peripheral tissues (Long et al, Chem Biol., 2009 Jul 31;16(7):744-53). Anandamide is hydrolyzed by fatty acid amide hydrolase (FAAH) (Piomelli, Nat Rev Neurosci, 2003, 4, 873-884). MGL exists in both soluble and membrane bound forms (Dinh et al, Proc Natl Acad Sci USA., 2002, Aug 6;99(16):10819- 24). In the brain MGL is located in presynaptic neurons (Straiker et al, Mol Pharmacol., 2009, Dec;76(6): 1220-7) and astrocytes (Walter et al, J Neurosci., 2004, Sep 15;24(37):8068-74) within regions associated with high CB1 receptor density. Compared to wild-type controls, genetic ablation of MGL expression produces 10-fold increase in brain 2-AG levels without affecting anandamide concentration (Schlosburg etal, Nat Neurosci., 2010, Sep;13(9): 1113-9).

Thus, MGL modulation offers an interesting strategy for potentiating the cannabinoid system. The primary advantage of this approach is that only brain regions where endocannabinoids are actively produced will be modulated, potentially minimizing the side effects associated with exogenous CB1 agonists. Pharmacological inactivation of MGL by covalent inhibitors in animals increase 2-AG content in brain and peripheral tissues and has been found to produce antinociceptive, anxiolytic and anti-inflammatory effects that are dependent on CB 1 and/or CB2 receptors (Long et al, Nat Chem Biol., 2009, Jan, 5(l):37-44; Ghosh et al, Life Sci., 2013, Mar 19, 92(8-9):498-505; Bedse etal, Biol Psychiatry., 2017, Oct 1, 82(7):488-499; Bemal-Chico etal, Glia., 2015, Jan, 63(1): 163-76; Patel etal. Neurosci Biobehav Rev., 2017, May, 76(Pt A):56-66; Betse et al, Transl Psychiatry., 2018, Apr 26, 8(1):92). In addition to the role of MGL in terminating 2-AG signaling, MGL modulation, including MGL inhibition also promotes CB 1/2-independent effects on neuroinflammation (Nomura etal, Science., 2011, Nov

11;334(6057):809-13). MGL modulation, including MGL inhibition leads to reduction in proinflammatory prostanoid signaling in animal models of traumatic brain injury (Katz et al, J Neurotrauma., 2015, Mar l;32(5):297-306; Zhang et al, J Cereb Blood Flow Metab., 2015, Mar 31 ;35(4): 443-453), neurodegeneration including Alzheimer’s disease (Piro et al, Cell Rep., 2012, Jun 28, l(6):617-23; Wenzel et al, Life Set, 2018, Aug 15, 207:314-322; Chen etal, Cell Rep., 2012, Nov 29, 2(5): 1329-39), Parkinson’s disease (Nomura et al, Science, 2011, Nov 11, 334(6057), 809-13; Pasquarelli etal, Neurochem Int., 2017, Nov, 110:14-24), amyotrophic lateral sclerosis (Pasquarelli etal, Neuropharmacology, 2017, Sep 15, 124:157-169), multiple sclerosis (Hernadez-Torres etal, Angew Chem Int Ed Engl., 2014, Dec 8, 53(50):13765-70; Bemal-Chico etal, Glia., 2015, Jan, 63(1): 163-76), Huntington’s disease (Covey etal, Neuropsychopharmacology , 2018, 43, 2056-2063), Tourette syndrome and status epilepticus (Terrone etal, Epilepsia., 2018, Jan, 59(1), 79-91; von Ruden etal, Neurobiol Dis., 2015, May;77:238-45).

Therefore, by potentiating the cannabinoid system and attenuating proinflammatory cascades, MGL modulation, including MGL inhibition offers a compelling therapeutic approach for the treatment of a vast array of complex diseases. Importantly, MGL modulation, including MGL inhibition in animals does not produces the full spectrum of neurobehavioral effects observed with A 9 -tetrahydrocannabinol and other CB1 agonists (Tuo et al, JMed Chem., 2017, Jan 12, 60(1), 4-46; Mulvihill etal, Life Set, 2013, Mar 19, 92(8-9), 492-7).

Endocannabinoid hypoactivity is a risk factor for the treatment of depression, anxiety and post-traumatic stress disorders. Millennia of human use of cannabis sativa, and a brief period in which humans were treated with the endocannabinoid antagonist, rimonabant, provide support for that hypothesis. 2-AG levels are decreased in individuals with major depression (Hill et al,

Pharmacopsychiatry., 2008, Mar; 41(2): 48-53; Hill etal, Psychoneuroendocrinology., 2009,

Sep; 34(8): 1257-1262.). Low circulating 2-AG levels predict rates of depression (Hauer et al,

Rev Neurosci., 2012, 23(5-6):681-90). Reduced circulating 2-AG has been found in patient with post-traumatic stress disorder (PTSD) (Hill etal, Psychoneuroendocrinology, 2013, 38 (12),

2952-2961). Healthy volunteers exposed to chronic stressors exhibited progressively diminished circulating 2-AG levels which correlated with the onset of reductions in measures of positive emotions (Yi et al, Progress in Neuro-Psychopharmacology and Biological Psychiatry, 2016,

67 (3), 92-97). The CB1 receptor inverse agonist/antagonist Rimonabant has been withdrawn from the market due to the high incidence of severe depression and suicidal ideation (Christensen et al, The Lancet, 2007, 370, 1706-1713). Therefore, MGL modulators are potentially useful for the treatment of mood disorders, anxiety, PTSD, autism spectrum disorders, and Asperger syndrome (Folkes et al., J Clin Invest. 2020;130(4):1728-1742, Jung et al., Nature Communications , 2012, 3, 1080; Wang et al., Mol Psychiatry , 2018 August, 23(8): 1798-1806).

Cannabinoid receptor agonists are clinically used to treat pain, spasticity, emesis, and anorexia (Di Marzo, etal, Annu Rev Med., 2006, 57:553-74; Ligresti et al, Curr Opin Chem Biol., 2009, Jun;13(3):321-31). Therefore, MGL modulators, including MGL inhibitors are also potentially useful for these indications. MGL exerts CB 1 -dependant antinociceptive effects in animal models of noxious chemical, inflammatory, thermal and neuropathic pain (Guindon etal, Br J Pharmacol, 2011, Aug; 163(7): 1464-78; Kinsey etal, J Pharmacol Exp Ther., 2009, Sep;330(3):902-10; Long et al, Nat Chem Biol, 2009, Jan;5(l):37-44). MGL blockade reduces mechanical and acetone induced cold allodynia in mice subjected to chronic constriction injury of the sciatic nerve (Kinsey etal, J Pharmacol Exp Ther., 2009, Sep;330(3):902-10). MGL inhibition produces opiate-sparing events with diminished tolerance, constipation, and cannabimimetic side effects (Wilkerson etal, J Pharmacol Exp Ther., 2016, Apr;357(l):145- 56). MGL blockade is protective in model of inflammatory bowel disease (Alhouayek et al, FASEB J., 2011, Aug;25(8):2711-21). MGL inhibition also reverse Paclitaxel-induced nociceptive behavior and proinflammatory markers in a mouse model of chemotherapy-induced neuropathy (Curry et al, J Pharmacol Exp Ther., 2018, Jul;366(l): 169-18). MGL inhibitors are also potentially useful for the treatment of chronic inflammatory condition of the urinary bladder like interstitial cystitis (Chinnadurai et al., 2019, Oct; 131: 109321).

Inhibition of 2-AG hydrolysis exerts anti-proliferative activity and reduction in prostate cancer cell invasiveness (Nithipatikom etal, Cancer Res., 2004, Dec 15, 64(24):8826- 30; Nithipatikom etal, Biochem Biophys Res Commun., 2005, Jul 15,332(4): 1028- 33; Nithipatikom et al, Prostaglandins Other Lipid Medial, 2011, Feb, 94(l-2):34-43). MGL is upregulated in aggressive human cancer cells and primary tumors where it has a unique role of providing lipolytic sources of free fatty acids for synthesis of oncogenic signaling lipids that promote cancer aggressiveness. Thus, beyond the physiological roles of MGL in mediated endocannabinoid signaling, MGL in cancer plays a distinct role in modulating the fatty acid precursor pools for synthesis of protumorigenic signaling lipids in malignant human cancer cells. MGL blockade shows anti-emetic and anti-nausea effects in a lithium chloride model of vomiting in shrews (Sticht etal., Br J Pharmacol., 2012, Apr, 165(8):2425-35).

MGL modulators, including MGL inhibitors may have utility in modulating drug dependence of opiates. MGL blockade reduce the intensity of naloxone-precipitated morphine withdrawal symptoms in mice. MGL blockade also attenuated spontaneous withdrawal signs in morphine- dependent mice (Ramesh etal, J Pharmacol Exp Ther., 2011, Oct, 339(1): 173-85).

MGL modulators are also potentially useful for the treatment of eye conditions, including but not limited to, glaucoma and disease states arising from elevated intraocular pressure (Miller etal, Pharmaceuticals , 2018, 11, 50).

SUMMARY OF THE INVENTION

Embodiments of the present invention relate to chemical entities, pharmaceutical compositions containing them, methods of making and purifying them, and methods for using them the treatment of diseases, disorders, and conditions associated with the MGL modulation. An additional embodiment of the invention is a method of treating a subject suffering from or diagnosed with a disease, disorder, or condition associated with the MGL modulation using at least one chemical entity of the invention.

Additional embodiments, features, and advantages of the invention will be apparent from the following detailed description and through practice of the invention.

Described herein are compounds of Formula (I): wherein X is CH 2 or O;

R 1 is H;

R 2a and R 2b are each independently selected from H and Ci-4alkyl;

R 3 is selected from:

(i) phenyl, benzyl, or monocyclic heteroaryl, each optionally substituted with one, two, or three substituents selected from: halo, Ci- 6 alkyl, Ci- 6 haloalkyl, Ci- 6 alkyl-OH, OCi- 6 alkyl, OCi- 6 haloalkyl, SCi- 6 alkyl, SFs, Si(CH 3 ) 3 , NR a R b , C 3 - 6 cycloalkyl, OC 3 - 6 cycloalkyl, phenyl, O-phenyl, and O-pyridyl, wherein each cycloalkyl, phenyl, or pyridyl is optionally substituted with one or two Ci-4alkyl, Ci-4haloalkyl, or halo groups; or two adjacent ring substituents on the phenyl, benzyl, or monocyclic heteroaryl, taken together with the atoms to which they are attached form a fused monocyclic C5- 6 cycloalkyl or heterocycloalkyl ring, each ring optionally substituted with one or two Ci-4alkyl, Ci- 4haloalkyl, or halo groups; wherein R a and R b are each independently H or Ci-4alkyl;

(ii) a bicyclic heteroaryl optionally substituted with Ci-4alkyl or halo; and

(iii) C 3 - 6 alkyl or C 3 - 6 cycloalkyl optionally substituted with Ci-4alkyl, Ci-4haloalkyl, or halo; R 4 is Ci- 6 alkyl; and pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers thereof.

In some embodiments are compounds of Formula (I): wherein

X is CH 2 or O;

R 1 is H;

R 2a and R 2b are each H;

R 3 is selected from: Ci- 6 alkyl (optionally C3- 6 alkyl); C3- 6 cycloalkyl; benzyl; phenyl; phenyl substituted with one or two members each independently selected from: halo, Ci- 6 alkyl, OCi- 6 alkyl, C 3 - 6 cycloalkyl, and C 3 - 6 cycloalkyl substituted with CH3 or CF3; 2, 3 -dihydro- 1H- inden-5-yl; bicyclo[4.2.0]octa-l(6),2,4-trien-3-yl; and 5,6,7,8-tetrahydronaphthalen-2-yl; and

R 4 is Ci- 6 alkyl; and pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers thereof. DETAILED DESCRIPTION OF THE INVENTION

As used herein, the terms "including", "containing" and “comprising” are used in their open, non-limiting sense.

Unless qualified specifically in particular instances of use, the term “alkyl” refers to a straight- or branched-chain alkyl group having from 1 to 8 carbon atoms in the chain. Examples of alkyl groups include methyl (Me), ethyl (Et), n-propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl (tBu), pentyl, isopentyl, tert-pentyl, hexyl, isohexyl, and groups that in light of the ordinary skill in the art and the teachings provided herein would be considered equivalent to any one of the foregoing examples. “Ci- 6 alkyl” refers to straight- or branched-chain alkyl group having from 1 to 6 carbon atoms in the chain. “Ci-4alkyl” refers to straight- or branched-chain alkyl group having from 1 to 4 carbon atoms in the chain.

The term “cycloalkyl” refers to a saturated or partially saturated, monocyclic, fused polycyclic, or spiro polycyclic carbocycle having from 3 to 12 ring atoms per carbocycle. Illustrative examples of cycloalkyl groups include the following entities, in the form of properly bonded moieties:

The term “halogen” or “halo” represents chlorine, fluorine, bromine, or iodine.

The term “haloalkyl” refers to a straight- or branched-chain alkyl group having from 1 to 6 carbon atoms in the chain optionally substituting hydrogens with halogens. The term “Ci-4 haloalkyl” as used here refers to a straight- or branched-chain alkyl group having from 1 to 4 carbon atoms in the chain, optionally substituting hydrogens with halogens. Examples of “haloalkyl” groups include trifluoromethyl (CF3), difluoromethyl (CF2H), monofluoromethyl (CFhF), pentafluoroethyl (CF2CF3), tetrafluoroethyl (CHFCF3), monofluoroethyl (CH2CH2F), trifluoroethyl (CH2CF3), tetrafluorotrifluoromethylethyl (CF(CF3)2), and groups that in light of the ordinary skill in the art and the teachings provided herein would be considered equivalent to any one of the foregoing examples.

The term "heteroaryl" as used herein, refers to an aromatic monocyclic or multicyclic ring system comprising 5 to 14 ring atoms, wherein from 1 to 4 of the ring atoms is independently O, N or S and the remaining ring atoms are carbon atoms. In one embodiment, a heteroaryl group has 5 to 10 ring atoms. In another embodiment, a heteroaryl group is monocyclic and has 5 or 6 ring atoms. In another embodiment, a heteroaryl group is monocyclic and has 5 or 6 ring atoms and at least one nitrogen ring atom. A heteroaryl group is joined via a ring carbon atom and any nitrogen atom of a heteroaryl can be optionally oxidized to the corresponding N-oxide. The term "heteroaryl" also encompasses a heteroaryl group, as defined above, which has been fused to a benzene ring.

The term “heterocycloalkyl” as used herein, refers to a ring system which is non aromatic, 1 to 4 of the ring atoms is independently O, N or S and the remaining ring atoms are carbon atoms, which may optionally be fused to another ring (aromatic or heteroaromatic). Non limiting examples of illustrative heterocycloalkyl include:

N ° .

The term “aryl” refers to a monocyclic, aromatic carbocycle (ring structure having ring atoms that are all carbon) having 6 atoms per ring (Carbon atoms in the aryl groups are sp2 hybridized.)

The term “phenyl” represents the following moiety:

The term “2,3-dihydro-lH-inden-5-yl” represents the following moiety:

The term “bicyclo[4.2.0]octa-l(6),2,4-trien-3-yl” represents the following moiety: The term “5,6,7,8-tetrahydronaphthalen-2-yl” represents the following moiety:

Those skilled in the art will recognize that the species of heteroaryl, heterocycloalkyl, cycloalkyl, or aryl groups listed or illustrated above are not exhaustive, and that additional species within the scope of these defined terms may also be selected. The term “substituted” means that the specified group or moiety bears one or more substituents. The term "unsubstituted" means that the specified group bears no substituents. The term “optionally substituted” means that the specified group is unsubstituted or substituted by one or more substituents. Where the term “substituted” is used to describe a structural system, the substitution is meant to occur at any valency-allowed position on the system.

The term “variable point of attachment” means that a group is allowed to be attached at more than one alternative position in a structure. The attachment will always replace a hydrogen atom on one of the ring atoms. In other words, all permutations of bonding are represented by the single diagram, as shown in the illustrations below.

Those skilled in the art will recognize that that if more than one such substituent is present for a given ring; the bonding of each substituent is independent of all of the others. The groups listed or illustrated above are not exhaustive.

The term “substituted” means that the specified group or moiety bears one or more substituents. The term "unsubstituted" means that the specified group bears no substituents. The term “optionally substituted” means that the specified group is unsubstituted or substituted by one or more substituents. Where the term “substituted” is used to describe a structural system, the substitution is meant to occur at any valency-allowed position on the system.

Any formula given herein is intended to represent compounds having structures depicted by the structural formula as well as certain variations or forms. In particular, compounds of any formula given herein may have asymmetric centers and therefore exist in different enantiomeric forms. All optical isomers and stereoisomers of the compounds of the general formula, and mixtures thereof, are considered within the scope of such formula. The compounds of this invention may possess one or more asymmetric centers; such compounds can therefore be produced as individual ( R )- or (A)-stereoi somers or as mixtures thereof. Thus, any formula given herein is intended to represent a racemate, one or more of its enantiomeric forms, one or more of its diastereomeric forms, and mixtures thereof. Additionally, any formula given herein is intended to refer also to any one of: hydrates, solvates, polymorphs and of such compounds, and mixtures thereof, even if such forms are not listed explicitly.

The term “R” at a stereocenter designates that the stereocenter is purely of the R- configuration as defined in the art; likewise, the term “S” means that the stereocenter is purely of the ^-configuration. As used herein, the term “RS” refers to a stereocenter that exists as a mixture of the R- and ^-configurations.

Compounds containing one stereocenter drawn without a stereo bond designation are a mixture of 2 enantiomers. Compounds containing 2 stereocenters both drawn without stereo bond designations are a mixture of 4 diastereomers. Compounds with 2 stereocenters both labeled “RS” and drawn with stereo bond designations are a 2-component mixture with relative stereochemistry as drawn. Unlabeled stereocenters drawn without stereo bond designations are a mixture of the R- and ^-configurations. For unlabeled stereocenters drawn with stereo bond designations, the absolute stereochemistry is as depicted.

Reference to a compound herein stands for a reference to any one of: (a) the actually recited form of such compound, and (b) any of the forms of such compound in the medium in which the compound is being considered when named. For example, reference herein to a compound such as R-COOH, encompasses reference to any one of: for example, R-COOH(s), R-

COOH(sol), and R-COO-(sol). In this example, R-COOH(s) refers to the solid compound, as it could be for example in a tablet or some other solid pharmaceutical composition or preparation;

R-COOH(sol) refers to the undissociated form of the compound in a solvent; and R-COO-(sol) refers to the dissociated form of the compound in a solvent, such as the dissociated form of the compound in an aqueous environment, whether such dissociated form derives from R-COOH, from a salt thereof, or from any other entity that yields R-COO- upon dissociation in the medium being considered. In another example, an expression such as “exposing an entity to compound of formula R-COOH” refers to the exposure of such entity to the form, or forms, of the compound

R-COOH that exists, or exist, in the medium in which such exposure takes place. In still another example, an expression such as “reacting an entity with a compound of formula R-COOH” refers to the reacting of (a) such entity in the chemically relevant form, or forms, of such entity that exists, or exist, in the medium in which such reacting takes place, with (b) the chemically relevant form, or forms, of the compound R-COOH that exists, or exist, in the medium in which such reacting takes place. In this regard, if such entity is for example in an aqueous environment, it is understood that the compound R-COOH is in such same medium, and therefore the entity is being exposed to species such as R-COOH(aq) and/or R-COO-(aq), where the subscript “(aq)” stands for “aqueous” according to its conventional meaning in chemistry and biochemistry. A carboxylic acid functional group has been chosen in these nomenclature examples; this choice is not intended, however, as a limitation but it is merely an illustration. It is understood that analogous examples can be provided in terms of other functional groups, including but not limited to hydroxyl, basic nitrogen members, such as those in amines, and any other group that interacts or transforms according to known manners in the medium that contains the compound. Such interactions and transformations include, but are not limited to, dissociation, association, tautomerism, solvolysis, including hydrolysis, solvation, including hydration, protonation, and deprotonation. No further examples in this regard are provided herein because these interactions and transformations in a given medium are known by any one of ordinary skill in the art.

Any formula given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds. Isotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number in an enriched form. Examples of isotopes that can be incorporated into compounds of the invention in a form that exceeds natural abundances include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2 H (or chemical symbol D), 3 H (or chemical symbol T), U C, 13 C, 14 C, 15 N, 18 0, 17 0, 31 P, 32 P, 35 S, 18 F, 36 C1, and 125 I, respectively. Such isotopically labelled compounds are useful in metabolic studies (preferably with 14 C), reaction kinetic studies (with, for example 2 H or 3 H), detection or imaging techniques [such as positron emission tomography (PET) or single photon emission computed tomography (SPECT)] including drug or substrate tissue distribution assays, or in radioactive treatment of patients. In particular, an 18 F or U C labeled compound may be particularly preferred for PET or SPECT studies. Further, substitution with heavier isotopes such as deuterium (i.e., 2 H, or D) may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements. Isotopically labeled compounds of this invention can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non- isotopically labeled reagent.

When referring to any formula given herein, the selection of a particular moiety from a list of possible species for a specified variable is not intended to define the same choice of the species for such variable appearing elsewhere. In other words, where a variable appears more than once, the choice of the species from a specified list is independent of the choice of the species for the same variable elsewhere in the formula, unless stated otherwise.

The term Cn-m alkyl refers to an aliphatic chain, whether straight or branched, with a total number N of carbon members in the chain that satisfies n < N < m, with m > n.

When the same plurality of substituents is assigned to various groups, the specific individual substituent assignment to each of such groups is meant to be independently made with respect to the specific individual substituent assignments to the remaining groups. By way of illustration, but not as a limitation, if each of groups Q and R can be H or F, the choice of H or F for Q is made independently of the choice of H or F for R, so the choice of assignment for Q does not determine or condition the choice of assignment for R, or vice-versa, unless it is expressly indicated otherwise. Illustrative claim recitation in this regard would read as “each of Q and R is independently H or F”, or “each of Q and R is independently selected H and F”.

Unless indicated otherwise, the description or naming of a particular compound in the specification and claims is intended to include both individual enantiomers and mixtures, racemic or otherwise, thereof. The methods for the determination of stereochemistry and the separation of stereoisomers are well-known in the art.

In another example, a zwitterionic compound would be encompassed herein by referring to a compound that is known to form a zwitterion, even if it is not explicitly named in its zwitterionic form. Terms such as zwitterion, zwitterions, and their synonyms zwitterionic compound(s) are standard IUP AC-endorsed names that are well known and part of standard sets of defined scientific names. In this regard, the name zwitterion is assigned the name identification CHEBT27369 by the Chemical Entities of Biological Interest (ChEBI) dictionary of molecular entities. As generally well known, a zwitterion or zwitterionic compound is a neutral compound that has formal unit charges of opposite sign. Sometimes these compounds are referred to by the term “inner salts”. Other sources refer to these compounds as “dipolar ions”, although the latter term is regarded by still other sources as a misnomer. As a specific example, aminoethanoic acid (the amino acid glycine) has the formula H2NCH2COOH, and it exists in some media (in this case in neutral media) in the form of the zwitterion + H3NCH2COO . Zwitterions, zwitterionic compounds, inner salts, and dipolar ions in the known and well- established meanings of these terms are within the scope of this invention, as would in any case be so appreciated by those of ordinary skill in the art. Because there is no need to name each and every embodiment that would be recognized by those of ordinary skill in the art, no structures of the zwitterionic compounds that are associated with the compounds of this invention are given explicitly herein. They are, however, part of the embodiments of this invention. No further examples in this regard are provided herein because the interactions and transformations in a given medium that lead to the various forms of a given compound are known by any one of ordinary skill in the art.

When referring to any formula given herein, the selection of a particular moiety from a list of possible species for a specified variable is not intended to define the same choice of the species for the variable appearing elsewhere. In other words, where a variable appears more than once, the choice of the species from a specified list is independent of the choice of the species for the same variable elsewhere in the formula, unless stated otherwise.

By way of a first example on substituent terminology, if substituent S Example is one of Si and S2, and substituent S 2 exampie is one of S3 and S4, then these assignments refer to embodiments of this invention given according to the choices S Example is Si and S 2 exampie is S3; S 'exam le is Si and S 2 example IS S4; S 'example is S2 and S 2 example IS S3; S 'example is S2 and S 2 example IS S4; and equivalents of each one of such choices. The shorter terminology “S Example is one of Si and S2, and S 2 exam P ie is one of S3 and S4” is accordingly used herein for the sake of brevity, but not by way of limitation. The foregoing first example on substituent terminology, which is stated in generic terms, is meant to illustrate the various substituent assignments described herein.

Furthermore, when more than one assignment is given for any member or substituent, embodiments of this invention comprise the various groupings that can be made from the listed assignments, taken independently, and equivalents thereof. By way of a second example on substituent terminology, if it is herein described that substituent Sexampie is one of Si, S2, and S3, this listing refers to embodiments of this invention for which Sexampie is Si; Sexampie is S2; Sexampie is S3 ^ Sexampie IS One of Si and S2; Sexampie IS One of Si and S3 ^ Sexampie IS One of S2 and S3 ^ Sexampie is one of Si, S2 and S3; and Sexampie is any equivalent of each one of these choices. The shorter terminology “Sexampie is one of Si, S2, and S3” is accordingly used herein for the sake of brevity, but not by way of limitation. The foregoing second example on substituent terminology, which is stated in generic terms, is meant to illustrate the various substituent assignments described herein.

The nomenclature “Ci-Q” with j > i, when applied herein to a class of substituents, is meant to refer to embodiments of this invention for which each and every one of the number of carbon members, from i to j including i and j, is independently realized. By way of example, the term C1-C3 refers independently to embodiments that have one carbon member (Ci), embodiments that have two carbon members (C2), and embodiments that have three carbon members (C3).

A "pharmaceutically acceptable salt” is intended to mean a salt of an acid or base of a compound represented by Formula (I) that is non-toxic, biologically tolerable, or otherwise biologically suitable for administration to the subject. See, generally, S.M. Berge, et al ., “Pharmaceutical Salts”, J. Pharm. Sci., 1977, 66:1-19, and Handbook of Pharmaceutical Salts, Properties, Selection, and Use, Stahl and Wermuth, Eds., Wiley-VCH and VHCA, Zurich, 2002. Preferred pharmaceutically acceptable salts are those that are pharmacologically effective and suitable for contact with the tissues of patients without undue toxicity, irritation, or allergic response.

A compound of Formula (I) may possess a sufficiently acidic group, a sufficiently basic group, or both types of functional groups, and accordingly react with a number of inorganic or organic bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt.

Examples of pharmaceutically acceptable salts include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogen-phosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-l,4-dioates, hexyne-1,6- dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, sulfonates, xylenesulfonates, phenyl acetates, phenylpropionates, phenylbutyrates, citrates, lactates, g-hydroxybutyrates, glycolates, tartrates, methane-sulfonates, propanesulfonates, naphthalene- 1 -sulfonates, naphthalene-2-sulfonates, and mandelates.

Compounds of Formula (I) may contain at least one nitrogen of basic character, so desired pharmaceutically acceptable salts may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, nitric acid, boric acid, phosphoric acid, and the like, or with an organic acid, such as acetic acid, phenylacetic acid, propionic acid, stearic acid, lactic acid, ascorbic acid, maleic acid, hydroxymaleic acid, isethionic acid, succinic acid, valeric acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, oleic acid, palmitic acid, lauric acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha-hydroxy acid, such as mandelic acid, citric acid, or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid, 2-acetoxybenzoic acid, naphthoic acid, or cinnamic acid, a sulfonic acid, such as laurylsulfonic acid, p-toluenesulfonic acid, methanesulfonic acid, ethanesulfonic acid, any compatible mixture of acids such as those given as examples herein, and any other acid and mixture thereof that are regarded as equivalents.

Compounds of Formula (I) may contain a carboxylic acid moiety, a desired pharmaceutically acceptable salt may be prepared by any suitable method, for example, treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), an alkali metal hydroxide, alkaline earth metal hydroxide, any compatible mixture of bases such as those given as examples herein, and any other base and mixture thereof that are regarded as equivalents or acceptable substitutes in light of the ordinary level of skill in this technology. Illustrative examples of suitable salts include organic salts derived from amino acids, such as glycine and arginine, ammonia, carbonates, bicarbonates, primary, secondary, and tertiary amines, and cyclic amines, such as benzylamines, pyrrolidines, piperidine, morpholine, piperazine, /V-methyl-glucamine and tromethamine and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium.

The compounds of the invention, including their pharmaceutically acceptable salts, whether alone or in combination, (collectively, “active agent” or "active agents") of the present invention are useful as MGL-modulators in the methods of the invention. Such methods for modulating MGL comprise the use of a therapeutically effective amount of at least one chemical entity of the invention.

In some embodiments, the MGL modulator is an inhibitor and is used in a subject diagnosed with or suffering from a disease, disorder, or condition associated with MGL receptor activity, such as those described herein. Symptoms or disease states are intended to be included within the scope of "disease, disorders or conditions."

Accordingly, the invention relates to methods of using the active agents described herein to treat subjects diagnosed with or suffering from a disease, disorder, or condition associated with the MGL receptor activity. The term "treat" or "treating" as used herein is intended to refer to administration of an active agent or composition of the invention to a subject for the purpose of effecting a therapeutic or prophylactic benefit through modulation of MGL receptor activity. Treating includes reversing, ameliorating, alleviating, inhibiting the progress of, lessening the severity of, or preventing a disease, disorder, or condition, or one or more symptoms of such disease, disorder or condition associated with the MGL modulation. The term "subject" refers to a mammalian patient in need of such treatment, such as a human.

The term “composition” refers to a product that includes the specified ingredients in therapeutically effective amounts, as well as any product that results, directly, or indirectly, from combinations of the specified ingredients in the specified amounts.

The term “MGL inhibitor” is intended to encompass a compound that interacts with MGL to substantially reduce or eliminate its catalytic activity, thereby increasing the concentrations of its substrate(s). The term “MGL-modulated” is used to refer to the condition of being affected by the modulation of the MGL enzyme including the condition of being affected by the inhibition of the MGL enzyme. The disclosure is directed to methods for treating, ameliorating and / or preventing diseases, conditions, or disorders associated with pain (including inflammatory pain), and also psychiatric disorders, neurological disorders, cancers and eye conditions by the administration of therapeutically effective amounts of MGL modulators to subjects in need thereof.

The term “modulators” include both inhibitors and activators, where "inhibitors” refer to compounds that decrease, prevent, inactivate, desensitize, or down-regulate the MGL expression or activity, and “activators” are compounds that increase, activate, facilitate, sensitize, or up- regulate MGL expression or activity. As used herein, unless otherwise noted, the term “affect” or “affected” (when referring to a disease, condition or disorder that is affected by inhibition of MGL) includes a reduction in the frequency and / or severity of one or more symptoms or manifestations of said disease, condition or disorder; and / or include the prevention of the development of one or more symptoms or manifestations of said disease, condition or disorder or the development of the disease, condition or disorder.

In treatment methods according to the invention, a therapeutically effective amount of at least one active agent according to the invention is administered to a subject suffering from or diagnosed as having such a disease, disorder, or condition. A "therapeutically effective amount" means an amount or dose sufficient to generally bring about the desired therapeutic or prophylactic benefit in subjects in need of such treatment for the designated disease, disorder, or condition. Effective amounts or doses of the active agents of the present invention may be ascertained by routine methods such as modeling, dose escalation studies or clinical trials, and by taking into consideration routine factors, e.g., the mode or route of administration or drug delivery, the pharmacokinetics of the agent, the severity and course of the disease, disorder, or condition, the subject's previous or ongoing therapy, the subject's health status and response to drugs, and the judgment of the treating physician. For a 70-kg human, an illustrative range for a suitable dosage amount is from about 1 to 1000 mg/day in single or multiple dosage units (e.g., BID, TID, QID or as required by modality).

Once improvement of the subject's disease, disorder, or condition has occurred, the dose may be adjusted for preventive or maintenance treatment. For example, the dosage or the frequency of administration, or both, may be reduced as a function of the symptoms, to a level at which the desired therapeutic or prophylactic effect is maintained. Of course, if symptoms have been alleviated to an appropriate level, treatment may cease. Subjects may, however, require intermittent treatment on a long-term basis upon any recurrence of symptoms.

In addition, the compounds of the invention are envisaged for use alone, in combination with one or more of other compounds of this invention, or in combination with additional active ingredients in the treatment of the conditions discussed below. The additional active ingredients may be co-administered separately with at least one compound of the invention, with active agents of the invention or included with such an agent in a pharmaceutical composition according to the invention. In an illustrative embodiment, additional active ingredients are those that are known or discovered to be effective in the treatment of conditions, disorders, or diseases associated with the MGL modulation, such as another MGL inhibitor or a compound active against another target associated with the particular condition, disorder, or disease. The combination may serve to increase efficacy (e.g., by including in the combination a compound potentiating the potency or effectiveness of an agent according to the invention), decrease one or more side effects, or decrease the required dose of the active agent according to the invention.

When referring to inhibiting the target, an “effective amount” means an amount sufficient to affect MGL modulation.

The active agents of the invention are envisaged for use, alone or in combination with one or more additional active ingredients, to formulate pharmaceutical compositions of the invention. A pharmaceutical composition of the invention comprises a therapeutically effective amount of at least one active agent in accordance with the invention.

Pharmaceutically acceptable excipients commonly used in pharmaceutical compositions are substances that are non-toxic, biologically tolerable, and otherwise biologically suitable for administration to a subject, such as an inert substance, added to a pharmacological composition or otherwise used as a vehicle, carrier, or diluent to facilitate administration of an agent and that is compatible therewith. Examples of such excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, and polyethylene glycols.

Delivery forms of the pharmaceutical compositions containing one or more dosage units of the active agents may be prepared using pharmaceutically acceptable excipients and compounding techniques known or that become available to those of ordinary skill in the art.

The compositions may be administered in the inventive methods by a suitable route of delivery, e.g., oral, parenteral, rectal, topical, or ocular routes, or by inhalation.

The preparation may be in the form of tablets, capsules, sachets, dragees, powders, granules, lozenges, powders for reconstitution, liquid preparations, or suppositories. The compositions may be formulated for any one of a plurality of administration routes, such as intravenous infusion, topical administration, or oral administration. Preferably, the compositions may be formulated for oral administration.

For oral administration, the active agents of the invention can be provided in the form of tablets or capsules, or as a solution, emulsion, or suspension. To prepare the oral compositions, the active agents may be formulated to yield a dosage of, e.g., for a 70-kg human, an illustrative range for a suitable dosage amount is from about 1 to 1000 mg/day in single or multiple dosage units.

Oral tablets may include the active ingredient(s) mixed with compatible pharmaceutically acceptable excipients such as diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavoring agents, coloring agents and preservative agents. Suitable inert fillers include sodium and calcium carbonate, sodium and calcium phosphate, lactose, starch, sugar, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol, and the like.

Exemplary liquid oral excipients include ethanol, glycerol, water, and the like. Starch, polyvinyl-pyrrolidone (PVP), sodium starch glycolate, microcrystalline cellulose, and alginic acid are exemplary disintegrating agents. Binding agents may include starch and gelatin. The lubricating agent, if present, may be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate to delay absorption in the gastrointestinal tract or may be coated with an enteric coating.

Capsules for oral administration include hard and soft gelatin or (hydroxypropyl)methyl cellulose capsules. To prepare hard gelatin capsules, active ingredient(s) may be mixed with a solid, semi-solid, or liquid diluent. Liquids for oral administration may be in the form of suspensions, solutions, emulsions, or syrups or may be lyophilized or presented as a dry product for reconstitution with water or other suitable vehicle before use. Such liquid compositions may optionally contain: pharmaceutically-acceptable excipients such as suspending agents (for example, sorbitol, methyl cellulose, sodium alginate, gelatin, hydroxyethyl cellulose, carboxymethylcellulose, aluminum stearate gel and the like); non-aqueous vehicles, e.g., oil (for example, almond oil or fractionated coconut oil), propylene glycol, ethyl alcohol, or water; preservatives (for example, methyl or propyl p-hydroxybenzoate or sorbic acid); wetting agents such as lecithin; and, if desired, flavoring or coloring agents.

The active agents of this invention may also be administered by non-oral routes. For example, compositions may be formulated for rectal administration as a suppository, enema or foam. For parenteral use, including intravenous, intramuscular, intraperitoneal, or subcutaneous routes, the agents of the invention may be provided in sterile aqueous solutions or suspensions, buffered to an appropriate pH and isotonicity or in parenterally acceptable oil. Suitable aqueous vehicles include Ringer's solution and isotonic sodium chloride. Such forms may be presented in unit-dose form such as ampules or disposable injection devices, in multi-dose forms such as vials from which the appropriate dose may be withdrawn, or in a solid form or pre-concentrate that can be used to prepare an injectable formulation. Illustrative infusion doses range from about 1 to 1000 pg/kg/minute of agent admixed with a pharmaceutical carrier over a period ranging from several minutes to several days.

For topical administration, the agents may be mixed with a pharmaceutical carrier at a concentration of about 0.01% to about 20% of drug to vehicle, preferably 0.1% to 10%. Another mode of administering the agents of the invention may utilize a patch formulation to affect transdermal delivery.

Active agents may alternatively be administered in methods of this invention by inhalation, via the nasal or oral routes, e.g., in a spray formulation also containing a suitable carrier.

In a further embodiment, the invention is directed to a method of treating a subject suffering from or diagnosed with a disease, disorder, or condition associated with MGL modulation, comprising administering to the subject in need of such treatment a therapeutically effective amount of the active agent.

The compounds of Formula (I) are useful in methods for treating, ameliorating and / or preventing a disease, a condition or a disorder that is affected by the inhibition of MGL. Such methods comprise administering to a subject, including an animal, a mammal, and a human in need of such treatment, amelioration and / or prevention, a therapeutically effective amount of a compound of Formula (I), or an enantiomer, diastereomer, solvate or pharmaceutically acceptable salt thereof.

In particular, the compounds of Formula (I), or pharmaceutically acceptable salts, isotopes, N-oxides, solvates and stereoisomers thereof, are useful for treating, ameliorating and / or preventing diseases, conditions, or disorders causing pain, psychiatric disorders, neurological disorders, cancers, and eyes conditions. More particularly, the compounds of Formula (I), or pharmaceutically acceptable salts, isotopes, N-oxides, solvates and stereoisomers thereof, are useful for treating, ameliorating and / or preventing inflammatory pain, major depressive disorder, treatment resistant depression, anxious depression or bipolar disorder by administering to a subject in need thereof a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, isotope, N-oxide, solvate or stereoisomer thereof as herein defined.

1) Pain

Examples of inflammatory pain include, but are not limited to, pain due to a disease, condition, disorder, or a pain state including inflammatory bowel disease, visceral pain, migraine, post-operative pain, osteoarthritis, rheumatoid arthritis, back pain, lower back pain, joint pain, abdominal pain, chest pain, labor, musculoskeletal diseases, skin diseases, toothache, pyresis, bum, sunburn, snake bite, venomous snake bite, spider bite, insect sting, neurogenic bladder, interstitial cystitis, urinary tract infection, rhinitis, contact dermatitis/hypersensitivity, itch, eczema, pharyngitis, mucositis, enteritis, irritable bowel syndrome, cholecystitis, pancreatitis, postmastectomy pain syndrome, menstrual pain, endometriosis, pain due to physical trauma, headache, sinus headache, tension headache, or arachnoiditis.

One type of inflammatory pain is inflammatory hyperalgesia / hypersensitivity.

Examples of inflammatory hyperalgesia include a disease, condition, disorder, or pain state including inflammation, osteoarthritis, rheumatoid arthritis, back pain, joint pain, abdominal pain, musculoskeletal diseases, skin diseases, post-operative pain, headaches, toothache, bum, sunburn, insect sting, neurogenic bladder, urinary incontinence, interstitial cystitis, urinary tract infection, cough, asthma, chronic obstructive pulmonary disease, rhinitis, contact dermatitis/hypersensitivity and/or dermal allergy, itch, eczema, pharyngitis, enteritis, irritable bowel syndrome, inflammatory bowel diseases including Crohn's Disease, ulcerative colitis, benign prostatic hypertrophy, and nasal hypersensitivity.

In an embodiment, the present invention is directed to a method for treating, ameliorating and / or preventing inflammatory visceral hyperalgesia in which an enhanced visceral irritability exists, comprising, consisting of, and/or consisting essentially of the step of administering to a subject in need of such treatment a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt, isotope, N-oxide, solvate or stereoisomer thereof. In a further embodiment, the present invention is directed to a method for treating inflammatory somatic hyperalgesia in which a hypersensitivity to thermal, mechanical and/or chemical stimuli exists, comprising administering to a subject in need of such treatment a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, isotope, N-oxide, solvate or stereoisomer thereof.

A further embodiment of the present invention is directed to a method for treating, ameliorating and / or preventing neuropathic pain. Examples of a neuropathic pain include pain due to a disease, condition, disorder, or pain state including cancer, neurological disorders, spine and peripheral nerve surgery, brain tumor, traumatic brain injury (TBI), spinal cord trauma, chronic pain syndrome, fibromyalgia, chronic fatigue syndrome, lupus, sarcoidosis, peripheral neuropathy, bilateral peripheral neuropathy, diabetic neuropathy, central pain, neuropathies associated with spinal cord injury, stroke, amyotrophic lateral sclerosis (ALS), Parkinson’s disease, multiple sclerosis, sciatic neuritis, mandibular joint neuralgia, peripheral neuritis, polyneuritis, stump pain, phantom limb pain, bony fractures, oral neuropathic pain, Charcot's pain, complex regional pain syndrome I and II (CRPS I/II), radiculopathy, Guillain-Barre syndrome, meralgia paresthetica, burning-mouth syndrome, optic neuritis, postfebrile neuritis, migrating neuritis, segmental neuritis, Gombaulf s neuritis, neuronitis, cervicobrachial neuralgia, cranial neuralgia, geniculate neuralgia, glossopharyngeal neuralgia, migrainous neuralgia, idiopathic neuralgia, intercostals neuralgia, mammary neuralgia, Morton's neuralgia, nasociliary neuralgia, occipital neuralgia, postherpetic neuralgia, causalgia, red neuralgia, Sluder's neuralgia, splenopalatine neuralgia, supraorbital neuralgia, trigeminal neuralgia, vulvodynia, vidian neuralgia or chemotherapy-induced neuropathy.

One type of neuropathic pain is neuropathic cold allodynia, which can be characterized by the presence of a neuropathy-associated allodynic state in which a hypersensitivity to cooling stimuli exists. Examples of neuropathic cold allodynia include allodynia due to a disease, condition, disorder or pain state including neuropathic pain (neuralgia), pain arising from spine and peripheral nerve surgery or trauma, traumatic brain injury (TBI), trigeminal neuralgia, postherpetic neuralgia, causalgia, peripheral neuropathy, diabetic neuropathy, central pain, stroke, peripheral neuritis, polyneuritis, complex regional pain syndrome I and II (CRPS I/II) and radiculopathy.

In a further embodiment, the present invention is directed to a method for treating, ameliorating and / or preventing neuropathic cold allodynia in which a hypersensitivity to a cooling stimuli exists, comprising administering to a subject in need of such treatment a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt, isotope, N-oxide, solvate or stereoisomer thereof.

2) Psychiatric disorders

Examples of psychiatric disorders include, but are not limited to, anxieties such as, social anxiety, post-traumatic stress disorder, phobias, social phobia, special phobias, panic disorder, obsessive-compulsive disorder, acute stress disorder, separation anxiety disorder, and generalized anxiety disorder, as well as depression such as, major depression, bipolar disorder, seasonal affective disorder, post-natal depression, manic depression, and bipolar depression, mood disorders and mood affective disorders that can be treated according to the present invention include, but are not limited to, bipolar disorder I depressed, hypomanic, manic and mixed form; bipolar disorder II; depressive disorders, such as single depressive episode or recurrent major depressive disorder, minor depressive disorder, treatment-resistant depression, anxious depression, bipolar disorder, depressive disorder with postpartum onset, depressive disorders with psychotic symptoms; persistent mood disorders, such as cyclothymia, dysthymia, euthymia; premenstrual dysphoric disorder; psychoses; and developmental disorders such as autism spectrum disorders, and Asperger syndrome.

3) Neurological disorders

Examples of neurological disorder include, but are not limited to, tremors, dyskinesias, dystonias, spasticity, Tourette’s Syndrome; neuromyelitis optica, Parkinson’s disease; Alzheimer’s disease; senile dementia; Huntington’s disease; Epilepsy/seizure disorders and sleep disorders.

4) Cancers

Examples of cancers include, but are not limited to, benign skin tumors, prostate tumors, ovarian tumors and cerebral tumors (glioblastomas, medulloepitheliomas, medulloblastomas, neuroblastomas, tumors of embryonic origin, astrocytomas, astroblastomas, ependymomas, oligodendrogliomas, neuroepitheliomas, epiphyseal tumor, ependymoblastomas, malignant meningiomas, sarcomatosis, malignant melanomas, schwannomas). 5) Eye conditions

Examples of eye conditions include, but are not limited to, ocular hypertension, glaucoma, degeneration, and apoptosis of retinal ganglion cells and neuroretinal cells. Other embodiments of this invention provide for a method for modulating MGL receptor activity, including when such receptor is in a subject, comprising exposing MGL receptor to a therapeutically effective amount of at least one compound selected from compounds of the invention.

In some embodiments of Formula (I), X is CEL. In some embodiments, X is O. In some embodiments, R 3 is tert-butyl. In some embodiments, R 3 is , , In some embodiments, R 3 is benzyl, phenyl, or phenyl substituted with one or two members each independently selected from: F, CFb, CH2CH3, CH(CH3)2, C(CH 3 ) 3 , OCH3, cyclopropyl, cyclopropyl substituted with CF3, and cyclobutyl. In some embodiments, R 3 is In some embodiments, R 3 is 3,5-dimethylphenyl, 3-ethyl-5-methylphenyl, 4-ethyl-3- methylphenyl, 3-isopropylphenyl, or 3-tert-butylphenyl.

In some embodiments, R 3 is phenyl; or phenyl substituted with one, two or three members each independently selected from: Cl, F, Ci- 6 alkyl, Ci- 6 haloalkyl, C(CH3)20H, OCi- 6 alkyl, OCi- 6 haloalkyl, SCH3, Si(CH 3 ) 3 , SFs, N(CH 3 )2, C 3 - 6 cycloalkyl, C 3 - 6 cycloalkyl substituted with CFb, OC 3 - 6 cycloalkyl, phenyl, O-phenyl, and O-pyridyl. In some embodiments, R 3 is phenyl substituted with one, two or three members each independently selected from: halo, Ci- 6 alkyl, Ci- 6 haloalkyl, OCi- 6 alkyl, OCi- 6 haloalkyl, SCFb, SFs, or Si(CH 3 ) 3. In some embodiments, R 3 is benzyl; tert-butyl; cyclohexyl; phenyl substituted with 1-methylcyclopropyl or 1-trifluoromethylcyclopropyl, or fused with a cyclobutenyl or cyclohexenyl ring; pyridyl optionally substituted with trifluoromethyl, fluoro, or methyl; pyrimidinyl optionally substituted with tert-butyl; or oxazolyl optionally substituted with tert-butyl. In some embodiments, R 3 is a bicyclic heteroaryl, optionally substituted as described herein. In some embodiments, R 3 is phenyl, optionally substituted as described herein.

In some embodiments, R 3 is

In some embodiments, R 3 is 4-trifluoromethylphenyl, 3-trifluoromethoxyphenyl, 3-tert- butylphenyl, 4-tert-butylphenyl, or 3-(l-methylcyclopropyl)phenyl.

In some embodiments, R 2a is H and R 2b is CFF. In other embodiments, R 2a and R 2b are each CFb. In some embodiments, R 2a and R 2b are each H.

In some embodiments, X is O, and R 2a and R 2b are each H. In some embodiments, X is CFh, and R 2a and R 2b are each H. In some embodiments, X is O and R 3 is phenyl substituted with one or two members each independently selected from: F, Ci- 6 alkyl, OCH3, cyclopropyl, cyclopropyl substituted with CFb or CF3, and cyclobutyl.

In some embodiments, R 4 is CFb or CH2CH3.

A further embodiment of the current invention is a compound as shown below in Table 1. Table 1. and pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers thereof.

A further embodiment of the current invention is a compound selected from: (2r,4S)-N-((ls,3S)-3-(3-(/er/-Butyl)phenyl)cyclobutyl)-N-met hyl-6-oxo-5-azaspiro[3.4]octane-2- carboxamide; (2s,4S)-N-((ls,3S)-3-(3,5-Dimethylphenyl)cyclobutyl)-N-methy l-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(3-Isopropylphenyl)cyclobutyl)-N-met hyl-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide;

(2s,4S)-N-((ls,3S)-3-(4-Isopropylphenyl)cyclobutyl)-N-met hyl-6-oxo-7-oxa-5- azaspiro[3 4]octane-2-carboxamide; and

(2s,4S)-N-((ls,3S)-3-(4-Ethylphenyl)cyclobutyl)-N-methyl- 6-oxo-7-oxa-5-azaspiro[3.4]octane-

2-carboxamide; and pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers thereof.

An additional embodiment of the invention is a pharmaceutical composition comprising:

(A) a therapeutically effective amount of at least one compound selected from compounds of Formula (I) and pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers of compounds of Formula (I); and

(B) at least one pharmaceutically acceptable excipient.

An additional embodiment of the invention is a pharmaceutical composition comprising a therapeutically effective amount of at least one compound selected from the compounds in Table 1, including pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers of compounds of Table 1, pharmaceutically acceptable prodrugs of compounds of Table 1, and pharmaceutically active metabolites of Table 1; and at least one pharmaceutically acceptable excipient.

Also within the scope of the invention are enantiomers and diastereomers of the compounds of Formula (I). Also within the scope of the invention are the pharmaceutically acceptable salts, N-oxides or solvates of the compounds of Formula (I). Also within the scope of the invention are the pharmaceutically acceptable prodrugs of compounds of Formula (I), and pharmaceutically active metabolites of the compounds of Formula (I).

Also within the scope of the invention are isotopic variations of compounds of Formula (I), such as, e.g., deuterated compounds of Formula (I). Also within the scope of the invention are the pharmaceutically acceptable salts, N-oxides or solvates of the isotopic variations of the compounds of Formula (I). Also within the scope of the invention are the pharmaceutically acceptable prodrugs of the isotopic variations of the compounds of Formula (I), and pharmaceutically active metabolites of the isotopic variations of the compounds of Formula (I).

An additional embodiment of the invention is a method of treating a subject suffering from or diagnosed with a disease, disorder, or condition mediated by MGL receptor activity, comprising administering to a subject in need of such treatment a therapeutically effective amount of at least one compound selected from compounds of Formula (I), and pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers thereof, including enantiomers and diastereomers of the compounds of Formula (I), isotopic variations of the compounds of Formula (I), and pharmaceutically acceptable salts of all of the foregoing. Also described herein is the use of a compound of Formula (I) or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof in the preparation of a medicament. In some embodiments, the medicament is for treatment of a disease, disorder, or condition mediated by MGL receptor activity. Also described herein is a compound of Formula (I), or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof, for use in a method of treating a disease, disorder, or condition mediated by MGL receptor activity.

Exemplary compounds useful in methods of the invention will now be described by reference to the illustrative synthetic schemes for their general preparation below and the specific examples that follow. Artisans will recognize that, to obtain the various compounds herein, starting materials may be suitably selected so that the ultimately desired substituents will be carried through the reaction scheme with or without protection as appropriate to yield the desired product. Alternatively, it may be necessary or desirable to employ, in the place of the ultimately desired substituent, a suitable group that may be carried through the reaction scheme and replaced as appropriate with the desired substituent. Unless otherwise specified, the variables are as defined above in reference to Formula (I). Reactions may be performed between the melting point and the reflux temperature of the solvent, and preferably between 0 °C and the reflux temperature of the solvent. Reactions may be heated employing conventional heating or microwave heating. Reactions may also be conducted in sealed pressure vessels above the normal reflux temperature of the solvent.

Abbreviations and acronyms used herein include the following: Table 2.

PREPARATIVE EXAMPLES

Exemplary compounds useful in methods of the invention will now be described by reference to the illustrative synthetic schemes for their general preparation below and the specific examples to follow. SCHEME 1

According to SCHEME 1, a compound of formula (V), where R a is Ci-4alkyl, is treated with hydroxylamine; using an additive such as sodium acetate (NaOAc), and the like; in a suitable solvent such as ethanol (EtOH), and the like; to provide a compound of formula (VI).

A compound of formula (VII) is prepared from a compound of formula (VI) using an oxidant such as hydrogen peroxide, urea-hydrogen peroxide, and the like; in the presence of an activator such as trifluoroacetic anhydride (TFAA), and the like; in the presence of a base such as dibasic sodium phosphate, and the like; in a solvent such as acetonitrile (ACN), and the like.

SCHEME 2

(XI) (X)

According to SCHEME 2, compounds of formula (Villa) and (VUIb) are prepared by reacting a compound of formula (VII), R a is Ci-4alkyl, with formaldehyde in the presence of a base such as triethylamine (TEA), and the like; in a solvent such as ACN, and the like. A compound of formula (IX) is prepared by hydrogenolysis of a compound of formula (VUIb) under an atmosphere of hydrogen gas (H2) in the presence of a catalyst such as palladium on carbon (Pd/C), and the like; in a solvent such as ethyl acetate (EtOAc), EtOH, and the like.

A compound of formula (X) is prepared by the reaction of a compound of formula (IX) with triphosgene in the presence of a base such as TEA, and the like; in a solvent such as tetrahydrofuran (THF), and the like. A compound of formula (XI) is prepared by the acidic deprotection of a compound of formula (X) using an acid such as trifluoroacetic acid (TFA), HC1 in dioxane, and the like.

SCHEME 3 According to SCHEME 3, compounds of formula (Xlla) and formula (Xllb) are prepared by a Michael-type reaction between a compound of formula (VII), where R a is ethyl, and methyl acrylate; in the presence of a base such as l,8-diazabicyclo[5.4.0]undec-7-ene (DBU), and the like; in a solvent such as ACN, and the like. Reductive ring closure of a compound of formula (Xlla) using a reducing agent such as sodium borohydride (NaBHr), and the like; an additive such as nickel(II) chloride hexahydrate, and the like; in a suitable solvent such as methanol (MeOH), and the like; provides a compound of formula (XIII), where X is CH2.

SCHEME 4 Magnesium Insertion

(XIV) iPrMgCI ( xv ) or

BuLi According to SCHEME 4, a commercially available or synthetically accessible aryl halide of formula (XIV), where X is Cl or Br, and R 3 is an aryl group suitably substituted with Ci- 6 alkyl, OCi- 6 alkyl, C 3 - 6 cycloalkyl, and C 3 - 6 cycloalkyl substituted with CF 3 ; and 5, 6,7,8- tetrahydronaphthalen-2-yl; is reacted with magnesium metal; LiCl as an additive; in a suitable solvent such as THF; DIBAL-H; at temperatures ranging from 0 °C to rt; employing conditions as described in Angew .Chem.Int.Ed. 2008, 47 , 6802-6806; to afford a compound of formula (XV), where M is MgLiCl.

A compound of formula (XV), where M is Mg, is prepared by magnesium-halogen exchange with a compound of formula (XIV), where X is Cl or Br, and R 3 is an aryl group suitably substituted with Ci- 6 alkyl, OCi- 6 alkyl, C 3 - 6 cycloalkyl, and C 3 - 6 cycloalkyl substituted with CF3; and 5,6,7,8-tetrahydronaphthalen-2-yl; employing iPrMgCl; in a suitable solvent such as THF, and the like; at temperatures of about 0 °C; for a period of 2 h.

A compound of formula (XV), where M is Li; is prepared by lithium-halogen exchange with a compound of formula (XIV), where X is Cl or Br, and R 3 a suitably substituted aryl group; by treatment with n- BuLi; in a suitable solvent such as THF, and the like; at temperatures of about -78 °C; for a period of 2 h.

SCHEME 5

3

According to SCHEME 5, /c/V-butyl methyl(3-oxocyclobutyl)carbamate is reacted with a arylmetal halide compound of formula (XV), where X is I, Cl, or Br, and R 3 is an aryl group suitably substituted with Ci- 6 alkyl, OCi- 6 alkyl, C3- 6 cycloalkyl, and C3- 6 cycloalkyl substituted with CF3; and 5,6,7,8-tetrahydronaphthalen-2-yl in a suitable solvent such as THF and the like; at temperatures ranging between -78 °C and room temperature; to provide a compound of formula (XVI). Subsequent ionic reduction of a compound of formula (XVI) employing conditions known to one skilled in the art, such as by treatment with triethylsilane; in a suitable solvent such as trifluoracetic acid, or a mixture of trifluoracetic acid and DCM; at a temperature between 0 °C and room temperature; provides a compound of formula (XVII), where R 4 is CH3. In the cases where incomplete reduction to the cyclobutyl ring is observed, reaction times of up to 20 h, excess amounts of triethylsilane and TFA are employed to provide a compound of formula (XVII) where R 4 is CH3.

SCHEME 6

According to SCHEME 6, treatment of a compound of formula (XVI), where R 3 is an aryl group suitably substituted with Ci- 6 alkyl, OCi- 6 alkyl, C3- 6 cycloalkyl, and C 3 - 6 cycloalkyl substituted with CF3; and 5,6,7,8-tetrahydronaphthalen-2-yl, with triethylsilane; in a suitable solvent such as trifluoracetic acid, or a mixture of trifluoracetic acid and DCM; provides a compound of formula (XVIII).

A compound of formula (XVIII) is hydrogenated under conditions known to one skilled in the art, or as previously described, to provide a compound of formula (XVII). For example, a compound of formula (XVIII) is treated with 10% Pd/C; under at atmosphere of H2 (30 bar); in a solvent such as MeOH and the like; at temperature of 50 °C; for a period of 16 h to provide a compound of formula (XVII), where R 4 is CH3. If necessary, a compound of formula (XVII) is further purified under conditions know to one skilled in the art by using a 3 -step sequence involving (1) N- Boc protection, (2) purification using flash column chromatography on silica and (3) A -Boc deprotection using TFA in DCM.

SCHEME 7

According to SCHEME 7, Wittig olefmation of /c/7-butyl methyl(3- oxocyclobutyl)carbamate using NaH as base; in a suitable solvent such as DMSO; at a temperature of 80 °C; for a period of 16 h; provides tert- butyl (3- benzylidenecyclobutyl)(methyl)carbamate. Hydrogenation of /c/V-butyl (3- benzylidenecyclobutyl)(methyl)carbamate, followed by /V-Boc cleavage, is achieved using conditions well known to one skilled in the art, or as previously described to provide 3-benzyl-N- methylcyclobutan- 1 -amine.

SCHEME 8

According to SCHEME 8, l-bromo-3-methyl-5-vinylbenzene is reacted in a formal ketene [2+2] reaction in the presence of Tf20, DMA, and 2,4,6-collidine to give an intermediate iminium salt which is hydrolyzed in situ to provide 3-(3-bromo-5-methylphenyl)cyclobutan-l- one. 3-(3-Bromo-5-methylphenyl)cyclobutan-l-one is subsequently reacted, in a Suzuki cross coupling reaction, with 2,4,6-trimethyl-l,3,5,2,4,6-trioxatriborinane; a palladium catalyst such as [l,l'-bis(di-tert-butylphosphino)fenOcene]dichloropalladium( II) (PdCl2(dtbpf)), tetrakis(triphenylphosphine)palladium(0) (Pd(PPh3)4), [1,1'- bis(diphenylphosphino)ferrocene]dichloropalladium(II) (PdCl2(dppf)), palladium(II)bis(triphenylphosphine) dichloride (Pd(PPh3)2Cl2), XPhos-Pd-G2 precatalyst (chloro(2-dicyclohexylphosphino-2',4',6'-triisopropyl-l, 1 '-biphenyl) [2-(2 '-amino- 1, 1 '- biphenyl)]palladium(II)), and the like; a base such as K3PO4, aq. Na2C03, Na2C03, CS2CO3, and the like, in a suitable solvent such as 1,2-dimethoxy ethane, 1,4-dioxane, DMF, water, or a mixture thereof, at a temperature ranging from 60 to 180 °C, employing microwave or conventional heating, for a period of about 30 min to 16 h, to provide a compound of formula (XX), where R 3 is a suitably substituted phenyl as described in claim 1.

SCHEME 9

According to SCHEME 9, a compound of formula (XX), where R 3 is Ci-6alkyl, C3-6cycloalkyl, or a suitably substituted phenyl as described in claim 1, is reacted under reductive amination conditions, with a suitable amine such as methylamine, A-benzhydrylethanamine, and the like; a reducing agent such as sodium cyanoborohydride (NaBH 3 CN), NaBH(OAc) 3 , NaBHi, and the like; in the presence of an acidic additive such as titanium(IV) isopropoxide, acetic acid, and the like; in a suitable solvent such as methanol, DCM, 1,2-dichloroethane, THF, or a mixture thereof; at room-temperature; for a period of 14-24 h; to provide a compound of formula (XVII). Wherein when A-benzhydrylethanamine is used in the reductive amination, subsequent hydrogenolysis employing Pd/C; a suitable solvent such as a mixture of MeOH and aq. HC1; under an atmosphere of hydrogen gas at 15 psi; provides a compound of formula (XVII), where R 4 is CH2CH3.

SCHEME 10

According to SCHEME 10, a compound of Formula (I), where R 1 is hydrogen and X is CH2 or O, is prepared by conventional amide bond forming techniques such as coupling reactions which are well known to those skilled in the art (such as HATU (1- [bi s(di methyl ami no)m ethylene]- 1//- 1 ,2,3-triazolo[4,5-/]pyridinium 3-oxid hexafluorophosphate), BOP (benzotriazol- 1 -yloxy-tris(dimethylamino)phosphonium hexafluorophosphate), or conversion of the acid to an acid chloride). For example, reaction of a commercially available or synthetically accessible compound of formula (XVII), where R 3 is Ci- 6 alkyl; benzyl; phenyl; phenyl substituted with one or two members each independently selected from: halo, Ci- 6 alkyl, OCi- 6 alkyl, C3- 6 cycloalkyl, and C3- 6 cycloalkyl substituted with CF3; and 5,6,7,8-tetrahydronaphthalen-2-yl; is reacted with a synthetically accessible suitably substituted carboxylic acid of formula (XIX) (which includes compounds of formulas (XI), and (XIII)); and where the acid is activated with an appropriate activating reagent, for example a carbodiimide, such as N,N'-dicyclohexylcarbodiimide (DCC) or l-ethyl-3-(3- dimethylaminopropyl)carbodiimide (EDC, ED AC or EDCI) optionally in the presence of hydroxybenzotriazole (HOBt) and/or a catalyst such as 4-dimethylaminopyridine (DMAP); a halotrisaminophosphonium salt such as (benzotriazol-l-yloxy)tris(dimethylamino)phosphonium hexafluorophosphate (BOP), or bromotripyrrolidinophosphonium hexafluorophosphate (PyBroP ® ); a suitable pyridinium salt such as 2-chloro-l -methyl pyridinium chloride; or another suitable coupling agent such as N, N, N A"-tetram ethyl -()-( 1 //-benzotri azol -1 -y 1 )uroni um hexafluorophosphate (HBTU), 1 -[bis(dimethylamino)methylene]-l//-l ,2,3-triazolo[4,5- Z]pyridinium 3-oxid hexafluorophosphate (HATU), 2,4,6-tripropyl-l,3,5,2,4,6- trioxatriphosphorinane-2, 4, 6-trioxide (T3P ® ) and the like. Coupling reactions are conducted in a suitable solvent such as DCM, THF, DMF and the like, optionally in the presence of a tertiary amine such as N-methylmorpholine, N-ethyldiisopropylamine (DIPEA), or triethylamine (TEA), at a temperature ranging from 0 °C to rt, to provide a compound of Formula (I).

Where the compounds according to this invention have at least one chiral center, they may accordingly exist as enantiomers. Where the compounds possess two or more chiral centers, they may additionally exist as diastereomers. It is to be understood that all such isomers and mixtures thereof are encompassed within the scope of the present invention.

Compounds prepared according to the schemes described above may be obtained as single forms, such as single enantiomers, by form-specific synthesis, or by resolution. Compounds prepared according to the schemes above may alternately be obtained as mixtures of various forms, such as racemic (1 : 1) or non-racemic (not 1 : 1) mixtures. Where racemic and non- racemic mixtures of enantiomers are obtained, single enantiomers may be isolated using conventional separation methods known to one of ordinary skill in the art, such as chiral chromatography, supercritical fluid chromatography (SFC), recrystallization, diastereomeric salt formation, derivatization into diastereomeric adducts, biotransformation, or enzymatic transformation. Where regioisomeric or diastereomeric mixtures are obtained, as applicable, single isomers may be separated using conventional methods such as chromatography or crystallization.

The following specific examples are provided to further illustrate the invention and various preferred embodiments. EXAMPLES

In obtaining the compounds described in the examples below and the corresponding analytical data, the following experimental and analytical protocols were followed unless otherwise indicated.

Unless otherwise stated, reaction mixtures were magnetically stirred at room temperature (rt) under a nitrogen atmosphere. Where solutions were “dried,” they were generally dried over a drying agent such as Na2SC>4 or MgSCU Where mixtures, solutions, and extracts were “concentrated”, they were typically concentrated on a rotary evaporator under reduced pressure. Reactions under microwave irradiation conditions were carried out in a Biotage Initiator or CEM (Microwave Reactor) Discover instrument.

For the reactions conducted under continuous flow conditions, “flowed through a LTF- VS mixer” refers to the use of a Chemyx Fusion 100 Touch Syringe Pump that is in line via 1/16” PTFE tubing to a LTF-VS mixer (Little Things Factory GmbH (http://www.ltf- gmbh.com), unless otherwise indicated.

Normal -phase silica gel chromatography (FCC) was performed on silica gel (S1O2) using prepacked cartridges.

Preparative reverse-phase high performance liquid chromatography (RP HPLC) was performed on either:

METHOD A An Agilent HPLC with an Xterra Prep RP18 column (5 mM, 30 x 100 or 50 x 150mm) or an XBridge C18 OBD column (5 pM, 30 x 100 or 50 x 150mm), and a mobile phase of 5% ACN in 20mM NH4OH was held for 2 min, then a gradient of 5-99% ACN over 15 min, then held at 99% ACN for 5 min, with a flow rate of 40 or 80 mL/min. or

METHOD B An Agilent HPLC with an Xterra Prep RP18 column (5 pM, 30 x 100 or 50 x 150mm) or an XBridge C18 OBD column (5 pM, 30 x 100 or 50 x 150mm), and a mobile phase of 5% ACN H2O (both with 0.05% TFA) was held for 2 min, then a gradient of 5-99% ACN over 15 min, then held at 99% ACN for 5 min, with a flow rate of 40 or 80 mL/min. or

METHOD C. An ACCQ Prep HPLC with an XBridge Cl 8 OBD column (5 pM, 50 x

100), mobile phase of 5% ACN in H2O (both with 0.05% TFA) was held for 1 min, then a gradient of 5-95% ACN over 12 min, then held at 95% ACN for 2 min, with a flow rate of 80 mL/min. or

METHOD D. Preparative reverse-phase high performance liquid chromatography using a Phenomenex Lux Cellulose-1 150 x 4.6 mm, 5 pm column, mobile phase of 40% Methanol + 0.1% Diethylamine and 60 % CO2), isocratic.

Preparative supercritical-fluid chromatography (SFC) was performed using:

METHOD E On an SFC instrument with a Phenomenex Lux Cellulose-1 150 x 4.6 mm, 5 pm column, mobile phase of 40% Methanol + 0.1% Diethylamine and 60 % CO2), isocratic.

Mass spectra (MS) were obtained on an Agilent series 1100 MSD using electrospray ionization (ESI) in positive mode unless otherwise indicated. Calculated (calcd.) mass corresponds to the exact mass.

Nuclear magnetic resonance (NMR) spectra were obtained on Bruker -spectrometers. For the 1H spectra, all chemical shifts are reported in parts per million (5) units and are relative to the residual signal at 7.26, 3.31, and 2.50 ppm for CDCh, CD3OD and DMSO-i¾, respectively. Definitions for multiplicity are as follows: s = singlet, d = doublet, t= triplet, q = quartet, m = multiplet, br = broad. It will be understood that for compounds comprising an exchangeable proton, said proton may or may not be visible on an NMR spectrum depending on the choice of solvent used for running the NMR spectrum and the concentration of the compound in the solution.

Chemical names were generated using ChemDraw Ultra 17.1 (Cambridge Soft Corp., Cambridge, MA) or OEMetaChem VI.4.0.4 (Open Eye).

Compounds designated as R* or S* are enantiopure compounds where the absolute configuration was not determined.

Intermediate 1: (2s.4s)-6-Oxo-7-oxa-5-azaspiror3.41octane-2-carboxylic acid.

Step A: tert- Butyl 3-hvdroxyiminocvclobutanecarboxylate. To a solution of /er/-butyl 3- oxocyclobutane-l-carboxylate (100 g, 588 mmol) in ethanol (EtOH) (1.8 L) was added sodium acetate (NaOAc) (192 g, 2340 mmol) and hydroxylamine hydrochloride (81 g, 1166 mmol). The reaction mixture was stirred at reflux for 4 h then filtered through a pad of Celite ® and the pad was washed with EtOH. The combined filtrates were evaporated and the residue was taken up in ethyl acetate (EtOAc) and washed with water and brine. The organic layer was dried over magnesium sulfate, filtered, and concentrated to give the title compound (108 g, 584 mmol, 99% yield) as a white solid. MS (ESI): mass calcd. for C9H15NO3 185.1; m/z found, 186.2 [M+H] + . Step B: fer/-Butyl 3-nitrocvclobutanecarboxylate. To a suspension of urea hydrogen peroxide (164 g, 1.74 mol) in acetonitrile (MeCN) (1 L) was added a solution of trifluoroacetic anhydride (TFAA) (245 mL, 1.75 mol) in MeCN (500 mL) dropwise over 1 h at -10 °C. The reaction mixture was stirred at room temperature for 1 h. The solution was added to a solution of tert- butyl 3-hydroxyiminocyclobutanecarboxylate (108 g, 0.584 mol) and sodium phosphate dibasic (911 g, 6.42 mol) in MeCN (1 L) dropwise over 30 min at 80 °C. The reaction mixture was stirred at 80 °C for 30 min then filtered through a pad of Celite ® and the pad was washed with MeCN. The combined filtrates were diluted with EtOAc. The mixture was washed with water and brine. The organic layer was dried over magnesium sulfate, filtered, and evaporated. The residue was purified by flash column chromatography (FCC) on silica (0-20% EtOAc in heptane) to give the title compound (89.6 g, 445 mmol, 76% yield) as a yellow oil as a 1.3:1 mixture of cisltrans isomers. Compound does not ionize with ESI + LCMS.

Step C: cis-tert- Butyl 3-(hvdroxymethvn-3-nitro-cvclobutanecarboxylate. To a solution of tert- butyl 3-nitrocyclobutanecarboxylate (89.6 g, 445 mmol) in ACN (1 L) was added formaldehyde (37 wt% in water, 73 mL, 971 mmol). To the reaction mixture was added TEA (62 mL, 444 mmol) dropwise at 0 °C and the reaction was stirred at room temperature for 2 h. The reaction mixture was evaporated under reduced pressure and the residue was purified by FCC on silica (0-30% EtOAc in heptane) to give the title compound (38.2 g, 37% yield) as a white powder. MS (ESI): mass calcd. for C10H17NO5 231.2; m/z found, 254.1 [M+Na] + . /ra//.s-/<2/7-Butyl 3- (hydroxymethyl)-3-nitro-cyclobutanecarboxylate was formed, but not isolated.

Step D: cis-tert- Butyl 3 -amino-3 - cl obutanecarboxylate. To a solution of cis- /<2/7-butyl 3 -(hydroxymethyl)-3-nitro-cy cl obutanecarboxylate (38.2 g, 165 mmol) in EtOAc (600 mL) was added 10% Pd/C (1.9 g). The reaction mixture was stirred at 50 °C for 1 h under Eh (10 bar). The reaction mixture was filtered through a pad of Celite ® . To the filtrate was added 10% Pd/C (1.9 g). The reaction mixture was stirred at 50 °C for 2 h under Eh (10 bar). The reaction mixture was filtered through a pad of Celite ® and the Celite ® was washed with EtOAc. The combined filtrates were evaporated under reduced pressure and the residue was triturated with diethyl ether (EhO) to give the title compound (18.6 g, 55% yield) as a white powder. MS (ESI): mass calcd. for C10H19NO3 201.1; m/z found, 202.2 [M+H] + . ¾ NMR (300 MHz, DMSO- e) d 5.26 - 3.98 (m, 1H), 3.74 - 2.94 (m, 4H), 2.70 - 2.57 (m, 1H), 2.20 - 2.07 (m, 2H), 1.97 - 1.82 (m, 2H), 1.39 (s, 9H).

Step E: cis-tert- Butyl 6-oxo-7-oxa-5-azaspiror3.41octane-2-carboxylate. To a solution of cis-tert- butyl 3 -amino-3 -(hydroxymethyl)cy cl obutanecarboxylate (18.6 g, 92.4 mmol) in THF (300 mL) was added TEA (26 mL, 186 mmol). To the mixture was added a solution of triphosgene (9.6 g, 32.4 mmol) in THF (200 mL) dropwise at -10 °C and stirred at room temperature for 1 h. The reaction mixture poured into saturated sodium bicarbonate (NaHCCh) (600 mL) and the mixture was extracted with EtOAc. The combined organic layers were dried over magnesium sulfate, filtered and evaporated under reduced pressure. The residue was triturated with EhO to give the title compound (17.7 g, 84% yield) as a white powder. MS (ESI): mass calcd. for C11H17NO4 227.1; m/z found, 228.2 [M+H] + .

Step F -6-Oxo-7-oxa-5-azaspiror3.41octane-2-carboxylic acid. To TFA (180 mL, 235 mmol) was added cis-tert- butyl 6-oxo-7-oxa-5-azaspiro[3.4]octane-2-carboxylate (17.7 g, 77.9 mmol) in portions at 0 °C. The reaction mixture was stirred at room temperature for 1 h. The reaction mixture was evaporated under reduced pressure and the residue was triturated with Et20 to afford the title compound (12.9 g, 96% yield) as a white powder. MS (ESI): mass calcd. for C7H9NO3 171.0; m/z found, 172.1 [M+H] + . ¾ NMR (500 MHz, DMSO- e) d 12.3 (br s, 1H), 8.08 (s, 1H), 4.34 (s, 2H), 2.79 - 2.66 (m, 1H), 2.43 - 2.29 (m, 4H). Intermediate 2: (2r.4s)-6-Qxo-5-azaspiror3.41octane-2-carboxylic acid.

Step A: Ethyl 3-nitrocvclobutanecarboxylate. The title compound was prepared in a manner analogous to Intermediate 1, Steps A-B; using ethyl 3-oxocyclobutane-l-carboxylate instead of /er/-butyl 3-oxocyclobutane-l-carboxylate in Step A. Compound does not ionize with ESI + LCMS. ¾ NMR (300 MHz, CDCb) d 5.02 - 4.70 (m, 1H), 4.20 (q, J= 7.2 Hz, 2H), 3.04 - 2.71 (m, 5H), 1.29 (t, J= 7.0 Hz, 3H).

Step B Ethyl 3-(3-methoxy-3-oxo-propyO-3-nitro-cvclobutanecarboxylate. To a solution of ethyl 3-nitrocyclobutanecarboxylate (16.6 g, 95.6 mmol) in ACN (145 mL) was added methyl acrylate (10.3 mL, 114 mmol). To the reaction mixture was added DBU (7.1 mL, 47.6 mmol) dropwise at 0 °C and the reaction mixture was stirred at 0 °C for 1 h. The reaction mixture was diluted with saturated ammonium chloride and EtOAc and the layers were separated. The organic layer was dried over magnesium sulfate, filtered, and evaporated under reduced pressure. The residue was purified by FCC on silica (0-15% EtOAc in heptane) to give the title compound (13.6 g, 55% yield) as a colorless liquid. MS (ESI): mass calcd. for C11H17NO6 259.1; m/z found, 282.1 [M+Na] + . ¾ NMR (300 MHz, Chloroform- ) d 4.17 (q, J = 7.1 Hz, 2H), 3.70 (s, 3H),

3.12 - 2.79 (m, 3H), 2.69 - 2.49 (m, 2H), 2.48 - 2.21 (m, 4H), 1.27 (t, J = 7.1 Hz, 3H).

Step C: (2r.4C)-6-Oxo-5-azaspiror3.41octane-2-carboxylic acid. To a solution of c/.s-ethyl 3-(3- m ethoxy-3 -oxo-propyl)-3-nitro-cy cl obutanecarboxylate (13.6 g, 52.5 mmol) in MeOH (133 mL) was added nickel(II) chloride hexahydrate (12.5 g, 52.6 mmol). To the reaction mixture was added NaBHi (10 g, 264 mmol) in small portions at -10 °C and the reaction mixture was stirred at 0 °C for 1 h. To the reaction mixture was added aqueous K2CO3 (47 mL, 141 mmol, 3 M) dropwise at 0 °C (pH 10) and the reaction mixture was stirred at 0 °C for 1 h. The reaction mixture was filtered through a pad of Celite ® and the pad was washed with EtOH. The combined filtrates were concentrated under reduced pressure. Saponification of the crude ester to the carboxylic acid was observed at this stage. The crude residue was purified by FCC on silica eluting with chloroform:methanol:acetic acid (100:0:0 9:1:1) to give the title compound (4.8 g, 53% yield) as an off-white powder. MS (ESI): mass calcd. for CsHiiNCb 169.1; m/z found, 170.1 [M+H] + . ¾NMR (300 MHz, DMSO- e) d 7.97 (br s, 1H), 4.01 - 2.94 (m, 1H), 2.82 - 2.65 (m, 1H), 2.36 - 2.01 (m, 8H).

Intermediate 3: 3-(4-(fe/V-Butyl (phenyl )-N-methylcvclobutan-l -amine trifluoroacetate salt.

Step A: fer/-Butyl G-iA-Ifc/V-butyl (phenyl )-3-hvdroxycvclobutyl ((methyl (carbamate. (4 - tert- Butyl)phenyl)magnesium bromide (2 M in THF, 376 pL, 753 pmol) was added dropwise to a 0 °C stirring solution of fe/7-butyl methyl(3-oxocyclobutyl)carbamate (100 mg, 502 pmol) in tetrahydrofuran (THF) (1.7 mL). After the end of the addition, the ice bath was removed and the reaction mixture was stirred at room temperature. After 30 min, the reaction mixture was quenched by the addition of sat’d aq. MHCl (10 mL) and the resulting aqueous mixture was extracted with EtOAc (3x10 mL). The combined organic extracts were dried over NaiSCri and concentrated in vacuo. Purification of the resulting crude product by flash column chromatography on silica (0-100% EtOAc/Hex) afforded the title product (107 mg, 321 pmol, 64% yield) as a yellow oil, and as a ca. 4:1 mixture of cis- and trans- isomers, which was used in step B without further purification. MS (ESI): mass calcd. for C20H31NO3, 333.2; m/z found, 260.1 [M-CTLHs-OHf.

Step B: 3-(4-(/c/7-Butyl (phenyl )-N-methylcvclobutan- l -amine trifluoroacetate salt tert- Butyl (3 -(4-(/<2/7-butyl )phenyl)-3 -hydroxycycl obutyl )(methyl )carbam ate (107 mg, 321 pmol) was dissolved in trifluoroacetic acid (TFA) (1.07 mL) and the resulting solution was stirred at rt for 5 min. Triethylsilane (369 pL, 2.25 mmol) was added dropwise. After the end of the addition, the reaction mixture was stirred vigorously at room temperature for 1 h. Solvent was subsequently removed in vacuo to give the crude title product which was used without further purification. MS (ESI): mass calcd. for C15H23N, 217.2; m/z found, 218.1 [M+H] + .

Intermediate d: 3-(3-(/c/7-Butyl (phenyl )-N-methylcvclobutan-l -amine. TFA salt. phenvOmagnesium bromide lithium chloride complex. A vial was charged with a stir bar and lithium chloride (112 mg, 2.64 mmol), sealed with a septum and dried under vacuum with a heat gun. The vial was cooled to room temperature and backfilled with N2. Magnesium turnings (128 mg, 5.28 mmol) were then added quickly and the vial was again evacuated and backfilled with N2. THF (5.3 mL) was added and the resulting mixture was stirred at room temperature vigorously until all LiCl had been dissolved (ca. 5 min). Diisobutylaluminum hydride (1M in THF, 21.1 pL) was added dropwise and the resulting pale yellow solution was stirred vigorously at rt for 5 min. The reaction vial was cooled to 0 °C and l-bromo-3-(/er/-butyl)benzene (363 pL, 2.11 mmol) was added dropwise. The ice bath was removed and the resulting pale yellow mixture was stirred at room temperature for 2 h. The resulting brown solution containing the title product was used directly, assuming a concentration of 0.37 M.

Step B: 3-(3- Butyl)phenyl )-N-methylcvclobutan- l -amine trifluoroacetate salt. The title compound was prepared in a manner analogous to Intermediate 3, using (3 -{tert- butyl)phenyl)magnesium bromide, lithium chloride complex instead of 4 -(tert- butyl)phenyl)magnesium bromide in step A. MS (ESI): mass calcd. for C15H23N, 217.2; m/z found, 218.1 [M+H] + . Dimethylphenyl )-N-methylcvclobutan-l -amine trifluoroacetate salt.

Step A: fer/-Butyl (3-(3.5-dimethylphenyl )-3-hvdroxycvclobutyl )(m ethyl icarbamate. (3,5- Dimethylphenyl)magnesium bromide (0.5 M in THF, 8.73 mL, 4.37 mmol) was added dropwise to a 0 °C stirring solution of tert- butyl methyl(3-oxocyclobutyl)carbamate (670 mg, 3.36 pmol) in tetrahydrofuran (THF) (1.7 mL). After the end of the addition, the ice bath was removed and the reaction mixture was stirred at room temperature. After lh, the reaction mixture was quenched by the addition of sat’d aq. MLCl (10 mL) and the resulting aqueous mixture was extracted with EtOAc (3x10 mL). The combined organic extracts were dried over Na2S04 and concentrated in vacuo. Purification of the resulting crude product by flash column chromatography on silica (0-100% EtO Ac/Hex) afforded the title product (646 mg, 2.12 mmol, 63% yield) as a yellow oil which was used in step B without further purification. MS (ESI): mass calcd. for C18H27NO3, 305.2; m/z found, 232.2 [M-OLHs-OHf.

Step B: 3- Dimethylphenyl )-N-methylcvclobutan- l -amine trifluoroacetate salt /tv/- Butyl (3-(3,5-dimethylphenyl)-3-hydroxycyclobutyl)(methyl)carbamat e (670 mg, 2.19 mmol) was dissolved in trifluoroacetic acid (TFA) (7.3 mL) and the resulting solution was stirred at rt for 5 min. Triethylsilane (3.5 mL, 21.9 mmol) was added dropwise. After the end of the addition, the reaction mixture was stirred vigorously at room temperature for 20 h. Solvent was subsequently removed in vacuo to give the crude title product which was used without further purification. MS (ESI): mass calcd. for C13H19N, 189.2; m/z found, 190.2 [M+H] + .

Intermediate 6: 3-(3-Isopropylphenyl )-N-methylcvclobutan- l -amine trifluoroacetate salt.

Step A. tert- Butyl (3-hvdroxy-3-(3-Isopropylphenvncvclobutvn(methvncarbamate. In a round bottom flask, n- BuLi (1.6 M in hexanes, 1.2 mL, 1.8 mmol, 1.2 equiv) was added dropwise to a solution of l-bromo-3-isopropylbenzene (300 mg, 1.5 mmol, 1 equiv) in dry THF (10 mL, 7.7 mL/mmol) at - 78 °C. The reaction mixture was stirred at -78 °C for 40 min. Then, a solution of /tv /-butyl -N- ethyl-N-(3-oxocyclobutyl (carbarn ate (300 mg, 1.5 mmol, 1 equiv) in dry THF (2 mL) was added dropwise and the reaction mixture was stirred at the same temperature for additional 1.5 h. The reaction mixture was warmed to room temperature, quenched with saturated MLCl aqueous solution. The mixture was extracted with EtOAc, and the organic phase washed with brine. The organic layer was dried over anhydrous MgSCri, filtered, and the solvents removed under vacuum to yield the title product (480 mg, crude) as brown oil which was used in step B without further purification. MS (ESI): mass calcd. for C19H29NO3, 319.2; m/z found, 246.1 [M-C H 8 -OH] + .

Step B 3-(3-Isopropylphenyl )-N-methylcvclobut-2-en- 1 -amine. TFA salt. In a round bottom flask, trifluoroacetic acid (0.11 mL, 1 equiv) was added to a mixture of /er/-butyl (3 -hydroxy-3 - (3-isopropylphenyl)cyclobutyl)(methyl)carbamate (480 mg, 1.5 mmol, 1 equiv), triethylsilane (1.68 mL, 7 equiv) and dichloromethane (8 mL) at room temperature. The reaction mixture was stirred at room temperature for 3 h, then additional trifluoroacetic acid (0.78 mL) was added to the reaction mixture and stirred at room temperature for 16 h. Solvent was subsequently removed in vacuo , and excess trifluoroacetic acid was co-distilled with toluene. The crude title product thus obtained (302 mg, crude), as a brown oil, was used in Step C without further purification. MS (ESI): mass calcd. for C14H19N, 201.2; m/z found, 202.1 [M+H] + .

Step C. 3-(3-Isopropylphenyl )-N-methylcvclobutan- l -amine. TFA salt. In a high-pressure vessel, Palladium over carbon (10 %, 15% m/m) was added to a solution of 3-(3- Isopropylphenyl)-N-methylcyclobut-2-en-l -amine, trifluoroacetate salt (300 mg, 1.5 mmol) in methanol (50 mL, 33 mL/mmol). The vessel was sealed, and charged with hydrogen (30 bar), and heated at 50 °C for 16 h. The system was cooled down and the reaction mixture was filtered through Celite ® . The solvent was removed under vacuum to afford the crude title product (300 mg, crude) as pale brown oil, which was used without further purification. MS (ESI): mass calcd. for C14H21N, 203.2; m/z found, 204.2 [M+H] + .

Intermediate 7: 3-(3-Methoxyphenyl )-N-methylcvclobutan- l -amine trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 6, using l-bromo-3- methoxybenzene, instead of l-bromo-3 -isopropylbenzene and isopropylmagnesium chloride instead of «-BuLi in step A. MS (ESI): mass calcd. for C12H17N, 191.1; m/z found, 192.1 [M+H] + .

Intermediate 8: 3-(4-Isopropylphenyl )-N-methylcvclobutan- l -amine trifluoroacetate salt.

Step A. tert- butyl (3 -hydroxy-3 -(4-IsopropylphenvOcv cl obutvOfmethvOcarbamate. In a round bottom flask, n- BuLi (1.6 M in hexanes, 784 pL, 1.26 mmol, 1.25 equiv) was added dropwise to a solution of l-bromo-4-isopropylbenzene (200 mg, 1.00 mmol, 1 equiv) in dry THF (4 mL) at - 78 °C. The reaction mixture was stirred at -78 °C for 1 h. Then, a solution of /cvV-butyl-N-methyl- N-(3-oxocyclobutyl)carbamate (200 mg, 1.00 mmol, 1 equiv) in dry THF (4 mL) was added dropwise and the reaction mixture was stirred at the same temperature for additional 2 h. The reaction mixture was warmed to room temperature, quenched with saturated MLCl aqueous solution. The mixture was extracted with EtOAc, and the organic phase washed with brine. The organic layer was dried over anhydrous MgSCri, filtered, and the solvents removed under vacuum to yield the title product (321 mg, crude) as brown oil which was used without further purification. MS (ESI): mass calcd. for C19H29NO3, 319.2; m/z found, 246.1 [M-CiHx-OHjT Step B 3-(4-Isopropylphenyl )-N-methylcvclobut-2-en- l -amine trifluoroacetate salt. In a round bottom flask, trifluoroacetic acid (150 pL, 1 equiv) was added to a mixture of tert- butyl (3- hydroxy-3-(4-isopropylphenyl)cyclobutyl)(methyl)carbamate (641 mg, 2.01 mmol, 1 equiv), triethylsilane (2.24 mL, 7 equiv) and dichloromethane (10 mL) at room temperature. The reaction mixture was stirred at room temperature for 3 h, then additional trifluoroacetic acid (1 mL) was added to the reaction mixture and stirred at room temperature for 16 h. Solvent was subsequently removed in vacuo , and excess trifluoroacetic acid was co-distilled with toluene.

The crude title product thus obtained (404 mg, crude) as a brown oil was used without further purification. MS (ESI): mass calcd. for C14H19N, 201.2; m/z found, 202.1 [M+H] + .

Step C. 3-(4-Isopropylphenyl )-N-methylcvclobutan- l -amine trifluoroacetate salt. In a high- pressure vessel, Palladium over carbon (10 %, 15% m/m) was added to a 0 °C solution of 3-(4- isopropylphenyl)-N-methylcyclobut-2-en-l-amine, trifluoroacetate salt (404 mg) in methanol (50 mL) under an atmosphere of N2. The vessel was sealed, and charged with hydrogen (30 bar), and heated at 50 °C for 16 h. The system was cooled down and the reaction mixture was filtered through Celite ® . The solvent was removed under vacuum to afford the crude title product (408 mg, crude) as pale brown oil, which was taken to step D without further purification. MS (ESI): mass calcd. for C14H21N, 203.2; m/z found, 204.2 [M+H] + .

Step D. fe/7-Butyl O-f -isopropylphenvDcvclobutylYmethvDcarbamate. 3-(4-Isopropylphenyl)- N-methylcyclobutan-1 -amine, trifluoroacetate salt (408 mg), from step C above was re-dissolved in dichloromethane (9 mL, 4.5 mL/mmol). DMAP (123 mg, 1 mmol, 0.5 equiv), triethylamine (420 pL, 3 mmol, 1.5 equiv), and di-/c/7-butyl dicarbonate (922 pL, 4 mmol, 2 equiv) were added sequentially. The reaction mixture was stirred at room temperature for 72 h. The mixture was diluted with DCM and washed with saturated aqueous NaHCCb solution and water. The organic layer was dried over MgSCB, filtered and concentrated under reduced pressure. The crude N- Boc product was purified by flash column chromatography on silica gel using Heptane/EtOAc (100:0 to 90:10) as eluents, to provide tert- butyl (3-(4- isopropylphenyl)cyclobutyl)(methyl)carbamate (168 mg, 555 pmol, 28% yield), as a colorless oil. MS (ESI): mass calcd. for C19H21NO2, 303.2; m/z found, 248.2 [M-C 4 H 8 +H] + .

Step E 3-(4-Isopropylphenyl )-N-methylcvclobutan- l -amine trifluoroacetate salt tert- Butyl (3- (4-isopropylphenyl)cyclobutyl)(methyl)carbamate (168 mg, 555 pmol) from step D above was redissolved in DCM (3 mL). Trifluoroacetic acid (494 pL, 6 mmol, 12 equiv) was added and the reaction mixture was stirred at room temperature for 16 hours. Solvent was subsequently removed under vacuum, and the excess the trifluoroacetic acid was co-distilled with toluene to yield the title product as a brown oil. The product was used without further purification. MS (ESI): mass calcd. for C14H21N, 203.2; m/z found, 204.1 [M+H] + .

Intermediate 9: 3-(3-(7c/7-Butyl -fluorophenyl )-N-methylcvclobutan- l -amine trifluoroacetate salt.

Step A fert-Butyl O-O-t/c/y-butyl )-4-fluorophenyl )-3-hvdroxycvclobutyl )(m ethyl icarbamate.

In a round bottom flask, n- BuLi (1.6 M in hexanes, 588 pL, 941 pmol, 1.25 equiv) was added dropwise to a solution of 4-bromo-2-(/c/7-butyl)-l -fluorobenzene (174 mg, 753 pmol, 1 equiv) in dry THF (4 mL) at - 78 °C. The reaction mixture was stirred at -78 °C for 1 h. Then, a solution of /er/-butyl-N-methyl-N-(3-oxocyclobutyl)carbamate (150 mg, 753 mihoΐ, 1 equiv) in dry THF (4 mL) was added dropwise and the reaction mixture was stirred at the same temperature for additional 2 h. The reaction mixture was warmed to room temperature, quenched with saturated NH4CI aqueous solution. The mixture was extracted with EtOAc, and the organic phase washed with brine. The organic layer was dried over anhydrous MgSCri, filtered, and the solvents removed under vacuum to yield the title product (265 mg, crude) as a gold oil which was used in step B without further purification. MS (ESI): mass calcd. for C20H30FNO3, 351.2; m/z found, 278.0 [M-C 4 H 8 -OH] + .

Step B 3-(3- Butyl )-4-fluorophenyl )-N-methylcvclobutan- 1 -amine trifluoroacetate salt. In a round bottom flask, trifluoroacetic acid (112 pL, 1 equiv) was added to a mixture of tert- butyl (3-(3-(fer/-butyl)-4-fluorophenyl)-3-hydroxycyclobutyl)(meth yl)carbamate (529 mg, 1.51 mmol, 1 equiv), triethylsilane (1.68 mL, 10.5 mmol, 7 equiv) and dichloromethane (8 mL) at room temperature. The reaction mixture was stirred at room temperature for 4 h, then additional amount of trifluoroacetic acid (800 pL) was added to the reaction mixture and stirred at room temperature for 72 h. Solvent was subsequently removed in vacuo , and excess trifluoroacetic acid was co-distilled with toluene. The crude title product thus obtained (354 mg, crude) as a brown oil, was used in Step C without further purification. MS (ESI): mass calcd. for C15H22FN, 235.2; m/z found, 236.2 [M+H] + .

Step C. fc/7- Butyl (l-fl-ftert-butvO-d-fluorophenvOcvclobutvOfmethvOcarbamate. 3 -(3 -{tert- Butyl)-4-fluorophenyl)-N-methylcyclobutan-l-amine, trifluoroacetate salt (522 mg), as prepared in step B above was redissolved in dichloromethane (13 mL). DMAP (135 mg, 1.00 mmol, 0.5 equiv), triethylamine (618 pL, 4.44 mmol, 2 equiv), and di-te/V-butyl dicarbonate (1.02 mL, 4.44 mmol, 2 equiv) were added sequentially. The reaction mixture was stirred at room temperature for 16 h. The mixture was diluted with DCM and washed with saturated aqueous NaHCCh solution and water. The organic layer was dried over MgSCri, filtered and concentrated under reduced pressure. The crude N- Boc product was purified by flash column chromatography on silica gel using Heptane/EtOAc (100:0 to 90:10) as eluents to provide tert- butyl (3-(4- isopropylphenyl)cyclobutyl)(methyl)carbamate (744 mg, 775 pmol, 35% yield) as a colorless oil. MS (ESI): mass calcd. for C20H30FNO2, 335.2; m/z found, 278.1 [M-C 4 H 8 +H] + .

Step D. 3-(3-(7 <2/7 -Butyl )-4-fluorophenyl )-N-methylcvclobutan- l -amine trifluoroacetate salt.

/ert-Butyl (3-(3-(tert-butyl)-4-fluorophenyl)cyclobutyl)(methyl)carbama te (260 mg, 775 pmol) from step C above was redissolved in DCM (4 mL). Trifluoroacetic acid (1.4 mL) was added and the reaction mixture was stirred at room temperature for 16 hours. Solvent was subsequently removed under vacuum, and the excess the trifluoroacetic acid was co-distilled with toluene to yield the title product as a brown oil. The product was used without further purification. MS (ESI): mass calcd. for C15H22FN, 235.2; m/z found, 236.2 [M+H] + .

Intermediate 10: A f -Methyl-3-(3-( l -(trifluorom ethyl level opropyl iphenyl )cvclobutan- l -amine trifluoroacetate salt.

Step A fc/7-Butyl (3-hvdroxy-3-(3-(T-

(trifluorom ethyl level opropyl iphenyl level obutyl )(m ethyl icarbamate. In a round bottom flask, n- BuLi (1.6 M in hexanes, 784 pL, 1.26 mmol, 1.2 equiv) was added dropwise to a solution of 1- bromo-3-(l-(trifluoromethyl)cyclopropyl)benzene (266 mg, 1.00 mmol, 1 equiv) in dry THF (4mL) at - 78 °C. The reaction mixture was stirred at -78 °C for 1 h. Then, a solution of tert- butyl-N-methyl-N-(3-oxocyclobutyl)carbamate (200 mg, 1.00 mmol, 1 equiv) in dry THF (4 mL) was added dropwise and the reaction mixture was stirred at the same temperature for additional 2 h. The reaction mixture was warmed to room temperature, quenched with saturated NH4CI aqueous solution. The mixture was extracted with EtOAc, and the organic phase washed with brine. The organic layer was dried over anhydrous MgSCri, filtered, and the solvents removed under vacuum to yield the title product (480 mg, crude) as a gold oil which was used in step B without further purification. MS (ESI): mass calcd. for C20H26F3NO3, 385.2; m/z found, 312.1 [M-C 4 H 8 -OH] + .

Step B A f -Methyl-3-(3-(l -(trifluoromethyl level opropyl iphenyl )cvclobut-2-en-l -amine trifluoroacetate salt. In a round bottom flask, trifluoroacetic acid (149 pL, 1 equiv) was added to a mixture of /er/-butyl (3 -hydroxy-3 -(3 -(1-

(trifluoromethyl)cyclopropyl)phenyl)cyclobutyl)(methyl)ca rbamate (774 mg, 2.01 mmol, 1 equiv), triethylsilane (2.24 mL, 7 equiv) and dichloromethane (10 mL) at room temperature. The reaction mixture was stirred at room temperature for 4 h, then additional trifluoroacetic acid (1 mL) was added to the reaction mixture and stirred at room temperature for 16 h. Solvent was subsequently removed in vacuo , and excess trifluoroacetic acid was co-distilled with toluene.

The crude title product thus obtained (536 mg, crude) as a brown oil was used in Step C without further purification. MS (ESI): mass calcd. for C15H16F3N, 267.1; m/z found, 268.2 [M+H] + .

Step C. A f -Methyl-3-(3- l -(trifluoromethyl)cvclopropyl jphenyl )cvclobutan- l -amine trifluoroacetate salt. In a high pressure vessel, Palladium over carbon (10 %, 15% m/m) was added to a solution of N-methyl-3-(3-(l-(trifluoromethyl)cyclopropyl)phenyl)cyclobu t-2-en-l- amine, intermediate from step B, (530 mg, 2 mmol) in methanol (50 mL, 25 mL/mmol). The vessel was sealed, and loaded with hydrogen (30 bar), and heated at 50 °C for 16 h. The system was cooled down, the reaction mixture was filtered through Celite ® . The solvent was removed under vacuum to give the crude title product, N-methyl-3-(3-(l-

(trifluoromethyl)cyclopropyl)phenyl)cyclobutan-l -amine, trifluoroacetate salt, as pale brown oil (540 mg, 2 mmol, 99% yield), which was purified as follows: The crude title product was redissolved in dichloromethane (9 mL, 4.5 mL/mmol), DMAP (122 mg, 1 mmol, 0.5 equiv), triethylamine (0.42 mL, 3 mmol, 1.5 equiv), and di-/er/-butyl dicarbonate (0.9 mL, 4 mmol, 2 equiv) were added sequentially. The reaction mixture was stirred at room temperature for 48 h. The mixture was diluted with DCM and washed with saturated aqueous NaHCCh solution and water. The organic layer was dried over MgSCri, filtered and concentrated under reduced pressure. The crude N-Boc product was purified by flash column chromatography on silica gel using Heptane/EtOAc (100:0 to 90:10) as eluents, to provide tert- butyl methyl(3-(3-(l- (trifluoromethyl)cyclopropyl)phenyl)cyclobutyl)carbamate as a colorless oil (186 mg, 0.5 mmol, 25% yield), which was redissolved in DCM (4 mL). Trifluoroacetic acid (0.45 mL, 6 mmol, 12 equiv) was added and the reaction mixture was stirred at room temperature for 16 hours. Solvent was subsequently removed under vacuum, and the excess the trifluoroacetic acid was co-distilled with toluene to yield the title product as a brown oil. The product was used in the next step without further purification. MS (ESI): mass calcd. for C15H18F3N, 269.1; m/z found, 270.1 [M+H] + .

Intermediate 11: /V-Methyl-3-(o-tolyl)cyclobutan-l -amine, trifluoroacetate salt.

Step A. tert- Butyl (3 -hydroxy-3 -(o-tolvDcv cl obutvOfmethvOcarbamate. In a round bottom flask, isopropylmagnesium chloride (1.1 mL, 2.2 mmol, 2 M, 1.5 equiv) was added dropwise to a solution of 2-iodotoluene (287 pL, 2.2 mmol, 1.5 equiv) in dry THF (15 mL, 7 mL/mmol) at 0 °C. The reaction mixture was stirred at 0 °C for 1 h. Then, a solution of /er/-butyl-N-methyl-N- (3-oxocyclobutyl)carbamate (300 mg, 1 equiv) in dry THF (3 mL) was added dropwise and the reaction mixture was stirred at the same temperature for additional 1.5 h. The reaction mixture was warmed to room temperature, quenched with saturated MLCl aqueous solution. The mixture was extracted with EtOAc and the organic phase washed with brine. The organic layer was dried over anhydrous MgSCri, filtered, and solvent removed under vacuum to afford the crude title product (439 mg), which was used in step B without further purification. MS (ESI): mass calcd. for C17H25NO3, 291.2; m/z found, 218.1 [M-C4Hs-OH] + .

Step B A f -Methyl-3-(o-tolyl )cvclobut-2-en- l -amine trifluoroacetate salt. In a round bottom flask, trifluoroacetic acid (112 pL, 1 equiv) was added to a mixture of tert- butyl (3 -hydroxy-3 - (o-tolyl)cyclobutyl)(methyl)carbamate (439 mg, 1.51 mmol, 1 equiv), triethylsilane (1.68 mL, 7 equiv) and dichloromethane (5 mL) at room temperature. The reaction mixture was stirred at room temperature for 3 h, then additional trifluoroacetic acid (800 pL) was added to the reaction mixture and stirred at room temperature for 72 h. Solvent was subsequently removed in vacuo , and excess trifluoroacetic acid was co-distilled with toluene. The crude title product thus obtained (261 mg, crude) as a brown oil was used in Step C without further purification. MS (ESI): mass calcd. for C12H15N, 173.1; m/z found, 174.1 [M+H] + .

Step C. A-Methyl-3-(o-tolvDcvclobutan-l -amine trifluoroacetate salt. In a high-pressure vessel, 10% palladium over carbon (40 mg, 15% m/m) was added to a solution of /V-m ethyl-3 -(o tolyl)cyclobut-2-en-l -amine, trifluoroacetate salt (261 mg, 1.51 mmol) in methanol (50 mL). The vessel was sealed, and charged with hydrogen (30 bar), and heated at 50 °C for 16 h. The system was cooled down and the reaction mixture was filtered through Celite ® . The solvent was removed under vacuum to afford the crude title product (264 mg, crude) as a brown oil, which was used without further purification. MS (ESI): mass calcd. for C12H17N, 175.1; m/z found, 176.1 [M+H] + .

Intermediate 12: N-Methyl-3-(ffl-tolv0cvclobutan-l -amine trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 6, using l-iodo-3- methylbenzene instead of l-bromo-3-isopropylbenzene and isopropylmagnesium chloride instead of n- BuLi in step A. MS (ESI): mass calcd. for C12H17N, 175.1; m/z found, 176.1 [M+H] + .

Intermediate 13: 3 -(3 -Ethyl phenyl )-N-methylcvclobutan- 1 -amine trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 8, using l-bromo-3- ethylbenzene instead of l-bromo-3-isopropylbenzene in step A. MS (ESI): mass calcd. for C13H19N, 189.2; m/z found, 190.2 [M+H] + .

Intermediate 14: 3-(2.3-Dimethylphenyl )-N-methylcvclobutan- 1 -amine trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 8, using l-bromo-2,3- dimethylbenzene instead of l-bromo-3-isopropylbenzene in step A. MS (ESI): mass calcd. for C13H19N, 189.2; m/z found, 190.2 [M+H] + .

Intermediate 15: 3-(4-Cvclopropylphenyl )-N-methylcvclobutan- 1 -amine trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 9, using l-bromo-4- cyclopropylbenzene instead of l-bromo-3-isopropylbenzene in step A. MS (ESI): mass calcd. for C14H19N, 201.2; m/z found, 202.2 [M+H] + .

Intermediate 16: 3-(3.4-Dimethylphenyl )-N-methylcvclobutan- 1 -amine trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 8, using 4-bromo-l,2- dimethylbenzene instead of l-bromo-3-isopropylbenzene in step A. MS (ESI): mass calcd. for C13H19N, 189.2; m/z found, 190.1 [M+H] + .

Intermediate 17: 3-(3-Fluoro-4-isopropylphenyl )-N-methylcvclobutan- 1 -amine trifluoroacetate salt. Step A /c/V-Butyl (3-(3-fluoro-4-isopropylphenvn-3-hvdroxycvclobutvn(methvncar bamate. In a round bottom flask, n- BuLi (1.6 M in hexanes, 784 pL, 1.26 mmol, 1.25 equiv) was added dropwise to a solution of 4-bromo-2-fluoro-l-isopropylbenzene (218 mg, 1.00 mmol, 1 equiv) in dry THF (10 mL) at - 78 °C. The reaction mixture was stirred at -78 °C for 1 h. Then, a solution of /c/7-butyl-N-methyl-N-(3-oxocyclobutyl (carbarn ate (300 mg, 1.50 mmol, 1 equiv) in dry THF (4 mL) was added dropwise and the reaction mixture was stirred at the same temperature for additional 2 h. Then, the reaction mixture was warmed to room temperature, quenched with saturated NH4CI aqueous solution. The mixture was extracted with EtOAc, and the organic phase washed with brine. The organic layer was dried over anhydrous MgSCri, filtered, and the solvents removed under vacuum to yield the title product (480 mg, crude) as a brown oil, which was used in step B without further purification. MS (ESI): mass calcd. for C19H28FNO3, 337.2; m/z found, 264.1 [M-C H 8 -OH] + .

Step B 3-(3-Fluoro-4-isopropylphenyl )-N-methylcvclobutan- 1 -amine trifluoroacetate salt. In a round bottom flask, trifluoroacetic acid (3.3 mL, 45 mmol, 45 equiv) was added dropwise to a solution of /er/-butyl (3-(3-fluoro-4-isopropylphenyl)-3-hydroxycyclobutyl)(methyl) carbamate (339 mg, 1 mmol, 1 equiv), triethylsilane (1.1 mL, 7 mmol, 7 equiv) in DCM (3.3 mL, 3.3 mL/mmol) at 0 °C. The reaction mixture was stirred at room temperature for 2.5 h. Then, the solvent was removed under reduced pressure, co-distilled with toluene, and the excess of Et3SiH and TFA removed under high vacuum for 30 min, to give crude title product which was used without further purification. MS (ESI): mass calcd. for C14H20FN, 221.2; m/z found, 222.2 [M+H] + .

Intermediate 18: 3-(4-Fluoro-3-isopropylphenyl )-N-methylcvclobutan- l -amine trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 17, using l-fluoro-4- iodo-2-isopropylbenzene instead of l-bromo-3-isopropylbenzene in step A. MS (ESI): mass calcd. for C14H20FN, 221.2; m/z found, 222.2 [M+H] + . Dimethylphenyl )-N-methylcvclobutan- l -amine trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 17, using l-bromo-2,4- dimethylbenzene instead of l-bromo-3-isopropylbenzene in step A. MS (ESI): mass calcd. for C13H19N, 189.2; m/z found, 190.2 [M+H] + . Intermediate 20: 3-(4-Ethylphenyl )-N-methylcvclobutan- 1 -amine trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 17, using l-bromo-4- ethylbenzene instead of l-bromo-3-isopropylbenzene in step A. MS (ESI): mass calcd. for C13H19N, 189.2; m/z found, 190.1 [M+H] + .

Intermediate 21: 3-(4-Ethyl-3-methylphenyl)-N-methylcvclobutan- l -amine trifluoroacetate salt. The title compound was prepared in a manner analogous to Intermediate 17, using 4-bromo-l- ethyl-2-methylbenzene instead of l-bromo-3-isopropylbenzene in step A. MS (ESI): mass calcd. for C14H21N, 203.2; m/z found, 204.2 [M+H] + .

Intermediate 22: 3-(3-Cvclobutylphenyl )-N-methylcvclobutan- 1 -amine trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 17, using l-bromo-3- cyclobutylbenzene instead of l-bromo-3-isopropylbenzene in step A. MS (ESI): mass calcd. for C15H21N, 215.2; m/z found, 216.2 [M+H] + . Intermediate 23 : 3 -(4-Cvclopropyl -3 - ethyl phenyl )-N-m ethyl cvclobutan-1 -amine. trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 17, using 4-bromo-l- cyclopropyl-2-methylbenzene instead of l-bromo-3-isopropylbenzene in step A. MS (ESI): mass calcd. for C15H21N, 215.2; m/z found, 216.2 [M+H] + .

Intermediate 24: 3 -(3 -Cvclopropyl -4-methyl phenyl )-N-m ethyl cvclobutan-1 -amine. trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 17, using 4-bromo-2- cyclopropyl- 1-m ethylbenzene instead of l-bromo-3-isopropylbenzene in step A. MS (ESI): mass calcd. for C15H21N, 215.2; m/z found, 216.2 [M+H] + .

Intermediate 25: N-Methyl-3-(5.6.7.8-tetrahvdronaphthalen-2-yl level obutan-1 -amine. trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 17, using 6-bromo- 1,2,3,4-tetrahydronaphthalene instead of l-bromo-3-isopropylbenzene in step A. MS (ESI): mass calcd. for C15H21N, 215.2; m/z found, 216.2 [M+H] + . Intermediate 26: 3-Benzyl-N-methylcvclobutan-l -amine trifluoroacetate salt. Step A. tert- Butyl (3-benzylidenecvclobutvD(methvDcarbamate. In a round bottom flask, a solution of sodium hydride (80 mg, 2.0 mmol) in dry DMSO (16 mL) was stirred at 80 °C for 20 min. The mixture was cooled to 0 °C, and a solution of benzyltriphenylphosphonium chloride (1.25 g, 3.2 mmol) in dry DMSO (7 mL) was added. The resulting mixture was stirred at room temperature for 30 min. Then, a solution of fe/7-butyl methyl(3-oxocyclobutyl)carbamate (400 mg, 2.0 mmol) in dry DMSO (7 mL) was added and the resulting reaction mixture was stirred at 80 °C for 16 h. The reaction mixture was diluted with EtOAc and washed with water. The organic layer was dried over MgS04, filtered and concentrated under reduced pressure to afford a dark orange oil. Purification by flash column chromatography using Heptane/EtOAc (100:0 to 90: 10) afforded the title product as a beige oil which was used in step B without further purification. MS (ESI): mass calcd. for C17H23NO2, 273.2; m/z found, 218.1 [M-C4H8+H] + .

Step B (S-BenzylcvclobutvDrinethvDcarbamic acid. In a high pressure vessel, Palladium over carbon (10 %, 15% m/m) was added to a solution of tert- butyl (3- benzylidenecyclobutyl)(methyl)carbamate (296 mg, 1.08 mmol) in methanol (50 mL). The vessel was sealed, and charged with hydrogen (30 bar), and heated at 50 °C for 16 h. The system was cooled down and the reaction mixture was filtered through Celite ® . The solvent was removed under vacuum to afford the crude title product (299 mg, crude) as a pale brown oil, which was used in step C without further purification. MS (ESI): mass calcd. for C17H25NO2, 219.1; m/z found, 220.2 [M+H] + .

Step C. 3-Benzyl-N-methylcvclobutan- l -amine trifluoroacetate salt. In a round bottom flask, trifluoroacetic acid (1.9 mL, 24 mmol, 24 equiv) was added to a solution of (3-benzyl- cyclobutyl)-methyl-carbamic acid tert- butyl ester (298 mg, 1 mmol, 1 equiv) in DCM (5.5 mL, 5.5 mL/mmol), and the reaction mixture was stirred at room temperature for 16 h. The solvent was removed under vacuum and the excess of trifluoroacetic acid was co-distilled with toluene to give (3-benzyl-cyclobutyl)-methyl-amine as a brown oil. The product was used without further purification. MS (ESI): mass calcd. for C12H17N, 175.1; m/z found, 176.2 [M+H] + .

Intermediate 27: 3- Butyl )-N-methylcvclobutan- l -amine.

In a round bottom flask, a mixture of 3-(fer/-butyl)cyclobutan-l-one (200 mg, 1.5 mmol, 1 equiv), methylamine (7.53 mL, 2 M in THF, 15.1 mmol, 10 equiv), titanium(IV) isopropoxide (0.92 mL, 3.0 mmol, 2 equiv), and 1,2-dichloroethane (32 mL, 21 mL/mmol) was stirred under nitrogen at room temperature for 16 h. Then, sodium triacetoxyborohydride (3.2 g, 15.0 mmol, 10 equiv) was added and the reaction mixture stirred at room temperature for additional 16 h. The reaction mixture was quenched with a saturated aqueous NLLCl solution and the product was extracted with EtOAc. The combined organic extracts were washed with water and brine, dried over MgSCri, filtered, and concentrated under reduced pressure to afford a beige solid which was purified by silica flash column chromatography using DCMMeOILNEb (100:0:0 to 98:2:0.1) as eluents to give (3 -/tv /-butyl -cyclobutyl)- ethyl -a ine, as a mixture of cis/trans isomers. MS (ESI): mass calcd. for C9H19N, 141.2; m/z found, 142.2 [M+H] + .

Intermediate 28: 3-(3-Chloro-5-methylphenyl )-N-methylcvclobutan- 1 -amine trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 17, using l-bromo-3- chloro-5-methylbenzene instead of 4-bromo-2-fluoro-l -isopropylbenzene in Step A. MS (ESI): mass calcd. for C12H16CIN, 209.1; m/z found, 210.1 [M+H] + . Dimethylphenyl )-N-ethylcvclobutan- 1 -amine. HC1 salt. Step A: 3-(3-Bromo-5-methylphenvOcvclobutan-l-one. T1¾0 (3.73 mL, 22.1 mmol) was added to a solution consisting of A,/V-dimethylacetamide (2.06 mL, 22.2 mmol) and 1,2-dichloroethane (8 mL). The resultant mixture was stirred at room -temperature for 30 minutes and then treated with a solution consisting of l-bromo-3-methyl-5-vinylbenzene (3.35 g, 17.0 mmol), 2,4,6- collidine (2.92 mL, 22.1 mmol), and 1,2-dichloroethane (8 mL). The reaction mixture was stirred at 90 °C for 16 hours before cooling to room-temperature and diluting with water (32 mL). The resultant mixture was stirred at 90 °C for another 18 hours before cooling to room-temperature, pouring it into LhO (100 mL), and extracting with dichloromethane (50 mL x 3). The combined organic extracts were dried over anhydrous Na2SC>4, filtered, and concentrated to dryness under reduced pressure, the resulting residue was purified by FCC (eluent: petroleum ether: ethyl acetate = 0: 1 to 5: 1) to afford the title compound (2.5 g, 62%), as a yellow oil. ¾ NMR (400 MHz, CDCb) d 7.26 - 7.23 (m, 2H), 7.03 (s, 1H), 3.67 - 3.57 (m, 1H), 3.53 - 3.43 (m, 2H), 3.28 - 3.18 (m, 2H), 2.35 (s, 3H).

Step B: 3- DimethylphenvDcvclobutan-l-one. A mixture of 3-(3-bromo-5- methylphenyl)cyclobutanone (1.00 g, 4.18 mmol), 2,4,6-trimethyl-l,3,5,2,4,6-trioxatriborinane (788 mg, 6.28 mmol), K2CO3 (1.73 g, 12.5 mmol), 1,4-dioxane (10 mL), and H2O (2.5 mL) was added to a 20 mL tube. The reaction mixture was sparged with Ar for 5 minutes and then treated with Pd(dppf)Ch*CH2Cb (342 mg, 0.419 mmol). The reaction mixture was sparged with Ar for another 5 minutes and the resultant mixture was stirred while heating at 90 °C for 2 hours before cooling to room -temperature, pouring it into H2O (100 mL), and extracting with ethyl acetate (50 mL x 3). The combined organic extracts were dried over anhydrous Na2SC>4, filtered, and concentrated to dryness under reduced pressure, the resulting residue was purified by FCC (eluent: petroleum ether: ethyl acetate = 0:1 to 5:1) to afford the title compound (180 mg, 25%) as a yellow oil. ¾ NMR (400 MHz, CDCb) d 6.95 - 6.91 (m, 3H), 3.67 - 3.58 (m, 1H), 3.53 - 3.42 (m, 2H), 3.30 - 3.20 (m, 2H), 2.34 (s, 6H).

Step C: (Ί s.3s)-N-Benzhydryl-3- dimethylphenyl )-N-ethylcvclobutan- l -amine.

NaBH(OAc)3 (2.43 g, 11.5 mmol) was added to a solution consisting of 3-(3,5- dimethylphenyl)cyclobutanone (1.0 g, 5.7 mmol), A-benzhydrylethanamine (1.21 g, 5.73 mmol), and dichloromethane (10 mL). Then, AcOH (0.25 mL) was added to the reaction mixture. The reaction mixture was stirred at room -temperature for 12 hours before pouring it into sat. NaHCCb (20 mL) and extracting with dichloromethane (30 mL x 3). The combined organic extracts were washed with brine (20 mL), dried over anhydrous Na2SC>4, filtered, and concentrated to dryness under reduced pressure, the resulting residue was purified by FCC (eluent: petroleum ether: ethyl acetate = 1:0 to 5:1) to afford the still-impure product (1.2 g, crude), as a clear oil. The post chromatographic product was combined with another crude batch of product (prepared separately from 180 mg of 3-(3,5-dimethylphenyl)cyclobutanone, following the same procedure from above) and further purified by preparative HPLC using a Phenomenex Gemini NX-C18 150 mm x 40 mm x 5 pm column (eluent: 85% to 90% (v/v) CTLCN and H2O with 0.04% NIL + 10 mM NH4HCO3). The thus purified product was suspended in water (10 mL), the mixture frozen using dry ice/acetone, and then lyophilized to dryness to afford the title product (200 mg). MS (ESI): mass calcd. for C27H31N, 369.3; m/z found, 370.3 [M+H] + .

Step D: DimethylphenvD-N-ethylcvclobutan-l-amine. HC1 salt. (Ts.3s)-N- Benzhydryl-3-(3,5-dimethylphenyl)-N-ethylcyclobutan-l-amine (200 mg, 0.541 mmol), methanol (30 mL), dry Pd/C (100 mg, 10 % dry Pd/C), and cone. HC1 (0.1 mL) were added to a 100 mL hydrogenation bottle. The resultant mixture was stirred under Eh (15 psi) at room- temperature for 16 hours. The suspension was filtered through a pad of Celite ® and the pad washed with methanol (100 mL). The mixture was concentrated to dryness under reduced pressure to give the product (200 mg), which was used in the next step without further purification. LC-MS (ESI): RT = 0.77 min, mass calcd. For C14H21N 203.17 m/z found 204.3 [M+H] + . MS (ESI): mass calcd. for C14H21N, 203.2; m/z found, 204.3 [M+H] + .

Intermediate 30: 3-Cvclohexyl-N-methylcvclobutan-l-amine.

The title compound was prepared in a manner analogous to Intermediate 27, using 3- cyclohexylcyclobutan-l-one instead of 3-(/er/-butyl)cyclobutan-l-one. MS (ESI): mass calcd. for C11H21N, 167.2; m/z found, 168.2 [M+H] + . Intermediate 31 : N-Methyl-3-(4-( l -methylcvclopropyl )phenyl )cvclobutan- l -amine trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 17, using l-bromo-4-(l- methylcyclopropyl)benzene instead of 4-bromo-2-fluoro-l-isopropylbenzene in Step A. MS (ESI): mass calcd. for C15H21N, 215.2; m/z found, 216.2 [M+H] + .

Intermediate 32: N-Methyl-3-(3-( l -methylcvclopropyl )phenyl)cvclobutan- l -amine trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 17, using l-bromo-3-(l- methylcyclopropyl)benzene instead of 4-bromo-2-fluoro-l-isopropylbenzene in Step A. MS (ESI): mass calcd. for C15H21N, 215.2; m/z found, 216.2 [M+H] + . Intermediate 33: 3 -(3 -Ethyl -5-methyl phenyl )-N-methylcvclobutan- l -amine trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 17, using l-bromo-3- ethyl-5-methylbenzene instead of 4-bromo-2-fluoro-l -isopropylbenzene in Step A. MS (ESI): mass calcd. for C14H21N, 203.2; m/z found, 204.1 [M+H] + .

Intermediate 34: 3-(3-Cvclopropyl-5-methylphenyl )-N-methylcvclobutan- l -amine trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 17, using l-bromo-3- cyclopropyl-5-methylbenzene instead of 4-bromo-2-fluoro-l -isopropylbenzene in Step A. MS (ESI): mass calcd. for C15H21N, 215.2; m/z found, 216.2 [M+H] + .

Intermediate 35: 3-(3-Isopropyl-5-methylphenyl )-N-methylcvclobutan- 1 -amine trifluoroacetate salt.

Step A: l-Bromo-3-methyl-5-(prop-l-en-2-vDbenzene. Nitrogen gas (N2) was bubbled through a solution of l-bromo-3-iodo-5-methylbenzene (1.0 g, 3.37 mmol) in 1,4-dioxane (7.5 mL) and water (2.5 mL) for 5 min in a pressure flask. While maintaining N2 bubbling, 4, 4,5,5- tetramethyl-2-(prop-l-en-2-yl)-l,3,2-dioxaborolane (0.62 mL, 3.37 mmol), cesium carbonate (2.37 g, 7.26 mmol), and dppfPdCh (404 mg, 495 pmol) were added sequentially. Bubbling of N2 was continued for 5 additional minutes, after which time the pressure cap was sealed with a screw cap and the reaction mixture was stirred at 90 °C. After 16 h, the reaction mixture was filtered through a pad of Celite ® . The filtrate was collected, diluted with EtOAc, and washed with water and brine. The organic phase was separated and dried over MgSCri, filtered and concentrated under reduced pressure, the resulting residue was purified by flash column chromatography using Heptane/EtOAc (100:0 to 90: 10) as eluents to afford the title compound (382 mg, 1.81 mmol) as a brownish oil. ¾ NMR (300 MHz, Chloroform- ): 7.39 (s, 1H), 7.23

(s, 1H), 7.18 (s, 1H), 5.22 (d, J = 73.9 Hz, 2H), 2.33 (s, 3H), 2.11 (s, 3H).

Step B: 3-(3-Isopropyl-5-methylphenvD-N-methylcvclobutan-l-amine. The title compound was prepared in a manner analogous to Intermediate 17, using l-bromo-3-methyl-5-(prop-l-en-2- yl)benzene instead of 4-bromo-2-fluoro-l -isopropylbenzene in Step A. MS (ESI): mass calcd. for C15H23N, 217.1; m/z found, 218.2 [M+H] + .

Intermediate 36: 3-(3-Cvclopropylphenyl )-N-methylcvclobutan- 1 -amine trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 17, using l-bromo-3- cyclopropylbenzene instead of 4-bromo-2-fluoro-l -isopropylbenzene in Step A. MS (ESI): mass calcd. for C14H19N, 201.2; m/z found, 202.1 [M+H] + . Intermediate 37: 3-(2.3-Dihydro- l H-inden-5-yl )-N-methylcvclobutan- 1 -amine trifluoroacetate salt.

The title compound was prepared in a manner analogous to Intermediate 17, using 5-bromo-2,3- dihydro-lH-indene instead of 4-bromo-2-fluoro-l -isopropylbenzene in Step A. MS (ESI): mass calcd. for C14H19N, 201.2; m/z found, 202.2 [M+H] + .

Intermediate 38: 3-(Bicvclor4.2.01octa- 1 (6).2.4-trien-3-yl )-N-methylcvclobutan- 1 -amine. trifluoroacetate salt. The title compound was prepared in a manner analogous to Intermediate 17, using 3- bromobicyclo[4.2.0]octa-l(6),2,4-triene instead of 4-bromo-2-fluoro-l -isopropylbenzene in Step A. MS (ESI): mass calcd. for C13H17N, 187.1; m/z found, 188.1 [M+H] + . Example 1: (2s.4S)-N-Methyl-6-oxo-N-(ns.3S)-3-phenylcvclobutvD-7-oxa-5- azaspiror3.41octane-2-carboxamide.

Triethylamine (110 pL, 794 pmol) was added dropwise to a stirring 0 °C mixture of N-methyl-3- phenylcyclobutan-1 -amine (ca. 4:1 cis:trans mixture, 40.0 mg, 248 pmol), (2s,4s)-6-oxo-7-oxa- 5-azaspiro[3.4]octane-2-carboxylic acid (Intermediate 1, 42.4 mg, 248 pmol) and HATU (117 mg, 273 pmol) in N,N-dimethylacetamide (2.5 mL). The reaction mixture was allowed to stir at room temperature for 14 h and subsequently diluted with water (1 mL). Purification by RP- HPLC (Method B, ACN/H2O, 0.05% TFA) afforded the title product, in 49% isolated yield, and (2s,4S)-N-Methyl-6-oxo-N-((lr,3R)-3-phenylcyclobutyl)-7-oxa- 5-azaspiro[3.4]octane-2- carboxamide (Example 2), in 9% yield. MS (ESI): mass calcd. for C18H22N2O3, 314.2; m/z found, 315.1 [M+H] + . ¾NMR (500 MHz, METHANOL- dt) d 7.36-7.11 (m, 5H), 4.79-4.70 (m, 0.5H), 4.52 (s, 1H), 4.48 (s, 1H), 4.45-4.34 (m, 0.5H), 3.24-3.04 (m, 2H), 2.95 (m, 3H), 2.63- 2.18 (m, 8H).

Example 2: -N-Methyl-6-oxo-N-(Y lr.3R)-3-phenylcvclobutvD-7-oxa-5- azaspiror3.41octane-2-carboxamide.

The title compound was isolated from Example 1. MS (ESI): mass calcd. for C18H22N2O3, 314.2; m/z found, 315.1 [M+H] + . Ή NMR (500 MHz, METHANOL-6/2) d 7.38-7.13 (m, 5H), 5.16-5.03 (m, 0.5H), 4.69-4.54 (m, 0.5H), 4.48 (s, 1H), 4.45 (s, 1H), 3.58-3.43 (m, 1H), 3.14- 2.99 (m, 4H), 2.86-2.62 (m, 2H), 2.58-2.31 (m, 5H). Example 3: -3-(3-Cvclobutylphenvncvclobutvn-N-methyl-6-oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 10, using 3-(3- cyclobutylphenyl)-N-methylcyclobutan-l -amine, trifluoroacetate salt (Intermediate 22) instead of 3-(3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C22H28N2O3, 368.2; m/z found, 369.2 [M+H] + . Ή NMR (400 MHz, Methanol - /2) d 7.29 - 7.13 (m, 1H), 7.10 - 7.00 (m, 3H), 4.82 - 4.68 (m, 0.5H), 4.50 (m, 2H), 4.45 - 4.32 (m, 0.5H), 3.61 - 3.46 (m, 1H), 3.25 - 3.03 (m, 2H), 2.95 (m, 3H), 2.63 - 1.97(m, 13H), 1.92 - 1.81 (m, 1H).

Example 4: (2s.4S)-N-(Yls.3S)-3-(4-( ' fer/-ButvDphenvDcvclobutvD-N-methyl-6-oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide. The title compound was prepared in a manner analogous to Example 1, using 3-(4-(tert- butyl)phenyl)-N-methylcyclobutan-l -amine, trifluoroacetate salt, (Intermediate 3, cis/trans mixture) instead of N-methyl-3-phenylcyclobutan-l -amine. Purification by RP-HPLC (Method B, ACN/H2O, 0.05% TFA) afforded the title product, in 72% isolated yield, and (2s,4S)-N- ((lr,3R)-3-(4-(/er/-Butyl)phenyl)cyclobutyl)-N-methyl-6-oxo- 7-oxa-5-azaspiro[3.4]octane-2- carboxamide (Example 5), in 5% isolated yield. MS (ESI): mass calcd. for C22H30N2O3, 370.2; m/z found, 371.3 [M+H] + . Ή NMR (500 MHz, METHANOL-6/2) d 7.42-7.31 (m, 2H), 7.19 (m, 2H), 4.80-4.71 (m, 0.4H), 4.54 (s, 1.1H), 4.50 (s, 0.9H), 4.44-4.35 (m, 0.6H), 3.25-3.07 (m, 2H), 2.97 (m, 3H), 2.65-2.44 (m, 6H), 2.42-2.33 (m, 1H), 2.29-2.19 (m, 1H), 1.32 (m, 9H).

Example 5: ButvOphenvOcvclobutvO-N-methyl-6-oxo-7-oxa-5- azaspirc>r3.41octane-2-carboxamide.

The title compound was isolated from Example 4. MS (ESI): mass calcd. for C22H30N2O3, 370.2; m/z found, 371.1 [M+H] + . Ή NMR (500 MHz, METHANOL-6/2) d 7.45-7.36 (m, 2H), 7.30-7.25 (m, 2H), 5.17-5.04 (m, 0.5H), 4.66-4.57 (m, 0.5H), 4.52-4.45 (m, 2H), 3.57-3.39 (m, 1H), 3.06 (m, 3H), 2.66-2.86 (m, 2H), 2.59-2.32 (m, 7H), 1.36-1.30 (m, 9H).

Example 6: (2s.4S)-N-( ' (Ts.3S)-3-(3-( ' fer/-ButvDphenvDcvclobutvD-N-methyl-6-oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 1, using 3 -(3 - tert- butyl)phenyl)-N-methylcyclobutan-l -amine, trifluoroacetate salt (Intermediate 4, cis/trans mixture) instead of N-methyl-3-phenylcyclobutan-l -amine. Purification by RP-HPLC (Method B, ACN/H2O, 0.05% TFA) afforded the title product in 89% isolated yield. MS (ESI): mass calcd. for C22H30N2O3, 370.2; m/z found, 371.3 [M+H] + . Ή NMR (500 MHz, METHANOL-6/2) d 7.30-7.19 (m, 3H), 7.12-7.00 (m, 1H), 4.78-4.69 (m, 0.5H), 4.54-4.46 (m, 2H), 4.44-4.35 (m, 0.5H), 3.25-3.03 (m, 2H), 2.95 (m, 3H), 2.65-2.43 (m, 6H), 2.40-2.31 (m, 1H), 2.28-2.17 (m, 1H), 1.31 (m, 9H).

Example 7: ButvOphenvOcvclobutvO-N-methyl-6-oxo-5- azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 6, using (2r,4s)-6-oxo-5- azaspiro[3.4]octane-2-carboxylic acid (Intermediate 2) instead of (2s,4s)-6-oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxylic acid (Intermediate 1). Purification by RP-HPLC (Method B, ACN/H2O, 0.05% TFA) afforded the title product. MS (ESI): mass calcd. for C23H32N2O2, 368.2; m/z found, 369.2 [M+H] + . 1 HNMR (500 MHz, METHANOL-A) d 7.29-7.18 (m, 3H), 7.13-7.00 (m, 1H), 4.76 (m, 0.5H), 4.48-4.35 (m, 0.5H), 3.28-3.09 (m, 2H), 2.96 (m, 3H), 2.65-2.54 (m, 2H), 2.52-2.18 (m, 10H), 1.31 (m, 9H). Example 8: DimethylphenvDcvclobutvD-N-methyl-6-oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 1, using 3-(3,5- dimethylphenyl)-N-methylcyclobutan-l -amine, trifluoroacetate salt (Intermediate 5, cis/trans mixture) instead of N-methyl-3-phenylcyclobutan-l -amine. Purification by RP-HPLC (Method

B, ACN/H2O, 0.05% TFA) afforded the title product and (2s,4S)-N-((lr,3R)-3-(3,5- dimethylphenyl)cyclobutyl)-N-methyl-6-oxo-7-oxa-5-azaspiro[3 .4]octane-2-carboxamide

(Example 9). MS (ESI): mass calcd. for C20H26N2O3, 342.2; m/z found, 343.2 [M+H] + . 1 HNMR (500 MHz, METHANOLS) d 6.90 - 6.77 (m, 3H), 4.77 - 4.67 (m, 0.5H), 4.50 (m, 2H), 4.43 - 4.30 (m, 0.5H), 3.23 - 3.04 (m, 2H), 2.99 - 2.90 (m, 3H), 2.61 - 2.40 (m, 6H), 2.38 - 2.11 (m, 8H).

In round bottom flask, propylphosphonic anhydride EtOAc solution (T3P ® ) (1 mL, 1.7 mmol,

50%, 1.25 equiv) and DIPEA (655 pL, 3.7 mmol, 2.5 equiv) was added to a solution of (2s, 4s)-

6-oxo-7-oxa-5-azaspiro[3.4]octane-2-carboxylic acid (Intermediate 1, 282 mg, 1.65 mmol, 1.2 equiv) in dry DMF (7.5 mL, 5 mL/mmol). The mixture was stirred for 10 min at room temperature, then a solution of 3-(3-Isopropylphenyl)-N-methylcyclobutan-l-amine

(Intermediate 6, 300 mg, 1.5 mmol, 1 equiv) in DMF (1 mL) was added, and the reaction mixture was stirred at room temperature for additional 16 h. The reaction mixture was diluted with

EtOAc and an aqueous saturated NaHC03 solution. The organic layer was dried over MgS04, filtered and the solvents removed under vacuum to get a dark brown oil. The crude was purified by flash column chromatography (silica, DCM/MeOH (9: 1) in DCM from 0% to 10%) to give the product as a mixture of the cis/trans isomers. This mixture was further purified by supercritical fluid chromatography (SFC) (Method D, Isocratic mode: 40% Methanol + 0.1% Diethylamine and 60% CO2) to yield the cis isomer, the title product (52.3 mg, 0.15 mmol, 10% yield). MS (ESI): mass calcd. for C21H28N2O3, 356.2; m/z found, 357.2 [M+H] + . ¾NMR (400 MHz, CD 3 OD) 6 7.31 - 7.15 (m, 1H), 7.14 - 6.99 (m, 3H), 4.80 - 4.66 (m, 0.5H), 4.50 (m, 2H), 4.45 - 4.32 (m, 0.5H), 3.26 - 3.01 (m, 2H), 2.96 (m, 3H), 2.93 - 2.81 (m, 1H), 2.67 - 2.40 (m, 6H), 2.43 - 2.28 (m, 1H), 2.29 - 2.16 (m, 1H), 1.24 (m, 6H).

Example 11 : -3-(3-Methoxyphenvncvclobutvn-N-methyl-6-oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 10, using 3-(3- methoxyphenyl)-N-methylcyclobutan-l-amine, trifluoroacetate salt (Intermediate 7) instead of 3- (3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6) and using HBTU instead of T3P ® as the coupling reagent. Purification by SFC (Method E) afforded the title product and (2s,4S)-N-((lr,3R)-3-(3-methoxyphenyl)cyclobutyl)-N-methyl-6 -oxo-7-oxa-5- azaspiro[3.4]octane-2-carboxamide (Example 12). MS (ESI): mass calcd. for C19H24N2O4,

344.2; m/z found, 345.2 [M+H] + . ¾NMR (400 MHz, Methanol-A) d 7.25 - 7.17 (m, 1H), 6.87 - 6.81 (m, 1H), 6.81 - 6.72 (m, 2H), 4.79 - 4.69 (m, 0.5H), 4.50 (m, 2H), 4.45 - 4.31 (m, 0.5H), 3.78 (m, 3H), 3.22 - 3.00 (m, 2H), 2.95 (m, 3H), 2.63 - 2.42 (m, 6H), 2.40 - 2.32 (m, 1H), 2.27 - 2.16 (m, 1H).

Example 12: (2s.4S)-N-(Tl r.3R)-3-(3-Methoxyphenyl level obutyl )-N-methyl-6-oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide. The title compound was prepared in a manner analogous to Example 10, using 3-(4- isopropylphenyl)-N-methylcyclobutan-l -amine, trifluoroacetate salt (Intermediate 8) instead of 3-(3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C21H28N2O3, 356.2; m/z found, 357.2 [M+H] + . 1 HNMR (400 MHz, Methanol-A) d 7.17 (m,

4H), 4.81 - 4.68 (m, 0.5H), 4.50 (m, 2H), 4.46 - 4.31 (m, 0.5H), 3.25 - 2.81 (m, 6H), 2.62 - 2.43 (m, 6H), 2.41 - 2.28 (m, 1H), 2.28 - 2.13 (m, 1H), 1.23 (m, 6H).

Example 14: (2s.4S)-N-((ls.3S)-3-(3-( ' fer/-ButvD-4-fluorophenvDcvclobutvD-N-methyl-6-oxo-7- oxa-5-azaspiror3.41octane-2-carboxamide. The title compound was prepared in a manner analogous to Example 10, using 3-(3-(/er/-butyl)- 4-fluorophenyl)-N-methylcyclobutan-l -amine, trifluoroacetate salt (Intermediate 9) instead of 3- (3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C22H29FN2O3, 388.2; m/z found, 389.1 [M+H] + . Ή NMR (400 MHz, Methanol - /2) d 7.21 - 7.05 (m, 2H), 7.04 - 6.89 (m, 1H), 4.80 - 4.64 (m, 0.5H), 4.50 (m, 2H), 4.46 - 4.32 (m, 0.5H), 3.26 -

3.00 (m, 2H), 2.95 (m, 3H), 2.68 - 2.39 (m, 6H), 2.41 - 2.25 (m, 1H), 2.27 - 2.09 (m, 1H), 1.37 (s, 9H).

Example 15: (2s.4S)-N-Methyl-6-oxo-N-((ls.3S)-3-(3-(l- ( ' trifluoromethvDcvclopropyDphenvDcvclobutvD-7-oxa-5-aza spiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 10, using N-methyl-3-(3-(l- (trifluoromethyl)cyclopropyl)phenyl)cyclobutan-l -amine, trifluoroacetate salt (Intermediate 10) instead of 3-(3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). Purification by SFC (Method E) afforded the title product and (2s,4S)-N-methyl-6-oxo-N-((lr,3R)-3-(3-(l- (trifluoromethyl)cyclopropyl)phenyl)cyclobutyl)-7-oxa-5-azas piro[3.4]octane-2-carboxamide (Example 16). MS (ESI): mass calcd. for C22H25F3N2O3, 422.2; m/z found, 423.2 [M+H] + . ¾ NMR (400 MHz, Methanol-A) d 7.38 - 7.18 (m, 4H), 4.77 - 4.67 (m, 0.5H), 4.50 (m, 2H), 4.47 - 4.32 (m, 0.5H), 3.27 - 3.0.3 (m, 2H), 2.95 (m, 3H), 2.71 - 2.12 (m, 8H), 1.38 - 1.27 (m, 2H), 1.06 (m, 2H).

Example 16: (2s^S)-N-Methyl-6-oxo-N-(Ylr.3R)-3-(3-(T-

( ' trifluoromethvDcvclopropyDphenvDcvclobutvD-7-oxa-5-aza spiror3.41octane-2-carboxamide.

The title compound was isolated from Example 15. MS (ESI): mass calcd. for C22H25F3N2O3, 422.1; m/z found, 423.1 [M+H] + . 1 HNMR (400 MHz, Methanol-A) d 7.43 (s, 1H), 7.39 - 7.26 (m, 3H), 5.17 - 5.00 (m, 0.5H), 4.67 - 4.51 (m, 1.5H), 4.46 (m, 2H), 3.66 - 3.43 (m, 1H), 3.18 -

3.05 (m, 4H), 2.92 - 2.65 (m, 2H), 2.60 - 2.32 (m, 6H), 1.42 - 1.29 (m, 2H), 1.16 - 0.98 (m,

2H).

Example 17: (2s.4S)-N-Methyl-6-oxo-N-(ns.3S)-3-(o-tolvncvclobutvn-7-oxa- 5- azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 10, using N-methyl-3-(o- tolyl)cyclobutan-l -amine, trifluoroacetate salt (Intermediate 11) instead of 3-(3- isopropylphenyl)-N-methylcyclobutan-l -amine (Intermediate 6). Purification by SFC (Method E) afforded the title product and (2s,4S)-N-methyl-6-oxo-N-((lr,3R)-3-(o-tolyl)cyclobutyl)-7- oxa-5-azaspiro[3.4]octane-2-carboxamide (Example 18). MS (ESI): mass calcd. for C19H24N2O3, 328.2; m/z found, 329.1 [M+H] + . ¾NMR (400 MHz, Methanol-A) d 7.33 - 6.93 (m, 4H), 4.84 - 4.71 (m, 0.5H), 4.51 (m, 2H), 4.46 - 4.35 (m, 0.5H), 3.27 - 3.0.1 (m, 2H), 2.94 (m, 3H), 2.71 - 2.12 (m, 11H).

Example 18: (2s.4S)-N-Methyl-6-oxo-N-(nr.3R)-3-(o-tolvncvclobutvn-7-oxa- 5- azaspiror3.41octane-2-carboxamide.

The title product was isolated from Example 17. MS (ESI): mass calcd. for C19H24N2O3, 328.2; m/z found, 329.1 [M+H] + . ¾NMR (400 MHz, Methanol-A) d 7.47 (d, J= 7.6 Hz, 1H), 7.29 - 7.01 (m, 3H), 5.15 - 4.95 (m, 0.5H), 4.66 - 4.51 (m, 1H), 4.46 (m, 2H), 3.72 - 3.53 (m, 1H),

3.17 - 3.04 (m, 4H), 2.89 - 2.62 (m, 2H), 2.60 - 2.25 (m, 6H), 2.21 (m, 3H).

Example 19: (2s.4S)-N-Methyl-6-oxo-N-((ls.3S)-3-(m-tolvDcvclobutvD-7-oxa -5- azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 10, using N-Methyl-3-(w- tolyl)cyclobutan-l -amine, trifluoroacetate salt (Intermediate 12) instead of 3-(3- isopropylphenyl)-N-methylcyclobutan-l -amine (Intermediate 6). Purification by SFC (Method E) afforded the title product and (2s,4S)-N-methyl-6-oxo-N-((lr,3R)-3-(w-tolyl)cyclobutyl)-7- oxa-5-azaspiro[3.4]octane-2-carboxamide (Example 20). MS (ESI): mass calcd. for

C19H24N2O3, 328.2; m/z found, 329.1 [M+H] + . 1 HNMR (400 MHz, Methanol-A) d 7.23 - 7.08 (m, 1H), 7.07 - 6.95 (m, 3H), 4.78 - 4.67 (m, 0.5H), 4.50 (m, 2H), 4.45 - 4.32 (m, 0.5H), 3.23 - 3.02 (m, 3H), 2.95 (m, 3H), 2.62 - 2.30 (m, 10H). Example 20: (2s.4S)-N-Methyl-6-oxo-N-(( 1 r.3R)-3-(m-tolyl level obutyl )-7-oxa-5- azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 10, using N-Methyl-3-(w- tolyl)cyclobutan-l -amine, trifluoroacetate salt (Intermediate 13) instead of 3-(3- isopropylphenyl)-N-methylcyclobutan-l -amine (Intermediate 6). MS (ESI): mass calcd. for C20H26N2O3, 342.2; m/z found, 343.2 [M+H] + . 1 HNMR (400 MHz, Methanol-A) d 7.30 - 7.09 (m, 1H), 7.10 - 6.96 (m, 3H), 4.79 - 4.67 (m, 0.5H), 4.50 ( m, 2H), 4.47 - 4.30 (m, 0.5H), 3.25 - 3.03 (m, 2H), 2.95 (m, 3H), 2.70 - 2.16 (m, 10H), 1.22 (m, 3H).

Example 22: DimethylphenvDcvclobutvD-N-methyl-6-oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide. The title compound was prepared in a manner analogous to Example 10, using 3 -(2,3- dimethylphenyl)-N-methylcyclobutan-l -amine, trifluoroacetate salt (Intermediate 14) instead of 3-(3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). Purification by SFC (Method E) afforded the title product and (2s,4S)-N-((lr,3R)-3-(2,3-dimethylphenyl)cyclobutyl)- N-methyl-6-oxo-7-oxa-5-azaspiro[3.4]octane-2-carboxamide (Example 23). MS (ESI): mass calcd. for C20H26N2O3, 342.2; m/z found, 343.2 [M+H] + . Ή NMR (400 MHz, Methanol - /2) d 7.14 - 7.02 (m, 2H), 7.02 - 6.95 (m, 1H), 4.81 - 4.71 (m, 0.5H), 4.50 (m, 2H), 4.46 - 4.34 (m, 0.5H), 3.42 - 3.33 (m, 1H), 3.28 - 3.14 (m, 0.5H), 3.14 - 3.00 (m, 0.5H), 2.91 (m, 3H), 2.69 - 2.39 (m, 6H), 2.39 - 2.28 (m, 1H), 2.26 (m, 3H), 2.17 (m, 4H).

Example 23: DimethylphenvDcvclobutvD-N-methyl-6-oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide. The title product was isolated from Example 22. MS (ESI): mass calcd. for C20H26N2O3, 342.2; m/z found, 343.2 [M+H] + . 1 HNMR (400 MHz, Methanol-A) d 7.30 (d, J= 7.6 Hz, 1H), 7.15 -

6.98 (m, 2H), 5.04 - 4.92 (m, 0.5H), 4.56 - 4.45 (m, 3H), 3.73 - 3.57 (m, 1H), 3.15 - 3.04 (m,

4H), 2.87 - 2.74 (m, 1H), 2.74 - 2.62 (m, 1H), 2.59 - 2.27 (m, 9H), 2.11 (m, 3H). Example 24: -3-(4-CvclopropylphenvDcvclobutvD-N-methyl-6-oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide. The title compound was prepared in a manner analogous to Example 10, using 3-(4- cyclopropylphenyl)-N-methylcyclobutan-l -amine, trifluoroacetate salt (Intermediate 15) instead of 3-(3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C21H26N2O3, 354.2; m/z found, 355.2 [M+H] + . Ή NMR (400 MHz, Methanol - /2) d 7.17 - 7.05 (m, 2H), 7.07 - 6.88 (m, 2H), 4.81 - 4.67 (m, 0.5H), 4.48 (m, 2H), 4.42 - 4.30 (m, 0.5H),

3.25 - 2.99 (m, 2H), 2.95 (m, 3H), 2.68 - 2.13 (m, 8H), 1.96 - 1.77 (m, 1H), 1.00 - 0.84 (m, 2H), 0.71 - 0.52 (m, 2H).

Example 25: -3-(3.4-Dimethylphenvncvclobutvn-N-methyl-6-oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 10, using 3 -(3, 4- dimethylphenyl)-N-methylcyclobutan-l -amine, trifluoroacetate salt (Intermediate 16) instead of 3-(3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C20H26N2O3, 342.2; m/z found, 343.1 [M+H] + . 1 HNMR (400 MHz, Methanol-A) d 7.09 - 6.91

(m, 3H), 4.81 - 4.63 (m, 0.5H), 4.50 (m, 2H), 4.44 - 4.30 (m, 0.5H), 3.25 - 3.02 (m, 2H), 2.95 (m, 3H), 2.61 - 2.41 (m, 6H), 2.40 - 2.09 (m, 10H).

Example 26: -3-(3-Fluoro-4-isopropylphenvDcvclobutvD-N-methyl-6-oxo-7- oxa-5-azaspiror3.41octane-2-carboxamide. The title compound was prepared in a manner analogous to Example 10, using 3-(3-Fluoro-4- isopropylphenyl)-N-methylcyclobutan-l -amine, trifluoroacetate salt, Intermediate 17, instead of 3-(3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C21H27FN2O3, 374.2; m/z found, 375.1 [M+H] + . Ή NMR (400 MHz, Methanol - /7) d 7.28-7.04 (m, 1H), 7.09 - 7.04 (m, 1H), 6.98 (d, J= 11.8 Hz, 1H), 4.85 - 4.39 (m, 0.5H), 4.56 (m, 2H),

4.50- 4.39 (m, 0.5H), 3.30 - 3.12 (m, 3H), 3.03 - 2.97 (m, 3H), 2.68 - 2.49 (m, 6H), 2.45 - 2.35 (m, 1H), 2.32 - 2.22 (m, 1H), 1.30 (m, 6H).

Example 27: -3-(4-Fluoro-3-isopropylphenvDcvclobutvD-N-methyl-6-oxo-7- oxa-5-azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 10, using 3-(4-Fluoro-3- isopropylphenyl)-N-methylcyclobutan-l -amine, trifluoroacetate salt (Intermediate 18) instead of 3-(3-Isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C21H27FN2O3, 374.2; m/z found, 375.2 [M+H] + . Ή NMR (400 MHz, Methanol - /7) d 7.17 - 7.02 (m, 2H), 7.01 - 6.88 (m, 1H), 4.79 - 4.65 (m, 0.5H), 4.50 (m, 2H), 4.46 - 4.32 (m, 0.5H), 3.26 - 3.02 (m, 3H), 2.95 (m, 3H), 2.65 - 2.13 (m, 8H), 1.25 (m, 6H). Example 28: -3-(2.4-Dimethylphenvncvclobutvn-N-methyl-6-oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide. The title compound was prepared in a manner analogous to Example 10, using 3 -(2,4- dimethylphenyl)-N-methylcyclobutan-l -amine, trifluoroacetate salt (Intermediate 19) instead of 3-(3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C20H26N2O3, 342.2; m/z found, 343.2 [M+H] + . 1 HNMR (400 MHz, Methanol-A) d 7.16 - 7.05 (m, 1H), 7.02 - 6.90 (m, 2H), 4.81 - 4.69 (m, 0.5H), 4.50 (m, 2H), 4.46 - 4.33 (m, 0.5H), 3.28 -

3.02 (m, 2H), 2.92 (m, 3H), 2.67 - 2.41 (m, 6H), 2.36 - 2.09 (m, 8H).

Example 29: -3-(4-EthylphenvDcvclobutvD-N-methyl-6-oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 10, using 3-(4- ethylphenyl)-N-methylcyclobutan-l -amine, trifluoroacetate salt (Intermediate 20) instead of 3- (3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C20H26N2O3, 342.2; m/z found, 343.1 [M+H] + . ¾NMR (400 MHz, Methanol-A) d 7.19 - 7.11 (m, 4H), 4.82 - 4.66 (m, 0.5H), 4.50 (m, 2H), 4.47 - 4.31 (m, 0.5H), 3.24 - 3.01 (m, 2H), 2.95

(m, 3H), 2.69 - 2.13 (m, 10H), 1.20 (t, J= 7.6 Hz, 3H).

Example 30: (2s.4S)-N-((ls.3S)-3-(4-Ethyl-3-methylphenvncvclobutvn-N-met hyl-6-oxo-7-oxa- 5-azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 10, using 3-(4-Ethyl-3- methylphenyl)-N-methylcyclobutan-l-amine, trifluoroacetate salt (Intermediate 21) instead of 3- (3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C21H28N2O3, 356.2; m/z found, 357.2 [M+H] + . ¾NMR (400 MHz, Methanol-A) d 7.09 - 6.97 (m, 3H), 4.78 - 4.65 (m, 0.5H), 4.54 - 4.46 (m, 2H), 4.42 - 4.30 (m, 0.5H), 3.24 - 3.02 (m, 2H), 2.95 (m, 3H), 2.65 - 2.14(m, 14H), 1.17 (m, 3H). Example 31 : -3-(4-Cvclopropyl-3-methylphenvDcvclobutvD-N-methyl-6-oxo- 7-oxa-5-azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 10, using 3-(4-cyclopropyl- 3-methylphenyl)-N-methylcyclobutan-l-amine, trifluoroacetate salt (Intermediate 23) instead of 3-(3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). The crude was purified by flash column chromatography using DCM/MeOH (100:0 to 98:2) and then purified further by reverse phase chromatography using Method D (25mM NH4HCO3 pH 8 / ACN: MeOH (1 : 1) (59:41 to 17:83) as eluents) to afford the title product. MS (ESI): mass calcd. for C22H28N2O3, 368.2; m/z found, 369.2 [M+H] + . ¾ NMR (400 MHz, Methanol-A) d 7.04 - 6.85 (m, 3H), 4.80 - 4.65 (m, 0.5H), 4.54 - 4.45 (m, 2H), 4.42 - 4.26 (m, 0.5H), 3.25 - 2.99 (m, 2H), 2.95 (m, 3H),

2.63 - 2.10 (m, 11H), 1.97 - 1.74 (m, 1H), 0.98 - 0.79 (m, 2H), 0.63 - 0.45 (m, 2H).

Example 32: -3-(3-Cvclopropyl-4-methylphenvDcvclobutvD-N-methyl-6-oxo- 7-oxa-5-azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 10, using 3-(3-cyclopropyl- 4-methylphenyl)-N-methylcyclobutan-l -amine, trifluoroacetate salt (Intermediate 24) instead of 3-(3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C22H28N2O3, 368.2; m/z found, 369.2 [M+H] + . ¾NMR (400 MHz, Methanol-A) d 7.12 - 7.00 (m, 1H), 7.02 - 6.93 (m, 1H), 6.82 (br s, 1H), 4.77 - 4.64 (m, 0.5H), 4.50 (m, 2H), 4.44 - 4.29 (m, 0.5H), 3.25 - 2.99 (m, 2H), 2.94 (m, 3H), 2.60 - 2.10 (m, 11H), 1.99 - 1.78 (m, 1H), 1.00 - 0.81 (m, 2H), 0.67 - 0.47 (m, 2H).

Example 33: (2s.4S)-N-Methyl-6-oxo-N-(Yls.3S)-3-(5.6.7.8-tetrahvdronapht halen-2- vDcvclobutvD-7-oxa-5-azaspiror3.41octane-2-carboxamide. The title compound was prepared in a manner analogous to Example 10, using N-methyl-3- (5,6,7,8-tetrahydronaphthalen-2-yl)cyclobutan-l -amine, trifluoroacetate salt (Intermediate 25) instead of 3-(3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C22H28N2O3, 368.2; m/z found, 369.2 [M+H] + . Ή NMR (400 MHz, Methanol - /2) d 6.99 - 6.86 (m, 4H), 4.78 - 4.66 (m, 0.5H), 4.50 (m, 2H), 4.42 - 4.30 (m, 0.5H), 3.26 - 3.00 (m, 2H), 2.95 (m, 3H), 2.79 - 2.67 (m, 4H), 2.63 - 2.12 (m, 8H), 1.85 - 1.71 (m, 4H). BenzylcvclobutvD-N-methyl-6-oxo-7-oxa-5- The title compound was prepared in a manner analogous to Example 10, using 3-benzyl-N- methylcyclobutan-1 -amine, trifluoroacetate salt (Intermediate 26) instead of 3-(3- isopropylphenyl)-N-methylcyclobutan-l -amine (Intermediate 6). Purification by SFC (Method E) afforded the title product and (2s,4S)-N-((ls,3R)-3-Benzylcyclobutyl)-N-methyl-6-oxo-7-oxa- 5-azaspiro[3.4]octane-2-carboxamide (Example 35). MS (ESI): mass calcd. for C19H24N2O3, 328.2; m/z found, 329.1 [M+H] + . ¾NMR (400 MHz, Methanol-A) d 7.32 - 7.20 (m, 2H), 7.20 - 7.06 (m, 3H), 4.69 - 4.53 (m, 0.5H), 4.48 (m, 2H), 4.28 - 4.14 (m, 0.5H), 3.19 - 2.99 (m, 1H), 2.92 - 2.79 (m, 3H), 2.72 (t, J= 7.0 Hz, 2H), 2.59 - 2.34 (m, 4H), 2.34 - 2.10 (m, 3H), 2.08 - 1.90 (m, 1H), 1.90 - 1.75 (m, 1H).

Example 35: (2s.4S)-N-(Yl s.3R)-3 -Benzyl cvclobutyl )-N-methyl-6-oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide. The title product was isolated from Example 34. MS (ESI): mass calcd. for C19H24N2O3, 328.2; m/z found, 329.1 [M+H] + . 1 HNMR (400 MHz, Methanol-A) d 7.37 - 7.08 (m, 5H), 5.15 - 4.93 (m, 0.5H), 4.48 (m, 2H), 4.43 - 4.26 (m, 0.5H), 3.17 - 2.76 (m, 6H), 2.62 - 2.21 (m, 7H), 2.11 - 1.81 (m, 2H). Example 36: (2s.4S)-N- Butyl icvclobutyl )-N-methyl-6-oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 10, using 3-(ter/-butyl)-N- methylcyclobutan-1 -amine (Intermediate 27) instead of 3-(3-isopropylphenyl)-N- methylcyclobutan-1 -amine (Intermediate 6). Purification by SFC (Method E) afforded the title product and (2s,4S)-N-((lr,3R)-3-(/er/-Butyl)cyclobutyl)-N-methyl-6-oxo- 7-oxa-5- azaspiro[3.4]octane-2-carboxamide (Example 37). MS (ESI): mass calcd. for C16H26N2O3, 294.2; m/z found, 295.2 [M+H] + . ¾NMR (400 MHz, Methanol-A) d 4.60 - 4.43 (m, 2.5H), 4.26 - 4.05 (m, 0.5H), 3.24 - 2.96 (m, 1H), 2.89 (m, 3H), 2.61 - 2.35 (m, 4H), 2.11 - 1.93 (m, 3H), 1.92 - 1.78 (m, 2H), 0.87 (m, 9H).

Example 37: ButvOcvclobutvO-N-methyl-6-oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide.

The title product was isolated from Example 36. MS (ESI): mass calcd. for C16H26N2O3, 294.2; m/z found, 295.1 [M+H] + . 1 HNMR (400 MHz, Methanol-A) d 4.93 - 4.78 (m, 0.5H), 4.51 - 4.42 (m, 2H), 4.40 - 4.23 (m, 0.5H), 3.14 - 3.01 (m, 1H), 2.99 (m, 3H), 2.58 - 2.37 (m, 4H), 2.37 - 2.26 (m, 1H), 2.26 - 1.96 (m, 4H), 0.90 (m, 9H).

Example 38: -3-CvclohexylcvclobutvD-N-methyl-6-oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 11, using 3-cyclohexyl-N- methylcyclobutan-1 -amine (Intermediate 30) instead of 3-(3-isopropylphenyl)-N- methylcyclobutan-1 -amine (Intermediate 6). Purification by SFC (Method: SFC-Lux Amylose- 2, Column Phenomenex Lux Amylose-2 150 x 4.6 mm, 5 pm, Isocratic mode: 13% Ethanol + 0.1% Diethylamine and 87 % CO2) afforded the title product and (2s,4S)-N-((lr,3R)-3- cyclohexylcyclobutyl)-N-methyl-6-oxo-7-oxa-5-azaspiro[3.4]oc tane-2-carboxamide (Example 39). MS (ESI): mass calcd. for C18H28N2O3, 320.2; m/z found, 321.2 [M+H] + . ¾NMR (400 MHz, Methanol Ά): 4.65 - 4.53 (m, 0.5H), 4.48 (d, J= 6.4 Hz, 2H), 4.24 - 4.13 (m, 0.5H), 3.18 - 2.99 (m, 1H), 2.88 (d, J= 9.9 Hz, 3H), 2.58 - 2.39 (m, 4H), 2.28 - 2.16 (m, 2H), 1.91 - 1.81 (m, 1H), 1.78 - 1.57 (m, 7H), 1.27 - 1.09 (m, 4H), 0.82 (dd, J= 23.3, 11.5 Hz, 2H).

Example 39: -3-Cvclohexylcvclobutvn-N-methyl-6-oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide.

The title product was isolated from Example 38. MS (ESI): mass calcd. for C18H28N2O3, 320.2; m/z found, 321.2 [M+H] + . ¾NMR (400 MHz, Methanol-A): 4.95 - 4.85 (m, 0.5H), 4.48 (d, J = 5.5 Hz, 2H), 4.46 - 4.35 (m, 0.5H), 3.14 - 3.00 (m, 1H), 2.96 (d, J= 7.6 Hz, 3H), 2.59 - 2.39 (m, 4H), 2.39 - 2.28 (m, 1H), 2.28 - 2.18 (m, 1H), 2.13 - 1.95 (m, 2H), 1.95 - 1.59 (m, 6H), 1.43 - 1.08 (m, 4H), 0.87 - 0.65 (m, 2H).

Example 40: (2s.4S)-N-Methyl-N-((ls.3S)-3-(4-n-methylcvclopropynphenvncv clobutvn-6- oxo-7-oxa-5-azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 11, using_N-methyl-3-(4-(l- methylcyclopropyl)phenyl)cyclobutan-l -amine, trifluoroacetate salt (Intermediate 31) instead of 3-(3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). Purification by preparative HPLC (Method: MAP4AC (25mM NH4HCO3) / (MeCN: MeOH 1:1); from 39/61 to 11/89). afforded the title product and (2s,4S)-N-((l s,3 S)-3-(4-(sec-butyl)phenyl)cyclobutyl)-N-methyl-6- oxo-7-oxa-5-azaspiro[3.4]octane-2-carboxamide (Example 41). MS (ESI): mass calcd. for C22H28N2O3, 368.2; m/z found, 369.2 [M+H] + . 1 HNMR (400 MHz, Methanol-A): 7.26 - 7.10 (m, 4H), 4.80 - 4.68 (m, 0.5H), 4.50 (d, J= 14.4 Hz, 2H), 4.43 - 4.32 (m, 0.5H), 3.24 - 3.02 (m, 2H), 2.95 (m, 3H), 2.62 - 2.43 (m, 6H), 2.39 - 2.28 (m, 1H), 2.26 - 2.15 (m, 1H), 1.37 (s, 3H), 0.84 - 0.78 (m, 2H), 0.72 - 0.65 (m, 2H). Example 41 : -3-(4-(Sec-butvDphenvDcvclobutvD-N-methyl-6-oxo-7-oxa-5- azaspirc>r3.41octane-2-carboxamide.

The title product was isolated from Example 40. MS (ESI): mass calcd. for C22H30N2O3, 370.2; m/z found, 372.2 [M+H] + . ¾NMR (400 MHz, Methanol-A): 7.20 - 7.08 (m, 4H), 4.79 - 4.68 (m, 0.5H), 4.50 (d, J= 14.9 Hz, 2H), 4.45 - 4.33 (m, 0.5H), 3.24 - 3.03 (m, 2H), 2.96 (d, J= 7.0

Hz, 3H), 2.64 - 2.42 (m, 7H), 2.35 (m, 1H), 2.22 (m, 1H), 1.68 - 1.51 (m, 2H), 1.24 - 1.18 (m, 3H), 0.84 - 0.75 (m, 3H).

Example 42: (2s.4S)-N-Methyl-N-((ls.3S)-3-(3-n-methylcvclopropynphenvncv clobutvn-6- oxo-7-oxa-5-azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 11, using N-methyl-3-(3-(l- methylcyclopropyl)phenyl)cyclobutan-l -amine, trifluoroacetate salt (Intermediate 32) instead of 3-(3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C22H28N2O3, 368.2; m/z found, 369.2 [M+H] + . ¾ NMR (400 MHz, Methanol-A): 7.24 - 7.17

(m, 1H), 7.14 - 7.02 (m, 3H), 4.80 - 4.68 (m, 0.5H), 4.50 (d, 7= 15.5 Hz, 2H), 4.43 - 4.32 (m, 0.5H), 3.23 - 3.02 (m, 2H), 2.95 (d, J= 7.2 Hz, 3H), 2.63 - 2.43 (m, 6H), 2.41 - 2.30 (m, 1H), 2.27 - 2.15 (m, 1H), 1.38 (s, 3H), 0.84 - 0.80 (m, 2H), 0.73 - 0.68 (m, 2H).

Example 43: (2s.4S)-N-((ls.3S)-3-(3-Ethyl-5-methylphenvncvclobutvn-N-met hyl-6-oxo-7-oxa- 5-azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 11, using 3-(3-ethyl-5- methylphenyl)-N-methylcyclobutan-l-amine, trifluoroacetate salt (Intermediate 33) instead of 3- (3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C21H28N2O3, 356.2; m/z found, 357.2 [M+H] + . ¾ NMR (400 MHz, Methanol-A): 6.89 - 6.81

(m, 3H), 4.80 - 4.67 (m, 0.5H), 4.50 (d, 7= 15.0 Hz, 2H), 4.45 - 4.32 (m, 0.5H), 3.25 - 3.02 (m, 2H), 2.95 (m, 3H), 2.65 - 2.39 (m, 8H), 2.39 - 2.30 (m, 1H), 2.29 (d, J= 2.5 Hz, 3H), 2.26 - 2.14 (m, 1H), 1.20 (m, 3H). Example 44: -3-(3-Cvclopropyl-5-methylphenvDcvclobutvD-N-methyl-6-oxo- 7-oxa-5-azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 11, using 3-(3-cyclopropyl- 5-methylphenyl)-N-methylcyclobutan-l -amine, trifluoroacetate salt (Intermediate 34) instead of 3-(3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C22H28N2O3, 368.2; m/z found, 369.2 [M+H] + . Ή NMR (400 MHz, Methanol -6/2): 6.83 (m, 1H), 6.74 (d, J= 4.0 Hz, 1H), 6.70 (m, 1H), 4.79 - 4.67 (m, 0.5H), 4.50 (d, J= 14.6 Hz, 2H), 4.42 - 4.31 (m, 0.5H), 3.24 - 3.02 (m, 2H), 2.95 (m, 3H), 2.60 - 2.42 (m, 6H), 2.38 - 2.28 (m, 1H), 2.27 (m, 3H), 2.24 - 2.14 (m, 1H), 1.90 - 1.78 (m, 1H), 0.97 - 0.85 (m, 2H), 0.67 - 0.58 (m, 2H).

Example 45 : -N-((T s.3 S)-3-(3-Isopropyl-5-methylphenvOcvclobutvO-N-methyl-6-oxo-7- oxa-5-azaspirc>r3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 11, using 3-(3-Isopropyl-5- methylphenyl)-N-methylcyclobutan-l-amine, trifluoroacetate salt (Intermediate 35) instead of 3- (3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C22H30N2O3, 370.2; m/z found, 371.2 [M+H] + . ¾ NMR (400 MHz, Methanol-A): 6.87 (m, 3H),

4.77 - 4.65 (m, 0.5H), 4.50 (d, J= 15.1 Hz, 2H), 4.45 - 4.29 (m, 0.5H), 3.25 - 3.02 (m, 2H),

2.95 (m, 3H), 2.89 - 2.77 (m, 1H), 2.62 - 2.42 (m, 6H), 2.42 - 2.30 (m, 1H), 2.30 (m 3H), 2.26 - 2.14 (m, 1H), 1.22 (m, 6H). Example 46: -3-(3-Chloro-5-methylphenvncvclobutvn-N-methyl-6-oxo-7- oxa-5-azaspirc>r3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 11, using 3-(3-chloro-5- methylphenyl)-N-methylcyclobutan-l-amine, trifluoroacetate salt (Intermediate 28) instead of 3- (3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C19H23CIN2O3, 362.1; m/z found, 363.1 [M+H] + . 1 HNMR (400 MHz, Methanol-A): 7.06 - 7.01 (m, 2H), 7.00 (m, 1H), 4.78 - 4.66 (m, 0.8H), 4.50 (d, J= 14.4 Hz, 2H), 4.46 - 4.33 (m, 0.5H),

3.24 - 3.02 (m, 2H), 2.94 (d, J= 8.1 Hz, 3H), 2.64 - 2.41 (m, 6H), 2.40 - 2.33 (m, 1H), 2.32 (m,

3H), 2.26 - 2.16 (m, 1H). Example 47 : -N-((T s 3 S)-3-(3-CvclopropylphenvOcvclobutvO-N-methyl-6-oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 11, using 3-(3- cyclopropylphenyl)-N-methylcyclobutan-l -amine, trifluoroacetate salt (Intermediate 36) instead of 3-(3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C21H26N2O3, 354.2; m/z found, 355.2 [M+H] + . Ή NMR (400 MHz, Methanol^): 7.22 - 7.10 (m, 1H), 7.07 - 6.98 (m, 1H), 6.98 - 6.92 (m, 1H), 6.92 - 6.83 (m, 1H), 4.80 - 4.67 (m, 0.5H), 4.50 (d, J= 15.1 Hz, 2H), 4.46 - 4.30 (m, 0.5H), 3.25 - 3.02 (m, 2H), 2.95 (m, 3H), 2.62 - 2.42 (m, 6H), 2.40 - 2.29 (m, 1H), 2.25 - 2.16 (m, 1H), 1.95 - 1.83 (m, 1H), 0.98 - 0.89 (m, 2H), 0.72 - 0.59 (m, 2H).

Example 48: Dihvdro-lH-inden-5-vDcvclobutvD-N-methyl-6-oxo-7- oxa-5-azaspiror3.41octane-2-carboxamide. The title compound was prepared in a manner analogous to Example 11, using 3-(2,3-dihydro- lH-inden-5-yl)-N-methylcyclobutan-l-amine, trifluoroacetate salt (Intermediate 37) instead of 3- (3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C21H26N2O3, 354.2; m/z found, 355.2 [M+H] + . ¾NMR (400 MHz, Methanol-A): 7.18 - 7.03

(m, 2H), 7.05 - 6.85 (m, 1H), 4.81 - 4.67 (m, 0.5H), 4.50 (d, J= 14.5 Hz, 2H), 4.42 - 4.31 (m, 0.5H), 3.27 - 3.02 (m, 2H), 2.95 (d, J= 7.5 Hz, 3H), 2.91 - 2.80 (m, 4H), 2.63 - 2.41 (m, 6H), 2.41 - 2.26 (m, 1H), 2.26 - 2.12 (m, 1H), 2.11 - 1.95 (m, 2H).

Example 49: (2s.4S)-N-((ls.3S)-3-(Bicvclor4 . 2 . 01octa-l(6).2.4-trien-3-vDcvclobutvD-N-methyl- 6-oxo-7-oxa-5-azaspiror3.41octane-2-carboxamide.

The title compound was prepared in a manner analogous to Example 11, 3-(bicyclo[4.2.0]octa- l(6),2,4-trien-3-yl)-N-methylcyclobutan-l -amine, trifluoroacetate salt (Intermediate 38) instead of 3-(3-isopropylphenyl)-N-methylcyclobutan-l-amine (Intermediate 6). MS (ESI): mass calcd. for C20H24N2O3, 340.2; m/z found, 341.2 [M+H] + . Ή NMR (400 MHz, Methanol^): 7.08 - 7.01 (m, 1H), 6.98 - 6.91 (m, 2H), 4.79 - 4.67 (m, 0.5H), 4.50 (d, J= 14.4 Hz, 2H), 4.43 - 4.32 (m, 0.5H), 3.22 - 3.03 (m, 6H), 2.95 (d, J= 7.6 Hz, 3H), 2.61 - 2.41 (m, 6H), 2.33 (td, J= 10.0, 2.8 Hz, 1H), 2.24 - 2.13 (m, 1H). Example 50: (2s.4S)-N-((ls.3S)-3-(3.5-DimethylphenvDcvclobutvD-N-ethyl-6 -oxo-7-oxa-5- azaspiror3.41octane-2-carboxamide.

T3P (0.440 mL, 50% purity in ethyl acetate, 0.739 mmol) was added to a 0 °C (ice/water) solution consisting of (ls,3s)-3-(3,5-dimethylphenyl)-N-ethylcyclobutan-l-amine, HC1 salt (Intermediate 29, 200 mg), (2s,4s)-6-oxo-7-oxa-5-azaspiro[3.4]octane-2-carboxylic acid (Intermediate 1, 84.0 mg, 0.491 mmol), triethylamine (0.81 mL, 5.94 mmol), and dichloromethane (5 mL). The resultant mixture was stirred for 2 hours with gradual warming to room-temperature before pouring it into water (50 mL) and extracting with dichloromethane (30 mL x 3). The combined organic extracts were dried over anhydrous Na2SC>4, filtered, and concentrated to dryness under reduced pressure to afford the crude product, which was purified by preparative HPLC using a Boston Prime C18 150 x 30 mm x 5 pm column (eluent: 50% to 80% (v/v) CPbCN and H2O with 0.05% NH3 + 10 mM NH4HCO3) to afford pure product. The product was suspended in water (10 mL), the mixture frozen using dry ice/acetone, and then lyophilized to dryness to afford the title compound (55.3 mg, 32%). MS (ESI): mass calcd. for C21H28N2O3, 356.2; m/z found, 357.2 [M+H] + . Ή NMR (400 MHz, Chlorofor -r/): 6.87 (m,

1H), 6.82 (m, 2H), 5.98 (m, 1H), 4.70 - 4.59 (m, 0.5H), 4.38 (d, J= 9.8 Hz, 2H), 4.15 - 4.02 (m, 0.5H), 3.54 - 3.42 (m, 1H), 3.36 - 3.27 (m, 1H), 3.21 - 2.96 (m, 2H), 2.75 - 2.58 (m, 4H), 2.56 - 2.46 (m, 2H), 2.36 - 2.22 (m, 7H), 2.17 - 2.06 (m, 1H), 1.13 (t, J= 6.9 Hz, 3H).

BIOLOGICAL DATA

The assay used to measure the in vitro activity of MGL is adapted from the assay used for another serine hydrolase (FAAH) described in Wilson et al., 2003 (A high-throughput- compatible assay for determining the activity of fatty acid amide hydrolase. Wilson SJ, Lovenberg TW, Barbier AJ. Anal Biochem. 2003 Jul 15;318(2):270-5.). The assay consists of combining endogenously expressed MGL from HeLa cells with test compounds, adding [glycerol-1, 3- 3 H]-oleoyl glycerol, incubating for one hour, and then measuring the amount of cleaved [l,3- 3 H]-glycerol that passes through an activated carbon filter. The amount of cleaved, tritiated glycerol passing through the carbon filter is proportional to the activity of the MGL enzyme in a particular well/test condition.

Standard conditions for this assay combine 300 nM [Glycerol-1, 3- 3 H]-oleoyl glycerol with human MGL from HeLa cells and test compounds for one hour, after which the reaction is filtered through activated carbon and tritium is measured in the flow through. The test compound concentration in screening mode is 10 pM, while the highest compound concentration in IC50 assays is determined empirically. MGL is the predominant hydrolase in HeLa cells/cell homogenates.

Table 3.

NT means not tested.