Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PIPERIDINE UREA DERIVATIVES FOR TREATMENT OF NEURODEGENERATIVE DISEASES
Document Type and Number:
WIPO Patent Application WO/2024/073680
Kind Code:
A1
Abstract:
Described herein are novel piperidine urea derived compounds and their pharmaceutical compositions for the treatment of neurodegenerative disorders and diseases mediated by soluble epoxide hydrolase.

Inventors:
KHANNA ISH (US)
PILLARISETTI SIVARAM (US)
Application Number:
PCT/US2023/075537
Publication Date:
April 04, 2024
Filing Date:
September 29, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
NEUROPN THERAPEUTICS INC (US)
International Classes:
A61K31/4025; A61K31/4155; A61K31/4184; A61K31/445; A61K31/497; A61K31/506; A61K31/5377; A61P25/00; A61P25/16; A61P25/28
Domestic Patent References:
WO2013057322A12013-04-25
Foreign References:
US20210161881A12021-06-03
US20220354839A12022-11-10
Other References:
WAGNER KAREN M ET AL: "Soluble epoxide hydrolase as a therapeutic target for pain, inflammatory and neurodegenerative diseases", PHARMACOLOGY & THERAPEUTICS, vol. 180, 19 July 2017 (2017-07-19), pages 62 - 76, XP085276381, ISSN: 0163-7258, DOI: 10.1016/J.PHARMTHERA.2017.06.006
TAKAI KENTARO ET AL: "Structure-based optimization of cyclopropyl urea derivatives as potent soluble epoxide hydrolase inhibitors for potential decrease of renal injury without hypotensive action", BIOORGANIC & MEDICINAL CHEMISTRY, ELSEVIER, AMSTERDAM, NL, vol. 22, no. 5, 31 January 2014 (2014-01-31), pages 1548 - 1557, XP028616666, ISSN: 0968-0896, DOI: 10.1016/J.BMC.2014.01.040
BIOMOLECULES, vol. 10, 2020, pages 703 - 724
PROC. NATL. ACAD. SCIENCES, vol. 115, 2018, pages E5815 - E5823
NEUROTHERAPEUTICS, vol. 17, no. 3, 2020, pages 1825 - 1835
PROC. NATL. ACAD. SCIENCES, vol. 105, no. 48, 2008, pages 18901 - 18906
PHARMACOLOGY & THERAPEUTICS, vol. 180, 2017, pages 62 - 76
NAT. REV. DRUG. DISCOV., vol. 8, no. 10, 2009, pages 794 - 805
CARDIOVASC. HEMATOL. AGENTS MED. CHEM., vol. 10, no. 3, September 2012 (2012-09-01), pages 212 - 22
PROSTAGLANDINS AND OTHER LIPID MEDIATORS, vol. 140, 2019, pages 31 - 39
PROGRESS IN NEUROBIOLOGY, vol. 172, 2019, pages 23 - 39
INFLAMM. ALLERGY DRUG TARGETS, vol. 11, no. 2, April 2012 (2012-04-01), pages 143 - 58
MOL. PAIN, vol. 4, 2011, pages 7 - 78
DRUG DISCOV TODAY., vol. 20, no. 11, November 2015 (2015-11-01), pages 1382 - 90
BIOMOLECULES, vol. 10, no. 5, 1 May 2020 (2020-05-01), pages 703
PROSTAGLANDINS OTHER LIPID MEDIAT., vol. 96, 2011, pages 76 - 83
PHARMACOL THER., vol. 0163-7258, no. 17, 19 June 2017 (2017-06-19), pages 30154 - 7
BIOCHIMIE, vol. 159, 2019, pages 59 - 65
J PHARMACOL EXP. THER., vol. 326, no. 2, August 2008 (2008-08-01), pages 614 - 22
J HISTOCHEM CYTOCHEM., vol. 56, no. 6, June 2008 (2008-06-01), pages 551 - 9
AM J PHYSIOL, vol. 263, 1992, pages H519 - 25
J NEUROCHEM, vol. 61, 1993, pages 150 - 9
PROSTAGLANDINS OTHER LIPID MEDIAT, vol. 91, 2010, pages 68 - 84
AM J PHYSIOL HEART CIRC PHYSIOL, vol. 296, 2009, pages H1352 - 63
EXPERT REV MOL MED, vol. 13, 2011, pages 7 - 12
EXPERT REV MOL MED, vol. 13, 1998, pages 7 - 12
EXPERT REV MOL MED, vol. 13, 2014, pages 7 - 12
NEUROPATHOL APPL NEUROBIOL, vol. 42, 2016, pages 607 - 620
PROC NATL ACAD SCI USA., vol. 112, 2015, pages 9082 - 9087
J NEUROSCI, vol. 27, 2007, pages 4642 - 9
J NEUROCHEM, vol. 117, 2011, pages 632 - 42
NEUROSCIENCE, vol. 223, 2012, pages 68 - 76
MOL NEUROBIOL., vol. 52, no. 1, 2015, pages 187 - 95
PROC NATL ACAD SCI USA., vol. 105, no. 48, 2 December 2008 (2008-12-02), pages 18901 - 6
EUR J PHARMACOL., vol. 700, no. 1-3, 30 January 2013 (2013-01-30), pages 93 - 101
J PAIN., vol. 15, no. 9, September 2014 (2014-09-01), pages 907 - 14
PROC NATL ACAD SCI USA., vol. 112, no. 29, 21 July 2015 (2015-07-21), pages 9082 - 7
BEHAV BRAIN RES, vol. 326, 30 May 2017 (2017-05-30), pages 69 - 76
PROSTAGLANDINS OTHER LIPID MEDIAT, October 2014 (2014-10-01), pages 113 - 115
J HUAZHONG UNIV SCI TECHNOLOG MED SCI, vol. 36, no. 6, December 2016 (2016-12-01), pages 785 - 790
FUTURE NEUROL, vol. 4, no. 2, 1 March 2009 (2009-03-01), pages 179 - 199
AM J PATHOL, vol. 174, no. 6, June 2009 (2009-06-01), pages 2086 - 95
PLOS ONE, vol. 9, no. 5, 13 May 2014 (2014-05-13), pages e97529
AM J PHYSIOL HEART CIRC PHYSIOL., vol. 305, no. 11, 1 December 2013 (2013-12-01), pages H1605 - 13
SCI TRANSL MED., vol. 12, no. 573, 9 December 2020 (2020-12-09), pages eabbl206
JOURNAL OF NEUROSCIENCE, no. 42, 2020, pages 8188 - 8203
JOURNAL OF NEUROINFLAMMATION, vol. 16, 2019
J AFFECT DISORD., vol. 270, 1 June 2020 (2020-06-01), pages 131 - 134
PROC NATL ACAD SCI U S A, vol. 113, no. 13, 29 March 2016 (2016-03-29), pages E1944 - 52
J CEREB BLOOD FLOW METAB, vol. 35, no. 2, 2015, pages 267 - 276
J NEUROSURG, vol. 121, no. 6, 2014, pages 1359 - 1366
NEUROCRITICAL CARE, vol. 22, 2015, pages 306 - 319
BRAIN HEMORRHAGES, May 2022 (2022-05-01)
NEUROCRIT CARE, 2021
J ATHEROSCLER THROMB., vol. 24, no. 12, 1 December 2017 (2017-12-01), pages 1258 - 1266
J. AM. CHEM. SOC., vol. 138, 2016, pages 13493 - 13496
FASEB J, vol. 22, no. 7, 2008, pages 2488 - 97
LIN ET AL., ELIFE, vol. 9, 26 June 2020 (2020-06-26), pages e54726
CALLIZOT ET AL., PLOS ONE, Retrieved from the Internet
SOLNICA-KREZE ET AL., DEVELOPMENT, vol. 123, December 1996 (1996-12-01), pages 37 - 46
BRETAUD ET AL., NEUROTOXICOL TERATOL, vol. 26, no. 6, November 2004 (2004-11-01), pages 857 - 64
J BASIC CLIN PHARM, vol. 7, no. 2, March 2016 (2016-03-01), pages 27 - 31
LANCET NEUROL, vol. 11, no. 11, November 2012 (2012-11-01), pages 986 - 98
MOVEMENT DISORDERS, vol. 35, no. 12, pages 2201 - 2210, Retrieved from the Internet
EMBO MOL MED, vol. 1, no. 2, May 2009 (2009-05-01), pages 99 - 111
Attorney, Agent or Firm:
LINEBERRY, Douglas L. et al. (US)
Download PDF:
Claims:
What is claimed is: 1. A method of treating a neurodegenerative disease or a disease associated with synucleinopathies, excluding Parkinsons disease, comprising: administering to a subject a therapeutically effective amount of at least one compound of Formula I: Formula I where R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2NHR2, or COR3; R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, or COR3; R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; X is selected from O, (CH2)p, NH and p is from 0-2; Y1-Y2 are selected from CH-CH2, CH-O, or C=CH, however when Y1-Y2 is CH-O, X is selected from O or NH or R1 is not hydrogen; and Y3 is selected from H or Me, its stereoisomers or pharmaceutically acceptable salts thereof. 2. A method of treating a neurodegenerative disease or a disease associated with synucleinopathies, excluding Parkinsons disease, comprising: administering to a subject a therapeutically effective amount of at least one compound of Formula I Formula I where R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2NHR2, or COR3; R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, or COR3; R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; X is selected from O, (CH2)p, NH and p is from 0-2, however when p = 0, Y1-Y2 is not CH-CH2 or CH-O, and R1 is not aryl; Y1-Y2 are selected from CH-CH2, CH-O, or C=CH, however when Y1-Y2 is CH-O, X is selected from O or NH and R1 is not hydrogen or alkyl; and Y3 is selected from H or Me, its stereoisomers or pharmaceutically acceptable salts thereof. 3. A method as claimed in claim 1 wherein Y3 is H and the compound is a compound according to Formula II: Formula II where R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2NHR2, or COR3; R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, COR3; R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; X is selected from O, (CH2)p, NH and p is from 0-2; Y1-Y2 are selected from CH-CH2, CH-O, or C=CH, however when Y1-Y2 is CH-O, X is selected from O or NH or R1 is not hydrogen; and its stereoisomers or pharmaceutically acceptable salts thereof. 4. A method as claimed in claim 2 wherein Y3 is H and the compound is a compound according to Formula II: Formula II where R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2NHR2, or COR3; R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, COR3; R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; X is selected from O, (CH2)p, NH and p is from 0-2, however when p = 0, Y1-Y2 is not CH-CH2 or CH-O, and R1 is not aryl; Y1-Y2 are selected from CH-CH2, CH-O, or C=CH, however when Y1-Y2 is CH-O, X is selected from O or NH, and R1 is not hydrogen or alkyl, its stereoisomers or pharmaceutically acceptable salts thereof. 5. A method as claimed in claim 1 wherein Y3 is H, Y1-Y2 is C=CH, and the compound is a compound according to Formula III Formula III where R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2R5¸ SO2NHR2, or COR3; R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5¸ SO2NHR2, COR3; R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; and X is selected from O, (CH2)p, NH and p is from 0-2, its stereoisomers or pharmaceutically acceptable salts thereof.

6. A method as claimed in claim 1 wherein Y3 is H, Y1-Y2 is CH-CH2, and the compound is a compound according to Formula IV where R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2R5¸ SO2NHR2, COR3; R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, COR3; R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl. Aryl or heteroaryl may optionally be substituted one or more times with groups or substituents such as alkyl, hydroxy, halogen, haloalkyl; and X is selected from O, (CH2)p, NH; wherein p is selected from 0-2, its stereoisomers or pharmaceutically acceptable salts thereof. 7. A method as claimed in claim 2 wherein Y3 is H, Y1-Y2 is CH-CH2, and the compound is a compound according to Formula IV Formula IV where R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2R5¸ SO2NHR2, COR3; R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, COR3; R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl. Aryl or heteroaryl may optionally be substituted one or more times with groups or substituents such as alkyl, hydroxy, halogen, haloalkyl; and X is selected from O, (CH2)p, NH; wherein p is selected from 0-2, however when p = 0, R1 is not aryl its stereoisomers or pharmaceutically acceptable salts thereof.

8. A method as claimed in claim 1, wherein the compound of Formula 1 is one or more of the following compounds

its stereoisomers or pharmaceutically acceptable salts thereof. 9. A method as claimed in claim 1, wherein the compound of Formula 1 is one of the following compounds: its stereoisomers or pharmaceutically acceptable salts thereof. 10. A method as claimed in claim 1, wherein the compound of Formula 1 is one or more of the following compounds:

its stereoisomers or pharmaceutically acceptable salts thereof. 11. A method as claimed in claim 1 wherein the compound inhibits soluble epoxide hydrolase at a concentration (IC50) of less than 10 µM. 12. A method as claimed in claim 1 wherein the compound inhibits soluble epoxide hydrolase at a concentration (IC50) of less than < 100 nM. 13. A method as claimed in claim 1 wherein the compound inhibits soluble epoxide hydrolase at a concentration (IC50) of less than < 100 nM and has at least 10-fold selectivity over inhibition of fatty acid amide hydrolase (IC50, (FAAH (SEQ ID NO: 3)).

14. A method as claimed in claim 1 wherein the compound inhibits soluble epoxide hydrolase at a concentration (IC50) of less than < 100 nM and inhibits fatty acid amide hydrolase (FAAH (SEQ ID NO: 3)) at a concentration (IC50) of > 1000 nM. 15. A method as claimed in claim 1 wherein the disease is selected from Gaucher’s disease, dementia with Lewy bodies, and Alzheimer’s disease. 16. A method as claimed in claim 1 wherein the compound is administered at a dose of about 1 mg/day to about 1,000 mg/day. 17. A method as claimed in claim 1, wherein the compound is administered at a dose of about 5 mg/d ay to about 500 mg/day. 18. A method as claimed in claim 1, wherein the compound is administered to treat one or more of neuronal loss, neuroinflammation, alpha-synculein aggregation and/ or lewy body formation. 19. A method as claimed in claim 1, wherein the compound is administered to treat impaired locomotor activity and other movement and non-movement related symptoms. 20. A method as claimed in claim 1, wherein the compound is administered to treat cognitive dysfunction or dementia. 21. A method of treating a Parkinsons disease with dementia (PDD), familial Parkinson’s disease related to PINK-1 (SEQ ID NO: 2) mutation, or familial Parkinson’s disease related to mutations in a glucocerebrosidase (GBA) gene comprising: administering to a subject a therapeutically effective amount of at least one compound of Formula I: Formula I Where R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2NHR2, or COR3; R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, or COR3; R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; X is selected from O, (CH2)p, NH and p is from 0-2; Y1-Y2 are selected from CH-CH2, CH-O, or C=CH, however when Y1-Y2 is CH-O, X is selected from O or NH or R1 is not hydrogen; and Y3 is selected from H or Me, its stereoisomers or pharmaceutically acceptable salts thereof. 22. A method of treating a Parkinsons disease with dementia (PDD), familial Parkinson’s disease related to PINK-1 (SEQ ID NO: 2) mutation, or familial Parkinson’s disease related to mutations in a glucocerebrosidase (GBA) gene comprising: administering to a subject a therapeutically effective amount of at least one compound of Formula I: Formula I where R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2NHR2, or COR3; R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, or COR3; R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; X is selected from O, (CH2)p, NH and p is from 0-2; Y1-Y2 are selected from CH-CH2, CH-O, or C=CH, however when Y1-Y2 is CH-O, X is selected from O or NH or R1 is not hydrogen; and Y3 is selected from H or Me, its stereoisomers or pharmaceutically acceptable salts thereof. 23. A method for treatment of Gaucher’s disease, Parkinsons disease with dementia (PDD), familial Parkinson’s disease related to PINK-1 (SEQ ID NO: 2) mutation, or familial Parkinson’s disease related to mutations in a glucocerebrosidase (GBA) gene comprising administering to a subject a therapeutically effective amount of a soluble epoxide hydrolase inhibitor. 24. A compound of Formula I: Formula I where R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2NHR2, or COR3; R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, or COR3; R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; X is selected from O, (CH2)p, NH and p is from 0-2, however when p = 0, Y1-Y2 is not CH-CH2 or CH-O, and R1 is not aryl; Y1-Y2 are selected from CH-CH2, CH-O, or C=CH, however when Y1-Y2 is CH-O, X is selected from O or NH and R1 is not hydrogen or alkyl; and Y3 is selected from H or Me. its stereoisomers or pharmaceutically acceptable salts thereof. for use in the treatment of a neurodegenerative disease or a disease associated with synucleinopathies, but not Parkinsons disease. 25. A compound of Formula I: Formula I where R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2NHR2, or COR3; R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, or COR3; R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; X is selected from O, (CH2)p, NH and p is from 0-2, however when p = 0, Y1-Y2 is not CH-CH2 or CH-O, and R1 is not aryl; Y1-Y2 are selected from CH-CH2, CH-O, or C=CH, however when Y1-Y2 is CH-O, X is selected from O or NH and R1 is not hydrogen or alkyl; and Y3 is selected from H or Me. its stereoisomers or pharmaceutically acceptable salts thereof. for use in the treatment of a neurodegenerative disease or a disease associated with synucleinopathies, but not Parkinsons disease. 26. A soluble epoxide hydrolase inhibitor for use in the treatment of Gaucher’s disease, Parkinsons disease with dementia (PDD), familial Parkinson’s disease related to PINK-1 (SEQ ID NO: 2) mutation, or familial Parkinson’s disease related to mutations in a glucocerebrosidase (GBA) gene.

AMENDED CLAIMS received by the International Bureau on 08 March 2024 (08.03.2024)

1. A method of treating a neurodegenerative disease or a disease associated with synucleinopathies, excluding Parkinsons disease, comprising: administering to a subject a therapeutically effective amount of at least one compound of Formula I: where

R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2NHR2, or COR3;

R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl;

R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy;

R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, or COR3;

R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine;

R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl;

X is selected from O, (CH2)p, NH and p is from 0-2; Y1-Y2 are selected from CH-CH2, CH-O, or C=CH, however when Y1-Y2 is CH-O, X is selected from O or NH or R1 is not hydrogen; and

Y3 is selected from H or Me, its stereoisomers or pharmaceutically acceptable salts thereof.

2. A method of treating a neurodegenerative disease or a disease associated with synucleinopathies, excluding Parkinsons disease, comprising: administering to a subject a therapeutically effective amount of at least one compound of Formula I

Formula I where

R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2NHR2, or COR3;

R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl;

R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy;

R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, or COR3;

R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine;

R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl;

X is selected from O, (CH2)p, NH and p is from 0-2, however when p 0, Y1-Y2 is not

CH-CH2 or CH-O, and R1 is not aryl; Y1-Y2 are selected from CH-CH2, CH-O, or C=CH, however when Y1-Y2 is CH-O, X is selected from O or NH and R1 is not hydrogen or alkyl; and

Y3 is selected from H or Me, its stereoisomers or pharmaceutically acceptable salts thereof.

3. A method as claimed in claim 1 wherein Y3 is H and the compound is a compound according to Formula II:

Formula II where

R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2NHR2, or COR3;

R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl;

R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy;

R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, COR3;

R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine;

R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl;

X is selected from O, (CH2)p, NH and p is from 0-2; Y1-Y2 are selected from CH-CH2, CH-O, or C=CH, however when Y1-Y2 is CH-O, X is selected from O or NH or R1 is not hydrogen; and its stereoisomers or pharmaceutically acceptable salts thereof.

4. A method as claimed in claim 2 wherein Y3 is H and the compound is a compound according to Formula II:

Formula II where

R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2NHR2, or COR3;

R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl;

R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy;

R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, COR3;

R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine;

R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl;

X is selected from O, (CH2)p, NH and p is from 0-2, however when p 0, Y1-Y2 is not

CH-CH2 or CH-O, and R1 is not aryl; Y1-Y2 are selected from CH-CH2, CH-O, or C=CH, however when Y1-Y2 is CH-O, X is selected from O or NH, and R1 is not hydrogen or alkyl, its stereoisomers or pharmaceutically acceptable salts thereof.

5. A method as claimed in claim 1 wherein Y3 is H, Y1-Y2 is C=CH, and the compound is a compound according to Formula III

Formula III where

R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2R5, SO2NHR2, or COR3;

R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl;

R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy;

R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, SO2NHR2, COR3;

R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine;

R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; and

X is selected from O, (CH2)p, NH and p is from 0-2, its stereoisomers or pharmaceutically acceptable salts thereof.

6. A method as claimed in claim 1 wherein Y3 is H, Y1-Y2 is CH-CH2, and the compound is a compound according to Formula IV

Formula IV where

R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2R5, SO2NHR2, COR3;

R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl;

R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy;

R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, COR3;

R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine;

R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl. Aryl or heteroaryl may optionally be substituted one or more times with groups or substituents such as alkyl, hydroxy, halogen, haloalkyl; and

X is selected from O, (CH2)p, NH; wherein p is selected from 0-2, its stereoisomers or pharmaceutically acceptable salts thereof.

7. A method as claimed in claim 2 wherein Y3 is H, Y1-Y2 is CH-CH2, and the compound is a compound according to Formula IV

Formula IV where

R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2R5, SO2NHR2, COR3;

R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl;

R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy;

R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, COR3;

R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine;

R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl. Aryl or heteroaryl may optionally be substituted one or more times with groups or substituents such as alkyl, hydroxy, halogen, haloalkyl; and

X is selected from O, (CH2)p, NH; wherein p is selected from 0-2, however when p = 0, R1 is not aryl its stereoisomers or pharmaceutically acceptable salts thereof.

8. A method as claimed in claim 1, wherein the compound of Formula 1 i more of the following compounds

its stereoisomers or pharmaceutically acceptable salts thereof.

9. A method as claimed in claim 1, wherein the compound of Formula 1 is one of the following compounds: its stereoisomers or pharmaceutically acceptable salts thereof.

10. A method as claimed in claim 1, wherein the compound of Formula 1 is one or more of the following compounds:

its stereoisomers or pharmaceutically acceptable salts thereof.

11. A method as claimed in claim 1 wherein the compound inhibits soluble epoxide hydrolase at a concentration (IC50) of less than 10 μM.

12. A method as claimed in claim 1 wherein the compound inhibits soluble epoxide hydrolase at a concentration (IC50) of less than < 100 nM.

13. A method as claimed in claim 1 wherein the compound inhibits soluble epoxide hydrolase at a concentration (IC50) of less than 100 nM and has at least 10-fold selectivity over inhibition of fatty acid amide hydrolase (IC50, (FAAH (SEQ ID NO: 3)).

14. A method as claimed in claim 1 wherein the compound inhibits soluble epoxide hydrolase at a concentration (IC50) of less than 100 nM and inhibits fatty acid amide hydrolase (FAAH (SEQ ID NO: 3)) at a concentration (IC50) of > 1000 nM.

15. A method as claimed in claim 1 wherein the disease is selected from Gaucher’s disease, dementia with Lewy bodies, and Alzheimer’s disease.

16. A method as claimed in claim 1 wherein the compound is administered at a dose of about 1 mg/day to about 1,000 mg/day.

17. A method as claimed in claim 1, wherein the compound is administered at a dose of about 5 mg/d ay to about 500 mg/day.

18. A method as claimed in claim 1, wherein the compound is administered to treat one or more of neuronal loss, neuroinflammation, alpha-synculein aggregation and/ or lewy body formation.

19. A method as claimed in claim 1, wherein the compound is administered to treat impaired locomotor activity and other movement and non-movement related symptoms.

20. A method as claimed in claim 1, wherein the compound is administered to treat cognitive dysfunction or dementia.

21. A method of treating a Parkinsons disease with dementia (PDD), familial

Parkinson’s disease related to PINK-1 (SEQ ID NO: 2) mutation, or familial Parkinson’s disease related to mutations in a glucocerebrosidase (GBA) gene comprising: administering to a subject a therapeutically effective amount of at least one compound of Formula I: Formula I

Where

R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2NHR2, or COR3;

R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl;

R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy;

R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, or COR3;

R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine;

R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl;

X is selected from O, (CH2)p, NH and p is from 0-2; Y1-Y2 are selected from CH-CH2, CH-O, or C=CH, however when Y1-Y2 is CH-O, X is selected from O or NH or R1 is not hydrogen; and

Y3 is selected from H or Me, its stereoisomers or pharmaceutically acceptable salts thereof.

22. A method of treating a Parkinsons disease with dementia (PDD), familial

Parkinson’s disease related to PINK-1 (SEQ ID NO: 2) mutation, or familial Parkinson’s disease related to mutations in a glucocerebrosidase (GBA) gene comprising: administering to a subject a therapeutically effective amount of at least one compound of Formula I:

Formula I where

R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2NHR2, or COR3;

R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl;

R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy;

R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, or COR3;

R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine;

R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl;

X is selected from O, (CH2)p, NH and p is from 0-2, however when p 0, Y1-Y2 is not

CH-CH2 or CH-O, and R1 is not aryl; Y1-Y2 are selected from CH-CH2, CH-O, or C=CH, however when Y1-Y2 is CH-O, X is selected from O or NH and R1 is not hydrogen or alkyl; and

Y3 is selected from H or Me, its stereoisomers or pharmaceutically acceptable salts thereof.

23. A method for treatment of Gaucher’s disease, Parkinsons disease with dementia

(PDD), familial Parkinson’s disease related to PINK-1 (SEQ ID NO: 2) mutation, or familial Parkinson’s disease related to mutations in a glucocerebrosidase (GBA) gene comprising administering to a subject a therapeutically effective amount of a soluble epoxide hydrolase inhibitor.

24. A compound of Formula I:

Formula I where

R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2NHR2, or COR3;

R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl;

R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy;

R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, or COR3;

R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine;

R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl;

X is selected from O, (CH2)p, NH and p is from 0-2; Y1-Y2 are selected from CH-CH2, CH-O, or C=CH, however when Y1-Y2 is CH-O, X is selected from O or NH or R1 is not hydrogen; and

Y3 is selected from H or Me. its stereoisomers or pharmaceutically acceptable salts thereof. for use in the treatment of a neurodegenerative disease or a disease associated with synucleinopathies, but not Parkinsons disease.

25. A compound of Formula I:

Formula I where

R1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R1 is aryl, heteroaryl or heterocycloalkyl, R1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2NHR2, or COR3;

R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl;

R3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy;

R4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R5, or COR3;

R5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine;

R6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl;

X is selected from O, (CH2)p, NH and p is from 0-2, however when p 0, Y1-Y2 is not

CH-CH2 or CH-O, and R1 is not aryl; Y1-Y2 are selected from CH-CH2, CH-O, or C=CH, however when Y1-Y2 is CH-O, X is selected from O or NH and R1 is not hydrogen or alkyl; and Y3 is selected from H or Me. its stereoisomers or pharmaceutically acceptable salts thereof. for use in the treatment of a neurodegenerative disease or a disease associated with synucleinopathies, but not Parkinsons disease.

26. A soluble epoxide hydrolase inhibitor for use in the treatment of Gaucher’s disease, Parkinsons disease with dementia (PDD), familial Parkinson’s disease related to

PINK-1 (SEQ ID NO: 2) mutation, or familial Parkinson’s disease related to mutations in a glucocerebrosidase (GBA) gene.

Description:
PIPERIDINE UREA DERIVATIVES FOR TREATMENT OF NEURODEGENERATIVE DISEASES

TECHNICAL FIELD

[0001] The subject matter disclosed herein is generally directed to methods and compositions for the treatment of neurodegenerative diseases and diseases associated with synucleinopathies, specifically, select piperidine urea-derived compounds for the treatment of neurodegenerative disease as well as to methods of treatment of conditions and diseases mediated by soluble epoxide hydrolase.

BACKGROUND

[0002] Epoxyeicosatrienoic acids (EETs), generated by the epoxidation of arachidonic acid by cytochrome P450 enzymes during inflammation and injury have potent antiinflammatory properties. In addition, more recent literature suggests that EETs reduce endoplasmic reticulum (ER) and mitochondrial stress in neuronal and other cells. By reducing inflammation and ER/mitochondrial stress EETs promote neuronal healing and survival. EETs levels are regulated by soluble epoxide hydrolase (sEH), the major enzyme responsible for their degradation and conversion to inactive or weakly active dihydroxyeicosatrienoic acids (DHETs). sEH, thereby, limits many of the biological actions of EETs. Inhibiting sEH increases the half-life of EETs which in turn translates into beneficial therapeutic effects.

[0003] sEH inhibitors may have utility in treatment of neuropathic and inflammatory pain, neurodegenerative diseases, diseases associated with synucleinopathies, acute respiratory distress syndrome (ARDS), rheumatoid arthritis (RA), inflammatory bowel disease (IBD), and Crohn’s disease [Biomolecules (2020), 10, 703-724; Proc. Natl. Acad. Sciences. (2018), 115, E5815-E5823,_Neurotherapeutics. (2020), 17(3), 900-916; Proc. Natl. Acad. Sciences. (2008), 105 (48), 18901-18906; Pharmacology & Therapeutics 180 (2017) 62-76; Nat. Rev. Drug. Discov. (2009), 8(10), 794-805; Cardiovasc. Hematol. Agents Med. Chem. (2012), Sep, 10(3), 212-22; Prostaglandins and Other Lipid Mediators 140 (2019) 31-39, Progress in Neurobiology, (2019), 172, 23-39, Inflamm. Allergy Drug Targets (2012) Apr, 11(2), 143-58; Mol. Pain (2011), 4, 7-78; Drug Discov Today. 2015 Nov; 20(11): 1382-90, Biomolecules. 2020 May l;10(5):703, Prostaglandins Other Lipid Mediat. (2011), 96, 76-83, Pharmacology & Therapeutics 180 (2017) 62-76; Pharmacol Ther. 2017 Jun 19, S0163-7258(17)30154-7]; Biochimie 159 (2019) 59-65. The sEH inhibitors reduce expression of inflammatory genes and show potential utility in inflammatory diseases (Inflamm. Allergy Drug Targets (2012) Apr, 11(2):143-58). The 14,15-EET is about 35-fold more potent than morphine and stimulates met- enkaphline in brain suggesting potential utility for analgesia (J Pharmacol Exp. Ther. (2008), Aug, 326(2), 614-22). [0004] Synucleins are a family of small, highly charged proteins expressed predominantly in neurons. Synuclein. a neuronal protein that regulates synaptic vesicle trafficking, is abundantly expressed in different regions of brain. One of these proteins, α-synuclein (α-syn), appears to be involved in many neurodegenerative disorders. These include Parkinson's disease (PD), dementia with Lewy bodies (DLB), Rapid Eye Movement Sleep Behavior Disorder (RBD), Lewy body variant of Alzheimer disease (LBAD), neurodegeneration with brain iron accumulation (NBAI-1) and Pure Autonomic Failure (PAF), i.e., collectively termed α- synucleinopathies. In addition, Gaucher disease (GD), the most common lysosomal storage disease caused by deficiency in glucocerebrosidase (GBA1) (SEQ ID NO: 1) and subsequent accumulation of toxic lipid substrates, has recently been recognized as the highest genetic risk factor for the development of α-synuclein aggregation disorders (synucleinopathies, including PD and DLB). Mutations in GBA result in three clinical manifestations. Type-1 Gaucher disease (GD) occurs in both children and adults and predominantly impacts non-neuronal organs, whereas type-2 and type-3 Gaucher disease have an onset in childhood and adolescence, respectively, and exhibit neurological deficits. Typical parkinsonism is among the neurological complications of Gaucher disease (including type-1). The neuropathology of Gaucher disease brains includes the typical hallmarks of Parkinson’s disease, such as cortical and brainstem Lewy bodies. [0005] All synucleinopathies demonstrate abnormal fibrillization and accumulation of proteinaceous, insoluble α-synuclein inclusions in neurons and glia, indicating a shared cellular pathology in the handling and clearance of α-synuclein. The mechanisms responsible for synuclein aggregation and pathology are not completely understood, however several processes such as neuronal inflammation, mitochondrial dysfunction and endoplasmic reticulum (ER) stress may contribute to the pathology. Mutations that contribute to mitochondrial dysfunction and synucleinopathies include PINK1 (SEQ ID NO: 2), encoding the PTEN-induced serine/threonine kinase 1. Thus, improving mitochondrial function and reducing ER stress and neuroinflammation is likely to have a positive impact on disorders associated with synucleinopathies. [0006] Several studies validate that EETs and sEH inhibition carry neuroprotective properties. sEH is highly expressed in brain and EETs production and metabolism in the brain spans many regions and extends to peripheral and central neurons, astroglia and oligodendrocytes, vascular endothelial and smooth muscle cells [J Histochem Cytochem.2008 Jun; 56(6):551-9, Am J Physiol 263: H519–25 1992, J Neurochem 61: 150–9 1993 Prostaglandins Other Lipid Mediat. 91: 68–84 2010]. Studies showed that EETs or sEH inhibition - i) prevent cytokine and oxidant mediated injury in neuronal cells; ii) prevent endoplasmic reticulum (ER) stress, a key contributor to loss of dopaminergic neurons; iii) augment astrocyte release of vascular endothelial growth factor and neuronal recovery after oxygen-glucose deprivation; and iv) promote axonal growth [Am J Physiol Heart Circ Physiol 296: H1352-63, 2009, Expert Rev Mol Med 13: 7-122011, Expert Rev Mol Med 13: 7-12, 1998, Expert Rev Mol Med 13: 7-12, 2014, Neuropathol Appl Neurobiol. 42:607-620, 2016, Proc Natl Acad Sci U S A. 112: 9082–90872015, J Neurosci 27: 4642-92007, J Neurochem. 117: 632-42 2011 and Neuroscience 223: 68-76 2012]. sEH deficiency attenuates dopaminergic neuronal cell loss in multiple animal models of Parkinson's disease [Mol Neurobiol.52(1):187-952015]. In multiple animal models, inhibition of sEH alleviates disease symptoms and promotes neuronal healing including diabetic neuropathic pain [Proc Natl Acad Sci U S A. 2008 Dec 2;105(48):18901-6, Eur J Pharmacol. 2013 Jan 30;700(1-3):93-101, J Pain.2014 Sep; 15(9):907-14, Proc Natl Acad Sci U S A.2015 Jul 21;112(29):9082-7, Behav Brain Res. 2017 May 30;326:69-76]. sEH levels are elevated in cortical brain tissue from subjects with cognitive impairment and sEH inhibition prevents H 2 O 2 -induced hyperphosphorylation of tau protein, a key factor in the pathogenesis of Alzheimer’s disease [Prostaglandins Other Lipid Mediat.2014 Oct; 113-115:30-7, J Huazhong Univ Sci Technolog Med Sci.2016 Dec; 36(6):785-790]. sEH inhibition is protective in rodent models of ischemic and diabetic stroke [Future Neurol. 2009 Mar 1;4(2):179-199, Am J Pathol. 2009 Jun; 174(6):2086-95, PLoS One.2014 May 13;9(5):e97529, Am J Physiol Heart Circ Physiol.2013 Dec 1;305(11):H1605-13]. [0007] Several studies validate that sEH inhibition could offer therapeutic utility in treatment of multiple neurodegenerative diseases such as Alzheimer’s disease [Sci Transl Med. 2020 Dec 9; 12(573): eabb1206, Journal of Neuroscience, 2020 (42):8188–8203, Journal of Neuroinflammation 16, Article 267 (2019) Neurotherapeutics volume 17, 1825–1835 (2020)], depression and related disorders, schizophrenia, stroke, subarachnoid hemorrhage, traumatic brain injury, and multiple system atrophy (MSA) [Progress in Neurobiology 172 (2019) 23– 39, J Affect Disord. 2020 June 01; 270: 131–134, Proc Natl Acad Sci U S A . 2016 Mar 29;113(13):E1944-52, J Cereb Blood Flow Metab, 35 (2) (2015), pp. 267-276, J Neurosurg, 121 (6) (2014), 1359-1366, Neurocritical Care volume 22, 306–319 (2015), Brain Hemorrhages May 2022, Neurocrit Care (2021), J Atheroscler Thromb.2017 Dec 1; 24(12): 1258–1266]. [0008] Accordingly, it is an object of the present disclosure to provide methods and compositions for the treatment of neurogenerative diseases and diseases associated with synucleinopathies including but not limited to dementia with Lewy bodies (DLB), Parkinson’s disease with dementia (PDD), multiple system atrophy (MSA), and Gaucher disease (GD). Select piperidine urea-derived compounds were presented for the treatment of dementia with Lewy bodies and Gaucher disease, as well as methods of treatment of conditions and diseases mediated by neurodegeneration and synucleinopathies. [0009] Citation or identification of any document in this application is not an admission that such a document is available as prior art to the present disclosure. SUMMARY [0010] The above objectives are accomplished according to the present disclosure by providing in one instance, a method of treating a neurodegenerative disease or a disease associated with synucleinopathies, excluding Parkinsons disease. The method may include administering to a subject a therapeutically effective amount of at least one compound of Formula I: [0012] Formula I [0013] where R 1 may be selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R 1 is aryl, heteroaryl or heterocycloalkyl, R 1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO 2 NHR 2 , or COR 3 ; [0014] R 2 may be selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; [0015] R 3 may be selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; [0016] R 4 may be selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO 2 R 5 , or COR 3 ; [0017] R 5 may be selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; [0018] R 6 may be selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; [0019] X may be selected from O, (CH 2 )p, NH and p is from 0-2; [0020] Y 1 -Y 2 may be selected from CH-CH 2 , CH-O, or C=CH, however when Y 1 -Y 2 is CH- O, X is selected from O or NH or R 1 is not hydrogen; and [0021] Y3 may be selected from H or Me; [0022] its stereoisomers or pharmaceutically acceptable salts thereof. [0023] In a further instance, a method of treating a neurodegenerative disease or a disease associated with synucleinopathies, excluding Parkinsons disease si provided. This method may include administering to a subject a therapeutically effective amount of at least one compound of Formula I [0025] Formula I [0026] where [0027] R 1 may be selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R 1 is aryl, heteroaryl or heterocycloalkyl, R 1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO 2 NHR 2 , or COR 3 ; [0028] R 2 may be selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; [0029] R 3 may be selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; [0030] R 4 may be selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO 2 R 5 , or COR 3 ; [0031] R 5 may be selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; [0032] R 6 may be selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; [0033] X may be selected from O, (CH 2 )p, NH and p is from 0-2, however when p = 0, Y 1 -Y 2 is not CH-CH 2 or CH-O, and R 1 is not aryl; [0034] Y 1 -Y 2 may be selected from CH-CH 2, CH-O , or C=CH, however when Y 1 -Y 2 is CH- O, X is selected from O or NH and R 1 is not hydrogen or alkyl; and [0035] Y3 may be selected from H or Me; [0036] its stereoisomers or pharmaceutically acceptable salts thereof. [0037] Further, Y 3 may be H and the compound is a compound according to Formula II: [0039] Formula II [0040] where [0041] R 1 may be selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R 1 is aryl, heteroaryl or heterocycloalkyl, R 1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO 2 NHR 2 , or COR 3 ; [0042] R 2 may be selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; [0043] R 3 may be selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; [0044] R 4 may be selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO 2 R 5 , COR 3 ; [0045] R 5 may be selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; [0046] R 6 may be selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; [0047] X may be selected from O, (CH 2 )p, NH and p is from 0-2; [0048] Y 1 -Y 2 may be selected from CH-CH 2, CH-O , or C=CH, however when Y 1 -Y 2 is CH-O, X is selected from O or NH or R 1 is not hydrogen; and [0049] its stereoisomers or pharmaceutically acceptable salts thereof. [0050] Still, when Y3 is H and the compound is a compound according to Formula II: [0053] where [0054] R 1 may be selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R 1 is aryl, heteroaryl or heterocycloalkyl, R 1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO 2 NHR 2 , or COR 3 ; [0055] R 2 may be selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; [0056] R 3 may be selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; [0057] R 4 may be selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO 2 R 5 , COR 3 ; [0058] R 5 may be selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; [0059] R 6 may be selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; [0060] X may be selected from O, (CH 2 )p, NH and p is from 0-2, however when p = 0, Y 1 -Y 2 is not CH-CH 2 or CH-O, and R 1 is not aryl; [0061] Y 1 -Y 2 may be selected from CH-CH 2 , CH-O, or C=CH, however when Y 1 -Y 2 is CH- O, X is selected from O or NH, and R 1 is not hydrogen or alkyl, [0062] its stereoisomers or pharmaceutically acceptable salts thereof. [0063] Still yet further, wherein Y3 is H, Y 1 -Y 2 is C=CH, and the compound is a compound according to Formula III [0064] [0065] [0066] Formula III [0067] where [0068] R 1 may be selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R 1 is aryl, heteroaryl or heterocycloalkyl, R 1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2R 5 ¸ SO 2 NHR 2 , or COR 3 ; [0069] R 2 may be selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; [0070] R 3 may be selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; [0071] R 4 may be selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO2R 5 ¸ SO 2 NHR 2 , COR 3 ; [0072] R 5 may be selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; [0073] R 6 may be selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; and [0074] X may be selected from O, (CH 2 )p, NH and p is from 0-2, [0075] its stereoisomers or pharmaceutically acceptable salts thereof. [0076] A method as claimed in claim 1 wherein Y3 is H, Y 1 -Y 2 is CH-CH 2 , and the compound is a compound according to Formula IV [0079] where [0080] R 1 may be selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R 1 is aryl, heteroaryl or heterocycloalkyl, R 1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO2R 5 ¸ SO2NHR 2 , COR 3 ; [0081] R 2 may be selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; [0082] R 3 may be selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; [0083] R 4 may be selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO 2 R 5 , COR 3 ; [0084] R 5 may be selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; [0085] R 6 may be selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl. Aryl or heteroaryl may optionally be substituted one or more times with groups or substituents such as alkyl, hydroxy, halogen, haloalkyl; and [0086] X may be selected from O, (CH 2 )p, NH; wherein p is selected from 0-2, [0087] its stereoisomers or pharmaceutically acceptable salts thereof. [0088] Morever, Y 3 may be H, Y 1 -Y 2 is CH-CH 2 , and the compound is a compound according to Formula IV [0090] Formula IV [0091] where [0092] R 1 may be selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R 1 is aryl, heteroaryl or heterocycloalkyl, R 1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO 2 R 5 ¸ SO 2 NHR 2 , COR 3 ; [0093] R 2 may be selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; [0094] R 3 may be selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; [0095] R 4 may be selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO 2 R 5 , COR 3 ; [0096] R 5 may be selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; [0097] R 6 may be selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl. Aryl or heteroaryl may optionally be substituted one or more times with groups or substituents such as alkyl, hydroxy, halogen, haloalkyl; and [0098] X may be selected from O, (CH 2 )p, NH; wherein p is selected from 0-2, however when p = 0, R 1 is not aryl; [0099] its stereoisomers or pharmaceutically acceptable salts thereof.

[0100] Even further, the compound of Formula 1 may be one or more of the following compounds [

[0102] [0103]

[0104] [0105] its stereoisomers or pharmaceutically acceptable salts thereof. [0106] Still further, the compound of Formula 1 may be one of the following compounds: [0107] [0108] its stereoisomers or pharmaceutically acceptable salts thereof. [0109] Further again, the compound of Formula 1 may be one or more of the following compounds:

[0110] [0111] its stereoisomers or pharmaceutically acceptable salts thereof. [0112] Yet again, the compound may inhibit soluble epoxide hydrolase at a concentration (IC50) of less than 10 µM. [0113] Further still, the compound may inhibit soluble epoxide hydrolase at a concentration (IC50) of less than < 100 nM. [0114] Still yet again, the compound may inhibit soluble epoxide hydrolase at a concentration (IC 50 ) of less than < 100 nM and has at least 10-fold selectivity over inhibition of fatty acid amide hydrolase (IC 50 , (FAAH (SEQ ID NO: 3)). [0115] Furthermore, the compound may inhibit soluble epoxide hydrolase at a concentration (IC 50 ) of less than < 100 nM and inhibits fatty acid amide hydrolase (FAAH (SEQ ID NO: 3)) at a concentration (IC 50 ) of > 1000 nM. [0116] Even further, the disease may be selected from Gaucher’s disease, dementia with Lewy bodies, and Alzheimer’s disease. [0117] Further again, the compound may be administered at a dose of about 1 mg/day to about 1,000 mg/day. [0118] Still yet again, the compound may be administered at a dose of about 5 mg/d ay to about 500 mg/day. [0119] Further still, the compound may be administered to treat one or more of neuronal loss, neuroinflammation, alpha-synculein aggregation and/ or lewy body formation. [0120] Yet further, the compound may be administered to treat impaired locomotor activity and other movement and non-movement related symptoms. [0121] Still yet further, the compound may be administered to treat cognitive dysfunction or dementia. [0122] Still further, the disclosure may provide a method of treating a Parkinson’s disease with dementia (PDD), familial Parkinson’s disease related to PINK-1 (SEQ ID NO: 2) mutation, or familial Parkinson’s disease related to mutations in a glucocerebrosidase (GBA) gene comprising: [0123] administering to a subject a therapeutically effective amount of at least one compound of Formula I: [0125] Formula I [0126] Where [0127] R 1 may be selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R 1 is aryl, heteroaryl or heterocycloalkyl, R 1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO 2 NHR 2 , or COR 3 ; [0128] R 2 may be selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; [0129] R 3 may be selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; [0130] R 4 may be selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO 2 R 5 , or COR 3 ; [0131] R 5 may be selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; [0132] R 6 may be selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; [0133] X may be selected from O, (CH 2 )p, NH and p is from 0-2; [0134] Y 1 -Y 2 may be selected from CH-CH 2 , CH-O, or C=CH, however when Y 1 -Y 2 is CH- O, X is selected from O or NH or R 1 is not hydrogen; and [0135] Y3 may be selected from H or Me, [0136] its stereoisomers or pharmaceutically acceptable salts thereof. [0137] Still further, the disclosure may provide a method of treating a Parkinsons disease with dementia (PDD), familial Parkinson’s disease related to PINK-1 (SEQ ID NO: 2) mutation, or familial Parkinson’s disease related to mutations in a glucocerebrosidase (GBA) gene comprising: [0138] administering to a subject a therapeutically effective amount of at least one compound of Formula I: [0140] Formula I [0141] Where [0142] R 1 may be selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R 1 is aryl, heteroaryl or heterocycloalkyl, R 1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO 2 NHR 2 , or COR 3 ; [0143] R 2 may be selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; [0144] R 3 may be selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; [0145] R 4 may be selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO 2 R 5 , or COR 3 ; [0146] R 5 may be selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; [0147] R 6 may be selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; [0148] X may be selected from O, (CH 2 )p, NH and p is from 0-2, however when p = 0, Y 1 -Y 2 is not CH-CH 2 or CH-O, and R 1 is not aryl; [0149] Y 1 -Y 2 may be selected from CH-CH 2 , CH-O, or C=CH, however when Y 1 -Y 2 is CH- O, X is selected from O or NH and R 1 is not hydrogen or alkyl; and [0150] Y 3 may be selected from H or Me, [0151] its stereoisomers or pharmaceutically acceptable salts thereof. [0152] Further yet again, the disclosure may provide a method for treatment of Gaucher’s disease, Parkinsons disease with dementia (PDD), familial Parkinson’s disease related to PINK-1 (SEQ ID NO: 2) mutation, or familial Parkinson’s disease related to mutations in a glucocerebrosidase (GBA) gene comprising administering to a subject a therapeutically effective amount of a soluble epoxide hydrolase inhibitor. [0153] A compound of Formula I: [0155] Formula I [0156] where [0157] R 1 may be selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R 1 is aryl, heteroaryl or heterocycloalkyl, R 1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO 2 NHR 2 , or COR 3 ; [0158] R 2 may be selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; [0159] R 3 may be selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; [0160] R 4 may be selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO 2 R 5 , or COR 3 ; [0161] R 5 may be selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; [0162] R 6 may be selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; [0163] X may be selected from O, (CH 2 )p, NH and p is from 0-2; [0164] Y 1 -Y 2 may be selected from CH-CH 2 , CH-O, or C=CH, however when Y 1 -Y 2 is CH- O, X is selected from O or NH or R 1 is not hydrogen; and [0165] Y 3 may be selected from [0166] its stereoisomers or pharmaceutically acceptable salts thereof. [0167] The disclosure may also provide a method for use in the treatment of a neurodegenerative disease or a disease associated with synucleinopathies, but not Parkinsons disease [0168] A compound of Formula I: [0170] Formula I [0171] where [0172] R 1 may be selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein when R 1 is aryl, heteroaryl or heterocycloalkyl, R 1 is unsubstituted or substituted with alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO 2 NHR 2 , or COR 3 ; [0173] R 2 may be selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl; [0174] R 3 may be selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy; [0175] R 4 may be selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO 2 R 5 , or COR 3 ; [0176] R 5 may be selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine; [0177] R 6 may be selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl; [0178] X may be selected from O, (CH 2 )p, NH and p is from 0-2, however when p = 0, Y 1 -Y 2 is not CH-CH 2 or CH-O, and R 1 is not aryl; [0179] Y 1 -Y 2 may be selected from CH-CH 2, CH-O , or C=CH, however when Y 1 -Y 2 is CH- O, X is selected from O or NH and R 1 is not hydrogen or alkyl; and [0180] Y3 may be selected from H or Me, [0181] its stereoisomers or pharmaceutically acceptable salts thereof. [0182] Even further, a method may be provided for use in the treatment of a neurodegenerative disease or a disease associated with synucleinopathies, but not Parkinsons disease. [0183] Still further, the disclosure may provide a soluble epoxide hydrolase inhibitor for use in the treatment of Gaucher’s disease, Parkinsons disease with dementia (PDD), familial Parkinson’s disease related to PINK-1 (SEQ ID NO: 2) mutation, or familial Parkinson’s disease related to mutations in a glucocerebrosidase (GBA) gene. [0184] These and other aspects, objects, features, and advantages of the example embodiments will become apparent to those having ordinary skill in the art upon consideration of the following detailed description of example embodiments. BRIEF DESCRIPTION OF THE DRAWINGS [0185] An understanding of the features and advantages of the present disclosure will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the disclosure may be utilized, and the accompanying drawings of which: [0186] FIG.1 shows Table 1, soluble epoxide hydrolase inhibitory potency of compounds of Formula I. [0187] FIG.2 shows neuroprotection of Compound A of Formula I in a synucleinopathy induced neuronal loss in cultured dopamine neurons. [0188] FIG. 3 shows efficacy (improvement in locomotor activity) of Compound A and Compound B of Formula I in synucleinopathy induced motor defects in a rodent model (alpha- syn + GBA1 (SEQ ID NO: 1) inhibition). [0189] FIG.4 shows efficacy of Compound A and Compound B of Formula I in blocking α-syn aggregation in a rodent model of synucleinopathy (alpha-syn + GBA1 (SEQ ID NO: 1) inhibition). [0190] FIG.5 shows neuroprotection of Compound A and Compound B of Formula I in a rodent model of synucleinopathy. [0191] FIG. 6 shows efficacy (blocking neuroinflammation) of Compound A and Compound B of Formula I in a rodent model of synucleinopathy. [0192] FIG. 7 shows efficacy (improvement in locomotor activity, bradykinesia) of Compound A and Compound B of Formula I in a zebrafish model of synucleinopathy. [0193] FIG.8 shows efficacy (improvement in dementia) of Compound A and Compound B of Formula I in a zebrafish model of synucleinopathy. [0194] FIG. 9 shows efficacy (improvement in locomotor activity) of Compound A of Formula I in a MPTP-induced Parkinsonism in a zebrafish model. [0195] FIG. 10 shows efficacy (blockage of neurodegeneration) of Compound A of Formula I in a MPTP-induced Parkinsonism in a zebrafish model. [0196] FIG.11 shows neuroprotection (TH+ neurons) of Compound A of Formula I in a MPTP-induced Parkinsonism in a zebrafish model. [0197] The figures herein are for illustrative purposes only and are not necessarily drawn to scale. DETAILED DESCRIPTION OF THE EXAMPLE EMBODIMENTS [0198] Before the present disclosure is described in greater detail, it is to be understood that this disclosure is not limited to particular embodiments described, and as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting. [0199] Unless specifically stated, terms and phrases used in this document, and variations thereof, unless otherwise expressly stated, should be construed as open ended as opposed to limiting. Likewise, a group of items linked with the conjunction “and” should not be read as requiring that each and every one of those items be present in the grouping, but rather should be read as “and/or” unless expressly stated otherwise. Similarly, a group of items linked with the conjunction “or” should not be read as requiring mutual exclusivity among that group, but rather should also be read as “and/or” unless expressly stated otherwise. [0200] Furthermore, although items, elements or components of the disclosure may be described or claimed in the singular, the plural is contemplated to be within the scope thereof unless limitation to the singular is explicitly stated. The presence of broadening words and phrases such as “one or more,” “at least,” “but not limited to” or other like phrases in some instances shall not be read to mean that the narrower case is intended or required in instances where such broadening phrases may be absent. [0201] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present disclosure, the preferred methods and materials are now described. [0202] All publications and patents cited in this specification are cited to disclose and describe the methods and/or materials in connection with which the publications are cited. All such publications and patents are herein incorporated by references as if each individual publication or patent were specifically and individually indicated to be incorporated by reference. Such incorporation by reference is expressly limited to the methods and/or materials described in the cited publications and patents and does not extend to any lexicographical definitions from the cited publications and patents. Any lexicographical definition in the publications and patents cited that is not also expressly repeated in the instant application should not be treated as such and should not be read as defining any terms appearing in the accompanying claims. The citation of any publication is for its disclosure prior to the filing date and should not be construed as an admission that the present disclosure is not entitled to antedate such publication by virtue of prior disclosure. Further, the dates of publication provided could be different from the actual publication dates that may need to be independently confirmed. [0203] As will be apparent to those of skill in the art upon reading this disclosure, each of the individual embodiments described and illustrated herein has discrete components and features which may be readily separated from or combined with the features of any of the other several embodiments without departing from the scope or spirit of the present disclosure. Any recited method can be carried out in the order of events recited or in any other order that is logically possible. [0204] Where a range is expressed, a further embodiment includes from the one particular value and/or to the other particular value. The recitation of numerical ranges by endpoints includes all numbers and fractions subsumed within the respective ranges, as well as the recited endpoints. Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the disclosure. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges and are also encompassed within the disclosure, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the disclosure. For example, where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the disclosure, e.g. the phrase “x to y” includes the range from ‘x’ to ‘y’ as well as the range greater than ‘x’ and less than ‘y’. The range can also be expressed as an upper limit, e.g. ‘about x, y, z, or less’ and should be interpreted to include the specific ranges of ‘about x’, ‘about y’, and ‘about z’ as well as the ranges of ‘less than x’, less than y’, and ‘less than z’. Likewise, the phrase ‘about x, y, z, or greater’ should be interpreted to include the specific ranges of ‘about x’, ‘about y’, and ‘about z’ as well as the ranges of ‘greater than x’, greater than y’, and ‘greater than z’. In addition, the phrase “about ‘x’ to ‘y’”, where ‘x’ and ‘y’ are numerical values, includes “about ‘x’ to about ‘y’”. [0205] It should be noted that ratios, concentrations, amounts, and other numerical data can be expressed herein in a range format. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. It is also understood that there are a number of values disclosed herein, and that each value is also herein disclosed as “about” that particular value in addition to the value itself. For example, if the value “10” is disclosed, then “about 10” is also disclosed. Ranges can be expressed herein as from “about” one particular value, and/or to “about” another particular value. Similarly, when values are expressed as approximations, by use of the antecedent “about,” it will be understood that the particular value forms a further aspect. For example, if the value “about 10” is disclosed, then “10” is also disclosed. [0206] It is to be understood that such a range format is used for convenience and brevity, and thus, should be interpreted in a flexible manner to include not only the numerical values explicitly recited as the limits of the range, but also to include all the individual numerical values or sub-ranges encompassed within that range as if each numerical value and sub-range is explicitly recited. To illustrate, a numerical range of “about 0.1% to 5%” should be interpreted to include not only the explicitly recited values of about 0.1% to about 5%, but also include individual values (e.g., about 1%, about 2%, about 3%, and about 4%) and the sub- ranges (e.g., about 0.5% to about 1.1%; about 5% to about 2.4%; about 0.5% to about 3.2%, and about 0.5% to about 4.4%, and other possible sub-ranges) within the indicated range. [0207] As used herein, the singular forms “a”, “an”, and “the” include both singular and plural referents unless the context clearly dictates otherwise. [0208] As used herein, "about," "approximately," “substantially,” and the like, when used in connection with a measurable variable such as a parameter, an amount, a temporal duration, and the like, are meant to encompass variations of and from the specified value including those within experimental error (which can be determined by e.g. given data set, art accepted standard, and/or with e.g. a given confidence interval (e.g. 90%, 95%, or more confidence interval from the mean), such as variations of +/-10% or less, +/-5% or less, +/-1% or less, and +/-0.1% or less of and from the specified value, insofar such variations are appropriate to perform in the disclosure. As used herein, the terms “about,” “approximate,” “at or about,” and “substantially” can mean that the amount or value in question can be the exact value or a value that provides equivalent results or effects as recited in the claims or taught herein. That is, it is understood that amounts, sizes, formulations, parameters, and other quantities and characteristics are not and need not be exact, but may be approximate and/or larger or smaller, as desired, reflecting tolerances, conversion factors, rounding off, measurement error and the like, and other factors known to those of skill in the art such that equivalent results or effects are obtained. In some circumstances, the value that provides equivalent results or effects cannot be reasonably determined. In general, an amount, size, formulation, parameter or other quantity or characteristic is “about,” “approximate,” or “at or about” whether or not expressly stated to be such. It is understood that where “about,” “approximate,” or “at or about” is used before a quantitative value, the parameter also includes the specific quantitative value itself, unless specifically stated otherwise. [0209] As used herein, a “biological sample” may contain whole cells and/or live cells and/or cell debris. The biological sample may contain (or be derived from) a “bodily fluid”. The present disclosure encompasses embodiments wherein the bodily fluid is selected from amniotic fluid, aqueous humour, vitreous humour, bile, blood serum, breast milk, cerebrospinal fluid, cerumen (earwax), chyle, chyme, endolymph, perilymph, exudates, feces, female ejaculate, gastric acid, gastric juice, lymph, mucus (including nasal drainage and phlegm), pericardial fluid, peritoneal fluid, pleural fluid, pus, rheum, saliva, sebum (skin oil), semen, sputum, synovial fluid, sweat, tears, urine, vaginal secretion, vomit and mixtures of one or more thereof. Biological samples include cell cultures, bodily fluids, and cell cultures from bodily fluids. Bodily fluids may be obtained from a mammal organism, for example by puncture, or other collecting or sampling procedures. [0210] As used herein, “agent” refers to any substance, compound, molecule, and the like, which can be administered to a subject on a subject to which it is administered to. An agent can be inert. An agent can be an active agent. An agent can be a primary active agent, or in other words, the component(s) of a composition to which the whole or part of the effect of the composition is attributed. An agent can be a secondary agent, or in other words, the component(s) of a composition to which an additional part and/or other effect of the composition is attributed. [0211] As used herein, “active agent” or “active ingredient” refers to a substance, compound, or molecule, which is biologically active or otherwise that induces a biological or physiological effect on a subject to which it is administered to. In other words, “active agent” or “active ingredient” refers to a component or components of a composition to which the whole or part of the effect of the composition is attributed. [0212] As used herein, “administering” refers to any suitable administration for the agent(s) being delivered and/or subject receiving said agent(s) and can be oral, topical, intravenous, subcutaneous, transcutaneous, transdermal, intramuscular, intra-joint, parenteral, intra-arteriole, intradermal, intraventricular, intraosseous, intraocular, intracranial, intraperitoneal, intralesional, intranasal, intracardiac, intraarticular, intracavernous, intrathecal, intravireal, intracerebral, and intracerebroventricular, intratympanic, intracochlear, rectal, vaginal, by inhalation, by catheters, stents or via an implanted reservoir or other device that administers, either actively or passively (e.g. by diffusion) a composition to the perivascular space and adventitia. For example, a medical device such as a stent can contain a composition or formulation disposed on its surface, which can then dissolve or be otherwise distributed to the surrounding tissue and cells. The term “parenteral” can include subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional, and intracranial injections or infusion techniques. Administration routes can be, for instance, auricular (otic), buccal, conjunctival, cutaneous, dental, electro-osmosis, endocervical, endosinusial, endotracheal, enteral, epidural, extra-amniotic, extracorporeal, hemodialysis, infiltration, interstitial, intra-abdominal, intra-amniotic, intra-arterial, intra- articular, intrabiliary, intrabronchial, intrabursal, intracardiac, intracartilaginous, intracaudal, intracavernous, intracavitary, intracerebral, intracisternal, intracorneal, intracoronal (dental), intracoronary, intracorporus cavernosum, intradermal, intradiscal, intraductal, intraduodenal, intradural, intraepidermal, intraesophageal, intragastric, intragingival, intraileal, intralesional, intraluminal, intralymphatic, intramedullary, intrameningeal, intramuscular, intraocular, intraovarian, intrapericardial, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrasinal, intraspinal, intrasynovial, intratendinous, intratesticular, intrathecal, intrathoracic, intratubular, intratumor, intratym panic, intrauterine, intravascular, intravenous, intravenous bolus, intravenous drip, intraventricular, intravesical, intravitreal, iontophoresis, irrigation, laryngeal, nasal, nasogastric, occlusive dressing technique, ophthalmic, oral, oropharyngeal, other, parenteral, percutaneous, periarticular, peridural, perineural, periodontal, rectal, respiratory (inhalation), retrobulbar, soft tissue, subarachnoid, subconjunctival, subcutaneous, sublingual, submucosal, topical, transdermal, transmucosal, transplacental, transtracheal, transtympanic, ureteral, urethral, and/or vaginal administration, and/or any combination of the above administration routes, which typically depends on the disease to be treated, subject being treated, and/or agent(s) being administered. [0213] As used herein, “control” can refer to an alternative subject or sample used in an experiment for comparison purpose and included to minimize or distinguish the effect of variables other than an independent variable. [0214] The term “optional” or “optionally” means that the subsequent described event, circumstance or substituent may or may not occur, and that the description includes instances where the event or circumstance occurs and instances where it does not. [0215] As used herein, “dose,” “unit dose,” or “dosage” can refer to physically discrete units suitable for use in a subject, each unit containing a predetermined quantity of a pharmaceutical formulation calculated to produce the desired response or responses in association with its administration. [0216] The term “molecular weight”, as used herein, can generally refer to the mass or average mass of a material. [0217] The terms “subject,” “individual,” and “patient” are used interchangeably herein to refer to a vertebrate, preferably a mammal, more preferably a human. Mammals include, but are not limited to, murines, simians, humans, farm animals, sport animals, and pets. Tissues, cells and their progeny of a biological entity obtained in vivo or cultured in vitro are also encompassed by the term “subject”. [0218] As used herein, "substantially pure" can mean an object species is the predominant species present (i.e., on a molar basis it is more abundant than any other individual species in the composition), and preferably a substantially purified fraction is a composition wherein the object species comprises about 50 percent of all species present. Generally, a substantially pure composition will comprise more than about 80 percent of all species present in the composition, more preferably more than about 85%, 90%, 95%, and 99%. Most preferably, the object species is purified to essential homogeneity (contaminant species cannot be detected in the composition by conventional detection methods) wherein the composition consists essentially of a single species. [0219] As used interchangeably herein, the terms “sufficient” and “effective,” can refer to an amount (e.g. mass, volume, dosage, concentration, and/or time period) needed to achieve one or more desired and/or stated result(s). For example, a therapeutically effective amount refers to an amount needed to achieve one or more therapeutic effects. [0220] As used herein, “tangible medium of expression” refers to a medium that is physically tangible or accessible and is not a mere abstract thought or an unrecorded spoken word. “Tangible medium of expression” includes, but is not limited to, words on a cellulosic or plastic material, or data stored in a suitable computer readable memory form. The data can be stored on a unit device, such as a flash memory or CD-ROM or on a server that can be accessed by a user via, e.g. a web interface. [0221] As used herein, the terms "treating" and "treatment" can refer generally to obtaining a desired pharmacological and/or physiological effect. The effect can be, but does not necessarily have to be, prophylactic in terms of preventing or partially preventing a disease, symptom or condition thereof, such as cancer and/or indirect radiation damage. The effect can be therapeutic in terms of a partial or complete cure of a disease, condition, symptom or adverse effect attributed to the disease, disorder, or condition. The term "treatment" as used herein covers any treatment of cancer and/or indirect radiation damage, in a subject, particularly a human and/or companion animal, and can include any one or more of the following: (a) preventing the disease or damage from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development; and (c) relieving the disease, i.e., mitigating or ameliorating the disease and/or its symptoms or conditions. The term "treatment" as used herein can refer to both therapeutic treatment alone, prophylactic treatment alone, or both therapeutic and prophylactic treatment. Those in need of treatment (subjects in need thereof) can include those already with the disorder and/or those in which the disorder is to be prevented. As used herein, the term "treating", can include inhibiting the disease, disorder or condition, e.g., impeding its progress; and relieving the disease, disorder, or condition, e.g., causing regression of the disease, disorder and/or condition. Treating the disease, disorder, or condition can include ameliorating at least one symptom of the particular disease, disorder, or condition, even if the underlying pathophysiology is not affected, such as treating the pain of a subject by administration of an analgesic agent even though such agent does not treat the cause of the pain. [0222] As used herein, the terms “weight percent,” “wt%,” and “wt. %,” which can be used interchangeably, indicate the percent by weight of a given component based on the total weight of a composition of which it is a component, unless otherwise specified. That is, unless otherwise specified, all wt% values are based on the total weight of the composition. It should be understood that the sum of wt% values for all components in a disclosed composition or formulation are equal to 100. Alternatively, if the wt% value is based on the total weight of a subset of components in a composition, it should be understood that the sum of wt% values the specified components in the disclosed composition or formulation are equal to 100. [0223] "Halogen or Halo" means fluorine, chlorine, bromine or iodine. [0224] "Alkyl" group means linear or branched alkyl groups. Exemplary alkyl groups include methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, t-butyl, n-pentyl, iso-pentyl, hexyl, heptyl, octyl and the like. Unless otherwise specified, an alkyl group typically has from about 1 to about 10 carbon atoms. [0225] "Cycloalkyl" group means a cyclic alkyl group which may be mono or bicyclic. Exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl and the like. Unless otherwise specified, a cycloalkyl group typically has from about 3 to about 10 carbon atoms. [0226] “Haloalkyl” group means linear or branched alkyl groups wherein at least one hydrogen is replaced by halogen or halo groups. Exemplary haloalkyl groups include trifluromethyl, chloroethyl, difluoromethyl, difluoroethyl, and the like. [0227] Hydroxyalkyl” group means a linear monovalent hydrocarbon radical of one to three carbon atoms or a branched monovalent hydrocarbon radical of three to five carbons substituted with one or two hydroxy groups, provided that if two hydroxy groups are present they are not both on the same carbon atom. Representative examples include hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropyl, 3-hydroxypropyl. [0228] “Alkoxyalkyl” means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with an alkoxy group, as defined above, e.g., methoxymethyl, 2-methoxyethyl, 1-, 2-, or 3- methoxypropyl. [0229] “Heterocycloalkyl” means a non-aromatic monocyclic or polycyclic ring comprising carbon and hydrogen atoms and at least one heteroatom, preferably, 1 to 4 heteroatoms selected from nitrogen, oxygen, and sulfur. A heterocycloalkyl group can have one or more carbon-carbon double bonds or carbon-heteroatoms double bonds in the ring as long as the ring is not rendered aromatic by their presence. Examples of heterocycloalkyl groups include pyrrolidinyl, pyrrolidino, piperidinyl, piperidino, piperazinyl, piperazino, morpholinyl, morpholino, tetrahydrofuranyl, tetrahydropyranyl, and pyranyl. [0230] "Alkoxy" means an -O (alkyl) group, where alkyl is as defined above. Exemplary alkoxyl groups include methoxy, ethoxy, propoxy, butoxy, iso-propoxy, iso-butoxy, and the like. Unless otherwise specified, an alkoxy group typically has from 1 to about 10 carbon atoms. [0231] "Amine" refers to a primary, secondary, or tertiary amino group. The secondary and tertiary amine may contain alkyl, cycloalkyl or aryl substitutions. Some examples of amines include NH 2 , NHMe, NMe2 NH(cyclopropyl). Unless otherwise specified, the alkyl or cycloalkyl group on an amine typically has from 1 to about 8 carbon atoms. [0232] "Aryl" means an optionally substituted monocyclic or polycyclic aromatic ring system of about 6 to about 14 carbon atoms. Exemplary aryl groups include phenyl, naphthyl, and the like. Unless otherwise specified, an aryl group typically has from 6 to about 14 carbon atoms. [0233] "Heteroaryl" means an aromatic monocyclic or polycyclic ring system of about 4 to about 12 carbon atoms, having at least one heteroatom or hetero group selected from -O-, - N-, -S-, -SO 2 , or -CO. Exemplary heteroaryl groups include one or more of pyrazinyl, isothiazolyl, oxazolyl, pyrazolyl, pyrrolyl, tetrazolyl, imidazolyl, triazolyl, pyridazinyl, thienopyrimidyl, furanyl, indolyl, isoindolyl, benzo[1,3]dioxolyl, benzimidazolyl, 1,3- benzoxathiolyl, pyrrolidine 2,4-dionyl, quinazolinyl, pyridyl, pyrimidinyl, thiophenyl and the like. Unless otherwise specified, a heteroaryl group typically has from 4 to about 10 carbon atoms. [0234] "5- to 6- member heteroaryl" is an aromatic monocyclic ring system of 5 or 6 ring atoms, having at least one heteroatom or hetero group selected from -O-, -N-, -S-, -SO 2 , or - CO. Exemplary "5- to 6- member heteroaryl" groups include one or more of pyrazinyl, isothiazolyl, oxazolyl, pyrazolyl, pyrrolyl, pyridazinyl, pyridyl, thienopyrimidyl, tetrazolyl, imidazolyl, triazolyl , furanyl and the like. [0235] "Optionally substituted" means that substitution is optional and, therefore, it is possible for the designated atom or molecule to be unsubstituted. In the event a substitution is desired, then such substitution means that any number of hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the normal valence of the designated atom is not exceeded, and that the substitution results in a sufficiently stable compound for use. [0236] "Salts" refers to any acid or base salt, pharmaceutically acceptable solvates, or any complex of the compound that, when administered to a recipient, is capable of providing (directly or indirectly) a compound as described herein. It should be appreciated, however, that salts that are not pharmaceutically acceptable also lie within the scope of the disclosure. The preparation of salts can be carried out using known methods. For example, pharmaceutically acceptable salts of compounds contemplated herein as being useful may be synthesized by conventional chemical methods using a parent compound containing a base or an acid functionality. Generally, such salts may be prepared, for example, by making free acid or base forms of the compounds and reacting with a stoichiometric quantity of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two. Generally, non- aqueous media such as one or more of solvents such as ether, ethyl acetate, ethanol, isopropanol or acetonitrile may be utilized. Examples of acid addition salts include one or more of mineral acid addition salts such as hydrochloride, hydrobromide, hydroiodide, sulphate, phosphate, and organic acid addition salts such as one or more of acetate, maleate, fumarate, citrate, oxalate, succinate, tartrate, malate, mandelate, methanesulphonate and p-toluenesulphonate. Examples of base addition salts include one or more of inorganic salts such as sodium, potassium, calcium, ammonium, magnesium, and lithium salts, and organic base salts such as one or more of ethylenediamine, ethanolamine, N,N-dialkyl-ethanolamine, triethanolamine, and basic amino acid salts. [0237] The phrase "therapeutically effective" indicates the capability of an agent or combination to prevent, or reduce the severity of, the disorder or the symptons of the disease, while generally avoiding adverse side effects. The therapeutically effective compositions of the present disclosure may include compounds of the present disclosure at doses of from about 1 to about 3000 mg. [0238] As used herein, “effective amount” or “therapeutically effective amount" means the dose or amount of a compound of the present disclosure administered to a subject and the frequency of administration to result in some therapeutic response. The dose or effective amount to be administered to a subject and the frequency of administration to the subject can be readily determined by one of ordinary skill in the art by the use of known techniques and by observing results obtained under analogous circumstances. In determining the effective amount or dose, a number of factors may be considered by the attending medical practitioner, including, but not limited to, the potency and duration of action of the compounds used; the nature and severity of the illness to be treated as well as the sex, age, weight, general health and individual responsiveness of the subject to be treated, and other relevant circumstances. [0239] The compounds described herein may be administered in admixture with one or more pharmaceutically acceptable excipients or carriers in the form of a pharmaceutical composition. A "composition" may contain one compound or a mixture of compounds. A "pharmaceutical composition" is any composition useful or potentially useful in producing physiological response in a subject to which such pharmaceutical composition is administered. [0240] The term "pharmaceutically acceptable," with respect to an excipient, is used to define non-toxic substances generally suitable for use in human or animal pharmaceutical products. The pharmaceutical composition may be in the forms normally employed, such as tablets, capsules, powders, syrups, solutions, suspensions, and the like. The pharmaceutical composition may contain flavorants, sweeteners, etc., in suitable solid or liquid carriers or diluents, or in suitable sterile media to form injectable solutions or suspensions. Such compositions typically contain from about 0.1 to about 50%, and in some embodiments, from about 1 to about 20%, by weight of active compound, the remainder of the composition being pharmaceutically acceptable carriers, diluents or solvents. [0241] Various embodiments are described hereinafter. It should be noted that the specific embodiments are not intended as an exhaustive description or as a limitation to the broader aspects discussed herein. One aspect described in conjunction with a particular embodiment is not necessarily limited to that embodiment and can be practiced with any other embodiment(s). Reference throughout this specification to “one embodiment”, “an embodiment,” “an example embodiment,” means that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment of the present disclosure. Thus, appearances of the phrases “in one embodiment,” “in an embodiment,” or “an example embodiment” in various places throughout this specification are not necessarily all referring to the same embodiment, but may. Furthermore, the particular features, structures or characteristics may be combined in any suitable manner, as would be apparent to a person skilled in the art from this disclosure, in one or more embodiments. Furthermore, while some embodiments described herein include some but not other features included in other embodiments, combinations of features of different embodiments are meant to be within the scope of the disclosure. For example, in the appended claims, any of the claimed embodiments can be used in any combination. [0242] All patents, patent applications, published applications, and publications, databases, websites and other published materials cited herein are hereby incorporated by reference to the same extent as though each individual publication, published patent document, or patent application was specifically and individually indicated as being incorporated by reference. KITS [0243] Any of the compounds and/or formulations described herein can be presented as a combination kit. As used herein, the terms "combination kit" or "kit of parts" refers to the compounds, compositions, formulations, particles, cells and any additional components that are used to package, sell, market, deliver, and/or administer the combination of elements or a single element, such as the active ingredient, contained therein. Such additional components include, but are not limited to, packaging, syringes, blister packages, bottles, and the like. When one or more of the compounds, compositions, formulations, particles, cells, described herein or a combination thereof (e.g., agent(s)) contained in the kit are administered simultaneously, the combination kit can contain the active agent(s) in a single formulation, such as a pharmaceutical formulation, (e.g., a tablet, liquid preparation, dehydrated preparation, etc.) or in separate formulations. When the compounds, compositions, formulations, particles, and cells described herein or a combination thereof and/or kit components are not administered simultaneously, the combination kit can contain each agent or other component in separate pharmaceutical formulations. The separate kit components can be contained in a single package or in separate packages within the kit. [0244] In some embodiments, the combination kit also includes instructions printed on or otherwise contained in a tangible medium of expression. The instructions can provide information regarding the content of the compounds and/or formulations, safety information regarding the content of the compounds and formulations (e.g., pharmaceutical formulations), information regarding the dosages, indications for use, and/or recommended treatment regimen(s) for the compound(s) and/or ph DETAILED DESCRIPTION OF THE DISCLOSURE [0245] Reference will now be made in detail to the embodiments of the disclosure, one or more examples of which are set forth below. Each example is provided by way of explanation of the disclosure, not a limitation of the disclosure. In fact, it will be apparent to those skilled in the art that various modifications can be made in the present disclosure without departing from the scope or spirit of the disclosure. For instance, features illustrated or described as part of one embodiment can be used on another embodiment to yield a still further embodiment. Thus it is intended that the present disclosure cover such modifications and variations as come within the scope of the appended claims and their equivalents. Other objects, features, and aspects of the present disclosure are disclosed in, or are obvious from, the following detailed description. [0246] For ease of reference, the present disclosure will be described in terms of administration to human subjects. It will be understood, however, that such descriptions are not limited to administration to humans, but will also include administration to other animals unless explicitly stated otherwise. [0247] Contemplated derivatives are those that may improve dissolution or increase the bioavailability of the compounds of this disclosure when such compounds are administered to a subject (e.g., by making an orally administered compound more easily absorbed). Compounds of Formula I may be amorphous, semi-crystalline, or crystalline and may either be given as parent compounds, its salts, and/or in solvated form. The solvate may be part of crystalline lattice or superficially associated. It is intended that all of these forms should be within the scope of the present disclosure. Methods of solvation are generally known within the art. Suitable solvates are pharmaceutically acceptable solvates. In one embodiment, the solvate is a hydrate. [0248] In one aspect, the present disclosure is directed to novel compounds of Formula I: [0250] its stereoisomers, stable label (e.g., deuterated variants), or pharmaceutically acceptable salts thereof; [0251] Wherein [0252] R 1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein R 1 may optionally be substituted one or more times with groups or substituents such as alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO 2 NHR 2 , COR 3 [0253] R 2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl [0254] R 3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy [0255] R 4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO 2 R 5 , COR 3 [0256] R 5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine [0257] R 6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl. aryl or heteroaryl may optionally be substituted one or more times with groups or substituents such as alkyl, hydroxy, halogen, haloalkyl [0258] X is selected from O, (CH 2 )p, NH; wherein p is selected from 0-2, however when p = 0, Y 1 -Y 2 is not CH-CH 2 or CH-O, and R 1 is not aryl; and [0259] Y 1 -Y 2 are selected from CH-CH 2 , CH 2 -O, or C=CH; however, when Y 1 -Y 2 is CH 2 - O, X is selected from O or NH and R 1 is not hydrogen or alkyl [0260] Y3 is selected from H or Me [0261] In another aspect, the present disclosure is directed to novel compounds of Formula II: [0263] its stereoisomers or pharmaceutically acceptable salts thereof; [0264] wherein [0265] R 1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein R 1 may optionally be substituted one or more times with groups or substituents such as alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO 2 NHR 2 , COR 3 [0266] R 2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl [0267] R 3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy [0268] R 4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO 2 R 5 , COR 3 [0269] R 5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine [0270] R 6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl. Aryl or heteroaryl may optionally be substituted one or more times with groups or substituents such as alkyl, hydroxy, halogen, haloalkyl [0271] X is selected from O, (CH 2 )p, NH; wherein p is selected from 0-2, however when p = 0, Y 1 -Y 2 is not CH-CH 2 or CH-O, and R 1 is not aryl; and [0272] Y 1 -Y 2 are selected from CH-CH 2 , CH-O, or C=CH; however, when Y 1 -Y 2 is CH 2 - O, X is selected from O or NH and R 1 is not hydrogen or alkyl.. [0273] In another aspect, the present disclosure is directed to novel compounds of Formula III: [0274] Formula III [0275] its stereoisomers or pharmaceutically acceptable salts thereof; [0276] wherein [0277] R 1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein R 1 may optionally be substituted one or more times with groups or substituents such as alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO 2 R 5 , SO 2 NHR 2 , COR 3 [0278] R2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl [0279] R 3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy [0280] R 4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO 2 R 5 , COR 3 [0281] R 5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine [0282] R 6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl. Aryl or heteroaryl may optionally be substituted one or more times with groups or substituents such as alkyl, hydroxy, halogen, haloalkyl [0283] X is selected from O, (CH 2 )p, NH; wherein p is selected from 0-2 [0284] In another aspect, the present invention is directed to novel compounds of Formula IV: [0285] Formula IV [0286] its stereoisomers or pharmaceutically acceptable salts thereof; [0287] wherein [0288] R 1 is selected from a group consisting of alkyl, hydrogen, haloalkyl, cycloalkyl, heterocycloalkyl, aryl, or heteroaryl and wherein R 1 may optionally be substituted one or more times with groups or substituents such as alkyl, hydroxy, halogen, haloalkyl, hydroxyalkyl, alkoxyalkyl, alkoxy, amine, SO 2 R 5 ¸ SO 2 NHR 2 , COR 3 [0289] R 2 is selected from a group consisting of hydrogen, alkyl, haloalkyl, or cycloalkyl [0290] R 3 is selected from a group consisting of alkyl, cycloalkyl, hydroxy, amine, alkyamine or alkoxy [0291] R 4 is selected from a group consisting of hydrogen, alkyl, halogen, haloalkyl, hydroxy, amine, alkoxy, SO 2 R 5 , COR 3 [0292] R 5 is selected from a group consisting of alkyl, haloalkyl, cycloalkyl, aryl or amine [0293] R 6 is selected from a group consisting of alkyl, cycloalkyl, aryl or heteroaryl. Aryl or heteroaryl may optionally be substituted one or more times with groups or substituents such as alkyl, hydroxy, halogen, haloalkyl [0294] X is selected from O, (CH 2 )p, NH; wherein p is selected from 0-2, however when p = 0, R 1 is not aryl [0295] Further, the composition may comprise one or more of the following compounds, its stereoisomers or pharmaceutically acceptable salts thereof;

[0296]

[0301] In an embodiment, the disclosure may comprise compounds of Formula I where the compounds are inhibitors of soluble epoxide hydrolase (sEH) with IC 50 of < 10 uM. [0302] In an embodiment, the disclosure may comprise compounds of Formula I where the compounds are inhibitors of soluble epoxide hydrolase (sEH) with >10-fold selectivity for fatty acid amide hydrolase (FAAH (SEQ ID NO: 3)). [0303] In another embodiment, the compounds of Formula I may inhibit soluble epoxide hydrolase with IC50 of <100 nM and inhibit fatty acid amide hydrolase (FAAH (SEQ ID NO: 3)) with IC 50 of >1000 nM [0304] In a further embodiment, the compounds of Formula I may inhibit soluble epoxide hydrolase with IC50 of <50 nM and inhibit fatty acid amide hydrolase (FAAH (SEQ ID NO: 3)) with IC 50 of >1000 nM [0305] In a further embodiment, the compounds of Formula I may inhibit soluble epoxide hydrolase with IC50 of <20 nM and inhibit fatty acid amide hydrolase (FAAH (SEQ ID NO: 3)) with IC50 of >1000 nM [0306] In some embodiments, the therapeutically effective amount of a compound of Formula I may be from about 0.5 mg/day to about 3,000 mg/day. In embodiments, the therapeutically effective amount of a compound of Formula I may be from about 1 mg/day to about 2,000 mg/day. In embodiments, the therapeutically effective amount of a compound of Formula I may be from about 2 mg/day to about 600 mg/day. In embodiments, the therapeutically effective amount of a compound of Formula I may be from 3 mg/day to about 500 mg/day. In embodiments, the therapeutically effective amount of a compound of Formula I may be from 4 mg/day to about 400 mg/day. In embodiments, the therapeutically effective amount of a compound of Formula I may be from 3 mg/day to about 400 mg/day. In embodiments, the therapeutically effective amount of a compound of Formula I may be from 10 mg/day to about 300 mg/day [0307] In some embodiments, the therapeutically effective amount of a compound of Formula I for an adult subject may be from about 2 mg to about 1,500 mg per day. In embodiments, the therapeutically effective amount of a compound of Formula I for an adult subject may be from about 4 mg to about 750 mg per day. In embodiments, the therapeuti-cally effective amount of a compound of Formula I for an adult subject may be from about 6 mg to about 600 mg per day. In embodiments, the therapeutically effective amount of a compound of Formula I for an adult subject may be from about 10 mg to about 500 mg per day. In embodiments, the therapeutically effective amount of a compound of Formula I for an adult subject may be from about 20 mg to about 400 mg per day. In embodiments, the therapeutically effective amount of a compound of Formula I for an adult subject may be from about 20 mg to about 300 mg per day. In embodiments, the therapeutically effective amount of a compound of Formula I for an adult subject may be from about 20 mg to about 200 mg per day. In embodiments, the therapeutically effective amount of a compound of Formula I for an adult subject may be from about 20 mg to about 120 mg per day. In embodiments, the therapeutically effective amount of a compound of Formula I for an adult subject may be from about 20 mg to about 100 mg per day. In embodiments, the therapeutically effective amount of a compound of Formula I for an adult subject may be from about 20 mg to about 75 mg per day. In embodiments, the therapeutically effective amount of a compound of Formula I for an adult subject may be from about 20 mg to about 60 mg per day. In embodiments, the therapeutically effective amount of a compound of Formula I for an adult subject may be from about 20 mg to about 50 mg per day. In embodiments, the therapeutically effective amount of a compound of Formula I for an adult subject may be from about 20 mg to about 40 mg per day. In embodiments, the therapeutically effective amount of a compound of Formula I for an adult subject may be from about 24 mg to about 40 mg per day. In embodiments, the therapeutically effective amount of a compound of Formula I for a non-adult subject may be from about 0.1 mg to about 800 mg per day. In embodiments, the therapeutically effective amount of a compound of Formula I for a non-adult subject may be from about 0.25 mg to about 350 mg per day. In embodiments, the therapeuti-cally effective amount of a compound of Formula I for a non-adult subject may be from about 0.5 mg to about 300 mg per day. In embodiments, the therapeuti-cally effective amount of a compound of Formula I for a non-adult subject may be from about 1 mg to about 200 mg per day. In embodiments, the therapeuti-cally effective amount of a compound of Formula I for a non-adult subject may be from about 2 mg to about 100 mg per day. In embodiments, the therapeuti-cally effective amount of a compound of Formula I for a non-adult subject may be from about 3 mg to about 80 mg per day. In embodiments, the therapeuti-cally effective amount of a compound of Formula I for a non- adult subject may be from about 4 mg to about 60 mg per day. In embodiments, the therapeuti-cally effective amount of a compound of Formula I for a non-adult subject may be from about 5 mg to about 80 mg per day. In embodiments, the therapeuti-cally effective amount of a compound of Formula I for a non-adult subject may be from about 6 mg to about 60 mg per day. In embodiments, the therapeuti-cally effective amount of a compound of Formula I for a non-adult subject may be from about 6 mg to about 50 mg per day. In embodiments, the therapeuti-cally effective amount of a compound of Formula I for a non-adult subject may be from about 6 mg to about 40 mg per day. In embodiments, the therapeuti-cally effective amount of a compound of Formula I for a non-adult subject may be from about 6 mg to about 30 mg per day. In embodiments, the therapeuti-cally effective amount of a compound of Formula I for a non-adult subject may be from about 7 mg to about 25 mg per day. [0308] In some embodiments, the therapeutically effective amount of a compound of Formula I may be administered in a single dose or a dose repeated one or several times after a certain time interval. In embodiments, the therapeutically effective amount is administered daily or every two or three days, or once a week. Administration may be one, twice, or three times daily, In embodiments, the therapeutically effective amount is administered daily or every other day for more than two weeks, ten weeks, thirty weeks, a year, or as long as symptoms or the disease are present. In embodiments, the therapeutically effective amount is administered daily or every other day for more than thirty weeks. [0309] In yet another aspect, the disclosure is directed to novel compounds of Formula I, its stereoisomers, stable label (e.g., deuterated) variants, tautomers and/or its pharmaceutically acceptable salts thereof which can be used as inhibitors of soluble epoxide hydrolase (sEH). [0310] In another aspect, the disclosure is directed to a method for the prevention and/or treatment of pain, neurodegenerative diseases, diseases associated with synucleinopathies, and inflammatory disorders in a subject in need of such treatment, the method comprising administering to the subject a therapeutically effective amount of a compound of Formula I, its stereoisomers and/or its pharmaceutically acceptable salts thereof. Still, the compounds of Formula I, its stereoisomers and/or pharmaceutically acceptable salts may be used for the prevention or treatment of diseases associated with synucleinopathies such as Lewy body dementia (LBD) and Gaucher disease (GD), Rapid Eye Movement Sleep Behavior Disorder (RBD), Lewy body variant of Alzheimer disease (LBAD), neurodegeneration with brain iron accumulation (NBAI-1) and Pure Autonomic Failure (PAF). [0311] Further still, the compounds of Formula I, its stereoisomers and/or pharmaceutically acceptable salts may be used for the prevention or treatment of Alzheimer’s disease, depression and related disorders, schizophrenia, stroke, subarachnoid hemorrhage, traumatic brain injury, and multiple system atrophy (MSA) in a subject in need of such treatment, the method comprising administering to the subject a therapeutically effective amount of a composition described herein [0312] In another embodiment, the stereoisomers and/or pharmaceutically acceptable salts of Formula I may be used for the treatment of neurodegenerative diseases such as dementia with Lewy bodies (DLB) and Gaucher disease. [0313] For ease of reference, the present disclosure will be described in terms of administration to human subjects. It will be understood, however, that such descriptions are not limited to administration to humans, but will also include administration to other animals unless explicitly stated otherwise. [0314] Contemplated derivatives are those that may improve dissolution or increase the bioavailability of the compounds of this disclosure when such compounds are administered to a subject (e.g., by making an orally administered compound more easily absorbed). Compounds of Formula I may be amorphous, semi-crystalline, or crystalline and may either be given as parent compounds, its salts, and/or in solvated form. The solvate may be part of crystalline lattice or superficially associated. It is intended that all of these forms should be within the scope of the present disclosure. Methods of solvation are generally known within the art. Suitable solvates are pharmaceutically acceptable solvates. In one embodiment, the solvate is a hydrate. [0315] In one embodiment, compounds of the present disclosure (those of Formula I) are useful for the treatment of inflammatory pain, neuropathic pain, rheumatoid arthritis, osteoarthritis, diabetic nephropathy, hypertension, diabetes, and/or metabolic syndrome. The compounds of Formula I may be useful in elevating epoxyeicosatrienoic acids (EETs) levels in a subject to prevent and treat inflammatory and/or pain conditions. [0316] Compounds of Formula I, their pharmaceutically acceptable salts, and/or solvates thereof can, therefore, be used in the prevention and/or treatment of a disease or condition discussed herein. Pharmaceutical compositions containing a therapeutically effective quantity of a compound of Formula I, its pharmaceutically acceptable salts, and/or solvates thereof, possibly together with pharmaceutically acceptable excipients, are additional aspects of the present disclosure. [0317] The therapeutically effective quantity of compounds of Formula I, their pharmaceutically acceptable salts and/or solvates that must be administered, and the dosage for treating a pathological state with said compounds, will depend on numerous factors, including age, the state of the patient, the severity of the disease, the route and frequency of administration, the modulator compound to be used, etc. [0318] Suitable pharmaceutically acceptable carriers may include solid fillers or diluents and sterile aqueous or organic solutions. The active ingredient may be present in such pharmaceutical compositions in the amounts sufficient to provide the desired dosage in the range as described above. Thus, for oral administration, the active ingredient may be combined with a suitable solid or liquid carrier or diluent to form capsules, tablets, powders, syrups, solutions, suspensions and the like. For parenteral administration, the active ingredient can be combined with sterile aqueous or organic media to form injectable solutions or suspensions. For example, solutions in sesame or peanut oil, aqueous propylene glycol and the like can be used, as well as aqueous solutions of water-soluble pharmaceutically-acceptable acid addition salts or salts with base of the compounds. Aqueous solutions with the active ingredient dissolved in pharmaceutically acceptable solvents such as polyhydroxylated castor oil may also be used for injectable solutions. The injectable solutions prepared in this manner can then be administered intravenously, intraperitoneally, subcutaneously, or intramuscularly, with intramuscular administration being generally preferred for humans. [0319] The following examples describe exemplary embodiments of the disclosure. Other embodiments within the scope of the claims herein will be apparent to one skilled in the art from consideration of the specification or practice of the disclosure as disclosed herein. It is intended that the specification, together with the examples, be considered to be exemplary only, with the scope and spirit of the disclosure being indicated by the claims which follow the examples. [0320] GENERAL SYNTHETIC PROCEDURES [0321] Compounds of present disclosure can be synthesized by following the procedures outlined in Schemes I-VIII. The suggested methodologies are not intended to be limiting. Variations of these synthetic methodologies or the methodologies reported in literature can be adopted to synthesize compounds within the scope of disclosure. [0322] [0323] Scheme I shows method of synthesis of compounds 7 of Formula I of the present disclosure. In the first step, substituted benzyl halide 1 (Z = Cl, Br) is reacted with trialkyl phosphate to yield substituted benzyl phosphonate 2. This reaction may be carried out by heating 1 with trialkyl phosphite at 120–150 ° C with or without solvent such as dimethylacetamide for 10-20 h. The substituted alkene 4 is synthesized by generating ylid from the intermediate 2 and reacting with substituted piperidone 3. The formation of ylid from 2 may be carried using a base such as sodium hydride or potassium hydride in presence of a crown ether in a solvent such as THF, dimethoxyethane, or diethyl ether. This reaction may be initiated at low temperature (0+5 o C), followed by warming of reaction mixture to approximately 20 ° C and stirring for additional 20-60 minutes. The reaction between ylid generated from 2 with 3 may be carried out in a solvent such as THF, dimethoxyethane, diethyl ether or toluene and initiating the reaction at low temperature (0+5 o C), followed by warming of reaction mixture to approximately 20-40 α C and stirring for 8-20 h. Deprotection of carbamate from 4 in presence of acid yields piperidine intermediate 5. This reaction may be carried out in a solvent such dichloromethane or dichloroethane and using acid such as trifluoroacetic acid by stirring reaction mixture at temperature of 0-25 ° C for 20-90 minutes. Reaction of 5 with substituted cyclopropane carbamate intermediate 6 gives the target compound 7. This reaction may be carried out using a solvent such as dimethyl sulfoxide or dimethylacetamide and a base such as triethylamine, diisopropylethylamine by heating the reaction components at 40-60 ° C over a period of 3-6 h. [0324] The substituted cyclopropane carbamate 6 may be synthesized from corresponding cyclopropylamine 8 by reacting with aryl chloroformate (R = Ph, or Ar) using solvent such as dichloromethane and a base such as diisopropylethylamine. The reaction may be initiated at low temperature (0+5 o C), followed by warming of reaction mixture to approximately 20-30 ° C and stirring for 20-40 minutes. The substituted benzyl halide 1 and the substituted piperidone 3 used in Scheme I may be purchased commercially or synthesized from easily accessible reagents. [0325] [0326] Scheme II shows a method of synthesis of compounds 11 of Formula I of the present disclosure. In the first step, compound 4 is synthesized using methodology described in Scheme I is hydrogenated to yield saturated compound 9. This reaction may be carried out using catalytic hydrogenation (using e.g.; Pd/C or Pt/C) in solvents such as methanol or ethanol in a Parr hydrogenation apparatus. Deprotection of the carbamate 9 in presence of an acid yields piperidine intermediate 10. This reaction may be carried out in a solvent such dichloromethane or dichloroethane and using acid such as trifluoroacetic acid by stirring reaction mixture at temperature of 0-25 ° C for 20-90 minutes. Reaction of 10 with substituted cyclopropane carbamate intermediate 6 gives the target compound 11. This reaction may be carried out using a solvent such as dimethyl sulfoxide or dimethylacetamide and a base such as triethylamine, diisopropylethylamine by heating the reaction components at 40-60 ° C over a period of 3-6 h. [0327] [0328] Scheme III shows method of synthesis of compounds 18 and 19 of the present disclosure. In the first step, compound 12 containing heteroaryl ring A (such as pyridine, pyrimidine, pyrazine) reacts with substituted phenol 13 to yield 14. This reaction may be carried out in a solvent such as dimethylacetamide, or dimethylformamide using a base such as potassium carbonate, sodium carbonate, or cesium carbonate and by heating the reaction mixture at 80-120 ° C for 3-6 h. The resulting substituted benzyl alcohol is converted to corresponding benzyl halide 15 by reaction with thionyl chloride. This reaction may be carried out using solvent such as dichloromethane and treating with thionyl chloride at 0-25 ° C for 1- 3 h. Substituted benzyl halide 15 is reacted with trialkyl phosphite to yield substituted benzyl phosphonate 16. This reaction may be carried out by heating 15 with trialkyl phosphite at 120– 150 ° C with or without solvent such as dimethylacetamide for 10-20 h. The substituted alkene 17 is synthesized by generating ylid from the intermediate 16 and reacting with substituted piperidone 3. The formation of ylid from 16 may be carried using a base such as sodium hydride or potassium hydride in presence of a crown ether in a solvent such as THF, dimethoxyethane, or diethyl ether. The reaction may be initiated at low temperature (0+5 o C), followed by warming of reaction mixture to approximately 20 ° C and stirring for additional 20-60 minutes. The reaction between ylid generated from 16 with 3 may be carried out in a solvent such as THF, dimethoxyethane, diethyl ether or toluene and initiating the reaction at low temperature (0+5 ° C), followed by warming of reaction mixture to approximately 20-40 o C and stirring for 8-20 h. Conversion of 17 to 18 may be carried out following steps of deprotection of carbamate in presence of acid and reaction with substituted cyclopropane carbamate intermediate 6 as described in Scheme I. Conversion of 17 to 19 may be accomplished using sequential steps involving hydrogenation, deprotection of carbamate and reaction with substituted cyclopropane carbamate intermediate 6 as described in Scheme II. [0329] [0330] Scheme IV shows method of synthesis of compounds 26 and 27 of the present disclosure. In the first step, compound 20 reacts with substituted phenol 21 to yield 22. This reaction may be carried out in a solvent such as dimethylacetamide or dimethylformamide using a base such as cesium carbonate by heating the reaction mixture at 80-120 ° C for 3-8 h. The resulting substituted benzyl alcohol is converted to corresponding benzyl halide 23 by reaction with thionyl chloride. This reaction may be carried out using solvent such as dichloromethane and treating with thionyl chloride at 0-25 ° C for 1-3 h. Substituted benzyl halide 23 is reacted with trialkyl phosphite to yield substituted benzyl phosphonate 24. This reaction may be carried out by heating 23 with trialkyl phosphite at 120–150 ° C with or without solvent such as dimethylacetamide for 10-20 h. The substituted alkene 25 is synthesized by generating ylid from the intermediate 24 and reacting with substituted piperidone 3. The formation of ylid from 24 may be carried using a base such as sodium hydride or potassium hydride in presence of a crown ether in a solvent such as THF, dimethoxyethane, or diethyl ether. The reaction may be initiated at low temperature (0+5 o C), followed by warming of reaction mixture to approximately 20 ° C and stirring for additional 20-60 minutes. The reaction between ylid generated from 24 with 3 may be carried out in a solvent such as THF, dimethoxyethane, diethyl ether or toluene and initiating the reaction at low temperature (0+5 ° C), followed by warming of reaction mixture to approximately 20-40 ° C and stirring for 8-20 h. Conversion of 25 to 26 may be carried out following steps of deprotection of carbamate in presence of acid and reaction with substituted cyclopropane carbamate intermediate 6 as described in Scheme I. Conversion of 25 to 27 may be accomplished using sequential steps involving hydrogenation, deprotection of carbamate and reaction with substituted cyclopropane carbamate intermediate 6 as described in Scheme II.

[0331] [0332] Scheme V shows method of synthesis of compounds 33 and 34 of the present disclosure. In the first step, substituted benzyl halide 28 is reacted with trialkyl phosphite to yield substituted benzyl phosphonate 29. This reaction may be carried out by heating 28 with trialkyl phosphite at 120–150 ° C with or without solvent such as dimethylacetamide for 10-20 h. The substituted alkene 30 is synthesized by generating ylid from the intermediate 29 and reacting with substituted piperidone 3. The formation of ylid from 29 may be carried using a base such as sodium hydride or potassium hydride in presence of a crown ether in a solvent such as THF, dimethoxyethane, or diethyl ether. The reaction may be initiated at low temperature (0+5 o C), followed by warming of reaction mixture to approximately 20 ° C and stirring for additional 20-60 minutes. The reaction between ylid generated from 29 with 3 may be carried out in a solvent such as THF, dimethoxyethane, diethyl ether or toluene and initiating the reaction at low temperature (0+5 o C), followed by warming of reaction mixture to approximately 20-40 ° C and stirring for 8-20 h. The intermediate 31 containing heterocyclyl ring A is synthesized from 30 by reacting with corresponding heterocycle such as pyrrolidine, morpholine, piperidine. This reaction may be carried out by treating 30 with ring A heterocycle using cesium carbonate and catalysts such as palladium acetate and 2,2′- bis(diphenylphosphino)-1,1′-binaphthy (BINAP). The reaction may be carried out using solvents such as 1,4-dioxane by treating the reaction mixture at 20-100 ° C for 5-20 h. Conversion of 31 to 33 may be carried out following steps of deprotection of carbamate in presence of acid and reaction with substituted cyclopropane carbamate intermediate 6 as described in Scheme I. [0333] Conversion of 31 to 32 may be accomplished using catalytic hydrogenation (Pd/C, H 2 ) as described in Scheme II. Similarly, conversion of 32 to 34 may be carried out following methodology involving deprotection of carbamate and reaction with substituted cyclopropane carbamate intermediate 6 as described in Scheme II. [0334] [0335] Scheme VI shows a method of synthesis of compounds 40 of the present disclosure. In the first step, compound 35 containing heteroaryl ring A (such as pyridine, pyrimidine, pyrazine) reacts with substituted phenol 36 to yield 37. This reaction may be carried out in a solvent such as dimethylacetamide or dimethylformamide using a base such as potassium carbonate, sodium carbonate, or cesium carbonate by heating the reaction mixture at 80-120 ° C for 3-8 h. Substituted phenol 37 is reacted with piperidine mesylate 38 to yield 39. This reaction may be carried out in a solvent such as dimethylacetamide, or dimethylformamide using a base such as potassium carbonate, sodium carbonate, cesium carbonate and heating the reaction mixture at 60-80 ° C for 6-8 h. Conversion of 39 to 40 may be carried out following steps of deprotection of carbamate in presence of acid and reaction with substituted cyclopropane carbamate intermediate 6 as described in Scheme I. [0336] [0337] Scheme VII shows method of synthesis of compounds of formula 45 and 46 of the present disclosure. In the first step, compound 41 containing heteroaryl boronate reacts with aryl halide 42 to yield 43. This reaction may be carried out by treating 41 and 42 in a solvent such as dimethylacetamide, dimethylformamide using 2N sodium carbonate solution and tetrakis(triphenylphosphine)palladium(0) at ambient temperature 20-25 ° C for 12-18 h. Conversion of 43 to 45 may be carried out following steps of deprotection of carbamate in presence of acid and reaction with substituted cyclopropane carbamate intermediate 6 as described in Scheme I. [0338] Conversion of 43 to 44 may be accomplished using catalytic hydrogenation (Pd/C, H 2 ) as described in Scheme II. Similarly, conversion of 44 to 46 may be carried out following methodology involving deprotection of carbamate and reaction with substituted cyclopropane carbamate intermediate 6 as described in Scheme II.

[0339] [0340] Scheme VIII shows method of synthesis of compounds of formula 54 of the present disclosure. In the first step, substituted benzyl halide 47 is reacted with trialkyl phosphite to yield substituted benzyl phosphonate 48. This reaction may be carried out by heating 47 with trialkyl phosphite at 120–150 °C for 10-20 h. The substituted alkene 49 is synthesized by generating ylid from the intermediate 48 and reacting with substituted piperidone 3. The formation of ylid from 48 may be carried using a base such as sodium hydride or potassium hydride in presence of a crown ether in a solvent such as THF, dimethoxyethane, or diethyl ether. The reaction may be initiated at low temperature (0+5 ° C), followed by warming of reaction mixture to approximately 20 ° C and stirring for additional 20-60 minutes. The reaction between ylid generated from 48 with 3 may be carried out in a solvent such as THF, dimethoxyethane, diethyl ether or toluene and initiating the reaction at low temperature (0+5 o C), followed by warming of reaction mixture to approximately 20-40 °C and stirring for 8-20 h. Conversion of 49 to 50 may be accomplished using catalytic hydrogenation (Pd/C, H 2 ). This reaction may be carried out using catalytic hydrogenation (using e.g.; Pd/C or Pt/C) in solvents such as methanol or ethanol in a Parr hydrogenation apparatus. In the next step, synthesis of 52 is accomplished by reacting 50 with substituted 1-fluoro-2-nitrobenzene 51 in a displacement reaction. This reaction may be carried out by treating 50 with 51 in solvent such as dimethylformamide and heating the reaction mixture in presence of cesium carbonate at 80- 120 ° C for 10-20 h. Synthesis of benzimidazole compound 53 may be accomplished by treating 52 with formic acid and sodium formate in presence of Pd/C at around 25 ° C and heating the mixture around 100-120 ° C for 12-20 h. Conversion of 53 to 54 may be carried out by treatment with substituted cyclopropane carbamate intermediate 6 as described in Scheme II. [0341] EXAMPLES [0342] An embodiment of the present disclosure provides for preparation of the novel compounds of Formula I using the procedures set forth in the following examples. Those skilled in the art will understand that known variations of the conditions and processes of the following preparative procedures can be used to prepare these compounds. Moreover, by utilizing the procedures described herein, one of ordinary skill in the art can prepare additional compounds of the present disclosure claimed herein. [0343] Example 1 - Synthesis of 4-(3-methoxy-benzylidene)-piperidine-1-carboxylic acid (2-phenyl-cyclopropyl)-amide: [0344] [0345] Step 1- To a mixture of (3-methoxy-phenyl)-methanol (20.0 g, 0.14 mol) and pyridine (5.8 mL, 0.72 mol) in benzene (120 mL) was added thionyl chloride (74 mL, 1.01 mol) dropwise while stirring reaction in an ice bath. After removal of ice both, the reaction mixture was allowed to stir at room temperature over a period of 2 h. The resulting reaction mixture was quenched with saturated sodium bicarbonate solution (100 mL), extracted with ethyl acetate (2X300 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of the volatiles was purified through silica gel (230-400) column (5% ethyl acetate in petroleum ether) to give product 2 as a yellow oil 20.01 g (88 %). 1 H NMR (300 MHz, CDCl3) δ (ppm): 3.84 (s, 3H), 4.58 (s, 2H), 6.86-6.90 (m, 1H), 6.95-7.00 (m, 2H), 7.26-7.32 (m, 1H). [0346] Step 2 - A solution of 2 (20 g, 0.12 mol) in triethyl phosphite (29.0 mL, 0.16 mol) was heated at 150 ° C over a period of 17 h. The reaction mixture was allowed to cool to room temperature and the crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (20% ethyl acetate in petroleum ether) to give product 3 as a colorless oil 27.0 g (81%). 1 H NMR (300 MHz, CDCl3) δ (ppm): 1.26 (t, J=7.2 Hz, 6H), 3.11 (s, 1H), 3.18 (s, 1H), 3.81 (s, 3H), 4.01-4.03 (m, 4H), 6.79-6.91 (m, 3H), 7.21-7.28 (m, 1H). [0347] Step 3 - To a solution of 3 (11.0 g, 43.0 mmol) in THF (44 mL) was added 15-crown ether (0.2 mL, 0.9 mmol). The reaction was cooled (ice bath) and NaH (580 mg, 24.2 mmol) added portion wise. The reaction mixture was allowed to stir at room temperature for 30 min and re-cooled using an ice bath. To the above reaction mixture a solution of 4-oxo-piperidine- 1-carboxylic acid tert-butyl ester 4 (8.5 g, 43.0 mmol) in THF (44 mL) was added at ice temperature and allowed to stir at room temperature over a period 16 h. The resulting reaction mixture was diluted with water (100 mL), extracted with ethyl acetate and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (25% ethyl acetate in petroleum ether) to give product 5 as a yellow color oil 7.0 g (54 %). 1 H NMR (300 MHz, CDCl3) δ (ppm): 1.49 (s, 9H), 2.32-2.36 (m, 2H), 2.44-2.50 (m, 2H), 3.42 (t, J=5.7 Hz, 2H), 3.52 (t, J=5.7 Hz, 2H), 3.84 (s, 3H), 6.35 (s, 1H), 6.75-6.81 (m, 3H), 7.25-7.25 (m, 1H). [0348] Step 4 - To a solution of 5 (1.0 g, 3.2 mmol) in dichloromethane (8.0 mL) was added trifluoroacetic acid (4.25 mL, 4.25 vol) at ice temperature and the reaction mixture stirred at room temperature over a period of 1h. The crude product obtained upon evaporation of volatiles was washed with diethyl ether to yield 6 as a white solid (600 mg, 89%). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 2.54-2.62 (m, 4H), 3.07-3.17 (m, 4H), 3.75 (s, 3H), 6.44 (s, 1H), 6.77- 6.84 (m, 3H), 7.27 (t, J=7.8 Hz, 1H), 8.79 (bs, 2H). [0349] Step 5 - To a suspension of trans-2-phenylcyclopropylamine 2A (3.5 g, 0.02 mol) in dichloromethane (35 mL) was added triethylamine (0.06 mol), phenyl chloroformate 1A (4.8 g, 0.03 mol) at ice bath temperature. Then ice bath was removed and reaction mixture allowed to stir at room temperature over a period of 30 min. The resulting reaction mixture was diluted with ethyl acetate (200 mL), washed with water (2X100 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230- 400) column (10% ethyl acetate in petroleum ether) to give product 7 as a white solid 2.6 g (50 %). mp: 113.6 –115.3°C. 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.14-1.26 (m, 2H), 2.03- 2.09 (m, 1H), 2.72-2.75 (m, 1H), 7.10-7.40 (m, 10H), 8.18 (bs, 1H). MS: 254 (M+H). [0350] Step 6: To a solution of amine 6 (300 mg, 0.94 mmol) in dimethyl sulfoxide (6 mL) was added diisopropylethylamine (0.5 mL, 2.82 mmol) and carbamate 7 (238 mg, 0.94 mmol) at 25 °C. The reaction mixture was allowed to stir at 55 °C over a period of 4 h. The resulting reaction mixture was diluted with ethyl acetate (250 mL), washed with water (4X75 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (50% ethyl acetate in petroleum ether) to give product 8 as a white solid 226 mg (65 %). mp: 104.7-106.4°C. 1 H NMR (300 MHz, CDCl3) δ (ppm): 1.15-1.27 (m, 2H), 2.02-2.09 (m, 1H), 2.40 (t, J=5.7 Hz, 2H), 2.54 (t, J=5.7 Hz, 2H), 2.87 (bs, 1H), 3.39 (t, J=5.7 Hz, 2H), 3.49 (t, J=5.7 Hz, 2H), 3.82 (s, 3H), 4.87 (s, 1H, -CONH- , exchangeable 1 H), 6.37 (s, 1H), 6.75-6.81 (m, 3H), 7.18-7.30 (m, 6H). 13 C NMR (75 MHz, CDCl3) δ (ppm): 16.44, 25.10, 29.14, 33.19, 35.72, 44.60, 45.60, 55.18, 111.88, 114.60, 121.36, 124.76, 125.95, 126.65, 128.28, 129.16, 137.90, 138.70, 140.88, 157.95, and 159.53. MS: 363 (M+H) [0351] Example 2 - Synthesis of 4-[3-(pyridin-2-yloxy)-benzylidene]-piperidine-1- carboxylic acid (2-phenyl-cyclopropyl)-amide

[0352] [0353] Step 1 - To a solution of 2-fluoro-pyridine (25.8 g, 0.27 mol) in DMF (300 mL) was added 3-hydroxyphenyl-methanol (30.0 g, 0.24 mol) and cesium carbonate (117.3 g, 0.36 mol) at room temperature. The reaction mixture was stirred at 100 ° C over a period of 5 h. Then the resulting mixture was allowed to reach room temperature, diluted with water (250 mL), extracted with ethyl acetate (3X500 mL) and the organic layer was dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230- 400) column (30% ethyl acetate in petroleum ether) to give product 2 as a pale yellow oil 34.5 g (71 %). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 4.49-4.51 (m, 2H), 5.24-5.30 (m, 1H), 6.98-7.15 (m, 5H), 7.33-7.40 (m, 1H), 7.82-7.89 (m, 1H), 8.14-8.16 (m, 1H). [0354] Step 2 - To a solution of 2 (34.5 g, 0.17 mol) in dichloromethane (345 mL) was added thionyl chloride (13.9 mL, 0.18 mol) dropwise while stirring reaction in an ice bath. After removal of ice-bath, the reaction mixture was allowed to stir at room temperature over a period of 1 h. Then volatiles were evaporated under reduced pressure and diluted with toluene (25 mL) and toluene was evaporated under reduced pressure. This azeotropic process was repeated 3 times to obtain product 3 as brown color oil (36.8 g, 98%). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 4.77 (s, 2H), 7.04-7.20 (m, 4H), 7.27-7.29 (m, 1H), 7.42 (t, J=8.4 Hz, 1H), 7.84-7.90 (m, 1H), 8.14-8.16 (m, 1H). [0355] Step 3 - A solution of 3 (36.7 g, 0.16 mol) in triethyl phosphite (41.6 mL, 0.26 mol) was heated at 150 ° C over a period of 6 h. The reaction mixture was allowed to reach room temperature and the crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (60% ethyl acetate in petroleum ether) to give product 4 as colorless oil 41.33 g. The product contained unused triethyl phosphate and was used in next step without additional purification. [0356] Step 4 - To a solution of [3-(pyridin-2-yloxy)-benzyl]-phosphonic acid diethyl ester 4 (30.0 g, 93.0 mmol) in THF (120 mL) was added 15-crown ether (0.41 g, 1.8 mmol). The reaction was cooled (ice bath) and NaH (3.35 g, 0.14 mol) was added portion wise. The reaction mixture was allowed to stir at room temperature for 30 min and again cooled to ice temperature. To the above reaction mixture, a solution of 4-oxo-piperidine-1-carboxylic acid tert-butyl ester 5 (18.6 g, 93.0 mmol) in THF (120 mL) was added at ice temperature and allowed to stir at room temperature over a period 16 h. The resulting reaction mixture was diluted with water (500 mL), extracted with ethyl acetate (3X500 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (3 % ethyl acetate in petroleum ether) to give product 6 as a yellow color oil 24.3 g (71 %). 1 H NMR (300 MHz, CDCl3) δ (ppm): 1.41 (s, 9H), 2.27 (t, J=5.4 Hz, 2H), 2.40 (t, J=5.4 Hz, 2H), 3.33 (bs, 2H), 3.40 (t, J=5.4 Hz, 2H), 6.37 (s, 1H), 6.95-7.15 (m, 4H), 7.37 (t, J=7.8 Hz, 2H), 7.83-7.88 (m, 1H), 8.14-8.16 (m, 1H). [0357] Step 5 - To a solution of 4-[3-(pyridin-2-yloxy)-benzylidene]-piperidine-1- carboxylic acid tert-butyl ester 6 (12.0 g, 33.0 mmol) in dichloromethane (120.0 mL) was added trifluoroacetic acid (51 mL) at ice temperature and the reaction mixture was stirred at room temperature over a period of 1h. The brown color oil 7 (13.7 g, 85%) obtained upon evaporation of volatiles was used to next step (13.7 g, 85%) without additional purification. 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 2.56-2.63 (m, 4H), 3.10-3.16 (m, 4H), 6.46 (s, 1H), 6.99-7.15 (m, 5H), 7.39 (t, J=7.5 Hz, 1H), 7.83-7.88 (m, 1H), 8.14-8.16 (m, 1H). [0358] Step 6 - To a solution of amine 7 (15.0 g, 26.0 mmol) in dimethyl sulfoxide (150 mL) was added diisopropylethylamine (13.6 mL, 78.0 mmol) and product of Step 5, Example 1 (6.7 g, 26.0 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 5 h. The resulting reaction mixture was diluted with ethyl acetate (1.2 L), washed with water (3X150 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (50% ethyl acetate in petroleum ether) to give product 8 as a pale yellow solid 9.1 g (81 %). mp: 52.3 - 54.1°C. 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.06-1.18 (m, 2H), 1.88 (bs, 1H), 2.26 (m, 2H), 2.28 (m, 2H), 2.69- 2.72 (m, 1H), 3.29-3.38 (m, 4H), 6.36 (s, 1H), 6.85-6.86 (bs, 1H, -CONH-, Exchangeable 1 H), 6.95-7.40 (m, 10H), 7.86 (t, J=6.3 Hz, 1H), 8.14-8.16 (m, 2H). 13 C NMR (75 MHz, DMSO- d6) δ (ppm): 15.99, 27.74, 29.40, 34.42, 36.08, 44.41, 45.47, 112.09, 119.31, 119.52, 121.60, 123.58, 125.24, 125.80, 126.41, 128.52, 129.88, 139.19, 140.09, 140.56, 142.42, 147.91, 154.42, 158.09 and 163.45. MS: 426 (M+H) [0359] Example 3 - Synthesis of 4-[3-(5-trifluoromethyl-pyridin-2-yloxy)-benzylidene]- piperidine-1-carboxylic acid (2-phenyl-cyclopropyl)-amide [0361] Step 1 - To a solution of 5-trifluoromethyl-2-chloro-pyridine (23.0 g, 0.12 mol) in DMF (230 mL) was added 3-hydroxyphenyl-methanol (17.4 g, 0.13 mol) and potassium carbonate (26.3 g, 0.19 mol) at room temperature. The reaction mixture was stirred at 100 °C over a period of 5 h. Then the resulting mixture was allowed to reach room temperature, diluted with water (200 mL), extracted with ethyl acetate (3X400 mL) and the organic layer was dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (12% ethyl acetate in petroleum ether) to give product 2 as a pale yellow oil 28.1 g (82 %). 1 H NMR (300 MHz, CDCl3) δ (ppm): 4.75 (s, 2H), 7.03- 7.10 (m, 2H), 7.19 (s, 1H), 7.26-7.28 (m, 1H), 7.44 (t, J=7.8 Hz, 1H), 7.90-7.94 (m, 1H), 8.45 (s, 1H). [0362] Step 2 - To a solution of 2 (28.0 g, 0.10 mol) in dichloromethane (280 mL) was added thionyl chloride (8.5 mL, 0.11 mol) drop wise while stirring reaction in an ice bath. After removal of ice-bath, the reaction mixture was allowed to stir at room temperature over a period of 1 h. Then volatiles were evaporated under reduced pressure, diluted with toluene (15 mL) and toluene was evaporated under reduced pressure. This azeotropic process was repeated 3 times to obtain product 3 as red color oil (29.6 g, 99%). 1 H NMR (300 MHz, CDCl 3 ) δ (ppm): 4.62 (s, 2H), 7.05 (d, J=8.7 Hz, 1H), 7.12-7.13 (m, 1H), 7.23-7.32 (m, 2H), 7.42-7.47 (m, 1H), 7.92-7.95 (m, 1H), 8.46 (s, 1H). [0363] Step 3 - A solution of 3 (29.0 g, 0.10 mol) in triethyl phosphite (26.2 mL, 0.15 mol) was heated at 150 ° C over a period of 6 h. The reaction mixture was allowed to reach room temperature and the mixture was added to n-heptane (150 mL) to obtain light orange color precipitate. The precipitate obtained was filtered and dried under vacuum to give product 4 as white solid (30.8 g, 94%) and it was used in next step without further purification. 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.16 (t, J=6.9 Hz, 6H), 3.24 and 3.31(2s, 2H), 3.90-4.00 (m, 4H), 7.07-7.10 (m, 2H), 7.17-7.25 (m, 2H), 7.39 (d, J=8.7 Hz, 1H), 8.23-8.26 (m, 1H), 8.55 (s, 1H). [0364] Step 4 - To a solution of ester 4 (25.0 g, 64.0 mmol) in THF (100 mL) was added 15-crown ether (0.28 g, 1.3 mmol). The reaction was cooled (ice bath) and NaH (2.3 g, 96.0 mmol) was added portion wise over a period of 5 min. The reaction mixture was allowed to stir at room temperature for 30 min and again cooled to ice temperature. To the above reaction mixture, a solution of 4-oxo-piperidine-1-carboxylic acid tert-butyl ester 5 (12.81 g, 64.0 mmol) in THF (100 mL) was added at ice temperature and allowed to stir at room temperature over a period 16 h. The resulting reaction mixture was diluted with water to obtain white precipitate. The precipitate was filtered and dried to give product 6 as a white solid (24.4 g, 87%). 1 H NMR (300 MHz, CD3OD) δ (ppm): 1.48 (s, 9H), 2.36 (t, J=5.1 Hz, 2H), 2.49 (t, J=5.4 Hz, 2H), 3.43 (t, J=5.7 Hz, 2H), 3.52 (t, J=5.7 Hz, 2H), 6.43 (s, 1H), 7.01-7.03 (m, 2H), 7.14 (d, J=8.4 Hz, 2H), 7.38-7.44 (m, 1H), 8.09-8.12 (m, 1H), 8.44 (bs, 1H). [0365] Step 5 - To a solution of 6 (10.0 g, 23.0 mmol) in dichloromethane (100 mL) was added trifluoroacetic acid (42.5 mL) at ice temperature and the reaction mixture was stirred at room temperature over a period of 1h. Then volatiles were removed under reduced pressure to obtain product as red color oil (10.7 g, 83%). The crude product 7 was used for next step without further purification. 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 2.60-2.64 (m, 4H), 3.11- 3.17 (m, 4H), 6.47 (s, 1H), 7.08-7.26 (m, 4H), 7.43 (t, J=8.1 Hz, 1H), 8.21-8.23 (m, 1H), 8.56 (s, 1H), 8.76 (bs, 1H). [0366] Step 6 - To a solution of amine 7 (10.5 g, 18.7 mmol) in dimethyl sulfoxide (10 mL) was added diisopropylethylamine (9.8 mL, 56.1 mmol) and product of Step 5, Example 1 (4.74 g, 18.7 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 5 h. The resulting reaction mixture was diluted with ethyl acetate (1.0 L), washed with water (3X150 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (40% ethyl acetate in petroleum ether) to give product 8 as a white solid 7.0 g (76 %). mp: 98.9–101.5 °C 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.08-1.18 (m, 2H), 1.85-1.90 (m, 1H), 2.77 (bs, 2H), 2.39 (bs, 2H), 2.69- 2.72 (m, 1H), 3.32-3.39 (m, 4H), 6.37 (s, 1H), 6.85 (s, 1H, D2O exchangeable 1 H), 7.04-7.24 (m, 9H), 7.41 (t, J=7.8 Hz, 1H), 8.22-8.25 (m, 1H), 8.58 (s, 1H). 13 C NMR (75 MHz, DMSO- d6) δ (ppm): 15.99, 24.74, 29.39, 34.44, 36.07, 44.37, 45.44, 112.25, 118.97, 119.81, 120.23, 120.66, 121.09, 121.53, 122.08, 122.57, 123.42, 125.79, 126.15, 126.38, 128.52, 129.76, 130.08, 137.98, 138.01, 139.42, 140.34, 142.42, 145.72, 145.77, 153.38, 158.08, 166.01. MS: 494 (M+H). [0367] Example 4 - Synthesis of 4-[3-(pyridin-2-yloxy)-benzyl]-piperidine-1-carboxylic acid (2-phenyl-cyclopropyl)-amide: [0369] Step 1 - To a solution of product of Step 4, Example 2 (6) in methanol (17 mL) was added 10% Pd/C (900 mg) at room temperature and the reaction mixture was stirred under hydrogen balloon pressure over a period of 1 h. The resulting reaction mixture was filtered through celite bed and the filtrate was concentrated under reduced pressure to give product 7 as a yellow oil (850 mg, 66%). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 0.98-1.04 (m, 2H), 1.37 (s, 9H), 1.43-1.56 (m, 3H), 2.60 (m, 4H), 3.88-3.92 (m, 2H), 6.91-7.02 (m, 2H), 7.10-7.17 (m, 2H), 7.25-7.31 (m, 2H), 7.84-7.89 (m, 1H), 8.14-8.16 (m, 1H). [0370] Step 2 - To a solution of ester 7 (800 mg, 2.17 mmol) in dichloromethane (8 mL) was added trifluoroacetic acid (3.4 mL, 4.25 vol) at ice temperature and the reaction mixture was stirred at room temperature over a period of 1h. Then volatiles were removed under reduced pressure to obtain product as brown color oil (800 mg, 74%). The crude product 8 was used in next step without further purification. [0371] Step 3 - To a solution of crude amine 8 (500 mg, 1.0 mmol) in dimethyl sulfoxide (10 mL) was added diisopropylethylamine (0.5 mL, 2.82 mmol) and product of Step 5, Example 1 (256 mg, 1.0 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 5 h. The resulting reaction mixture was diluted with ethyl acetate (250 mL), washed with water (4X75 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (60% ethyl acetate in petroleum ether) to give product 9 as a white solid 230 mg (53 %). mp: 126.8 - 128.6 °C. 1 H NMR (300 MHz, CDCl3) δ (ppm): 1.12-1.27 (m, 4H), 1.68-1.71 (m, 3H), 1.89-2.06 (m, 1H), 2.56 (d, J=6.6 Hz, 2H), 2.73 (t, J=11.4 Hz, 2H), 2.82-2.86 (m, 1H), 3.92 (d, J=13.5 Hz, 2H), 4.84 (s, 1H, -CONH-, Exchangeable 1 H), 6.90-7.03 (m, 5H), 7.15-7.35 (m, 6H), 7.68-7.74 (m, 1H), 8.21-8.23 (m, 2H). 13 C NMR (75 MHz, CDCl3) δ (ppm): 16.45, 25.06, 29.65, 31.82, 33.18, 37.88, 42.86, 44.24, 111.61, 118.48, 118.61, 121.66, 125.36, 125.88, 126.66, 128.24, 129.40, 139.37, 141.02, 141.99, 147.80, 154.28, 158.21 and 163.73. MS: 428 (M+H) [0372] Example 5 - Chiral separation of racemic product of Example 2 [0373] The chiral column (CHIRALPACK IA 250 mm x10 mm 5μm) was equilibrated using mobile phase (n-hexane: isopropyl alcohol; 80:20 v/v) for 15 column volumes prior to the compound elution. Then 500 µL of the stock solution, prepared by dissolving 500 mg of the product of Example 2 in 5 mL of n-hexane and isopropyl alcohol (8:2), was injected and fractions collected based on the separation seen in the chromatogram. The fraction F1 was the first eluted fraction (retention time: 11.5 min to 13.00 min) and F2 the second eluted fraction (retention time: 13.50 min to 15.50 min) from chiral column. The injections were repeated to complete separation of the remaining (4.5 mL) stock solution. Then solvents of F-1 and F-2 were removed separately under reduced pressure to yield the chiral products 5A (entA) (140 mg) and 5B (entB) (150 mg), respectively. [0374] 5A 4-[3-(pyridin-2-yloxy)-benzylidene]-piperidine-1-carboxylic acid {(1S,2R)-2- phenyl-cyclopropyl)}-amide - HPLC: 99.98% (Chiral purity: 98.55%). mp: 45.0 - 47.1 °C. 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.06-1.10 (m, 1H), 1.15-1.18 (m, 1H), 1.87 (m, 1H), 2.24-2.28 (m, 2H), 2.38-2.40 (m, 2H), 2.69-2.72 (m, 1H), 3.29-3.40 (m, 4H), 6.36 (s, 1H), 6.85- 6.86 (m, 1H), 6.95-7.27 (m, 10H), 7.37 (t, J=7.8 Hz, 1H), 7.83-7.85 (m, 1H), 8.14-8.16 (m, 1H). MS: 426 (M+H). [0375] 5B 4-[3-(pyridin-2-yloxy)-benzylidene]-piperidine-1-carboxylic acid {(1R,2S)-2- phenyl-cyclopropyl)}-amide - HPLC: 99.89% (Chiral purity: 98.93%). mp: 51.0 - 54.3 °C. 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.06-1.08 (m, 1H), 1.15-1.18 (m, 1H), 1.87 (m, 1H), 2.24-2.26 (m, 2H), 2.38-2.40 (m, 2H), 2.69-2.72 (m, 1H), 3.29-3.40 (m, 4H), 6.36 (s, 1H), 6.84- 6.85 (m, 1H), 6.95-7.27 (m, 10H), 7.37 (t, J=7.8 Hz, 1H), 7.83-7.85 (m, 1H), 8.14-8.16 (m, 1H MS: 426 (M+H). [0376] [0377] Example 6 - Synthesis of 4-[3-(pyrimidin-2-yloxy)-benzylidene]-piperidine-1- carboxylic acid (2-phenyl-cyclopropyl)-amide [0378] [0379] Step 1 - To a solution of 3-hydroxyphenyl-methanol (500 mg, 4.0 mmol) in DMF (5 mL) was added cesium carbonate (2.6 g, 8.0 mmol) and 2-chloropyrimidine (680 mg, 6.0 mmol) at room temperature. The reaction mixture was stirred at 100 ° C over a period of 5 h. Then the resulting reaction mixture was allowed to cool to room temperature, filtered to remove cesium carbonate, the filtrate was diluted with water (50 mL), extracted with ethyl acetate (100 mL) and the organic layer was dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (50% ethyl acetate in petroleum ether) to give product 1 as a pale yellow oil 440 mg (54 %). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 4.52 (s, 2H), 5.28 (bs, 1H), 7.03-7.99 (m, 5H), 8.63 (d, J = 1.2 Hz, 2H). [0380] Step 2 - To a solution of 1 (440 mg, 2.1 mmol) in dichloromethane (8 mL) was added thionyl chloride (0.19 mL, 2.6 mmol) dropwise while stirring reaction in an ice bath. After removal of ice-bath, the reaction mixture was allowed to stir at room temperature over a period of 1 h. The resulting reaction mixture was quenched with ice cold water (10 mL) and extracted with ethyl acetate (100 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (25% ethyl acetate in petroleum ether) to give product 2 as a pale pink solid 400 mg (83 %). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 4.78 (s, 2H), 7.17-7.19 (m, 1H), 7.28-7.34 (m, 3H), 7.42-7.45 (m, 1H), 8.65 (d, J = 4.5 Hz, 2H). [0381] Step 3 - A solution of 2-(3-chloromethyl-phenoxy)-pyrimidine 2 (400 mg, 1.8 mmol) in triethyl phosphite (0.45 mL, 2.7 mmol) was heated at 150 ° C over a period of 6 h. The reaction mixture was allowed to reach room temperature and the crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (50% ethyl acetate in petroleum ether) to give product 3 as white solid 400 mg (69%). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.16 (t, J = 6.9 Hz, 6H), 3.23 (s, 1H), 3.30 (s, 1H), 3.90-4.00 (m, 4H), 7.06-7.09 (m, 2H), 7.16-7.18 (m, 1H), 7.25-7.28 (m, 1H), 7.37 (t, J = 7.5 Hz, 1H), 8.64 (d, J = 4.8 Hz, 2H). [0382] Step 4 - To a solution of ester 3 (400 mg, 1.2 mmol) in THF (2.5 mL) was added 15-crown ether (5 µL, 0.02 mmol). The reaction was cooled (ice bath) and NaH (44 mg, 1.8 mmol) added portion wise. The reaction mixture was allowed to stir at room temperature for 30 min and again cooled to ice temperature. To the above reaction mixture, a solution of 4- oxo-piperidine-1-carboxylic acid tert-butyl ester (250 mg, 1.2 mmol) in THF (2.5 mL) was added at ice temperature and allowed to stir at room temperature over a period 16 h. The resulting reaction mixture was diluted with water (10 mL), extracted with ethyl acetate (100 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (25 % ethyl acetate in petroleum ether) to give product 4 as a white solid 360 mg (80 %). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.41 (s, 9H), 2.28 (t, J = 5.4 Hz, 2H), 2.41 (t, J = 5.7 Hz, 2H), 3.33-3.41 (m, 4H), 6.38 (s, 1H), 7.03- 7.13 (m, 3H), 7.26 (t, J = 4.8 Hz, 1H), 7.39 (t, J = 7.8 Hz, 1H), 8.65 (d, J = 4.8 Hz, 2H). [0383] Step 5 - To a solution of 4 (360 mg, 0.97 mmol) in dichloromethane (4.0 mL) was added trifluoroacetic acid (1.7 mL) at ice temperature and the reaction mixture was stirred at room temperature over a period of 1h. The crude product obtained upon evaporation of the solvent was washed with diethyl ether to yield product 5 as an off-white solid 0.31 g (83 %) and was taken to next without additional purification. [0384] Step 6 - To a solution of amine 5 (310 mg, 0.6 mmol) in dimethyl sulfoxide (4.0 mL) was added diisopropylethylamine (0.5 mL, 3.1 mmol) and product of Step 5, Example 1 (0.58 g, 2.4 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 5 h. The resulting reaction mixture was diluted with ethyl acetate (100 mL), washed with water (3X50 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (60% ethyl acetate in petroleum ether) to give product 6 as a white solid 210 mg (77 %). mp: 52.7 – 57.6 °C. 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.04-1.06 (m, 1H), 1.14-1.20 (m, 1H), 1.85-1.88 (m, 1H), 2.27 (bs, 2H), 2.39 (bs, 2H), 2.70-2.71 (m, 1H), 3.32-3.39 (m, 4H), 6.37 (s, 1H), 6.85 (bs, 1H, CONH exchangeable 1 H), 7.03-7.15 (m, 6H), 7.22-7.28 (m, 3H), 7.39 (t, J = 7.8 Hz, 1H), 8.65 (d, J = 7.8 Hz, 2H). 13 C NMR (75 MHz, DMSO-d6) δ (ppm): 16.03, 24.76, 29.37, 34.50, 36.10, 44.37, 45.41, 117.37, 119.95, 122.23, 123.49, 125.80, 126.00, 126.34, 128.54, 129.91, 139.19, 140.21, 142.44, 153.20, 158.05, 160.47 and 165.21. MS: 427 (M+H). [0385] Example 7 - Synthesis of 4-{3-[1-(2-phenyl-cyclopropylcarbamoyl)-piperidin-4- ylidenemethyl]-phenoxy}-benzoic acid methyl ester: [0387] Step 1 - To a solution of 3-hydroxyphenyl-methanol 2 (5.0 g, 40.2 mmol) in DMF (50 mL) was added cesium carbonate (26.2 g, 80.5 mmol) and ester 1 (7.5 g, 48.3 mmol) at room temperature. The reaction mixture was stirred at 100 ° C over a period of 5 h. Then the resulting reaction mixture was allowed to reach room temperature and filtered to remove cesium carbonate. The filtrate was diluted with water (200 mL), extracted with ethyl acetate (2X250 mL) and the organic layer dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (30% ethyl acetate in petroleum ether) to give product 3 as a pale yellow oil 3.2 g (31 %). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 3.81 (s, 3H), 4.51 (d, J = 5.7 Hz, 2H), 5.28 (t, J = 5.7 Hz, 1H), 6.98- 7.06 (m, 4H), 7.17-7.19 (m, 1H), 7.38-7.40 (m, 1H), 7.95-7.98 (m, 2H). [0388] Step 2 - To a solution of alcohol 3 (3.2 g, 12.3 mmol) in dichloromethane (50 mL) was added thionyl chloride (1.7 mL, 14.8 mmol) dropwise while stirring reaction in an ice bath. After removal of ice-bath, the reaction mixture was allowed to stir at room temperature over a period of 1 h. The resulting reaction mixture was quenched with ice cold water (50 mL) and extracted with ethyl acetate (250 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (5% ethyl acetate in petroleum ether) to give product 4 as colorless oil 2.3 g (67 %). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 3.83 (s, 3H), 4.78 (s, 2H), 7.06-7.12 (m, 3H), 7.21 (bs, 1H), 7.30- 7.33 (m, 1H), 7.44-7.49 (m, 1H), 7.98 (d, J = 8.7 Hz, 2H). [0389] Step 3 - A solution of compound 4 (2.3 g, 7.9 mmol) in triethyl phosphite (2.3 mL, 11.9 mmol) was heated at 150 ° C over a period of 6 h. The reaction mixture was allowed to reach room temperature and the crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (50% ethyl acetate in petroleum ether) to give product 5 as pale yellow oil 3.5 g (91%). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.15 (t, J = 6.9 Hz, 6H), 3.23 (s, 1H), 3.30 (s, 1H), 3.83 (s, 3H), 3.89-3.99 (m, 4H), 7.00-7.06 (m, 4H), 7.15-7.17 (m, 1H), 7.39 (t, J = 7.8 Hz, 1H), 7.97 (d, J = 8.7 Hz, 2H). [0390] Step 4 - To a solution of 5 (3.5 g, 9.2 mmol) in THF (20 mL) was added 15-crown ether (40 µL, 0.18 mmol). The reaction was cooled (ice bath) and NaH (560 mg, 13.8 mmol) was added portion wise. The reaction mixture was allowed to stir at room temperature for 30 min and again cooled to ice temperature. To the above reaction mixture, a solution of 4-oxo- piperidine-1-carboxylic acid tert-butyl ester 6 (1.9 g, 9.2 mmol) in THF (15 mL) was added at ice temperature and allowed to stir at room temperature over a period 16 h. The resulting reaction mixture was quenched with saturated ammonium chloride (50 mL), extracted with ethyl acetate (500 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (10 % ethyl acetate in petroleum ether) to give product 7 as a white solid 1.8 g (46 %). 1 H NMR (300 MHz, DMSO- d6) δ (ppm): 1.40 (s, 9H), 2.27-2.40 (m, 4H), 3.40-3.60 (m, 4H), 3.83 (s, 3H), 6.37 (s, 1H), 6.94-6.99 (m, 2H), 7.05-7.12 (m, 3H), 7.39-7.41 (m, 1H), 7.98 (d, J = 8.7 Hz, 2H). [0391] Step 5 - To a solution of tert-butyl ester 7 (1.8 g, 4.2 mmol) in dichloromethane (18.0 mL) was added trifluoroacetic acid (9.0 mL) at ice temperature and the reaction mixture was stirred at room temperature over a period of 1h. The crude product obtained upon evaporation of the solvent was washed with n-hexane to obtain product 8 as thick black liquid 1.5 g (83 %). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 2.63-2.72 (m, 4H), 3.16-3.28 (m, 4H), 3.91 (s, 3H), 6.49 (s, 1H), 6.86 (s, 1H), 6.95-7.02 (m, 4H), 7.36 (t, J = 8.1 Hz, 1H), 8.03 (t, J = 8.4 Hz, 2H). [0392] Step 6 -To a solution of amine 8 (1.5 g, 3.4 mmol) in dimethyl sulfoxide (15.0 mL) was added diisopropylethylamine (2.0 mL, 10.2 mmol) and the product of Step 5, Example 1 (0.86 g, 3.4 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 5 h. The resulting reaction mixture was diluted with ethyl acetate (250 mL), washed with water (2X100 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (25% ethyl acetate in petroleum ether) to give product 9 as a white solid 0.9 g (54 %). mp: 48.5 - 53.2 °C. 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.10-1.20 (m, 2H), 1.88 (bs, 1H), 2.26 (bs, 2H), 2.37 (bs, 2H), 2.70-2.71 (m, 1H), 3.31-3.38 (m, 4H), 3.83 (s, 3H), 6.36 (s, 1H), 6.84 (bs, 1H, CONH exchangeable 1 H), 6.94-7.26 (m, 10H), 7.42 (t, J = 7.8 Hz, 1H), 7.97 (d, J = 8.4 Hz, 2H). 13 C NMR (75 MHz, DMSO-d6) δ (ppm): 16.02, 24.76, 29.40, 34.49, 36.07, 44.32, 45.38, 52.44, 117.81, 118.27, 120.49, 123.40, 124.51, 125.61, 125.79, 126.33, 128.53, 130.59, 132.02, 139.85, 140.45, 142.43, 155.40, 158.04, 161.73 and 166.11. MS: 483 (M+H). [0393] Example 8 - Synthesis of 4-{3-[1-(2-phenyl-cyclopropylcarbamoyl)-piperidin-4- ylidenemethyl]-phenoxy}-benzoic acid: [0395] To a solution of product of Step 6, Example 7 (0.5 g, 1.03 mmol) in methanol (3.0 mL) was added a solution of sodium hydroxide (120 mg, 3.1 mmol) in water (2.0 mL) at 25 °C. The reaction mixture was allowed to stir at 55 °C over a period of 16 h. The crude product obtained upon evaporation of the solvent was diluted with water (20.0 mL) and the aqueous layer washed with ethyl acetate (2X20 mL). Then aqueous layer was acidified (pH=2, 1.0 N HCl), saturated with solid NaCl and the product was extracted with ethyl acetate (2X150 mL). The combined organic layers were dried over sodium sulphate and concentrated to yield the product as a white solid 350 mg (72%). mp: 99.4 - 102.5 °C. 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.15-1.29 (m, 2H), 1.88 (bs, 1H), 2.26 (bs, 2H), 2.37 (bs, 2H), 2.71 (bs, 1H), 3.32- 3.38 (m, 4H), 6.36 (s, 1H), 6.84 (bs, 1H, CONH exchangeable 1 H), 6.93-7.44 (m, 11H), 7.95 (d, J = 8.4 Hz, 2H), 12.83 (bs, 1H). 13 C NMR (75 MHz, DMSO-d6) δ (ppm): 16.02, 24.75, 29.40, 34.51, 36.07, 44.31, 45.37, 117.76, 118.17, 120.38, 123.43, 125.47, 125.79, 126.32, 128.54, 130.57, 132.13, 139.81, 140.41, 144.44, 155.59, 158.03, 163.36 and 167.22. MS: 467 (M-H). [0396] Example 9 - Synthesis of 4-(3-pyrrolidin-1-yl-benzyl)-piperidine-1-carboxylic acid-(2-phenyl-cyclopropyl)-amide: [0397] [0398] Step 1 - A solution of 3-bromobenzyl bromide (6.0 g, 24.0 mmol) in triethyl phosphite (6.2 mL, 36.0 mmol) was heated at 130 ° C over a period of 16 h. The reaction mixture was cooled and allowed to reach room temperature and the crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (30 % ethyl acetate in petroleum ether) to give product 2 as colorless oil 6.5 g (89%). 1 H NMR (300 MHz, DMSO- d6) δ (ppm): 1.17 (t, J= 7.2 Hz, 6H), 3.24 (s, 1H), 3.31 (s, 1H), 3.91-4.01 (m, 4H), 7.28-7.29 (m, 2H), 7.43-7.50 (m, 2H). MS: 307.0 (M+) and 309.0 (M+2). [0399] Step 2 - To a solution of 2 (3.0 g, 9.7 mmol) in THF (20 mL) was added 15-crown ether (0.04 mL, 0.19 mmol). The reaction was cooled (ice bath) and NaH (0.58 g, 14.6 mmol) added portion wise. The reaction mixture was allowed to stir at room temperature for 30 min and again cooled to ice temperature. To the above reaction mixture, a solution of 4-oxo- piperidine-1-carboxylic acid tert-butyl ester (1.95 g, 9.7 mmol) in THF (10 mL) was added at ice temperature and allowed to stir at room temperature over a period of 16 h. The resulting reaction mixture was diluted with water (100 mL), extracted with ethyl acetate (3X100 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (5 % ethyl acetate in petroleum ether) to give product 3 as a yellow color oil 1.8 g (53 %). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.41 (s, 9H), 2.28 (bs, 2H), 2.38 (bs, 2H), 3.34-3.41 (m, 4H), 6.36 (s, 1H), 7.25-7.30 (m, 2H), 7.41- 7.43 (m, 2H). MS: 252.0 (M-BOC). [0400] Step 3 - To a solution of compound 3 (2.0 g, 7.0 mmol) in 1,4-dioxane (20.0 mL) were added pyrrolidine (0.9 mL, 10.6 mmol), cesium carbonate (7.0 g, 21.2 mmol), racemic BINAP (0.9 g, 1.4 mmol) and palladium acetate (0.95 g, 1.4 mmol) under argon atmosphere at room temperature. The reaction mixture was stirred at room temperature for 30 min, followed by stirring under reflux for a period of 16 hours. The resulting reaction mass was filtered through a celite pad, washed with ethyl acetate (250 mL). The ethyl acetate layer was washed with water (2X100 mL), dried over sodium sulphate and concentrated. The crude product obtained was purified by silica gel column chromatography (15% ethyl acetate in petroleum ether) to obtain product 4 as a pale yellow oil 0.6 g (32%). 1 H NMR (300 MHz, CDCl3) δ (ppm): 1.49 (s, 9H), 2.01 (bs, 4H), 2.33-2.35 (m, 2H), 2.50-2.53 (m, 2H), 3.27-3.29 (m, 4H), 3.39-3.49 (m, 2H), 3.50-3.54 (m, 2H), 6.36-6.53 (m, 4H), 7.19 (t, J= 8.1 Hz, 1H). MS: 343.7 (M+H). [0401] Step 4 - To a solution of compound 4 (0.6 g, 1.7 mmol) in tetrahydrofuran (10.0 mL) was added 10% Pd/C (240 mg). The reaction mass was stirred at room temperature under hydrogen gas pressure (1 kg/cm 2 ) over a period of two hours. After releasing hydrogen pressure, reaction mixture was filtered through celite pad, washed with tetrahydrofuran and filtrate concentrated to give product 5 as a pale yellow liquid (0.6 g). The crude product was taken to next step without further purification. 1 H NMR (300 MHz, DMSO-d 6 ) δ (ppm): 1.10- 1.21 (m, 2H), 1.49 (s, 9H), 1.64-1.72 (m, 3H), 1.98-2.03 (m, 4H), 2.49 (d, J= 6.9 Hz, 2H), 2.65 (t, J= 12.3 Hz, 2H). 3.27-3.31 (m, 2H), 4.07-4.15 (m, 2H), 6.35 (s, 1H), 6.42-6.47 (m, 2H), 7.14 (t, J= 7.8 Hz, 1H). MS: 345.7 (M+H). [0402] Step 5 - To a solution of compound 5 (0.6 g, 1.7 mmol) in dichloromethane (6.0 mL) was added trifluoroacetic acid (3 mL) at ice temperature and the reaction mixture was stirred at room temperature for a period of 1 h. The brown colored oil 6 (0.6 g) obtained upon evaporation of volatiles was used in next step without additional purification. [0403] Step 6 - To a solution of amine 6 (600 mg, 1.9 mmol) in dimethyl sulfoxide (6.0 mL) was added N,N-diisopropylethylamine (1.1 mL, 5.8 mmol) and product of Step 5, Example 1 (0.5 g, 1.9 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 5 h. The resulting reaction mixture was diluted with ethyl acetate (500 mL), washed with water (3X100 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (40% ethyl acetate in petroleum ether) and by preparative HPLC (phenomenex 250x221.20 mm, 10 µM, 0.1% TFA in water and acetonitrile mobile phase) to give product 7 as a pale yellow solid 160 mg (23 %). mp: 145.6 °C – 151.5 °C. IR: 3334, 2842, 1620, 1600, 1545, 1252 and 752 cm -1 . 1 H NMR: (300 MHz, DMSO-d6) δ (ppm): 0.95-1.19 (m, 3H), 1.13-1.17 (m, 1H), 1.50-1.55 (m, 2H), 1.63-1.66 (m, 1H), 1.81-1.87 (m, 1H), 1.93 (bs, 4H), 2.40-2.42 (m, 2H), 2.56-2.60 (m, 2H), 2.67-2.68 (m, 1H), 3.19 (bs, 4H), 3.90 (d, J= 12.9 Hz, 2H), 6.32-6.40 (m, 3H), 6.71 (bs, 1H), 7.02-7.15 (m, 4H), 7.21-7.26 (m, 2H). MS: 404.5 (M+H). [0404] Example 10 - Synthesis of 4-(3-morpholin-4-yl-benzyl)-piperidine-1-carboxylic acid (2-phenyl-cyclopropyl)-amide: [0406] Step 1 - To a solution of product of Step 2, Example 9 (1.5 g, 4.2 mmol) in 1,4- dioxane (15.0 mL) were added morpholine (0.45 mL, 5.1 mmol), cesium carbonate (4.1 g, 12.6 mmol), racemic BINAP (0.52 g, 0.84 mmol) and palladium acetate (0.56 g, 0.84 mmol) under argon atmosphere at room temperature. The reaction mixture was stirred at room temperature for 30 min, followed by stirring under reflux for 16 hours. The resulting reaction mass was cooled and filtered through celite pad and washed with ethyl acetate (250 mL). The ethyl acetate layer was washed with water (2X100 mL), organic layer dried over sodium sulphate and concentrated. The crude product obtained was purified by silica gel column chromatography (15% ethyl acetate in petroleum ether) to obtain the crude product 2 as a pale yellow oil 0.35 g. [0407] Step 2 - To a solution of compound 2 (0.85 g, 2.37 mmol) in tetrahydrofuran (10.0 mL) was added 10% Pd/C (350 mg). The reaction mass was stirred at room temperature under hydrogen gas pressure (1 kg/cm 2 ) for a period of two hours. After releasing hydrogen pressure, reaction mixture was filtered through celite pad, filtrate was concentrated to get the product 3 as a pale yellow liquid (0.8 g). The product was taken to next step without further purification. [0408] Step 3 - To a solution of crude compound 3 (0.8 g, 2.2 mmol) in dichloromethane (8.0 mL) was added trifluoroacetic acid (4 mL) at ice temperature and the reaction mixture was stirred at room temperature for a period of 1h. The brown color oil 4 (0.8 g,) obtained upon evaporation of volatiles was taken to next step without additional purification. [0409] To a solution of amine 4 (800 mg, 2.3 mmol) in dimethyl sulfoxide (8.0 mL) was added diisopropylethylamine (1.3 mL, 7.0 mmol) and the product of Step 5, Example 1 (0.6 g, 2.3 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C for a period of 5 h. The resulting reaction mixture was diluted with ethyl acetate (500 mL), washed with water (3X100 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (50% ethyl acetate in petroleum ether) to give product 5 as a pale yellow solid 420 mg. mp: 182.3 °C - 186.0 °C. IR: 3330, 2841, 1620, 1600, 1545, 1247 and 756 cm -1 . 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 0.95-1.08 (m, 3H), 1.11- 1.18 (m, 1H), 1.49-1.53 (m, 2H), 1.61-1.67 (m, 1H), 1.85-1.88 (m, 1H), 2.43-2.61 (m, 4H), 2.67-2.68 (m, 1H), 3.08 (t, J= 4.5 Hz, 4H), 3.74 (t, J= 4.5 Hz, 4H), 3.90 (d, J= 12.6 Hz, 2H), 6.61 (d, J= 7.2 Hz, 1H), 6.70-6.76 (m, 3H, one 1 H is D 2 O exchangeable), 7.07-7.15 (m, 4H), 7.22-7.27 (m, 2H). 13 C NMR (75 MHz, DMSO-d6) δ (ppm): 16.01, 24.75, 32.08, 34.56, 37.96, 43.17, 43.98, 49.00, 66.61, 113.09, 116.43, 120.52, 125.77, 126.29, 128.53, 129.12, 141.31, 142.51, 151.50, 158.28. MS: 420.2 (M+H). [0410] Example 11 - Synthesis of 4-[3-(pyridin-2-yloxy)-phenoxy]-piperidine-1- carboxylic acid (2-phenyl-cyclopropyl)-amide: [0411] [0412] Step 1 - To a solution of 1,3-dihydroxybenzene (1.0 g, 9.0 mmol) in DMF (10.0 mL) was added Cs2CO3 (5.92 g, 18.0 mmol) and 2-fluoropyridine (0.8 mL, 9.0 mmol). The reaction mixture was heated to 100 °C for a period of 16 h. Then the resulting mixture was allowed to reach room temperature, diluted with water (250 mL), extracted with ethyl acetate (3X500 mL) and the organic layer dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (10% ethyl acetate in petroleum ether) to give product 3 as a pale yellow oil 400 mg (23 %). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 6.20 (s, 1H), 6.48-6.53 (m, 1H), 6.61 (d, J= 8.1 Hz, 1H), 6.98 (d, J= 8.1 Hz, 1H), 7.11-7.21 (m, 2H), 7.84 (t, J= 8.1 Hz, 1H), 8.18 (bs, 1H), 9.61 (s, 1H, D2O exchangeable 1 H). MS: 187.9 (M+H). [0413] Step 2 - To a solution of compound 3 (400 mg, 2.14 mmol) in DMF (8.0 mL) was added Cs2CO3 (1.4 g, 4.2 mmol) at room temperature. The reaction mixture was stirred for 5 min, then compound 4 (600 mg, 2.14 mmol) in DMF (2.0 mL) was added to the reaction mixture at room temperature and the reaction mixture was stirred at 65 °C for a period of 8 h. The reaction mixture was diluted with ethyl acetate (300 mL), washed with water (3X50 mL) and ethyl acetate layer dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (50% ethyl acetate in petroleum ether) to give product 5 as a pale yellow oil 540 mg (67 %). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.40 (s, 9H), 1.48-1.51 (m, 2H), 1.89-1.92 (m, 2H), 3.14-3.16 (m, 2H), 3.63-3.68 (m, 2H), 4.56-4.58 (m, 1H), 6.65-6.68 (m, 1H), 6.75 (s, 1H), 6.80-6.84 (m, 1H), 6.99- 7.02 (m, 1H), 7.14 (d, J= 5.1 Hz, 1H), 7.29 (t, J= 7.8 Hz, 1H), 7.85 (t, J= 7.5 Hz, 1H), 8.17 (d, J= 5.1 Hz, 1H). MS: 371.4 (M+H). [0414] Step 3 - To a solution of compound 5 (0.4 g, 0.8 mmol) in dichloromethane (8.0 mL) was added trifluoroacetic acid (2.0 mL) at ice temperature and the reaction mixture was stirred at room temperature over a period of 1h. The brown color oil 6 obtained upon evaporation of volatiles was used in next step without additional purification. [0415] To a solution of amine 6 (390 mg, 0.78 mmol) in dimethyl sulfoxide (5.0 mL) were added diisopropylethylamine (0.67 mL, 3.9 mmol) and the product of Step 5, Example 1 (198 mg, 0.78 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C for a period of 5 h. The resulting reaction mixture was diluted with ethyl acetate (250 mL), washed with water (3X10 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (70% ethyl acetate in petroleum ether) to give product as an off-white low melting solid. The product obtained was further purified by preparative HPLC to obtain product 7 as an off-white low melting solid 200 mg. mp: 43.9 °C – 46.8 °C. IR: 3313, 1621, 1586, 1423 and 1235 cm -1 . 1 H NMR (300 MHz, DMSO- d6) δ (ppm): 1.07-1.09 (m, 1H), 1.14-1.18 (m, 1H), 1.46-1.49 (m, 2H), 1.86 (bs, 3H), 2.68-2.69 (m, 1H), 3.08 (t, J= 9.6 Hz, 2H), 3.64-3.68 (m, 2H), 4.53 (bs, 1H), 6.64-6.67 (m, 1H), 6.74 (bs, 1H), 6.80 (bs, 1H), 6.83 (bs, 1H, D2O exchangeable 1 H), 7.00 (d, J= 8.0 Hz, 1H), 7.08-7.14 (m, 4H), 7.21-7.31 (m, 3H), 8.82 (t, J= 7.2 Hz, 2H), 8.16 (d, J= 5.1 Hz, 1H). MS: 430.4 (M+H). [0416] Example 12 - Synthesis of 4-[3-(1H-pyrazol-4-yl)-benzyl]-piperidine-1-carboxylic acid (-2-phenyl-cyclopropyl)-amide: [0417] [0418] Step 1 - To a cooled (0-5 °C) solution of compound 1 (2.0 g, 10.3 mmol, Sigma Aldrich) in dimethylformamide (20.0 mL) was added 4-dimethylaminopyridine (0.25 g, 2.0 mmol) and di-tert-butyl dicarbonate (3.0 mL, 15.4 mmol). The resulting reaction mass was stirred at room temperature for a period of 12 hours. The reaction was quenched with water (50.0 mL) and extracted with ethyl acetate (200 mL). The ethyl acetate layer was dried over anhydrous sodium sulphate and concentrated under reduced pressure. The crude product obtained was purified by silica gel column chromatography (15% ethyl acetate in petroleum ether) to obtain product 2 as an off-white solid 1.25 g (40%). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.27 (s, 12H), 1.58 (s, 9H), 7.87 (s, 1H), 8.37 (s, 1H). MS: 195.3 (M-BOC+H). [0419] Step 2 - To a solution compound product of Step 2, Example 9 (1.0 g, 2.8 mmol) in DMF (10 mL) was added compound 2 (1.9 g, 5.6 mmol) and 2N sodium carbonate solution (4.3 mL, 8.5 mmol) at room temperature. Reaction mixture was stirred under argon atmosphere over a period of 10 minutes. Then tetrakis(triphenylphosphine)palladium(0) (0.33 g, 0.28 mmol) was added to the reaction mixture under argon. The resulting reaction mixture was stirred at room temperature for a period of 16 hours. Reaction mixture was quenched with water (100 mL) and extracted with ethyl acetate (500 mL), washed with water (2X250 mL) and brine (100 mL). Ethyl acetate layer was dried over sodium sulphate and concentrated. The crude product obtained upon evaporation of the solvent under reduced pressure was purified by silica gel column chromatography (40% ethyl acetate in petroleum ether) to obtain the product 3 as an off-white solid 550 mg (57%). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.42 (s, 9H), 2.28- 2.31 (m, 2H), 2.42-2.44 (m, 2H), 3.34-3.44 (m, 4H), 6.39 (s, 1H), 7.03 (d, J= 7.5 Hz, 1H), 7.31 (t, J= 7.5 Hz, 1H), 7.44-7.47 (m, 2H), 7.92 (s, 1H), 8.19 (s, 1H), 12.94 (s, 1H). MS: 338.1 (M- H). [0420] Step 3 - To a solution of compound 3 (0.5 g, 1.4 mmol) in 20% methanol in chloroform (10.0 mL) was added 10% Pd/C (200 mg). The reaction mass was stirred at room temperature under hydrogen gas pressure (1 kg/cm 2 ) over a period of 24 hours. Reaction mass was filtered through a celite pad, the filtrate was concentrated to get the product 4 as a pale yellow liquid (0.5 g). The crude product was taken for next step without further purification. [0421] Step 4 - To a solution of compound 4 (0.5 g, 1.6 mmol) in dichloromethane (5.0 mL) was added trifluoroacetic acid (2.5 mL) at ice temperature and the reaction mixture was stirred at room temperature over a period of 1h. The brown color oil 5 (0.5 g,) obtained upon evaporation of volatiles was used to next step without additional purification. [0422] Step 5 - To a solution of amine 5 (500 mg, 1.0 mmol) in dimethyl sulfoxide (5.0 mL) was added N,N-diisopropylethylamine (0.8 mL, 4.2 mmol) and the product of Step 5, Example 1 (270 mg, 1.0 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 5 h. The resulting reaction mixture was diluted with ethyl acetate (500 mL), washed with water (3X100 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (3% methanol in chloroform) to give product 6 as an off-white solid 210 mg. mp: 157.4 °C – 163.4 °C. IR: 3330, 1619, 1545, 1475 and 753 cm -1 . 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.01-1.17 (m, 4H), 1.51-1.55 (m, 2H), 1.82 (bs, 1H), 1.83-1.85 (m, 1H), 2.54-2.68 (m, 5H), 3.90 (d, J= 12.6 Hz, 2H), 6.71 (d, J= 2.7 Hz, 1H, D2O exchangeable 1 H), 6.98 (d, J= 7.5 Hz, 1H), 7.07-7.15 (m, 3H), 7.21-7.27 (m, 3H), 7.41-7.43 (m, 2H), 7.90 (s, 1H), 8.17 (s, 1H), 12.91 (s, 1H, D 2 O exchangeable 1 H). 13 C NMR (75 MHz, DMSO-d6) δ (ppm): 16.00, 24.74, 32.06, 34.53, 37.88, 42.81, 43.98, 121.69, 123.17, 125.77, 126.31, 127.05, 128.52, 128.99, 133.22, 141.06, 142.50, 158.22. MS: 401.3 (M+H). [0423] Example 13 - Synthesis of 4-[3-(1-methyl-1H-pyrazol-4-yl)-benzyl]-piperidine-1- carboxylic acid (2-phenyl-cyclopropyl)-amide: [0424] [0425] Step 1 - To a cooled (0-5 °C) solution of product of Step 2, Example 12 (0.7 g, 2.0 mmol) in THF (25 mL) was added sodium hydride (0.25 g, 6.1 mmol) and methyl iodide (0.4 mL, 6.1 mmol). The resulting reaction mass was allowed to stir at room temperature over a period of 1 hour. Reaction mixture was quenched with water (100 mL) and extracted with ethyl acetate (500 mL). Organic layer was washed with water (2X250 mL) and brine (100 mL). Ethyl acetate layer was dried over anhydrous sodium sulphate and concentrated. The crude product obtained was purified by silica gel column chromatography (20% ethyl acetate in petroleum ether) to get the product 1 as an off-white solid 600 mg (83%). 1 H NMR (300 MHz, DMSO- d6) δ (ppm): 1.45 (s, 9H), 2.30-2.31 (m, 2H), 2.42-2.44 (m, 2H), 3.35-3.44 (m, 4H), 3.89 (s, 3H), 6.39 (s, 1H), 7.02-7.05 (m, 1H), 7.30 (t, J= 7.5 Hz, 1H), 7.39-7.43 (m, 2H), 7.85 (s, 1H), 8.14 (s, 1H). MS: 298.0 (M-t-butyl+H). [0426] Step 2 - To a solution of compound 1 (0.6 g, 1.7 mmol) in 20% methanol in chloroform (10.0 mL) was added 10% Pd/C (240 mg). The reaction mass was stirred at room temperature under hydrogen gas pressure (1 kg/cm 2 ) over a period of 16 hours. Reaction mass was filtered through celite pad, filtrate was concentrated to get the product 2 as a pale yellow liquid (0.6 g, 98%). The product obtained was taken to next step without further purification. 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.02-1.09 (m, 2H), 1.38 (s, 9H), 1.54-1.58 (m, 2H), 1.68-1.70 (m, 1H), 2.50-2.52 (m, 1H), 2.62-2.73 (m, 2H), 3.85-3.93 (m, 2H), 3.89 (s, 3H), 6.98 (d, J= 7.2 Hz, 1H), 7.25 (t, J= 7.5 Hz, 1H), 7.36 (s, 2H), 7.83 (s, 1H), 8.11 (s, 1H). MS: 300.2 (M - t-butyl + H). [0427] Step 3 - To a solution of compound 2 (0.6 g, 1.6 mmol) in dichloromethane (6.0 mL) was added trifluoroacetic acid (3.0 mL) at ice temperature and the reaction mixture was stirred at room temperature over a period of 1h. The brown color oil 3 (0.6 g,) obtained upon evaporation of volatiles was used in next step without additional purification. [0428] Step 4 - To a solution of amine 3 (600 mg, 1.6 mmol) in dimethyl sulfoxide (6.0 mL) was added diisopropylethylamine (1.2 mL, 6.4 mmol) and the product of Step 5, Example 1 (370 mg, 1.4 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C for a period of 5 h. The resulting reaction mixture was diluted with ethyl acetate (500 mL), washed with water (3X100 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (3% methanol in chloroform) to give product 4 as an off-white solid 350 mg (53 %). mp: 158.4 °C – 160.3 °C. IR: 3353, 1619, 1544, 1473 and 752 cm -1 . 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.01-1.15 (m, 4H), 1.51-1.55 (m, 2H), 1.82-1.83 (m, 1H), 1.84-1.86 (m, 1H), 2.57-2.68 (m, 4H), 3.34- 3.37 (m, 1H), 3.85 (s, 3H), 3.90 (d, J= 13.2 Hz, 2H), 6.71 (d, J= 2.7 Hz, 1H, D2O exchangeable 1 H), 6.98 (d, J= 7.5 Hz, 1H), 7.07-7.15 (m, 3H), 7.21-7.27 (m, 3H), 7.36-7.38 (m, 2H), 7.83 (s, 1H), 8.11 (s, 1H). 13 C NMR (75 MHz, DMSO-d6) δ (ppm): 16.00, 24.74, 32.05, 34.53, 37.89, 42.79, 43.99, 122.44, 122.97, 125.77, 126.10, 126.31, 127.15, 128.19, 128.52, 129.05, 132.91, 136.43, 141.11, 142.50, 158.21. MS: 415.0 (M+H). [0429] Example 14 - Synthesis of 4-(3-benzoimidazol-1-yl-benzyl)-piperidine-1- carboxylic acid (2-phenyl-cyclopropyl)-amide:

[0430] [0431] Step 1 - A solution of compound 1 (5.0 g, 23.14 mmol) in triethyl phosphite (5.9 mL, 37.7 mmol) was heated at 130 o C over a period of 16 h. The reaction mixture was cooled to room temperature and the crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (50 % ethyl acetate in petroleum ether) to give product 2 as a pale yellow oil 4.9 g (89%). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.09-1.21 (m, 6H), 3.44 (s, 1H), 3.51 (s, 1H), 3.93-4.07 (m, 4H), 7.60-7.66 (m, 1H), 7.73-7.76 (m, 1H), 8.11-8.14 (m, 1H), 8.19 (s, 1H). MS: 274.1 (M+1). [0432] Step 2 - To a solution of compound 2 (4.9 g, 20.3 mmol) in THF (50 mL) was added 15-crown ether (0.08 mL, 0.04 mmol). The reaction was cooled (ice bath) and NaH (1.22 g, 30.5 mmol) added portion wise. The reaction mixture was allowed to stir at room temperature for 30 min and again cooled to ice temperature. To the above reaction mixture, a solution of 4- oxo-piperidine-1-carboxylic acid tert-butyl ester (4.05 g, 20.3 mmol) in THF (10 mL) was added at ice temperature and allowed to stir at room temperature over a period 4 h. The resulting reaction mixture was diluted with water (100 mL), extracted with ethyl acetate (3X250 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (5 % ethyl acetate in petroleum ether) to give product 3 as a yellow color oil 4.0 g (62 %). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.42 (s, 9H), 2.33 (t, J= 5.7 Hz, 2H), 2.41 (t, J= 5.7 Hz, 2H), 3.38-3.41 (m, 2H), 3.44 (t, J= 5.7 Hz, 2H), 6.50 (s, 1H), 7.61-7.71 (m, 2H), 8.02 (s, 1H), 8.07-8.10 (m, 1H). MS: 219.2 (M-BOC+H), 263.1 (M-t-Butyl+1). [0433] Step 3 - To a solution of compound 3 (4.0 g, 12.5 mmol) in 90% methanol in dichloromethane (40.0 mL) was added 10% Pd/C (3.0 g). The reaction mass was stirred at room temperature under hydrogen gas pressure (1 kg/cm 2 ) over a period of 20 hours. Reaction mass was filtered through celite pad, filtrate was concentrated to get the product 4 as a pale yellow liquid (1.5 g, 41%). The product was taken to next step without further purification. 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 0.81-1.00 (m, 2H), 1.38 (s, 9H), 1.77-1.80 (m, 2H), 2.28-2.34 (m, 2H), 2.60-2.72 (m, 1H), 3.90 (t, J= 12.6 Hz, 2H) 4.92 (s, 2H), 6.28-6.38 (m, 3H), 6.90 (t, J= 12.6 Hz, 1H). MS: 191.1 (M-BOC+1). [0434] Step 4 - To a solution of compound 4 (1.5 g, 3.4 mmol) in DMF (8.0 mL) was added Cs 2 CO 3 (8.9 g, 25.7 mmol) at room temperature. The reaction mixture was stirred for 5 min and 1-fluoro-2-nitrobenzene (0.8 g, 3.7 mmol) in DMF (2.0 mL) was added to the reaction mixture at room temperature and the mixture stirred at 100 °C for a period of 16 h. The reaction mixture was diluted with ethyl acetate (500 mL), washed with water (3 X 100 mL). The ethyl acetate layer was dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (7 % ethyl acetate in petroleum ether) to give product 5 as a reddish yellow oil 500 mg (35 %). 1 H NMR (300 MHz, DMSO- d6) δ (ppm): 0.97-1.08 (m, 2H), 1.38 (s, 9H), 1.54-1.58 (m, 2H), 1.65-1.68 (m, 1H), 2.40-2.52 (m, 2H), 2.64-2.73 (m, 2H), 3.91 (d, J = 12.9 Hz, 2H), 6.87 (t, J = 7.5 Hz, 1H), 7.01 (d, J = 7.8 Hz, 1H), 7.13-7.20 (m, 3H), 7.30-7.35 (m, 1H), 7.50 (t, J = 6.6 Hz, 1H), 8.11 (d, J = 7.5 Hz, 1H), 9.35 (s, 1H). MS: 410.0 (M-H). [0435] Step 5 - To a solution of compound 5 (500 mg, 1.2 mmol) in formic acid (10 mL) were added sodium formate (290 mg, 4.3 mmol), and Pd/C (10 mol %, 120 mg, 0.01 mmol) at room 25 °C. Then the reaction mixture was allowed to stir at 110 °C for a period of 18 h. The reaction mixture was allowed to cool to room temperature and then filtered through celite with the aid of 20 mL of formic acid. The crude product obtained upon evaporation of the volatiles was dissolved in 5% methanol in dichloromethane (50 mL) and filtered to remove inorganic salts. The filtrate was concentrated to obtain product 6 as an off-white solid (350 mg). The crude product was taken to next without purification. [0436] Step 6 - To a solution of amine 6 (350 mg, 1.2 mmol) in dimethyl sulfoxide (5.0 mL) were added diisopropylethylamine (1.03 mL, 6.0 mmol) and the product of Step 5, Example 1 (186 mg, 1.2 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C for a period of 6 h. The resulting reaction mixture was diluted with ethyl acetate (300 mL), washed with water (3X50 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (1.5% methanol in dichloromethane) to give product 7 as an off-white solid 170 mg (31 %). mp: 72.5 °C – 76.4° C. IR: 3347, 2931, 16161, 1542, and 742 cm -1 . 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.00- 1.14 (m, 4H), 1.54-1.57 (m, 2H), 1.71-1.79 (m, 1H), 1.80-1.84 (m, 1H), 2.48-2.65 (m, 5H), 3.92 (d, J= 12.9 Hz, 2H), 6.72 (d, J= 2.7 Hz, 1H, D 2 O exchangeable 1 H), 7.07-7.15 (m, 3H), 7.21-7.26 (m, 3H), 7.30-7.35 (m, 3H), 7.50-7.54 (m, 3H), 7.60-7.62 (m, 1H), 7.77-7.79 (m, 1H), 8.55 (s, 1H). MS: 451.0 (M+H). [0437] Example 15 - Synthesis of 4-(3-pyrrolidin-1-yl-benzylidene)-piperidine-1- carboxylic acid (2-phenyl-cyclopropyl)-amide: [0439] Step 1 - To a solution of product of Step 2, Example 9 (2.0 g, 7.0 mmol) in 1,4- dioxane (20.0 mL) were added pyrrolidine (0.9 mL, 10.6 mmol), cesium carbonate (7.0 g, 21.2 mmol), racemic BINAP (0.9 g, 1.4 mmol) and palladium acetate (0.95 g, 1.4 mmol) under argon atmosphere at room temperature. The reaction mixture was stirred at room temperature for 30 min, followed by stirring under reflux for a period of 16 hours. The resulting reaction mass was filtered through a celite pad, washed with ethyl acetate (250 mL). The ethyl acetate layer was washed with water (2X100 mL), dried over sodium sulphate and concentrated. The crude product obtained was purified by silica gel column chromatography (15% ethyl acetate in petroleum ether) to obtain the product 2 as a pale yellow oil 0.6 g (32%). 1 H NMR (300 MHz, CDCl 3 ) δ (ppm): 1.49 (s, 9H), 2.01 (bs, 4H), 2.33-2.35 (m, 2H), 2.50-2.53 (m, 2H), 3.27- 3.29 (m, 4H), 3.39-3.49 (m, 2H), 3.50-3.54 (m, 2H), 6.36-6.53 (m, 4H), 7.19 (t, J= 8.1 Hz, 1H). MS: 343.7 (M+1). [0440] Step 2 - To a solution of compound 2 (0.6 g, 1.7 mmol) in dichloromethane (6.0 mL) was added trifluoroacetic acid (3 mL) at ice temperature and the reaction mixture was stirred at room temperature over a period of 1h. The brown color oil 3 (0.6 g) obtained upon evaporation of volatiles was used in next step without additional purification. [0441] Step 3 - To a solution of amine 3 (600 mg, 1.9 mmol) in dimethyl sulfoxide (6.0 mL) was added diisopropylethylamine (1.1 mL, 5.8 mmol) and the product of Step 5, Example 1 (0.5 g, 1.9 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 5 h. The resulting reaction mixture was diluted with ethyl acetate (500 mL), washed with water (3X100 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (40% ethyl acetate in petroleum ether) to give product 4 as an off-white solid 300 mg (42 %). mp: 145.7 °C – 147.4 °C . IR: 3292, 2963, 1626, 1533, 1263 and 746 cm -1 . 1 H NMR (300 MHz, CDCl3) δ (ppm): 1.22-1.26 (m, 2H), 2.02 (bs, 5H), 2.38-2.41 (m, 2H), 2.56-2.59 (m, 2H), 2.88 (bs, 1H), 3.30 (bs, 4H), 3.36-3.40 (m, 2H), 3.47-3.51 (m, 2H), 4.87 (bs, 1H, D 2 O exchangeable 1 H), 6.38-6.52 (m, 3H), 7.19-7.46 (m, 7H). 13 C NMR (75 MHz, CDCl 3 ) δ (ppm): 16.01, 24.75, 32.08, 34.56, 37.96, 43.17, 43.98, 49.00, 66.61, 113.09, 116.43, 120.52, 125.77, 126.29, 128.53, 129.12, 141.31, 142.51, 151.50, 158.28. MS: 402.7 (M+H). [0442] Example 16 - Synthesis of phenyl N-[(1R,2S)-2-phenylcyclopropyl] carbamate:

[0443] [0444] A general methodology described in literature (WO 2013/057322) was used to synthesize the chiral intermediate above. [0445] Step 1 - To a suspension of trans-2-phenyl-cyclopropylamine hydrochloride (100 g, 0.59 mol) in water (500 mL) was added saturated sodium bicarbonate solution at 0-5 °C over a period of 20 min and basified to pH > 7. The reaction mixture was stirred at 25-30 °C over a period of 2h. The reaction mixture was extracted with dichloromethane (3X700 mL), separated organic phase was dried over sodium sulphate and concentrated to yield 2-phenyl- cyclopropylamine as an off-white solid 2 (71.2 g, 92 %). [0446] Step 2 - To a solution of trans-2-phenyl-cyclopropylamine (70 g, 0.52 mol) in ethanol (700 mL) was added L (+) tartaric acid (79 g, 0.52 mol) at 0- 5 °C and stirred at 25-30 °C for 1h. After reaction completion, the solid was filtered and dried to yield 2-phenyl- cyclopropylamine as tartrate salt (133 g). Isopropanol: water (3:1) (1.3 L) was added to the above salt (130 g) and stirred at 70 °C over a period of 2 h. The reaction mixture was allowed to cool to room temperature over a period of 1h. The solid separated was collected by filtration to yield (1R,2S)-N-{[(2R,3R)-3-carboxy-2,3-dihydroxypropanoyl]oxy}-2- phenylcyclopropan-1-aminium (3) as a white solid (60 g, 90%). [0447] Step 3 - To a solution of (1R,2S)-N-{[(2R,3R)-3-carboxy-2,3- dihydroxypropanoyl]oxy}-2-phenylcyclopropan-1-aminium (3) (60 g, 0.19 mol) in water (200 mL) was added 1.0 M sodium hydroxide (194 mL, 0.19 mol) at 0-5 °C over a period of 20 min and stirred for 1 h. The aqueous phase was extracted with ethyl acetate (2X700 mL). The combined extracts were washed with water (2X400mL), brine (400 mL), dried over sodium sulphate and concentrated under reduced pressure to yield (1R,2S)-2-phenyl-cyclopropylamine as pale yellow solid 4 (25 g, 87%). [0448] Step 4 - To a suspension of amine 4 (15.0 g, 88.0 mmol) in dichloromethane (150 mL) was added triethylamine (36.0 mL, 0.26 mol), phenyl chloroformate (20.7 g, 0.13 mol) at ice bath temperature. Then ice bath was removed and reaction mixture was allowed to stir at room temperature over a period of 1 h. The resulting reaction mixture was diluted with ethyl acetate (1.0 L), washed with water (2X200 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (10% ethyl acetate in petroleum ether) to give product 5 as a white solid 16.0 g (71 %). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 1.15-1.25 (m, 2H), 2.04-2.08 (m, 1H), 2.72-2.75 (m, 1H), 7.10-7.40 (m, 10H), 8.17 (bs, 1H). MS (M+H) 254.3. [0449] Step 5 - To a stirring solution of (1R,2S)-2-phenyl-cyclopropylamine 4 (25.0 g, 0.19 mol) in diethyl ether (150 mL) was added 2.0 M HCl in ether (140 mL, 0.28 mol) at 0-5 °C. The reaction mixture was allowed to stir at 20-25 °C over a period of 30 min. The reaction mixture was concentrated under reduced pressure. The resulted reaction mass was washed with diethyl ether (2X100 mL) to yield product 6, hydrochloric acid salt of (1R,2S)-2-phenyl- cyclopropylamine as an off-white solid 30.0 g (95%). mp: 179.2 - 180.1 °C; IR: 3643, 3054, 1979, 1501, 1160, 799, 743, 696 cm -1 . 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 1.14-1.19 (m, 1H), 1.43 – 1.48 (m, 1H), 2.38 - 2.43(m, 1H), 2.72-2.76 (m, 1H), 7.09 - 7.24 (m, 3H), 7.22 - 7.33 (m, 2H), 8.81 (bs, 3H). MS (M+H) 134.3. Chiral HPLC purity: 100 %. The chirality of 6 was further confirmed by matching analytical and spectral data with authentic sample, (1R,2S)- 2-phenylcyclopropylamine hydrochloride, purchased from Sigma-Aldrich. [0450] Example 17 – Synthesis of 4-[3-(5-trifluoromethyl-pyridin-2-yloxy)-benzylidene]- piperidine-1-carboxylic acid [(1S,2R)-2-phenyl-cyclopropyl)]-amide:

[0451] [0452] Step 1 - To a suspension of (1S,2R)-2-phenylcyclopropan-1-amine 2 (500 mg, 2.95 mmol) in dichloromethane (5.0 mL) was added triethylamine (1.21 mL, 8.85 mol), phenyl chloroformate 1 (0.41 mL, 3.3 mol) at ice bath temperature. Then ice bath was removed and reaction mixture allowed to stir at room temperature over a period of 1 h. The resulting reaction mixture was diluted with ethyl acetate (1.0 L), washed with water (2X200 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (10 % ethyl acetate in petroleum ether) to give product 3 as a white solid 500 mg (71 %). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 1.19-1.26 (m, 2H), 2.05-2.08 (m, 1H), 2.72-2.75 (m, 1H), 7.11-7.38 (m, 10H), 8.18 (bs, 1H). MS (M+H) 254.5. [0453] Step 2 - To a solution of product of Step 5, Example 3 (1.0 g, 2.2 mmol) in dimethyl sulfoxide (10 mL) was added diisopropylethylamine (1.2 mL, 6.6 mmol) and 3 (556 mg, 2.2 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 4 h. The resulting reaction mixture was diluted with ethyl acetate (300 mL), washed with water (3X100 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (40% ethyl acetate in petroleum ether) to give product 4 as a white solid 770 mg (70 %). mp: 100.2 °C -101.0 °C. IR: 3329, 1622, 1531, 1487, 1329, 1076 cm -1 . 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 1.08-1.18 (m, 2H), 1.85-1.90 (m, 1H), 2.27 (t, J=5.6 Hz, 2H), 2.40 (t, J=5.2 Hz, 2H), 2.69-2.72 (m, 1H), 3.32 (t, J=6.0 Hz, 2H), 3.38 (t, J=6.0 Hz, 1H), 6.37 (s, 1H), 6.85 (d, J=3.2 Hz, 1H), 7.04-7.41 (m, 10H), 8.22-8.25 (m, 1H), 8.58 (bs, 1H). MS: 494.3 (M+H). HPLC purity: 99.78%. Chiral HPLC purity: 100%. [0454] Example 18 – Synthesis of 4-[3-(5-trifluoromethyl-pyridin-2-yloxy)-benzylidene]- piperidine-1-carboxylic acid [(1R,2S)-2-phenyl-cyclopropyl)]-amide: [0455] [0456] Step 1 – To a suspension of (1R,2S)-2-phenyl-cyclopropylamine 2 (500 mg, 2.95 mmol) in dichloromethane (5.0 mL) was added triethylamine (1.21 mL, 8.85 mol), phenyl chloroformate 1 (0.41 mL, 3.3 mol) at ice bath temperature. Then ice bath was removed and reaction mixture was allowed to stir at room temperature over a period of 1 h. The resulting reaction mixture was diluted with ethyl acetate (250 mL), washed with water (2X100 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (10 % ethyl acetate in petroleum ether) to give product 3 as a white solid 495 mg (70 %). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 1.15-1.25 (m, 2H), 2.04-2.08 (m, 1H), 2.72-2.75 (m, 1H), 7.10-7.40 (m, 10H), 8.17 (bs, 1H). MS (M+H) 254.3. [0457] Step 2 - To a solution of product of Step 5, Example 3 (1.0 g, 2.2 mmol) in dimethyl sulfoxide (10 mL) was added diisopropylethylamine (1.2 mL, 6.6 mmol) and 3 (556 mg, 2.2 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 4 h. The resulting reaction mixture was diluted with ethyl acetate (300 mL), washed with water (3X100 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (40% ethyl acetate in petroleum ether) to give product 4 as a white solid 715 mg (65 %). mp: 101.8 °C -103.2 °C. IR: 3329, 1623, 1531, 1388, 1329, 1076, 697 cm -1 . 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 1.07-1.18 (m, 2H), 1.85-1.90 (m, 1H), 2.25 – 2.40 (m, 4H), 2.67 – 2.75 (m, 2H), 2.69-2.72 (m, 1H), 3.31 (t, J=6.0 Hz, 2H), 3.38 (t, J=5.6 Hz, 1H), 6.36 (s, 1H), 6.85 (s, 1H), 7.04-7.15 (m, 6H), 7.22 -7.41 (m, 4H), 8.22- 8.24 (m, 1H), 8.58 (bs, 1H). MS: 494.3 (M+H). HPLC purity: 99.96%. Chiral HPLC purity: 100%. [0458] Example 19 - Synthesis of 4-({3-[(5-methylpyridin-2-yl)oxy]phenyl} methylidene)-N-[2-phenylcyclopropyl]piperidine-1-carboxamide : [0460] Step 1 - To a solution of 2-fluoro-5-methyl-pyridine 1 (14.76 g, 0.13 mol) in DMF (150 mL) was added 3-hydroxyphenyl-methanol (15.0 g, 0.12 mol) and cesium carbonate (59.0 g, 0.18 mol) at room temperature. The reaction mixture was stirred at 100 ° C over a period of 5 h. Then the resulting mixture was cooled and allowed to reach room temperature, diluted with water (250 mL), extracted with ethyl acetate (3X500 mL) and the organic layer was dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (30% ethyl acetate in petroleum ether) to give product 2 as a pale yellow oil 6.0 g (23 %). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 2.25 (s, 3H), 4.40 (d, J=6.0 Hz, 2H), 5.22 (t, J=6.0 Hz ,1H), 6.91- 6.94 (m, 2H), 7.01 (s, 1H), 7.11 (d, J=7.5 Hz,1H), 7.34 (t, J=7.5 Hz, 1H), 7.66 – 7.69 (m, 1H), 7.98 (d, J=2.1 Hz, 1H). MS: (M+H) 216.2. [0461] Step 2 - To a solution of [3-(5-methyl-pyridin-2-yloxy)-phenyl]-methanol 2 (6.0 g, 0.027 mol) in dichloromethane (60 mL) was added thionyl chloride (2.3 mL, 0.03 mol) drop wise while stirring reaction in an ice bath. After removal of ice-bath, the reaction mixture was allowed to stir at room temperature over a period of 1 h. Then volatiles were evaporated under reduced pressure and diluted with toluene (25 mL) and toluene was evaporated under reduced pressure. This azeotropic process was repeated 3 times to obtain product 3 as pale brown color oil (6.2 g, 95%). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 2.25 (s, 3H), 4.76 (s, 2H), 6.95 – 6.98 (m, 2H), 7.15 – 7.26 (m, 2H), 7.38 – 7.40 (m, 1H), 7.66 – 7.69 (m, 1H), 7.99 – 8.0 (m, 1H). MS: (M+H) 234.3. [0462] Step 3 - A solution of 2-(3-chloromethyl-phenoxy)-5-methyl-pyridine (6.2 g, 0.026 mol) in triethyl phosphite (7.3 mL, 0.042 mol) was heated at 150 ° C over a period of 6 h. The reaction mixture was allowed to reach room temperature and the crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (60% ethyl acetate in petroleum ether) to give product 4 as a pale yellow oil 8.3 g. The product contained unused triethyl phosphate and was used in next step without additional purification. 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.13 – 1.23 (m, 6H), 2.25 (s, 3H), 3.20 – 3.27 (m, 2H), 3.89 – 3.99 (m, 4H), 6.91- 6.99 (m, 3H), 7.09 (d, J=7.4 Hz, 1H), 7.32 (t, J=8.1 Hz, 1H), 7.66 – 7.69 (m, 1H), 7.98 – 8.32 (m, 1H). MS: (M+H) 336.1. [0463] Step 4 - To a solution of [3-(5-methyl-pyridin-2-yloxy)-benzyl]-phosphonic acid diethyl ester 4 (8.3 g, 0.024 mol) in THF (40 mL) was added 15-crown ether (0.1 g, 0.48 mmol). The reaction was cooled (ice bath) and NaH (1.44 g, 0.036 mol) was added portion wise. The reaction mixture was allowed to stir at room temperature for 30 min and again cooled to ice temperature. To the above reaction mixture, a solution of 4-oxo-piperidine-1-carboxylic acid tert-butyl ester (4.9 g, 0.024 mol) in THF (40 mL) was added at ice temperature and allowed to stir at room temperature over a period 16 h. The resulting reaction mixture was diluted with water (250 mL), extracted with ethyl acetate (3X500 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (3 % ethyl acetate in petroleum ether) to give product 5 as a pale yellow color oil (6.7 g, 76 %). 1 H NMR (300 MHz, CDCl3) δ (ppm): 1.41 (s, 9H), 2.25 – 2.29 (m, 5H), 2.39 (t, J=6.0 Hz, 2H), 3.36 – 3.42 (m, 4H), 6.36 (s, 1H), 6.90 – 6.95 (m, 3H), 7.02 -7.05 (m, 1H), 7.35 (t, J=7.8 Hz, 1H), 7.66 -7.70 (m, 1H), 7.98 (d, J=2.4 Hz, 1H). MS: (M+H) 381.2. [0464] Step 5 - To a solution of 4-[3-(5-methyl-pyridin-2-yloxy)-benzylidene]-piperidine- 1-carboxylic acid tert-butyl ester 5 (6.7 g, 0.017 mol) in dichloromethane (67.0 mL) was added trifluoroacetic acid (27 mL) at ice temperature and the reaction mixture was stirred at room temperature over a period of 1h. The product 6 (6.96 g) obtained upon evaporation of volatiles was used to next step without additional purification. 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 2.24 (s, 3H), 2.61 (t, J = 6.1 Hz, 2H), 3.10 – 3.30 (m, 4H), 6.36 (s, 1H), 6.90 – 6.95 (m, 3H), 7.02 -7.05 (m, 1H), 7.35 (t, J=7.8 Hz, 1H), 7.66 -7.70 (m, 1H), 7.98 (d, J=2.4 Hz, 1H), 8.70 (bs, 2H). MS: (M+H) 281.3. [0465] Step 6 - To a solution of amine 6 (3.0 g, 7.0 mmol) in dimethyl sulfoxide (30 mL) was added diisopropylethylamine (4.2 mL, 22.0 mmol) and the product of Step 5, Example 1 (1.93 g, 7.0 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 5 h. The resulting reaction mixture was diluted with ethyl acetate (500 mL), washed with water (3X150 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (50% ethyl acetate in petroleum ether) to give product 7 as an off-white solid 2.33 g (70 %). mp: 87.8 °C - 91.0 °C. IR: 3250, 2895, 1624, 1425, 1263, 848, 774 cm -1 . 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.05-1.18 (m, 2H), 1.80 – 1.95 (m, 1H), 2.23 – 2.33 (m, 5H), 2.40 (t, J = 5.1 Hz, 2H), 2.71 (m, 1H), 3.31 (t, J = 5.5 Hz, 2H), 3.38 (t, J = 5.5 Hz, 2H), 6.37 (s, 1H), 6.86 - 6.97 (m, 4H), 7.04 -7.22 (m, 3H), 7.23 – 7.33 (m, 3H), 7.40 (td, J = 7.9, 2.0 Hz, 1H), 7.70 (dt, J = 8.3, 2.5 Hz, 1H), 7.99 (bs, 1H). MS: (M+H) 440.5. [0466] Example 20 - Synthesis of 4-[3-(pyridin-2-yloxy)-benzylidene]-piperidine-1- carboxylic acid [2-(4-methyl)phenyl-cyclopropyl]-amide:

[0467] [0468] Step 1 - Ammonium acetate (13.4 g, 0.17 mol) was added to acetic acid (100 mL) and stirred until fully dissolved. Then nitromethane (30.46 g, 0.49 mol) was added to reaction mixture followed by addition of 4-methyl-benzaldehyde (9.82 mL, 0.083 mol). The reaction mixture was refluxed at 100 °C for 6 h. The reaction mixture was allowed to stir at room temperature over a period of 16 h. The resulting reaction mixture was quenched with aq.2M sodium hydroxide solution (pH = 7) and extracted with ethyl acetate (2X300 mL), dried over sodium sulphate and concentrated under reduced pressure. The crude was washed with hexane to give product 2 as a yellow solid (10 g, 74%). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 2.361(s, 3H), 7.303(d, 2H), 7.755(d, 2H), 8.072-8.213(m, 2H). MS (M-H) 162.9. [0469] Step 2 - To a solution of sodium hydride 60% dispersion in mineral oil (0.98 g, 0.024 mol) in dimethyl sulfoxide (10 mL) was added trimethyloxosulphonium iodide (6.7 g, 0.03 mol) and stirred for 30 min at room temperature. Then 2 (2 g, 0.012 mol) in dimethyl sulfoxide (10 mL) was added and reaction mixture stirred at room temperature over a period of 1 h. The resulting reaction mixture was quenched with water (100 mL), extracted with ethyl acetate (2X300 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400 mesh) column (2% ethyl acetate in hexanes) to give product 3 as a pale yellow oil (300 mg, 14%). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.653-1.704 (m, 1H), 2.203-2.273(m, 1H), 2.353(s, 3H), 3.112-3.157(m, 1H), 4.370-4.417 (m, 1H), 7.015-7.042 (d, 2H), 7.14-7.166 (d, 2H). MS (M+H) 178.1. [0470] Step 3 - To a solution of 3 (0.3 g, 0.0016 mol) in isopropyl alcohol (12 mL) was added hydrochloric acid (6.2 mL of 2.7 N solution, 0.0169 mol) followed by zinc dust (1.1 g, 0.0169 mol) portion wise. Reaction mass allowed to stir at room temperature over a period of 16 h. The reaction mass was neutralized with 10% sodium hydroxide solution and filtered through celite bed. The filtrate was diluted with ethyl acetate (150 mL), washed with water (50 mL) and brine solution (50 mL), dried over sodium sulphate and concentrated under reduced pressure. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400 mesh) column (2% methanol in chloroform) to give product 4 as a yellow oil (150 mg, 60%). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 0.879 (m, 2H), 1.667 (m, 1H), 2.229 (s, 3H), 2.293-2.331 (m, 1H), 7.018(d, J=8.1, 2H), 6.879 (d, J=8.1, 2H). MS (M+H) 148.2. [0471] Step 4 - To a solution of 4 (90 mg, 0.0006 mol) in dichloromethane (2 mL) was added triethylamine (0.17 mL, 0.0012 mol) and phenyl chloroformate (115 mg, 0.0007 mol) at 0 °C. Reaction mass was allowed to stir at room temperature over a period of 1 h. The resulting reaction mass was diluted with ethyl acetate (150 mL), washed with water (50 mL), dried over sodium sulphate and concentrated under reduced pressure. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400 mesh) column (10% ethyl acetate in hexanes) to give product 5 as a white solid (30 mg, 18%). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.115-1.123 (m, 2H), 2.245 (s, 3H), 7.000-7.120 (m, 6H), 7.181-7.356 (m, 1H), 7.361-7.395 (m, 2H). MS (M+H) 268.3. [0472] Step 5 - To a solution of product of Step 4, Example 2 (1.0 g, 0.002 mol) in dichloromethane (10 mL) was added trifluoroacetic acid (4 mL) at 0 °C and the reaction mixture was allowed to stir at room temperature over a period of 1 h. The product 7 (1.3 g, 97%) obtained upon evaporation of volatiles was used for next step without additional purification. [0473] Step 6 - To a solution of 7 (0.29 g, 0.0006 mol) in dimethyl sulfoxide (3 mL) was added diisopropylethylamine (0.59 mL, 0.0034 mol) and 5 (0.16 g, 0.0006 mol) at room temperature. The reaction mass was allowed to stir at 60 °C over a period of 5 h. The resulting reaction mass was diluted with ethyl acetate (200 mL), washed with water (3X50 mL) and dried over sodium sulphate and concentrated under reduced pressure. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400 mesh) column (40% ethyl acetate in hexanes) to give product 8 as an off-white solid (180 mg, 69%). mp: 87.8 -91.0 °C. IR: 3250, 3013, 1624, 1573, 1425, 1263, 1117, 775 cm -1 . 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.001-1.046 (m, 1H), 1.085-1.133 (m, 1H), 1.836-1.845 (m, 1H), 2.249 (s, 5H), 2.382-2.44 (m, 2H), 2.643-2.665 (m, 1H), 3.29-3.326 (m, 2H), 3.365-3.401 (m, 2H), 6.360 (s, 1H), 6.952-7.082 (m, 8H), 7.117-7.157 (m, 1H), 7.348-7.401 (m, 1H), 7.830-7.888 (m, 1H), 8.156-8.166 (m, 1H). MS (M+H) 440.4. [0474] Example 21 - Synthesis of 4-[3-(pyrimidin-2-yloxy)-benzylidene]-piperidine-1- carboxylic acid [(1R, 2S)-2-phenyl-cyclopropyl]-amide: [0475] [0476] Step 1 - To a solution of amine 1, the product of Step 5, Example 6 (3.02 g, 7.89 mmol) in dimethyl sulfoxide (30.0 mL) was added diisopropylethylamine (4.13 mL, 23.6 mmol) and 2, the product of Step 4, Example 16 (2.0 g, 7.89 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 5 h. The resulting reaction mixture was diluted with ethyl acetate (300 mL), washed with water (3X150 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (60% ethyl acetate in petroleum ether) to give product 3 as a white solid 2.3 g (70 %). mp: 62.8 - 65.2 °C. IR: 3627, 3310, 1732, 1629, 1570, 1526, 1310, 1249, 1148,753, 696 cm -1 . 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 1.04 - 1.18 (m, 2H), 1.85 – 1.88 (m, 1H), 2.27 (t, J = 5.7 Hz, 2H), 2.39 (t, J = 5.8 Hz, 2H), 2.71 (dt, J=7.4, 3.7 Hz, 1H), 3.31(d, J = 5.9 Hz, 2H), 3.39 (d, J = 5.9 Hz, 2H), 6.37 (s, 1H), 6.84 (d, J=3.1 Hz, 1H), 7.03 - 7.12 (m, 6H), 7.20 -7.30 (m, 3H), 7.39 (t, J = 4.7 Hz, 1H), 8.64 (dd, J = 4.7 Hz, 1.1 Hz, 2H). MS: 427.4 (M+H). HPLC purity: 99.79 %. Chiral HPLC purity: 99.92 %. Optical Rotation: -1.190. Specific Optical Rotation: -111.71. [0477] Example 22 - Synthesis of 4-[3-(5-methyl-pyridin-2-yloxy)-benzylidene]- piperidine-1-carboxylic acid ((1R,2S)-2-phenyl-cyclopropyl)-amide: [0478] [0479] Step 1 - To a solution of amine 1, the product of Step 5, Example 19 (3.0 g, 7.0 mmol) in dimethyl sulfoxide (30 mL) was added diisopropylethylamine (4.2 mL, 22.0 mmol) and the product of Step 4, Example 16 (1.93 g, 7.0 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 5 h. The resulting reaction mixture was diluted with ethyl acetate (500 mL), washed with water (3X150 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (50% ethyl acetate in petroleum ether) to give product 3 as a pale yellow solid 2.7 g (81 %). mp: 53.1-53.9 °C. IR: 3321, 3024, 1628, 1526, 1475,1249, 752, 695 cm -1 . 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.06-1.21 (m, 2H), 1.80 – 1.95 (m, 1H), 2.23 – 2.33 (m, 5H), 2.40 (t, J = 5.6 Hz, 2H), 2.71 (m, 1H), 3.31 (t, J = 5.5 Hz, 2H), 3.38 (t, J = 5.5 Hz, 2H), 6.37 (s, 1H), 6.86 - 6.97 (m, 4H), 7.04 -7.22 (m, 3H), 7.23 – 7.33 (m, 3H), 7.40 (td, J = 7.9, 2.0 Hz, 1H), 7.70 (dt, J = 8.3, 2.5 Hz, 1H), 7.99 (d, J = 2.8 Hz, 1H). MS (M+H) 440.5. HPLC purity: 98.5%. Chiral HPLC purity: 100 %. [0480] Example 23 - Synthesis of 4-[3-(pyrimidin-2-yloxy)-benzylidene]-piperidine-1- carboxylic acid methyl-((1R,2S)-2-phenyl-cyclopropyl)-amide: [0481] [0482] Step 1 - To a solution of 1, product of Example 21 (150 mg, 7.89 mmol) in dimethylformamide (30.0 mL) were added sodium hydride (4.13 mL, 23.6 mmol) and methyl iodide (2.0 g, 7.89 mmol) at 0-5 °C. The reaction mixture was allowed to stir at 25-30 °C over a period of 1 h. The resulting reaction mixture was diluted with water (50 mL), extracted with ethyl acetate (2X100 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (25 % ethyl acetate in petroleum ether) to give 2 as a pale yellow solid 77 mg (50 %). 1 H NMR (300 MHz, DMSO- d6) δ (ppm): 1.24 (dt, J = 7.8 Hz, 5.9 Hz, 2H), 2.02 – 2.07 (m, 1H), 2.27 (q, J = 5.9 Hz, 2H), 2.38 – 2.41 (m, 2H), 2.79 (s, 4H), 3.12 - 3.33 (m, 4H), 6.34 (s, 1H), 6.97 -7.21 (m, 6H), 7.20- 7.31 (m, 3H), 7.39 (t, J = 7.9 Hz, 1H), 8.65 (d, J = 4.8 Hz, 2H). MS: 441.5 (M+H). HPLC purity: 98.1 %. [0483] Example 24 - Synthesis of 4-[3-(5-methyl-pyrazin-2-yloxy)-benzylidene]- piperidine-1-carboxylic acid ((1R,2S)-2-phenyl-cyclopropyl)-amide:

[0485] Step 1 - To a solution of 2-chloro-5-methyl-pyrazine (10 g, 0.078 mol) in DMF (100 mL) was added 3-hydroxyphenyl-methanol (11.6 g, 0.094 mol) and cesium carbonate (76.0 g, 0.23 mol) at room temperature. The reaction mixture was stirred at 100 ° C over a period of 5 h. Then the resulting mixture was allowed to reach room temperature, diluted with water (250 mL), extracted with ethyl acetate (3X500 mL) and the organic layer was dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (30% ethyl acetate in petroleum ether) to give product 1 as a pale yellow oil 6.8 g (40%). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 2.45 (s, 3H), 4.50 (d, J=4.5 Hz, 2H), 5.26 (t, J=5.26 Hz ,1H), 7.0 – 7.18 (m,3H), 7.37 (t, J=7.5 Hz, 1H), 8.1 (s, 1H), 8.4 (d, J=1.2 Hz, 1H). MS (M+H) 217.2. [0486] Step 2 - To a solution of 1 (6.0 g, 0.027 mol) in dichloromethane (60 mL), thionyl chloride (2.3 mL, 0.03 mol) was added in a dropwise fashion while stirring reaction mixture in an ice bath. After removal of ice-bath, the reaction mixture was allowed to stir at room temperature over a period of 1 h. Then volatiles were evaporated under reduced pressure and diluted with toluene (25 mL) and toluene was evaporated under reduced pressure. This azeotropic process was repeated 3 times to obtain product 2 as a pale brown color oil (6.2 g). The crude product was used for next step without further purification. MS (M+H) 235.3. [0487] Step 3 - A solution of 2 (6.2 g, 0.026 mol) in triethyl phosphite (7.3 mL, 0.042 mol) was heated at 150 ° C over a period of 6 h. The reaction mixture was cooled and allowed to reach room temperature and the crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (60% ethyl acetate in petroleum ether) to give product 3 as a pale yellow oil 8.3 g. The product contained unused triethylphosphate and was used in next step without additional purification. 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 1.16 (t, J=7.0 Hz, 6H), 2.45 (s, 3H), 3.23 (s, 1H), 3.23 (s, 1H), 3.94 (dq, J=8.2 Hz, 7.0 Hz, 4H), 7.0 - 7.12 (m, 2H), 7.13 - 7.15 (m, 1H), 7.36 (t, J=7.8 Hz, 1H), 8.09 (dd, J=1.4 Hz, 0.7 Hz, 1H), 8.39 (d, J=1.4 Hz, 1H). MS (M+H) 337.1. [0488] Step 4 - To a solution of 3 (8.3 g, 0.024 mol) in THF (40 mL) was added 15-crown ether (0.1 g, 0.48 mmol). The reaction was cooled (ice bath) and NaH (1.44 g, 0.036 mol) was added portion wise. The reaction mixture was allowed to stir at room temperature for 30 min and again cooled to ice temperature. To the above reaction mixture, a solution of 4-oxo- piperidine-1-carboxylic acid tert-butyl ester 5 (4.9 g, 0.024 mol) in THF (40 mL) was added at ice temperature and allowed to stir at room temperature over a period 16 h. The resulting reaction mixture was diluted with water (250 mL), extracted with ethyl acetate (3X500 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles to give product 4 as a pale yellow color oil (6.7 g). MS (M+H) 382.3. The product 4 was used in next step without further purification. [0489] Step 5 - To a solution of 4 (6.7 g, 0.017 mol) in dichloromethane (67.0 mL) was added trifluoroacetic acid (27 mL, 4V) at ice-cold temperature and the reaction mixture was stirred at room temperature over a period of 1 h. The product 6 (6.96 g, 90%) obtained upon evaporation of volatiles was used to next step without additional purification. 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 2.45 (s, 3H), 2.60 – 2.67 (m, 4H), 3.10 – 3.17 (m, 4H), 6.46 (s, 1H), 7.04 -7.12 (m, 4H), 7.38 (t, J=7.8 Hz, 1H), 8.09 (s, 1H), 8.40 (d, J=4.4 Hz, 1H), 8.61 (bs, 2H). MS (M+H) 282.3. [0490] Step 6 - To a solution of amine 6 (1.0 g, 2.5 mmol) in dimethyl sulfoxide (30 mL) was added diisopropylethylamine (1.4 mL, 7.5 mmol) and the carbamate product of Step 4, Example 16 (0.7 g, 2.75 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 5 h. The resulting reaction mixture was diluted with ethyl acetate (500 mL), washed with water (3X150 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (60% ethyl acetate in petroleum ether) to give product 7 as a pale yellow solid 0.7 g (68 %). mp: 50.8 °C. IR: 3305, 2923, 1627, 1528, 1473, 1337, 1266, 695 cm -1 . 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 1.02-1.20 (m, 2H), 1.82 – 1.84 (m, 1H), 2.21 – 2.41 (m, 4H), 2.46 (s, 3H), 2.60 – 2.70 (m, 1H), 3.26 – 3.34 (m, 4H), 6.32 (s, 1H), 6.80 – 7.22 (m, 9H), 7.35 (t, J = 7.6, 1H), 8.06 (bs, 1H), 8.37 (bs, J = 8.3, 1H). MS: 441.4 (M+H). HPLC: 99.91 %. [0491] Example 25 - Synthesis of 4-[3-pyrazin-2-yloxy)-benzylidene]-piperidine-1- carboxylic acid ((1R,2S)-2-phenyl-cyclopropyl)-amide The target compound 7 is prepared following the synthetic methodology described for product of Example 24 starting with 2-chloro-pyrazine instead of 2-chloro-5-methyl-pyrazine as described in Example 24. [0492] Example 26 - Synthesis of 4-[5-methyl-3-(pyrimidin-2-yloxy)-benzylidene]- piperidine-1-carboxylic acid (2-phenyl-cyclopropyl)-amide

The target compound 6 is prepared following the synthetic methodology described for product of Example 6 starting with 2-chloro-5-methyl-pyrimidine instead of 2-chloro-pyrimidine as described in Example 6. [0493] Example 27 - Synthesis of 4-[3-(5-chloro-pyridin-2-yloxy)-benzylidene]- piperidine-1- carboxylic acid ((1R,2S)-2-phenyl-cyclopropyl)-amide.

[0495] Step 1 - To a solution of 5-chloro-2-fluoropyridine (10.0 g, 0.0760 mol) in DMSO (100 mL) were added 3-hydroxyphenyl-methanol (9.42 g, 0.0760 mol) and cesium carbonate (29.72 g, 0.0912 mol) at room temperature. The reaction mixture was stirred at 100 °C over a period of 6 h. Reaction was monitored by TLC. The resulting mixture was cooled to room temperature, diluted with water (200 mL), extracted with ethyl acetate (2X400 mL) and the organic layer was dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (12% ethyl acetate in petroleum ether) to give product 1 as a pale-yellow oil 12.0 g (67 %). 1 H NMR (400 MHz, DMSO-d6) δ(ppm): 8.17 (t, J=2.4 Hz, 1H), 7.94-7.91 (m, 1H), 7.33 (t, J=8.0 Hz, 1H), 7.14-7.1 (m, 1H), 7.06-7.03 (m, 2H), 6.98 – 6.95 (m, 1H), 5.22 (t, J=5.6 Hz, 1H), 4.48 (d, J=5.6 Hz, 2H). MS m/z (M+H): 236.0 [0496] Step 2: To a solution of 1 (12.0 g, 0.0509 mol) in dichloromethane (120 mL) was added thionyl chloride (4.1 mL, 0.0560 mol) in a dropwise fashion while stirring reaction in an ice bath. After removal of ice-bath, the reaction mixture was allowed to stir at room temperature over a period of 1 h. After complete consumption of starting material, the volatiles were evaporated under reduced pressure, diluted with ethyl acetate (250 mL) and organic layer was washed with saturated sodium bicarbonate solution and water. Organic layer was dried over anhydrous sodium sulphate and concentrated. The crude product 2 obtained upon evaporation was directly taken to next step without further purification (12.5 g, 96%). 1 H NMR (400 MHz, DMSO-d6) δ(ppm): 8.20 (d, J=2.4 Hz, 1H), 8.19-7.95 (m, 1H), 7.42 (t, J=7.6 Hz, 1H), 7.29 (d, J=8.0 Hz, 1H), 7.21 (t, J=2.0 Hz, 1H), 7.12-7.09 (m, 2H), 4.75 (s, 1H). MS m/z (M+H): 254.1 [0497] Step 3: A solution of 2 (12.5 g, 0.0494 mol) in triethyl phosphite (20.0 mL, 0.1235 mol) was heated at 150 °C over a period of 6 h. The reaction mixture was allowed to reach room temperature and the crude product obtained after removal of the volatiles was added to n-heptane (150 mL) to yield a light orange color precipitate. The precipitate obtained was filtered and dried under vacuum to give product 3 as an off-white solid (16.5 g, 91%) and was used in the next step without further purification. 1 H NMR (400 MHz, DMSO-d6) δ(ppm): 8.19-8.18 (m, 1H), 7.97-7.94 (m, 1H), 7.34 (t, J=7.6 Hz, 1H), 7.13-7.06 (m, 1H), 7.03-6.99 (m, 3H), 3.95 (m, 4H), 3.27 and 3.21(2s, 2H), 1.15 (t, J=4.4 Hz, 6H). MS m/z (M+H): 356.2 [0498] Step 4: To a solution of 3 (15.5 g, 0.0435 mol) in THF (100 mL) was added 15- crown ether (0.19 g, 0.87 mmol). The reaction was cooled (ice bath) and NaH (2.07 g, 0.0870 mol) added portion wise over a period of 5 min. The reaction mixture was allowed to stir at room temperature for 30 minutes and again cooled to ice temperature. A solution of 4-oxo- piperidine-1-carboxylic acid tert-butyl ester 5 (8.66 g, 0.0435 mol) in THF (50 mL) was added at ice temperature and allowed to stir at room temperature over a period 16 h. The resulting reaction mixture was quenched with water and extracted with ethyl acetate. Organic layer was dried over anhydrous sodium sulphate and concentrated. The crude product obtained after evaporation of the volatiles were purified by silica-gel column chromatography to obtain the product 4 as a light yellow liquid (13.1 g, 75%). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 8.19 (d, J=2.0 Hz, 1H), 7.96-7.93 (m, 1H), 7.36 (t, J=7.6 Hz, 1H), 7.09-7.06 (m, 2H), 6.99-6.96 (m, 2H), 6.35 (s, 1H), 3.40-3.32 (m, 4H), 2.38 (t, J=5.2 Hz, 2H), 2.26 (t, J=6.0 Hz, 2H), 1.39 (s, 9H). MS m/z (M+Na): 423.2 [0499] Step 5: To a solution of 4 (13.0 g, 0.0325 mol) in dichloromethane (130 mL) was added trifluoroacetic acid (52.0 mL) at ice temperature and the reaction mixture was stirred at room temperature over a period of 2 h. Reaction was monitored by TLC. After complete consumption of the starting material, volatiles were removed under reduced pressure to get product as a pale brown oil. The crude product obtained was washed with ether (3X50 mL) to give 6 as a pale brown thick liquid (13.8 g crude). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 8.82 (bs, 2H), 8.19 (d, J=2.8 Hz, 1H), 7.96-7.93 (m, 1H), 7.38 (t, J=8.0 Hz, 1H), 7.09 (d, J=8.8 Hz, 2H), 7.02-7.00 (m, 2H), 6.44 (s, 1H), 3.38-3.33 (m, 4H), 2.60 (t, J=5.6 Hz, 2H), 2.53-2.48 (m, 2H). MS m/z (M+H): 301.2 [0500] Step 6: To a solution of 6 (15.8 g, 0.0381 mol) in dimethyl sulfoxide (78 mL,) was added diisopropyl-ethyl-amine (20.34 mL, 0.1149 mol) and the carbamate product of Step 4, Example 16 (10.62 g, 0.0419 mol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 6 h. The resulting reaction mixture was diluted with ethyl acetate (500 mL), washed with water (3X200 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (40% ethyl acetate in petroleum ether) to give product 7 as a light yellow fluffy solid (11.6 g, 66 %). melting range (MR): 44.8-62.6 °C 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 8.18 (d, J=2.4 Hz, 1H), 7.94-7.91 (m, 1H), 7.35 (t, J=7.6 Hz, 1H), 7.23-7.19 (m, 2H), 7.12-7.05 (m, 5H), 6.95- 6.82 (m, 2H), 6.32 (s, 1H), 3.37-3.26 (m, 4H), 2.71-2.69 (m, 1H), 2.36-2.33 (t, J=5.2 Hz, 2H), 2.24-2.22 (t, J=5.6 Hz, 2H), 1.85 (m, 1H), 1.13 (d, J=4.8 Hz, 1H), 1.04 (d, J=7.6 Hz, 1H). 13 C NMR: (100 MHz, DMSO-d6): δ 161.69, 157.57, 153.49, 145.56, 141.99, 139.92, 139.75, 138.80, 129.56, 128.08, 125.85, 125.34, 125.21, 123.04, 121.29, 119.02, 113.05, 44.93, 43.89, 35.63, 34.07, 28.91, 24.32 and 15.58. MS m/z (M+H): 460.32, HPLC purity: 99.36%, Chiral purity: 99.71%. [0501] Example 28 - Synthesis of 4-[3-(5-fluoro-pyridin-2-yloxy)-benzylidene]- piperidine-1-carboxylic acid ((1R,2S)-2-phenyl-cyclopropyl)-amide [0503] Step 1: To a solution of 2,5-difluoropyridine (8.2 g, 0.0719 mol) in DMSO (80 mL) were added 3-hydroxyphenyl-methanol (8.9 g, 0.0719 mol) and cesium carbonate (28.12 g, 0.0863 mol) at room temperature and the reaction mixture stirred at 85 °C over a period of 6 h. Reaction was monitored by TLC. The resulting mixture was cooled to reach room temperature, diluted with water (200 mL), extracted with ethyl acetate (3X400 mL) and the organic layer dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (12% ethyl acetate in petroleum ether) to give product 1 as a pale yellow oil 4.3 g (28 %). 1 H NMR (400 MHz, DMSO-d6) δ(ppm): 8.17 (t, J=2.4 Hz, 1H), 7.84-7.78 (m, 1H), 7.35 (t, J=10.4 Hz, 1H), 7.15-6.95 (m, 4H), 5.25 (t, J=7.6 Hz, 1H), 4.50 (d, J=7.6 Hz, 2H). MS m/z (M+H): 220.0 [0504] Step 2: To a solution of 1 (6.5 g, 0.0296 mol) in dichloromethane (65 mL) was added thionyl chloride (2.4 mL, 0.0326 mol) dropwise while stirring reaction in an ice bath. After removal of ice-bath, the reaction mixture was allowed to stir at room temperature over a period of 2 hours. After complete consumption of starting material, the volatiles were evaporated under reduced pressure, diluted with ethyl acetate (200 mL), and organic layer washed with saturated sodium bicarbonate solution and water. Organic layer was dried over anhydrous sodium sulphate and concentrated. The crude product 2 obtained upon evaporation was directly taken to next step without further purification (6.7 g, 95%). 1 H NMR (400 MHz, DMSO-d6) δ(ppm): 8.15 (d, J=2.4 Hz, 1H), 7.83-7.80 (m, 1H), 7.40 (t, J=7.6 Hz, 1H), 7.27- 7.25 (m, 1H), 7.19-7.06 (m, 3H), 4.75 (s, 1H). MS m/z (M+H): 238.0 [0505] Step 3: A solution of 2 (6.5 g, 0.0274 mol) in triethyl phosphite (12.6 mL, 0.0685 mol) was heated at 150 °C over a period of 6 h. The reaction mixture was cooled to reach room temperature and the mixture obtained after evaporation of the volatiles was purified by silica- gel (230-400) column chromatography to obtain product 3 as a pale yellow liquid (9.0 g, 95%). 1 H NMR (400 MHz, DMSO-d6) δ(ppm): 8.15 (d, J=4.0 Hz, 1H), 7.86-7.80 (m, 1H), 7.34 (t, J=10.4 Hz, 1H), 7.12-6.97 (m, 4H), 4.02-3.89 (m, 4H), 3.28 and 3.21(2s, 2H), 1.15 (t, J=9.2 Hz, 6H). MS m/z (M+H): 340.2 [0506] Step 4: To a solution of 3 (9.0 g, 0.0256 mol) in THF (60 mL) was added 15-crown ether (0.12 g, 0.53 mmol). The reaction was cooled (ice bath) and 60% NaH (1.26 g, 0.0530 mol) was added portion wise over a period of 5 min. The reaction mixture was allowed to stir at room temperature for 30 min and again cooled to ice temperature. A solution of 4-oxo- piperidine-1-carboxylic acid tert-butyl ester 5 (5.28 g, 0.0256 mol) in THF (30 mL) was added at ice temperature and allowed to stir at room temperature over a period 16 h. The resulting reaction mixture was quenched with water and extracted with ethyl acetate. Organic layer was dried over anhydrous sodium sulphate and concentrated. The crude product obtained after evaporation of the volatiles were purified by silica-gel column chromatography to get product 4 as a pale yellow solid (8.0 g, 78%). %). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 8.15 (d, J=3.2 Hz, 1H), 7.84-7.79 (m, 1H), 7.35 (t, J=8.0 Hz, 1H), 7.12-7.04 (m, 2H), 6.96-6.92 (m, 2H), 6.35 (s, 1H), 3.40-3.32 (m, 4H), 2.37 (t, J=5.6 Hz, 2H), 2.25 (t, J=5.2 Hz, 2H), 1.39 (s, 9H). MS m/z (M+Na): 407.2 [0507] Step 5: To a solution of 4 (8.2 g, 0.0213 mol) in dichloromethane (82 mL) was added trifluoroacetic acid (32.5 mL) at ice temperature and the reaction mixture was stirred at room temperature over a period of 1 h. Reaction was monitored by TLC. After complete consumption of starting material, volatiles were removed under reduced pressure to obtain product as a red oil. The crude product obtained was washed with ether (3X50 mL) to yield 6 as thick brown oil (9.0 g Crude). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 8.73 (bs, 2H), 8.15 (d, J=3.2 Hz, 1H), 7.84-7.79 (m, 1H), 7.37 (t, J=8.0 Hz, 1H), 7.12-7.06 (m, 2H), 7.00-6.96 (m, 2H), 6.44 (s, 1H), 3.15-3.09 (m, 4H), 2.59 (t, J=6.0 Hz, 2H), 2.49-2.48 (m, 2H). MS m/z (M+H): 285.4 [0508] Step 6: To a solution of 6 (8.4 g, 0.021 mol) in dimethyl sulfoxide (42 mL) was added diisopropyl-ethyl-amine (11.1 mL, 0.063 mol) and the carbamate product of Step 4, Example 16 (5.8 g, 0.023 mol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 6 h. The resulting reaction mixture was diluted with ethyl acetate (400 mL), washed with water (3X100 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (40% ethyl acetate in petroleum ether) to give product 7 as a light yellow fluffy solid (7.15 g, 66 %). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 8.15 (d, J=2.4 Hz, 1H), 7.82-7.80 (m, 1H), 7.35 (t, J=8.0 Hz, 1H), 7.24-7.21 (m, 2H), 7.14-7.04 (m, 5H), 6.96-6.94 (m, 2H), 6.84 (d, J=2.8 Hz, 1H), 6.34 (s, 1H), 3.38-3.28 (m, 4H), 2.71-2.69 (m, 1H), 2.34 (t, J=5.2 Hz, 2H), 2.23 (t, J=4.8 Hz, 2H), 1.86 (m, 1H), 1.13 (d, J=4.8 Hz, 1H), 1.04 (d, J=7.6 Hz, 1H). 13 C NMR: (100 MHz, DMSO-d6): δ 159.15, 157.57, 156.01 (d, J=244.7 Hz), 154.08, 141.99, 139.69, 138.76, 134.13 (d, J=26.3 Hz), 129.54, 128.09, 127.85 (d, J=20.9 Hz), 125.86, 125.35, 124.91, 123.09, 120.97, 118.71, 113.01, 44.94, 43.90, 35.63, 34.07, 28.91, 24.32 and 15.58. MS m/z (M+H): 444.3, HPLC purity: 99.21%, Chiral HPLC: 99.37%. [0509] Example 29: Synthesis of 6-{3-[1-((1R,2S)-2-phenyl-cyclopropylcarbamoyl)- piperidin-4-ylidenemethyl]-phenoxy}-nicotinic acid methyl ester [0510] [0511] Step 1: To a solution of methyl 6-chloropyridine-3-carboxylate (50.0 g, 0.29 mol) in dimethyl-acetamide (500 mL) were added 3-hydroxyphenyl-methanol (39.79 g, 0.32 mol) and potassium carbonate (60.4 g, 0.43 mol) at room temperature. The reaction mixture was stirred at 100 °C over a period of 6 h. Reaction was monitored by TLC. The resulting mixture was cooled to room temperature, diluted with water (300 mL), extracted with ethyl acetate (2X500 mL) and the organic layer dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (12% ethyl acetate in petroleum ether) to give product 1 as a pale yellow oil (30.0 g, 40 %). 1 H NMR (400 MHz, DMSO-d 6 ) δ(ppm): 8.82 (d, J=2.0 Hz, 1H), 8.30-8.27 (m, 1H), 7.42 (t, J=8.0 Hz, 1H), 7.27-7.19 (m, 2H), 7.09-7.07 (m, 1H), 6.96 (d, J=8.8 Hz, 1H), 4.73 (s, 2H), 3.93 (d, J=3.6 Hz, 3H). MS m/z (M+H): 259.8 [0512] Step 2: To a solution of 1 (30.0 g, 0.115 mol) in dichloromethane (300 mL) was added thionyl chloride (9.4 mL, 0.127 mol) dropwise while stirring reaction in an ice bath. After removal of ice-bath, the reaction mixture was allowed to stir at room temperature over a period of 2 hours. After complete consumption of starting material, the volatiles were evaporated under reduced pressure, diluted with ethyl acetate (500 mL), organic layer was washed with saturated sodium bicarbonate (200 mL) solution and water. Organic layer was dried over anhydrous sodium sulphate and concentrated. The crude product obtained upon evaporation was purified by silica gel (230-400) column chromatography to obtain product 2 as a pale yellow liquid (28.0 g, 87%). 1 H NMR (400 MHz, DMSO-d6) δ(ppm): 8.82 (d, J=0.8 Hz, 1H), 8.31-8.28 (m, 1H), 7.43 (t, J=8.0 Hz, 1H), 7.29-7.27 (m, 1H), 7.21 (t, J=2.0 Hz, 1H), 7.14-7.11 (m, 1H), 6.98-6.96 (m, 1H), 4.61 (s, 2H), 3.93 (s, 3H). MS m/z (M+H): 278.0 [0513] Step 3: A solution of 2 (28.0 g, 0.10 mol) in triethyl phosphite (41.0 mL, 0.25 mol) was heated at 150 °C over a period of 6 h. The reaction mixture was cooled to reach room temperature and the mixture obtained after evaporation of the volatiles was purified by silica- gel column chromatography to get 3 as a pale yellow liquid (32.0 g, 84%). 1 H NMR (400 MHz, DMSO-d6) δ(ppm): 8.65-8.64 (m, 1H), 8.30-8.27 (m, 1H), 7.35 (t, J=8.0 Hz, 1H), 7.16-7.03 (m, 4H), 3.96-3.88 (m, 4H), 3.82 (s, 3H), 3.27 and 3.21(2s, 2H), 1.15-1.11 (m, 6H). MS m/z (M+H): 380.2 [0514] Step 4: To a solution of 3 (35.5 g, 0.093 mol) in THF (200 mL) was added 15- crown ether (0.41 g, 1.8 mmol). The reaction was cooled (ice bath) and 60% NaH (5.5 g, 0.14 mol) added portion wise over a period of 5 minutes. The reaction mixture was allowed to stir at room temperature for 30 min and again cooled to ice temperature. A solution of 4-oxo- piperidine-1-carboxylic acid tert-butyl ester 5 (18.7 g, 0.093 mol) in THF (150 mL) was added at ice temperature and allowed to stir at room temperature over a period 16 h. The resulting reaction mixture was quenched with saturated ammonium chloride and extracted with ethyl acetate. Organic layer was dried over anhydrous sodium sulphate and concentrated. The crude product (30.0 g) obtained after evaporation of the volatiles was dissolved in methanol (300 mL) and aqueous lithium hydroxide solution (3.0 g, 0.0707 mol) was added at ice temperature. Resulting reaction mass was stirred at 50 °C for 2 hours. Reaction was monitored by TLC. The crude product obtained after evaporation of the volatiles was dissolved in water (200 mL) and washed with methyl tert-butyl ether (2x200 mL). Aqueous layer was acidified to pH 2.0 using 1.0 N aqueous HCl solution. The product precipitated was filtered and dried to get 4 as an off- white solid (23.0 g, 61%). 1 H NMR (400 MHz, DMSO-d 6 ) δ(ppm): 13.19 (bs, 1H), 8.66-8.65 (m, 1H), 8.28-8.25 (m, 1H), 7.39 (t, J=8.0 Hz, 1H), 7.11-7.00 (m, 4H), 6.37 (s, 1H), 3.40-3.32 (m, 4H), 2.39 (t, J=5.6 Hz, 2H), 2.27 (t, J=5.2 Hz, 2H), 1.39 (s, 9H). MS m/z (M+H): 433.2 [0515] Step 5: To a solution of 4 (13.0 g, 0.0317 mol) in methanol (130 mL) was added trimethyl-silyl chloride (8.9 mL, 0.0697 mol) at ice temperature and the reaction mixture was stirred at room temperature over a period of 12 h and monitored by TLC. After complete consumption of starting material, volatiles were removed under reduced pressure. The crude product obtained was diluted with saturated sodium bicarbonate solution and extracted with ethyl acetate. Organic layer was washed with water, dried over anhydrous sodium sulphate, filtered and concentrated. The crude product obtained after evaporation of the volatiles were purified by silica-gel (230-400) column chromatography to obtain the product 6 as a pale yellow liquid (5.2 g, 51%). 1 H NMR (400 MHz, DMSO-d6) δ(ppm): 8.68-8.67 (m, 1H), 8.31- 8.28 (m, 1H), 7.39-7.36 (m, 1H), 7.13-6.97 (m, 4H), 6.24 (s, 1H), 3.84 (s, 3H), 2.78-2.65 (m, 4H), 2.34 (t, J=5.2 Hz, 2H), 2.21 (t, J=5.2 Hz, 2H). MS m/z (M+H): 325.3 [0516] Step 6: To a solution of 6 (5.2 g, 0.016 mol) in dimethyl sulfoxide (52 mL, 10V) was added diisopropyl-ethyl-amine (8.9 mL, 0.048 mol) and the carbamate product of Step 4, Example 16 (4.0 g, 0.016 mol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 6 h. Reaction was monitored by TLC. The resulting reaction mixture was diluted with ethyl acetate (300 mL), washed with water (3X100 mL) and dried over anhydrous sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (40% ethyl acetate in petroleum ether) to give product 7 as a light yellow fluffy solid (5.0 g, 65 %).melting point range (MR): 52.6-72.8 °C. 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 8.68 (d, J=2.0 Hz, 1H), 8.31-8.28 (m, 1H), 7.38 (t, J=8.0 Hz, 1H), 7.24- 7.20 (m, 2H), 7.13-7.01 (m, 7H), 6.84-6.83 (m, 1H), 6.35 (s, 1H), 3.84 (s, 3H), 3.38-3.30 (m, 4H), 2.71-2.69 (m, 1H), 2.37 (t, J=5.2 Hz, 2H), 2.25 (t, J=5.2 Hz, 2H), 1.86 (m, 1H), 1.15 (d, J=4.8 Hz, 1H), 1.05 (d, J=6.0 Hz, 1H). 13 C NMR: (100 MHz, DMSO-d6): δ 165.87, 164.78, 157.57, 152.92, 149.45, 141.99, 140.87, 139.87, 138.88, 129.66, 128.08, 125.85, 125.67, 125.34, 122.97, 121.66, 120.97, 119.39, 111.21, 52.23, 44.92, 43.88, 35.63, 34.07, 28.90, 24.31 and 15.57. MS m/z (M+H): 484.3, HPLC purity: 98.65%, Chiral HPLC: 99.08% [0517] Example 30: Synthesis of 6-{3-[1-((1R,2S)-2-phenyl-cyclopropylcarbamoyl)- piperidin-4-ylidenemethyl]-phenoxy}-nicotinic acid [0518] [0519] To a solution of 1, product of Example 27 (1.8 g, 0.0038 mol) in methanol (18 mL) was added aqueous lithium hydroxide (0.32 g, 0.0076 mol) dropwise at ice temperature. The reaction mixture was allowed to stir at room temperature over a period of 3 h. Reaction was monitored by TLC. The crude product obtained after evaporation of the volatiles were diluted with water (10 mL) and the aqueous layer washed with methyl tert-butyl ether. The resulting aqueous layer was acidified with 1.5 N HCl to pH 2. The product precipitated, was filtered and dried to give 2 as an off-white solid 1.52 g (87 %). Melting range (MR) 141-159 °C. 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 13.19 (bs, 1H), 8.66-8.65 (m, 1H), 8.28-8.26 (m, 1H), 7.38 (t, J=8.0 Hz, 1H), 7.24-7.21 (m, 2H), 7.13-7.01 (m, 7H), 6.83 (d, J=3.2 Hz, 1H), 6.35 (s, 1H), 3.38-3.28 (m, 4H), 2.71-2.69 (m, 1H), 2.37 (t, J=5.2 Hz, 2H), 2.25 (t, J=5.2 Hz, 2H), 1.86 (m, 1H), 1.15 (d, J=4.4 Hz, 1H), 1.05 (d, J=7.6 Hz, 1H). 13 C NMR: (100 MHz, DMSO-d6): δ 165.84, 165.69, 157.57, 153.03, 149.57, 142.0, 141.05, 139.84, 138.86, 129.64, 128.09, 125.85, 125.59, 125.34, 123.0, 122.02, 121.66, 119.40, 111.06, 44.92, 43.89, 35.64, 34.08, 28.91, 24.30 and 15.57. MS m/z (M+H): 470.3, HPLC purity: 99.88%, Chiral HPLC: 99.50%, [0520] Example 31: Synthesis of 4-[3-(5-hydroxymethyl-pyridin-2-yloxy)-benzylidene]- piperidine-1-carboxylic acid ((1R,2S)-2-phenyl-cyclopropyl)-amide [0522] Step 1: To a solution of 1, product of step 4 Example 27 (3.6 g, 0.0085 mol) in dimethoxyethane (35 mL) was added N-methyl morpholine (1.4 mL, 0.0128 mol) and isobutyl chloroformate (1.21 mL, 0.0093 mol) at ice temperature and the reaction mixture was stirred at room temperature over a period of 30 minutes. Sodium borohydride (1.9 g, 0.0512 mol) was added portion wise to the reaction mass and stirred for 12 hours. Reaction was monitored by TLC. After complete consumption of the starting material 1, reaction mass was quenched with water (100 mL) and extracted with ethyl acetate (300 mL). Organic layer was dried over anhydrous sodium sulphate and concentrated. The crude product obtained was purified by silica-gel (230-400) column chromatography to obtain the product 2 as an off-white solid (3.2 g, 92%). 1 H NMR (400 MHz, DMSO-d 6 ) δ(ppm): 8.06 (m, 1H), 7.79-7.76 (m, 1H), 7.34 (t, J=8.0 Hz, 1H), 7.05-6.95 (m, 4H), 6.35 (s, 1H), 5.24 (t, J=6.0 Hz, 1H), 4.45 (d, J=5.6 Hz, 1H), 3.40-3.32 (m, 4H), 2.38 (t, J=5.6 Hz, 2H), 2.25 (t, J=5.2 Hz, 2H), 1.39 (s, 9H). MS m/z (M+H): 397.3 [0523] Step 2: To a solution of 2 (3.2 g, 0.08 mol) in dichloromethane (32 mL) was added trifluoroacetic acid (12.8 mL) at ice temperature and the reaction mixture was stirred at room temperature over a period of 1 h. Reaction was monitored by TLC. After complete consumption of starting material, volatiles were removed under reduced pressure to obtain crude product as brown red oil. The crude product obtained was washed with ether (3X50 mL) to yield 3 as an off-white solid (3.3 g crude). MS m/z (M+H): 297.17 [0524] Step 3: To a solution of 3 (3.3 g, 0.08 mmol) in dimethyl sulfoxide (30 mL) was added diisopropyl-ethyl-amine (4.2 mL, 0.024 mol) and the carbamate product of Step 4, Example 16 (2.0 g, 0.08 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 6 h. The resulting reaction mixture was diluted with ethyl acetate (300 mL), washed with water (3X100 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (40% ethyl acetate in petroleum ether) to give 4 as a light yellow fluffy solid (1.82 g, 58 %). Melting range (MR): 51-65 °C. 1 H NMR (400 MHz, DMSO-d 6 ) δ(ppm): 8.07 (d, J=2.0 Hz, 1H), 7.80-7.77 (m, 1H), 7.34 (t, J=8.0 Hz, 1H), 7.26-6.92 (m, 7H), 6.83 (d, J=3.2 Hz, 1H), 6.35 (s, 1H), 5.25 (t, J=6.0 Hz, 1H), 4.45 (d, J=7.2 Hz, 1H), 3.40-3.32 (m, 4H), 2.38 (t, J=5.6 Hz, 2H), 2.25 (t, J=5.2 Hz, 2H), 1.87 (m, 1H), 1.16 (d, J=6.0 Hz, 1H), 1.05 (d, J=7.6 Hz, 1H). 13 C NMR: (100 MHz, DMSO-d6): δ162.03, 157.55, 154.11, 145.64, 141.94, 139.57, 139.09, 138.65, 132.95, 129.43, 128.04, 125.84, 125.31, 124.66, 123.11, 120.95, 118.69, 111.17, 60.09, 44.92, 43.87, 35.59, 34.00, 28.88, 24.26 and 15.52. MS m/z (M+H) 456.3, HPLC purity: 98.99%, Chiral HPLC: 98.95%. [0525] Example 32: Synthesis of 4-[3-(5-methoxymethyl-pyridin-2-yloxy)-benzylidene]- piperidine-1-carboxylic acid ((1R,2S)-2-phenyl-cyclopropyl)-amide

[0527] Step 1: To a solution of 1, product of step 1 of Example 29, (3.2 g, 8.0 mmol) in tetrahydrofuran (32 mL) was added 60% NaH (0.97 g, 0.024 mol) at ice temperature and the reaction mixture was stirred at room temperature over a period of 10 minutes. Methyl iodide (1.56 mL, 0.024 mol) was added to the reaction mass at the same ice temperature and continued the stirring for 12 h. Reaction was monitored by TLC. After complete consumption of starting material, reaction was quenched with saturated ammonium chloride solution (100 mL) and extracted with ethyl acetate (300 mL). Organic layer further washed with water, dried over anhydrous sodium sulphate, filtered and concentrated under reduced pressure. The crude product obtained was purified by silica-gel (230-400) column chromatography to obtain the product 2 as an off-white solid (2.6 g, 78%). 1 H NMR (400 MHz, DMSO-d6) δ(ppm): 8.08 (d, J=2.0 Hz, 1H), 7.80-7.78 (m, 1H), 7.35 (t, J=8.0 Hz, 1H), 7.06-6.93 (m, 4H), 6.35 (s, 1H), 4.37 (s, 2H), 3.40-3.32 (m, 4H), 3.26 (s, 3H), 2.38 (t, J=5.6 Hz, 2H), 2.25 (t, J=5.6 Hz, 2H), 1.39 (s, 9H). MS m/z (M+H): 411.3 [0528] Step 2: To a solution of 2 (2.6 g, 6.3 mmol) in dichloromethane (26.0 mL) was added trifluoroacetic acid (10.4 mL) at ice temperature and the reaction mixture was stirred at room temperature over a period of 2 h. Reaction was monitored by TLC. After complete consumption of starting material, volatiles were removed under reduced pressure to obtain product as brow red oil. The crude product obtained was washed with ether (3X50 mL) to yield 3 as a pale yellow thick liquid (2.9 g crude). 1 H NMR (400 MHz, DMSO-d 6 ) δ(ppm): 8.69 (bs, 2H), 8.09-8.08 (m, 1H), 7.81-7.78 (m, 1H), 7.38 (t, J=7.6 Hz, 1H), 7.08-6.97 (m, 4H), 6.45 (s, 1H), 4.37 (s, 2H), 3.27 (s, 3H), 3.15-3.09 (m, 4H), 2.60 (t, J=5.6 Hz, 2H), 2.45 (t, J=5.6 Hz, 2H). MS m/z (M+H): 311.3 [0529] Step 3: To a solution of 3 (2.9 g, 6.8 mmol) in dimethyl sulfoxide (30 mL, 10V) was added diisopropyl-ethyl-amine (3.5 mL, 0.0205 mol) and the carbamate product 5 of Step 4, Example 16 (1.7 g, 6.8 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 6 h. Reaction was monitored by TLC. The resulting reaction mixture was diluted with ethyl acetate (300 mL), washed with water (3X100 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of the volatiles were purified through silica gel (230-400) column (30% ethyl acetate in n-hexane) to give product 4 as a light yellow gummy solid (2.6 g, 86 %). 1 H NMR (400 MHz, DMSO-d6) δ(ppm): 8.08 (d, J=2.0 Hz, 1H), 7.81-7.78 (m, 1H), 7.33 (t, J=7.6 Hz, 1H), 7.25-7.21 (m, 2H), 7.09-6.93 (m, 7H), 6.83 (d, J=3.2 Hz, 1H), 6.34 (s, 1H), 4.37 (s, 2H), 3.38-3.28 (m, 4H), 3.27 (s, 3H), 2.71-2.69 (m, 1H), 2.36 (t, J=5.6 Hz, 2H), 2.25 (t, J=5.6 Hz, 2H), 1.85 (m, 1H), 1.13 (d, J=4.8 Hz, 1H), 1.04 (d, J=7.6 Hz, 1H). 13 C NMR: (100 MHz, DMSO-d6): δ162.60, 157.57, 153.87, 146.76, 142.00, 140.15, 139.65, 138.71, 129.52, 128.87, 128.10, 125.86, 125.35, 124.90, 123.12, 121.21, 118.96, 111.26, 70.58, 57.49, 44.94, 43.90, 35.63, 34.08, 28.92, 24.31 and 15.57. MS m/z (M+H): 470.3, HPLC Purity: 99.57%, Chiral HPLC: 99.60%. [0530] Example 33: Synthesis of 4-({3-[(5-methylpyrimidin-2-yl)oxy]phenyl} methylidene)-N-[(1R,2S)-2-phenylcyclopropyl]piperidine-1-car boxamide: [0532] Step 1 - To a solution of 2-chloro-5-methylpyrimidine 1 (33.0 g, 0.256 mol) in DMF (330 mL) were added 3-(hydroxymethyl)phenol (31.86 g, 0.256 mol) and cesium carbonate (100.36 g, 0.308 mol) at room temperature. The reaction mixture was stirred at 100 o C over a period of 5 h. Then the resulting mixture was allowed to cool to room temperature, diluted with dichloromethane (330 mL), washed with water (2X330 mL), 1N KOH solution (2X165mL), brine and the organic layer was dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (8% ethyl acetate in hexane) to give product 2 as a white solid (22.0 g, 40%). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 8.4 (s, 2H), 7.3 (t, J = 8.1 Hz, 1H), 7.16 (d, J = 7.5 Hz, 1H), 7.07 (s, 1H), 7.0 (d, J = 7.5 Hz, 1H), 5.26 (m, 1H), 4.49 (d, J = 5.4, 2H), 2.19 (s, 3H). MS m/z (M+H): 217.1 [0533] Step 2 - To a solution of {3-[(5-methylpyrimidin-2-yl)oxy]phenyl}methanol 2 (22.0 g, 0.101 mol) in dichloromethane (220 mL,) was added thionyl chloride (8.1 mL, 0.111 mol) dropwise while stirring reaction in an ice bath. After removal of ice-bath, the reaction mixture was allowed to stir at room temperature over a period of 1 h. The volatiles were evaporated under reduced pressure and diluted with ethyl acetate (220 mL). Organic layer was washed with saturated sodium bicarbonate solution (110 mL) and water (2X220 mL). Organic layer was dried over anhydrous sodium sulphate and concentrated to yield product 3 as white solid (21.5 g, 90%). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 8.45 (s, 2H), 7.4 (t, J = 8 Hz, 1H), 7.28 (d, J = 7.6 Hz, 1H), 7.2 (t, J = 2 Hz, 1H), 7.13-7.10 (m, 1H), 4.75 (s, 2H), 2.18 (s, 3H). MS m/z (M+H): 235.3 [0534] Step 3 - A solution of 2-[3-(chloromethyl)phenoxy]-5-methylpyrimidine 3 (21.0 g, 0.0894 mol) in triethyl phosphite (25.0mL, 0.143 mol) was heated at 130 o C over a period of 16 h. The reaction mixture was allowed to reach room temperature and the crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (86% ethyl acetate in hexane) to give product 4 as a pale green oil (21.6 g, 72%). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 8.44 (s, 2H), 7.3 (t, J = 8 Hz, 1H), 7.12 (d, J = 7.2 Hz, 1H), 7.02 (t, J = 8 Hz, 2H), 3.95-3.88 (m, 4H), 3.31-3.20 (m, 2H), 2.18 (s, 3H), 1.15-1.11 (m, 6H). MS m/z (M+H): 337.3 [0535] Step 4 - To a solution of diethyl ({3-[(5-methylpyrimidin-2-yl)oxy]phenyl} methyl)phosphonate 4 (21.0 g, 0.062 mol) in THF (147 mL) was added 15-crown-5 ether (0.275 g, 0.0012 mol) at room temperature. The reaction was cooled (ice bath) and 60% NaH (3.73 g, 0.093 mol) added portion wise. The reaction mixture was allowed to stir at room temperature for 30 min and again cooled to ice temperature. A solution of tert-butyl 4- oxopiperidine-1-carboxylate (12.4 g, 0.0624 mol) in THF (63 mL) was added at ice temperature and allowed to stir at room temperature over a period 16 h. The resulting reaction mixture was diluted with ethyl acetate (210 mL) and washed with water (3X210 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (15% ethyl acetate in hexane) to give product 5 as a white solid (17.0 g, 72%). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 8.44 (s, 2H), 7.34 (t, J = 8 Hz, 1H), 7.06 (d, J = 7.6 Hz, 1H), 6.99-6.97 (m, 2H), 6.34 (s, 1H), 3.38 (t, J = 5.6 Hz, 2H), 2.37 (t, J = 5.6 Hz, 2H), 2.24 (t, J = 5.6 Hz, 2H), 2.18 (s, 3H), 1.38 (s, 9H). MS m/z (M+H): 382.3 [0536] Step 5 - To a solution tert-butyl 4-({3-[(5-methylpyrimidin-2-yl)oxy]phenyl} methylidene)piperidine-1-carboxylate 5 (17.0 g, 0.0445 mol) in dichloromethane (170 mL) was added trifluoroacetic acid (68.0 mL) at ice temperature and the reaction mixture was stirred at room temperature over a period of 1 h. The resulting mixture was evaporated of volatiles under reduced pressure to obtain crude product as yellow oil. The crude product obtained was washed with diethyl ether (3X50 mL) to give product 6 as an off white solid (15.8 g, Crude). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 8.83 (bs, 2H), 8.45 (s, 2H), 7.37 (t, J = 8.4 Hz, 1H), 7.08 (d, J = 7.6 Hz, 1H), 7.02 (m, 2H), 6.43 (s, 1H), 3.11 (d, J = 24 Hz, 4H), 2.59 (t, J = 5.6 Hz, 2H), 2.18 (s, 3H). MS m/z (M+H): 282.33 [0537] Step 6 - To a solution of trifluoroacetic acid salt of 5-methyl-2-{3-[(piperidin-4- ylidene)methyl]phenoxy}pyrimidine 6 (15.0 g, 0.0379 mol) in dimethyl sulfoxide (150 mL) were added diisopropylethylamine (20.0 mL, 0.113 mol) and the carbamate product 5 of Step 4, Example 16 (10.57 g, 0.0417 mol) at 25-30 °C . The reaction mixture was allowed to stir at 60 °C over a period of 5 h. The resulting reaction mixture was diluted with ethyl acetate (150 mL), washed with water (3X150 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (70% ethyl acetate in hexane) to give product 7 as a pale yellow solid (12.0 g, 72%). Melting range: 58° C – 67.5° C. 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 8.4 (s, 2H), 7.3 (t, J = 7.6 Hz, 1H), 7.23-7.19 (m, 2H), 7.12-7.06 (m, 4H), 6.9 (d, J = 8 Hz, 2H), 6.8 (s, 1H), 6.3 (s, 1H), 3.36-3.29 (m, 4H), 2.6 (s, 2H), 2.3 (s, 2H), 2.2-2.1(m, 5H), 1.87-1.84 (m, 1H), 1.16-1.06 (m, 2H). MS m/z (M+H): 441.4, HPLC purity: 99.67% [0538] Example 34 : Synthesis of N-[(2S)-2-phenylcyclopropyl]-4-{[3-(pyrazin-2- yloxy)phenyl] methylidene}piperidine-1-carboxamide:

[0540] Step 1: To a solution of 2-chloropyrazine 1 (15.0 g, 0.130 mol) in DMF (150mL) were added 3-(hydroxymethyl)phenol (16.25 g, 0.130 mol) and cesium carbonate (51.2 g, 0.157 mol) at room temperature. The reaction mixture was stirred at 100 o C over a period of 5 h. Then the resulting mixture was allowed to reach room temperature, diluted with water (250 mL), extracted with ethyl acetate (3X500 mL) and the resulting organic layer was washed with 1N KOH (2X250 mL) solution and the separated organic layer was dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230- 400) column (10% ethyl acetate in hexane) to give product 2 as an off white solid (9.3 g, 35%). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 8.50 (d, J = 1.2 Hz, 1H), 8.34 (d, J = 2.4 Hz, 1H), 8.18-8.17 (m, 1H), 7.36 (t, J = 7.6 Hz, 1H), 7.17 (d, J = 7.6 Hz, 1H), 7.1 (s, 1H),7.03 (dd, J = 6 Hz, J = 2 Hz, 1H), 5.25 (t, J = 5.6 Hz, 1H), 4.49 (d, J = 6 Hz, 2H). MS m/z (M+1): 203.2 [0541] Step 2: To a solution of {3-[(pyrazin-2-yl)oxy]phenyl}methanol 2 (10.0 g, 0.049 mol) in dichloromethane (100mL, 10V) was added thionyl chloride (3.94 mL, 0.054 mol) dropwise while stirring reaction in an ice bath. After removal of ice-bath, the reaction mixture was allowed to stir at room temperature over a period of 1h. The volatiles were evaporated under reduced pressure and diluted with ethyl acetate (250 mL). Organic layer was washed with saturated sodium bicarbonate solution (150 mL) and water (2X200 mL). Organic layer was dried over anhydrous sodium sulphate and concentrated to obtain the product 3 as yellow solid (9.0 g, 82%). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 8.54-8.54 (m, 1H), 8.36 (d, J = 2.8, 1H), 8.19-8.18 (m, 1H), 7.43 (t, J = 7.6, 1H), 7.31-7.14 (m, 3H), 4.75 (s, 2H). MS m/z (M+1): 221.2. [0542] Step 3: A solution of 2-[3-(chloromethyl)phenoxy]pyrazine 3 (10.0 g, 0.045 mol) in triethyl phosphite (12.3 mL, 0.072 mol) was heated at 130 o C over a period of 6 h. The reaction mixture was allowed to reach room temperature and the crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (60% ethyl acetate in hexane) to give product 4 as colorless oil (9.49 g 64.5%). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 8.51 (d, J = 1.6 Hz, 1H), 8.35 (d, J = 2.8 Hz, 1H), 8.18-8.17 (m, 1H), 7.35 (t, J = 7.6 Hz, 1H), 7.14(d, J = 7.6 Hz, 1H), 7.08-7.05 (m, 2H), 3.95-3.88 (m, 4H), 3.26 (2s, 2H), 1.13 (t, J = 3.2 Hz, 6H). MS m/z (M+1): 323.2 [0543] Step 4: To a solution of diethyl {[3-(pyrazin-2-yloxy)phenyl]methyl} phosphonate 4 (10.0 g, 0.031 mol) in THF (70 mL) was added 15-crown-5 ether (136 mg, 0.62 mmol) at room temperature. To the above reaction mixture was added 60% NaH (1.86 g, 0.046 mol) portion wise at 0-5 °C. The reaction mixture was allowed to stir at room temperature for 30 min and again cooled to ice temperature. A solution of tert-butyl 4-oxopiperidine-1-carboxylate (6.18 g, 0.031 mol) in THF (30 mL) was added at ice temperature and allowed to stir at room temperature over a period 16 h. The resulting reaction mixture was diluted with water (500 mL), extracted with ethyl acetate (3X500 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (15% ethyl acetate in hexane) to give product 5 as an off white solid (9.1 g, 80%). 1 H NMR (300 MHz, DMSO-d6) δ (ppm): 8.51 (d, J = 1.2 Hz, 1H), 8.34 (d, J = 2.4 Hz, 1H), 8.18- 8.17 (m, 1H), 7.37 (t, J = 7.6 Hz, 1H), 7.04-7.02 (m, 3H), 6.34 (s, 1H), 3.37 (t, J = 5.2 Hz, 2H), 3.32-3.29 (m, 2H), 2.37 (t, J = 5.6 Hz, 2H), 2.24 (t, J = 5.6 Hz, 2H), 1.37 (s, 9H). MS m/z (M+1): 368.4 [0544] Step 5: To a solution of tert-butyl 4-{[3-(pyrazin-2-yloxy)phenyl]methylidene} piperidine-1-carboxylate 5 (10.0 g, 0.027 mol) in dichloromethane (100 mL) was added trifluoroacetic acid (40 mL) at ice temperature and the reaction mixture was stirred at room temperature over a period of 1 h. The resulting mixture was evaporated of volatiles under reduced pressure to obtain crude product as red oil. The crude product obtained was washed with ether (3X50mL) to yield 6 as an off white solid (9.5 g, 92%). 1 H NMR (300 MHz, DMSO- d6) δ (ppm): 8.72 (bs, 2H), 8.52 (d, J = 1.6 Hz, 1H), 8.35 (d, J = 2.8 Hz, 1H), 8.18-8.17 (m, 1H), 7.42-7.38 (m, 1H), 7.12-7.06 (m, 3H), 6.44 (s, 1H), 3.13-3.07 (m, 4H), 2.58 (t, J = 5.6 Hz, 2H). MS m/z (M+1): 268.3 [0545] Step 6: To a solution of trifluoroacetic acid salt of 2-[3-(piperidin-4-ylidenemethyl) phenoxy]pyrazine 6 (10.0 g, 0.026 mol) in dimethyl sulfoxide (100mL, 10V) were added diisopropylethylamine (13.7 mL, 0.078 mol) and the carbamate product 5 of Step 4, Example 167.3 g, 0.028 mol) at 25-30 °C. The reaction mixture was allowed to stir at 60 °C over a period of 5 h. The resulting reaction mixture was diluted with ethyl acetate (500mL), washed with water (3X150 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (50% ethyl acetate in hexane) to give product 7 as a pale yellow solid (8.0 g, 71%). Melting range: 41.7° C – 52.6° C; 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 8.52 (s, 1H), 8.34 (d, J = 2.4 Hz, 1H), 8.18 (d, J = 1.2 Hz, 1H), 7.37 (t, J = 7.6 Hz, 1H), 7.21 (t, J = 7.6 Hz, 2H), 7.12-7.01 (m, 6H), 6.83 (d, J = 2.4 Hz, 1H), 6.33 (s, 1H) 3.37-3.27 (m, 4H), 2.67 (d, J = 3.2 Hz, 1H), 2.35 (s, 1H), 2.23 (s, 1H), 1.87-1.82 (m, 1H), 1.16-1.01 (m, 2H). MS m/z (M+1): 427.4. HPLC purity: 99.49% [0546] Example 35: Synthesis of 4-[3-(5-hydroxy-pyridin-2-yloxy)-benzylidene]- piperidine-1-carboxylic acid ((1R,2S)-2-phenyl-cyclopropyl)-amide: [0548] Step 1: To a solution of 5-bromo-2-fluoropyridine 1 (7.72 g, 44.3 mmol) in DMSO (40 mL, 8V) was added 3-hydroxymethyl phenol (5 g, 40.3 mmol) and cesium carbonate (15.75 g, 48.3 mmol) at room temperature. The reaction mixture was stirred at 100 o C for 8 h. Then the resulting mixture was allowed to reach room temperature, diluted with water (70 mL), extracted with ethyl acetate (3x100 mL). The combined organic layer was dried over sodium sulphate and the solvent evaporated using a rotovap under reduced pressure. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (30% ethyl acetate in n-hexane) to give product 2 as a pale yellow oil (7.1 g , 63% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 8.19 (dd, J = 2.6, 0.6 Hz, 1H), 7.77 (dd, J = 8.7, 2.6 Hz, 1H), 7.38 (t, J = 7.8 Hz, 1H), 7.20 (ddd, J = 7.6, 1.7, 0.9 Hz, 1H), 7.13 (d, J = 2.1 Hz, 1H), 7.03 (ddd, J = 8.1, 2.5, 1.0 Hz, 1H), 6.84 (dd, J = 8.7, 0.7 Hz, 1H), 4.68 (s, 2H). MS m/z (M): 280.21 [0549] Step 2: To a solution of {3-[(5-bromopyridin-2-yl)oxy]phenyl}methanol 2 (5.2 g, 18.57 mmol) in dichloromethane (52 mL) was added thionyl chloride (2.43 g, 20.4 mmol) dropwise while stirring reaction in an ice bath. After removal of ice-bath, the reaction mixture was allowed to stir at room temperature over a period of 1 h. The volatiles were evaporated under reduced pressure and diluted with ethyl acetate (80 mL). Organic layer was washed with saturated sodium bicarbonate solution (25 mL) and water (25 mL). Organic layer was dried over anhydrous sodium sulphate and concentrated to obtain the product 3 as white solid (4.12 g, 75% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 8.22 (dd, J = 2.6, 0.7 Hz, 1H), 7.79 (dd, J = 8.7, 2.6 Hz, 1H), 7.40 (t, J = 7.9 Hz, 1H), 7.29 – 7.21 (m, 2H), 7.17 (t, J = 2.1 Hz, 1H), 7.08 (m, 1H), 6.86 (dd, J = 8.8, 0.6 Hz, 1H), 4.59 (s, 2H). MS m/z (M+2): 299.9 [0550] Step 3: A solution of 5-bromo-2-[3-(chloromethyl)phenoxy]pyridine 3 (4 g, 13.5 mmol) in triethyl phosphite (5.78 mL, 33.7 mmol) was heated at 150 o C over a period of 6 h. The reaction mixture was allowed to reach room temperature and the crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (60% ethyl acetate in petroleum ether) to give product 4 as a colorless oil (3.88 g, 72% yield ).1H NMR (400 MHz, CDCl3) δ 8.20 (dd, J = 2.6, 0.7 Hz, 1H), 7.76 (dd, J = 8.7, 2.6 Hz, 1H), 7.40 – 7.29 (m, 1H), 7.16 (dt, J = 7.3, 1.7 Hz, 1H), 7.07 (q, J = 2.3 Hz, 1H), 7.02 (dtd, J = 8.1, 2.3, 1.0 Hz, 1H), 6.83 (dd, J = 8.7, 0.7 Hz, 1H), 4.03 (dqd, J = 8.7, 7.1, 1.6 Hz, 4H), 3.16 (d, J = 21.6 Hz, 2H), 1.25 (t, J = 7.1 Hz, 6H). MS m/z (M+1): 401 [0551] Step 4: To a solution of diethyl ({3-[(5-bromopyridin-2-yl)oxy]phenyl} methyl)phosphonate 4 (3.8 g, 9.52 mmol) in THF (19 mL) was added 15-crown ether (41 mg, 0.190 mmol). The reaction was cooled (ice bath) and 60% NaH (342 mg, 14.2 mol) was added portion wise. The reaction mixture was allowed to stir at room temperature for 30 min and again cooled to ice temperature. A solution of 4-oxo-piperidine-1-carboxylic acid tert-butyl ester (1.9 g, 9.52 mmol) in THF (19 mL) was added at ice temperature and allowed to stir at room temperature over a period 16 h. The resulting reaction mixture was diluted with water (50mL), extracted with ethyl acetate (3X40 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (5% ethyl acetate in n-hexane) to give product 5 as a white solid (2.58 g, 61 % yield). 1 H NMR (400 MHz, CDCl 3 ) δ 8.22 (dd, J = 2.6, 0.7 Hz, 1H), 7.77 (dd, J = 8.7, 2.6 Hz, 1H), 7.35 (t, J = 7.8 Hz, 1H), 7.04 (ddt, J = 7.7, 1.7, 0.8 Hz, 1H), 7.01 – 6.92 (m, 2H), 6.84 (dd, J = 8.7, 0.7 Hz, 1H), 6.34 (s, 1H), 3.45 (m, 4H), 2.39 (m, 4H), 1.47 (s, 9H). MS m/z (M+Na): 469.21 [0552] Step 5: The experiment was carried using methodology reported in literature (reference: J. Am. Chem. Soc. 2016, 138, 13493−13496). To a degassed solution of 4-[3-(5- bromo-pyridin-2-yloxy)-benzylidene]-piperidine-1-carboxylic acid tert-buty ester 5 (2 g, 4.49 mmol) in DMSO (40 mL) and water (2 mL), LiOH.H 2 O (396 mg, 9.43 mmol) and ligand, L-1 (110 mg, 0.337 mmol) and Cu(acac)2 (88 mg, 0.337 mmol) were added successively and continued degassing for 10 min. The resulting reaction mixture was heated at 85 °C for 48 h. Progress of reaction was monitored using TLC. The reaction mixture was cooled to ambient temperature, quenched by adding 5 % aq NH4Cl (12 mL) and then diluted with ethyl acetate (25 mL). Organic layer was separated, washed with brine (15 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to get crude product. The crude product obtained was further purified by silica gel flash chromatography using 40- 45 % ethyl acetate in hexanes to afford 4-[3-(5-hydroxy-pyridin-2-yloxy)-benzylidene]-piperidine-1- carboxylic acid tert-butyl ester (6) as a pale brown solid (738 mg, 43% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 9.69 (bs, 1H), 7.83 (s, 1H), 7.28 (d, J = 7.8 Hz, 1H), 7.25 (d, J = 4.9 Hz, 1H), 6.92 (d, J = 7.8 Hz, 2H), 6.81 (d, J = 8.7 Hz, 2H), 6.30 (s, 1H), 3.48 (t, J = 5.7 Hz, 2H), 3.35 (t, J = 5.9 Hz, 2H), 2.41 (t, J = 5.9 Hz, 2H), 2.29 (t, J = 5.8 Hz, 2H), 1.47 (s, 9H). MS m/z (M+Na): 405.44 [0553] Step 6: To a solution of 6 ( 600 mg, 1.57 mmol) in dichloromethane (6 mL) was added trifluoroacetic acid (2.4 mL) at ice temperature and the reaction mixture was stirred at room temperature over a period of 1 h. Reaction was monitored by TLC. After complete consumption of starting material, volatiles were removed under reduced pressure to obtain crude product 7 as a red oil. The crude product obtained was washed with ether (3 x 5 mL) to yield trifluoroacetic acid salt of 6-{3-[(piperidin-4-ylidene)methyl]phenoxy}pyridin-3-ol as a brown oil (400 mg crude). MS m/z (M+1): 283.21 [0554] Step 7: To a solution of 7 (300 mg, 1.06 mmol) in dimethyl sulfoxide (3 mL) was added diisopropylethylamine (1.48 mL, 8.51 mmol) and the carbamate product 5 of Step 4, Example 16 (269 mg, 1.06 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 4 h. Reaction was monitored by TLC. The resulting reaction mixture was diluted with ethyl acetate (20 mL), washed with water (3 x 20 mL) and dried over anhydrous sodium sulphate. The crude product obtained upon evaporation of volatiles was purified by reverse phase HPLC, collected fractions were concentrated, obtained residue was lyophilized to get product 8 as an off white solid (93 mg, 20 %). 1 H NMR (400 MHz, DMSO-d 6 ) δ 9.70 (s, 1H), 7.77 – 7.67 (m, 1H), 7.37 – 7.15 (m, 4H), 7.17 – 7.07 (m, 3H), 6.98 (d, J = 7.6 Hz, 1H), 6.93 – 6.79 (m, 4H), 6.33 (s, 1H), 3.37 (d, J = 6.0 Hz, 2H), 3.32 – 3.25 (m, 2H), 2.75 – 2.61 (m, 1H), 2.30 (dt, J = 42.8, 5.8 Hz, 4H), 1.88 (ddd, J = 9.4, 6.1, 3.2 Hz, 1H), 1.16 (dt, J = 9.6, 5.1 Hz, 1H), 1.09 – 1.02 (m, 1H).MS m/z (M+1): 442.4; HPLC purity: 96.70%; Chiral HPLC purity: 96.0% [0555] Example 36: Synthesis of 4-[3-(4-hydroxy-pyridin-2-yloxy)-benzylidene]- piperidine-1-carboxylic acid ((1R,2S)-2-phenyl-cyclopropyl)-amide:

[0557] Step 1: To a solution of 4-bromo-2-fluoropyridine 1 (7.72 g, 44.3 mmol) in DMSO (40 mL) was added 3-hydroxymethyl phenol (5 g, 40.3 mmol) and cesium carbonate (15.75 g, 48.3 mmol) at room temperature. The reaction mixture was stirred at 100 o C over a period of 8 h. Then the resulting mixture was allowed to reach room temperature, diluted with water (70 mL), extracted with ethyl acetate (3 x100 mL) and the organic layer was dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (30% ethyl acetate in n-hexane) to give product 2 as a pale yellow oil (4.8 g, 43% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 8.00 (dd, J = 5.4, 1.8 Hz, 1H), 7.40 (td, J = 7.8, 1.8 Hz, 1H), 7.28 – 7.22 (m, 1H), 7.15 (dt, J = 5.3, 2.0 Hz, 2H), 7.11 (d, J = 1.8 Hz, 1H), 7.05 (dd, J = 8.0, 2.3 Hz, 1H), 4.72 (s, 2H). MS m/z (M+2): 281.9 [0558] Step 2: To a solution of {3-[(4-bromopyridin-2-yl)oxy]phenyl}methanol 2 (4.8 g 17.1 mmol) in dichloromethane (48 mL) was added thionyl chloride ( 2.24 g, 18.8 mmol) dropwise while stirring reaction in an ice bath. After removal of ice-bath, the reaction mixture was allowed to stir at room temperature over a period of 1 h. The volatiles were evaporated under reduced pressure and diluted with ethyl acetate (80 mL). Organic layer was washed with saturated sodium bicarbonate solution (25 mL) and water (25 mL). Organic layer was dried over anhydrous sodium sulphate and concentrated to obtain the product 3 as white solid (4.05 g, 80% yied). 1 H NMR (400 MHz, CDCl 3 ) δ 8.01 (d, J = 5.5 Hz, 1H), 7.40 (t, J = 7.9 Hz, 1H), 7.25 (q, J = 2.0, 1.3 Hz, 1H), 7.21 – 7.14 (m, 2H), 7.13 – 7.05 (m, 2H), 4.59 (s, 2H). MS m/z (M+2): 299.9 [0559] Step 3: A solution of 3 (4 g, 13.5 mmol) in triethyl phosphite (5.78 mL, 33.7 mmol) was heated at 150 o C over a period of 6 h. The reaction mixture was allowed to reach room temperature and the crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (60% ethyl acetate in petroleum ether) to give product 4 as colorless oil (3.88 g, 72% yield ). 1 H NMR (400 MHz, Chloroform-d) δ 8.00 (d, J = 5.4 Hz, 1H), 7.36 (m, 1H), 7.20 – 7.13 (m, 2H), 7.07 (t, J = 1.9 Hz, 2H), 7.03 (m, 1H), 4.03 (m, 4H), 3.16 (d, J = 21.7 Hz, 2H), 1.25 (t, J = 7.1 Hz, 6H). MS m/z (M+H): 401.20 [0560] Step 4: To a solution of 4 (3.8 g, 9.52 mmol) in THF (19 mL) was added 15-crown ether (41 mg, 0.190 mmol). The reaction was cooled (ice bath) and 60% NaH (342 mg, 14.2 mol) was added portion wise. The reaction mixture was allowed to stir at room temperature for 30 min and again cooled to ice temperature. A solution of 4-oxo-piperidine-1-carboxylic acid tert-butyl ester (1.9 g, 9.52 mmol) in THF (19 mL) was added at ice temperature and allowed to stir at room temperature over a period 16 h. The resulting reaction mixture was diluted with water (50 mL), extracted with ethyl acetate (3x40 mL) and dried over anhydrous sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230 - 400 mesh) column (5% ethyl acetate in n-hexane) to give product 5 as a white solid (2.75 g, 65 % yield). 1 H NMR (400 MHz, CDCl3) δ 8.02 (d, J = 5.4 Hz, 1H), 7.35 (t, J = 7.8 Hz, 1H), 7.15 (dd, J = 5.4, 1.6 Hz, 1H), 7.10 (d, J = 1.6 Hz, 1H), 7.05 (d, J = 7.7 Hz, 1H), 7.00 – 6.93 (m, 2H), 6.34 (s, 1H), 3.45 (dt, J = 40.2, 5.9 Hz, 5H), 2.50 – 2.27 (m, 4H), 1.47 (s, 9H).MS m/z (M+Na): 467.6 [0561] Step 5: To a degassed solution of 5 (2 g, 4.49 mmol) in DMSO (40 mL) and water (2 mL), LiOH.H 2 O (396 mg, 9.43 mmol) and ligand, L-1 (110 mg, 0.337 mmol) and Cu(acac) 2 (88 mg, 0.337 mmol) were added successively and continued degassing for 10 min. The resulting reaction mixture was heated at 85 °C for 48 h. Progress of reaction was monitored using TLC. The reaction mixture from above was cooled ambient temperature, quenched by adding 5 % aq NH4Cl (12 mL) and then diluted with ethyl acetate (25 mL). Organic was separated, washed with brine (15 mL), dried over Na2SO4 and concentrated under reduced pressure to get crude product. The crude product obtained was further purified by silica gel flash chromatography using 40-45% ethyl acetate in hexane to afford 4-[3-(5-hydroxy-pyridin- 2-yloxy)-benzylidene]-piperidine-1-carboxylic acid tert-butyl ester (6) as a pale brown solid (686 mg, 40% yield). 1 H NMR (400 MHz, Chloroform-d) δ 8.02 (d, J = 5.4 Hz, 1H), 7.35 (t, J = 7.8 Hz, 1H), 7.15 (dd, J = 5.4, 1.6 Hz, 1H), 7.10 (d, J = 1.6 Hz, 1H), 7.05 (d, J = 7.7 Hz, 1H), 7.00 – 6.93 (m, 2H), 6.34 (s, 1H), 3.45 (dt, J = 40.2, 5.9 Hz, 5H), 2.50 – 2.27 (m, 4H), 1.47 (s, 9H). MS m/z (M+1): 383.2 [0562] Step 6: To a solution of 4-[3-(4-hydroxy-pyridin-2-yloxy)-benzylidene]-piperidine- 1-carboxylic acid tert-butyl ester 6 (400 mg, 1.04 mmol) in dichloromethane (4 mL) was added trifluoroacetic acid (1.2 mL) at ice temperature and the reaction mixture was stirred at room temperature over a period of 1h. Reaction was monitored by TLC. After complete consumption of starting material, volatiles were removed under reduced pressure to obtain product 7 as a red oil. The crude product obtained was washed with ether (3 x 50 mL) to yield trifluoroacetic acid salt of 6-{3-[(piperidin-4-ylidene)methyl]phenoxy}pyridin-3-ol as a brown oil (400 mg Crude). MS m/z (M+1): 283.2 [0563] Step 7: To a solution of trifluoroacetic acid salt of 6-{3-[(piperidin-4- ylidene)methyl]phenoxy}pyridin-3-ol 7 (280 mg, 0.992 mmol) in dimethyl sulfoxide (2.8 mL, 10V) was added diisopropylethylamine (1.38 mL, 7.94 mmol) and the carbamate product 5 of Step 4, Example 16 (251 mg, 0.992 mmol) at 25 °C. The reaction mixture was allowed to stir at 60 °C over a period of 4 h. Reaction was monitored by TLC. The resulting reaction mixture was diluted with ethyl acetate (20 mL), washed with water (3X20 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified by reverse phase HPLC to give product 8 as an off white solid (137 mg , 32 %). 1 H NMR (400 MHz, DMSO-d 6 ) δ 10.79 (s, 1H), 7.86 (d, J = 5.7 Hz, 1H), 7.35 (t, J = 7.9 Hz, 1H), 7.24 (tt, J = 7.9, 1.3 Hz, 2H), 7.15 – 7.08 (m, 3H), 7.04 (d, J = 7.7 Hz, 1H), 6.95 – 6.88 (m, 2H), 6.83 (d, J = 3.2 Hz, 1H), 6.55 (dd, J = 5.7, 2.1 Hz, 1H), 6.35 (s, 1H), 6.28 (d, J = 2.0 Hz, 1H), 3.38 (t, J = 5.8 Hz, 2H), 3.32 (m, 2H), 2.70 (tt, J = 6.9, 3.6 Hz, 1H), 2.37 (t, J = 5.8 Hz, 2H), 2.29 – 2.22 (m, 2H), 1.88 (ddd, J = 9.4, 6.1, 3.2 Hz, 1H), 1.16 (dt, J = 9.6, 5.1 Hz, 1H), 1.07 (dt, J = 7.6, 5.8 Hz, 1H). MS m/z (M+H): 442.19, HPLC purity: 98.50%; Chiral HPLC purity: 97.9% [0564] Example 37: Synthesis of 4-[3-(6-hydroxy-pyridin-2-yloxy)-benzylidene]- piperidine-1-carboxylic acid ((1R,2S)-2-phenyl-cyclopropyl)-amide:

[0566] Step 1: To a solution of 2-bromo-6-fluoropyridine 1 (2.5 g, 14.2 mmol) in DMF (25 mL, 10V) was added 3-hydroxymethyl phenol (2.64 g, 21.3 mmol) and cesium carbonate (5.55 g, 17.0 mmol) at room temperature. The reaction mixture was stirred at 100 o C over a period of 8 h. Then the resulting mixture was allowed to reach room temperature, diluted with water (50 mL), extracted with ethyl acetate (3 x 50 mL) and the organic layer was dried over anhydrous sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (30% ethyl acetate in n-hexanes) to give product 2 as a pale yellow oil (2.48 g , 62% yield). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 7.79 (t, J = 7.9 Hz, 1H), 7.39 (t, J = 7.9 Hz, 2H), 7.19 (dt, J = 7.7, 1.2 Hz, 1H), 7.09 (t, J = 1.9 Hz, 1H), 7.04 – 6.97 (m, 2H), 5.29 (t, J = 5.8 Hz, 1H), 4.52 (d, J = 5.8 Hz, 2H). MS m/z (M): 280. [0567] Step 2: To a solution of {3-[(6-bromopyridin-2-yl)oxy]phenyl}methanol 2 (2.48 g, 8.85 mmol) in dichloromethane (25 mL) was added thionyl chloride (0.7 mL, 9.7 mmol) drop wise while stirring reaction in an ice bath. After removal of ice-bath, the reaction mixture was allowed to stir at room temperature over a period of 1 h. The volatiles were evaporated under reduced pressure and diluted with ethyl acetate (60 mL). Organic layer was washed with saturated sodium bicarbonate solution (25 mL) and water (25 mL). Organic layer was dried over anhydrous sodium sulphate and concentrated to obtain the product 3 as brown oil (2.56 g, 86%). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 7.86 – 7.76 (m, 1H), 7.50 – 7.36 (m, 2H), 7.33 (dt, J = 7.7, 1.3 Hz, 1H), 7.25 (t, J = 2.1 Hz, 1H), 7.161– 7.071 (m, J = 8.1, 2.4, 1.1 Hz, 1H), 7.06 (d, J = 8.1 Hz, 1H), 4.79 (s, 2H). MS m/z (M): 298.19 [0568] Step 3: A solution of 2-bromo-6-[3-(chloromethyl)phenoxy]pyridine 3 (2.55 g, 8.50 mmol) in triethyl phosphite (3.67 mL, 21.0 mmol) was heated at 150 o C over a period of 6 h. The reaction mixture was allowed to reach room temperature and the crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (60% ethyl acetate in petroleum ether) to give product 4 as pale brown oil (2.97 g, crude). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 7.88 – 7.72 (m, 1H), 7.52 – 6.96 (m, 6H), 4.011– 3.90 (m, J = 14.7, 7.3, 4.0 Hz, 4H), 1.16 (tt, J = 7.7, 5.3 Hz, 6H). MS m/z (M+Na): 422.5 [0569] Step 4: To a solution of diethyl ({3-[(6-bromopyridin-2-yl)oxy]phenyl}methyl) phosphonate 4 (2.96 g, 7.39 mmol) in THF (20 mL) was added 15-crown ether (32 mg, 0.147 mmol). The reaction was cooled (ice bath) and 60% NaH (266 mg, 11.0 mmol) was added portion wise. The reaction mixture was allowed to stir at room temperature for 30 min and again cooled to ice temperature. A solution of 4-oxo-piperidine-1-carboxylic acid tert-butyl ester (1.76 g, 8.86 mmol) in THF (15 mL) was added at ice temperature and allowed to stir at room temperature over a period 16 h. The resulting reaction mixture was diluted with water (50 mL), extracted with ethyl acetate (3X40 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified through silica gel (230-400) column (5% ethyl acetate in n-hexanes) to give product 5 as a yellow oil (2.11 g, 65% yield). 1 H NMR (400 MHz, DMSO) δ (ppm): 7.84 – 7.77 (m, 1H), 7.44 – 7.35 (m, 2H), 7.14 – 6.99 (m, 4H), 6.39 (s, 1H), 3.47 – 3.36 (m, 3H), 3.35 (s, 2H), 2.42 (t, J = 5.8 Hz, 2H), 2.30 – 2.24 (m, 2H), 1.40 (s, 9H). MS m/z (M+Na): 467.6 [0570] Step 5: To a degassed solution of 4-[3-(6-bromo-pyridin-2-yloxy)-benzylidene]- piperidine-1-carboxylic acid tert-butyl ester 5 (1.8 g, 4.04 mmol) in DMSO (36 mL, 20 V) and water (1.8 mL,), LiOH.H 2 O (356 mg, 8.49 mmol) and ligand, L-1 (99.6 mg, 0.303 mmol) and Cu(acac)2 (79 mg, 0.303 mmol) were successively added and continued degassing with for 10 min. The resulting reaction mixture was heated at 100 °C for 4 h under MW radiation. Progress of reaction was monitored using TLC. The reaction mixture from above was cooled ambient temperature, quenched by adding 5 % aq NH4Cl (12 mL) and then diluted with ethyl acetate (25 mL). Organic was separated, washed with brine (15 mL), dried over Na2SO4 and concentrated under reduced pressure to get crude product. The crude product obtained was further purified by silica gel flash chromatography using 40- 45 % Ethyl acetate in hexane to afford 4-[3-(6-hydroxy-pyridin-2-yloxy)-benzylidene]-piperidine-1-c arboxylic acid tert-butyl ester (6) as pale brown solid (730 mg, 47% yield). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 10.80 (bs, 1H), 7.64 (t, J =8.0 Hz, 1H), 7.53 (t, J =8.0 Hz, 1H), 7.03 (d, J =7.6 Hz, 1H), 6.96 – 6.90 (m, 2 H), 6.36 – 6.33 (m, 3H), 3.40 (t, J = 5.2 Hz, 1H), 3.38 – 3.31(m, 2H), 2.39 (t, J = 5.6 Hz, 2H), 2.27 (t, J = 5.6 Hz, 2H), 1.45 (s, 9H), MS m/z (M+Na): 405.17 [0571] Step 6: To a solution of 4-[3-(6-hydroxy-pyridin-2-yloxy)-benzylidene]-piperidine- 1-carboxylic acid tert-butyl ester 6 (720 mg, 1.88 mmol) in dichloromethane (5.6 mL) was added trifluoroacetic acid (1.44 mL) at ice temperature and the reaction mixture was stirred at room temperature over a period of 1h. Reaction was monitored by TLC. After complete consumption of starting material, volatiles were removed under reduced pressure to obtain product as red oil. The crude product obtained was washed with ether (3X5 mL) to yield trifluoroacetic acid salt of 6-{3-[(piperidin-4-ylidene)methyl]phenoxy}pyridin-2-ol 7 as a brown oil (750 mg Crude). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 8.64 (s, 2H), 7.65 (t, J = 7.9 Hz, 1H), 7.38 (t, J = 7.9 Hz, 1H), 7.29 – 6.89 (m, 5H), 6.46 (s, 1H), 6.35 (dd, J = 7.8, 3.7 Hz, 2H), 3.36-3.08 (m, 4H), 2.60 (t, J = 6.0 Hz, 2H, 2.39 (t, J = 6.0 Hz, 2H).MS m/z (M+H): 283.33 [0572] Step 7: To a solution of trifluoroacetic acid salt of 2-bromo-6-{3-[(piperidin-4- ylidene) methyl] phenoxy}pyridine 7 (730 mg, 2.5 mmol) in dimethyl sulfoxide (7.3 mL) was added diisopropylethylamine (1.9 mL, 12.9 mmol) and the carbamate product 5 of Step 4, Example 16 (570 mg, 2.25 mmol) at 25 °C . The reaction mixture was allowed to stir at 60 °C over a period of 5 h. Reaction was monitored by TLC. The resulting reaction mixture was diluted with ethyl acetate (70 mL), washed with water (3x20 mL) and dried over sodium sulphate. The crude product obtained upon evaporation of volatiles was purified 2 times by reverse phase HPLC to give product 8 as an off white solid (120 mg, 11%). 1 H NMR (400 MHz, DMSO-d6) δ (ppm): 10.83 (s, 1H), 7.64 (t, J = 7.9 Hz, 1H), 7.36 (t, J = 7.9 Hz, 1H), 7.24 (dd, J = 8.2, 6.9 Hz, 2H), 7.17 – 7.00 (m, 4H), 6.99 – 6.81 (m, 3H), 6.35 (s, 3H), 3.38 (t, J = 5.7 Hz, 3H), 3.30 (t, J = 5.9 Hz, 2H), 2.70 (ddd, J = 7.9, 5.5, 2.4 Hz, 1H), 2.37 (t, J = 5.8 Hz, 2H), 2.26 (t, J = 5.7 Hz, 2H), 1.88 (ddd, J = 9.4, 6.1, 3.2 Hz, 1H), 1.16 (m, 1H), 1.07 (m, 1H).MS m/z (M+H): 442.7, HPLC purity: 98.48%; Chiral HPLC: 98.6% [0573] Example 38 - Soluble Epoxide Hydrolase (sEH) Inhibition Assay: [0574] The sEH enzyme inhibition assay was performed using a commercially available kit from Cayman Chemical Company of Ann Arbor, Michigan (Cayman Cat. No.10011671). The assay uses 3-phenyl-cyano(6-methoxy-2-naphthalenyl)-methyl ester-2-oxiraneacetic acid as the substrate for sEH. Hydrolysis of the substrate yields a highly fluorescent product that can be monitored at excitation and emission wavelengths of 330 and 465 nm, respectively. The assay mixture consists of 185-190 μL assay buffer, 5 μL of sEH enzyme in a 96 well plate. Compounds at different concentrations (in 5 μL DMSO) or DMSO (vehicle) alone were added and the reaction was initiated by the addition of 5 µl of substrate. The plate was incubated for 15 min at 25 ° C. Data analysis was performed and the percentage inhibition determined. [0575] Example 39 – Fatty Acid Amide Hydrolase (FAAH (SEQ ID NO: 3)) Inhibition Assay: [0576] The FAAH enzyme inhibition assay was performed using a commercially available kit from Cayman Chemical Company of Ann Arbor, Michigan (Cayman Cat. No. 10010183). The assay uses AMC arachidonoyl amide (7-amino-4-methyl-2H-1-benzopyran-2- one-5Z,8Z,11Z,14Z-eicosatetraenamide, Cayman Chemical Cat# 10005098) as a substrate for FAAH. FAAH hydrolyzes AMC arachidonoyl amide resulting in the release of the fluorescent product, 7-amino-4-methylcoumarin (AMC). The fluorophore can be easily analyzed using an excitation wavelength of 340-360 nm and an emission wavelength of 450-465 nm. Compounds at different concentrations (in DMSO) or DMSO (vehicle) alone were added. After FAAH addition, the plates were incubated at ambient temperature for 20 minutes. Assays were initiated by the rapid addition of AMC amide arachidonoyl amide and reactions were performed at ambient temperature for 60 minutes, during which time the liberation of AMC was monitored by the concomitant increase in fluorescence intensity (excitation wavelength of 340-360 nm and an emission wavelength of 450-465 nm). Fluorescence intensity measurements were performed in a kinetic manner and reaction rates were calculated from the linear portion of the reaction using linear regression analysis. [0577] % Initial Activity = (Inhibited fluorescence/100% Activity fluorescence)*100 [0578] % Inhibition = 100 – (% Initial Activity) [0579] Compounds that inhibit soluble epoxide hydrolase at concentration (IC50) of less than 10 µM are considered active sEH inhibitors. Inhibitory activity of compounds of Formula I for sEH and FAAH enzymes is given in Table 1, see FIG.1. The selectivity for sEH inhibition is determined by dividing inhibitory potency of FAAH vs sEH [IC50 FAAH/IC50, sEH] [0580] The efficacy of compounds of general Formula I in neurodegenerative diseases can be evaluated using animal models known in literature. [0581] Example 40 - The method (in vitro model of GBA1 (SEQ ID NO: 1) inhibition) described below can be used to evaluate potential of compounds of Formula 1 for neuroprotection and to block α-syn aggregation associated with synucleinopathies including in Gaucher’s disease, see FIG.2. [0582] In this in vitro model, dopaminergic neurons were isolated from rat midbrain and cultured as described in literature (FASEB J. 2008; 22(7):2488-97). To mimic synucleinopathies the cells were injured with α-syn protofibrils and with conduritol B epoxide (CBE, 20 µM), a covalent inhibitor of GBA1 (SEQ ID NO: 1). This model reproduces the essential neuropathological features of synucleinopathies (e.g. loss of dopaminergic neurons and neurite network as assessed by staining with tyrosine hydroxylase (TH), a marker for dopaminergic neurons in the nervous system). [0583] Treatment with test compound (Compound A) - On day 6 of culture, Compound A was dissolved into DMSO (100 %, stock solution) and then diluted with culture medium to get the needed concentration. Compound A was added to the culture for 1 h, before CBE and 2 hours before the α-syn application. [0584] Treatment with CBE: On day 6 of culture, conduritol B epoxide (CBE, 20 µM) was added to the culture for 1 h, before α-syn injury (to be confirmed/changed). [0585] Treatment with α-syn: On day 3, human α-syn was agitated for 3 days at 37 °C in order to generate protofibrils (Lin et al., Elife. 2020 Jun 26;9:e54726). On day 6, α-syn (250 nM) was directly applied on cells, in presence of CBE. The injury was renewed on day 8 for an additional 48 h still in presence of the test compound. [0586] Treatment of neurons with CBE and α-syn induced a significant decrease in neuronal survival and neurite network. Compound A treatment resulted in a large neuroprotective effect from 50 nM to 500 nM. At 500 nM concentration, Compound A fully prevented the loss of dopaminergic neurons, fully preserved the neurite network (as assessed by TH staining). [0587] Example 41 - The animal model of GBA1 (SEQ ID NO: 1) inhibition and synucleinopathy to evaluate potential of compounds of Formula 1 to treat diseases associated with synucleinopathy including Gaucher Disease. [0588] Aged mice (18 months) were used as model of neurodegenerative disease of ageing and Parkinson’s disease. Injury with alpha-synuclein (α-syn) oligomers/protofibrils (precisely characterized by western blot) were carried out by intra-nigral (substantia nigra, pars compacta, SNpc) injections. These animals were treated with CBE with or without Compound A or Compound B for 4 weeks. Chronic inhibition of GBA1 (SEQ ID NO: 1) (CBE) and α-syn protofibrils mediate toxicity that occurs via several intracellular mechanisms such as mitochondrial and endoplasmic reticulum (ER) stress and an impaired autophagy-lysosomal pathway (Callizot et al., Plos One. https://doi.org/10.1371/journal.pone.0215277). [0589] Assessment of synucleinopathies induced motor defects by bar test (FIG.3) - Three weeks post-surgery, motor coordination of the mice was assessed with the bar test. The principle of the test is based on the capacity of the mice to cross a horizontal bar connected to a platform (18 mm of diameter, 60 cm long). The test requires good coordination between front and hind limbs, and good balance. Mice are placed at one extremity of the bar and need to reach a platform located at the other side of the bar. Motor coordination was studied after 2 training sessions (one and two days) before the test. Sessions were recorded by a video camera system. The time to cross the bar, the number of steps and the number of failed strides were measured. [0590] As shown in FIG. 3, the time to reach platform on a bar test was significantly increased in mice treated with CBE and α-syn (no compound vs. control). Treatment with Compound A or Compound B normalized the behavior to control levels. [0591] Tissue collection and immunostaining - At the end of the experiment (on day 28 post-surgery), mice were deeply anesthetized, and perfused with cold PBS (3 minutes), and cold paraformaldehyde (PFA) 4% in PBS (3 minutes). Brains were dissected and further fixed in PFA 4%, overnight at 4 °C. After, brains were placed in 30% sucrose in tris-phosphate saline (TBS) solution at 4 °C. Coronal sections, including the SNpc, of 40 μm-thickness were cut using a freezing microtome (4 sections per mouse, each 100 mm apart). For immunostaining, free-floating sections were incubated in TBS with 0.25% bovine serum albumin, 0.3% Triton X-100 and 1% goat serum, for 1 hour at room temperature. This incubation will block unspecific binding sites and will permeabilize the tissues. Select number of sections per animal were processed and incubated for 24 hours at 4 °C or 2 hours at room temperature with selected antibodies: TH: Chicken polyclonal antibody anti-tyrosine hydroxylase (1/1000); α-syn: Rabbit polyclonal antibody anti-alpha synuclein (1/200); IBA1: Goat polyclonal antibody anti-IBA1 (1/200). These antibodies were revealed with Alexa Fluor 488 anti-rabbit IgG, Alexa Fluor 568 anti-chicken IgG, at the dilution 1/500, in TBS with 0.25% donkey serum albumin, 0.3% Triton X-100 and 1% goat serum. Images were acquired with a confocal laser-scanning microscopy. [0592] The following read-outs were assessed: Aggregation of α–syn in the SNpc (FIG.4) – Treatment with CBE and α-syn (no compound vs. control) significantly increased α–syn aggregation (Control vs. no compound). Treatment with sEH inhibitor Compound B significantly decreased α–syn aggregation. [0593] Number of TH positive cells in the SNpc (FIG.5) - α-syn/CBE induced synuclein aggregation was associated with a significant loss of TH (+) neurons in the SNpc, this loss was associated with strong neuroinflammation (Ib1+, FIG.6). [0594] Treatment with Compound A or Compound B reduced the neurodegeneration preventing the neuronal death. FIG. 5. In addition a significant reduction of the Iba1+ microglial cell was observed translating a positive effect of the molecule on the neuroinflammation. FIG.6. [0595] Treatment with Compound A or Compound B showed strong protection of dopaminergic neurons (survival and α-syn aggregation) and fully prevented the neuroinflammation. In addition a full prevention of the motor impairment was observed. [0596] Example 426 – PINK1 (SEQ ID NO: 2) -null/α-syn zebrafish model of synucleinopathies was used to evaluate potential of compounds of Formula 1 to treat diseases associated with PINK1 (SEQ ID NO: 2) (PTEN-induce putative kinase) mutations linked to familial Parkinson's disease. Mitochondrial quality control is important in neurological diseases, and mutations in PINK results in reduced autophagy and mitophagy - implicated in aging, age related diseases such as PD. PINK1 (SEQ ID NO: 2) mutations cause autosomal recessive PD and certain clinical features are more common in patients with PINK1 (SEQ ID NO: 2) mutations, e.g. the age of onset that in this case is around 30 years of age. [0597] The genetic method as per Solnica-Kreze et al., Development. 1996 Dec; 123:37- 46. PMID: 9007227 was employed to generate PINK-null Zebrafish. Adult zebrafish males were subjected to the ENU chemical mutagen at 5 mM to induce random mutagenesis and screened for cognitive and locomotor deficits. Genotyping was performed through fin clips and PINK1+/- mutants were identified. (Gene ID: 30214). The F1 mutants carrying the PINK1+/- mutation were inbred and the resulting F2 embryos (henceforth addressed as PINK1- /-) were employed for the study purpose. α-Synuclein was delivered to PINK-null Zebrafish larvae via intracranial microinjection. Synucleinopathies-induced behavioral changes were monitored 7-days after α-synuclein challenge by assessing swim velocity (bradykinesia – FIG. 7) and dementia (predator avoidance test – FIG.8). The total swim velocity was derived from the video recordings and data analysis containing the time and the distance travelled by the zebrafish. [0598] The PINK1-/- mutant fish exhibited significant (P<0.0001) reduction in swim velocity in comparison to the wild type (WT) affirming the presence of bradykinesia (FIG.7 - wild type vs no compound). The groups treated with sEH inhibitors (Compound A of Formula I) exhibited a significant increase in swim velocity, thereby implying a rescue of bradykinesia. [0599] In memory test - Wild type group exhibited spatial memory wherein 100% of wild type zebrafish avoided the predator when compared to the PINK1-/- α- syn model group. Treatment with Compound A restored the memory to the wild type zebrafish levels (FIG.8) [0600] Example 43 – MPTP induced Parkinsonism in zebrafish model was used to evaluate potential of compounds of Formula I to treat defects in motor activity and rescue neuronal damage. [0601] The study was carried following protocol published by Bretaud et al. Neurotoxicol Teratol. 2004 Nov-Dec; 26(6), 857-64. For MPTP (1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine) induced Parkinsonism, adult zebrafish were injected with 3 ul of 60 ug/g of MPTP as an intramuscular injection along the lateral muscles. The fish were sedated with ice cold water to restrict movement during injection. The injection was held at 40º to the injection site and the respective volume of MPTP delivered as intra muscular injection. The test compounds were dosed orally by mixing with the fish feed pellets. Fishes were conditioned to consume three pellets a day. The test Compound A was evaluated both in a preventive and a therapeutic mode (after 5-days of MPTP treatment). MPTP-induced behavioral changes and nerve damage were monitored by – i) assessing distance travelled to track locomotor activity, ii) peripheral nerve damage assessed by twitch response) and iii) brain neuronal damage by brain pathology. [0602] Locomotor Activity: The observation tank was divided into four zones by drawing three vertical lines at equal distances, at a length of 6.25 cm per zone. The number of lines that adult zebrafish crossed over a period of 5 minutes was counted. Fishes were allowed to acclimatize in measurement tanks for a period of 30 minutes before readings were taken. Measurements were separated by a period of 4 hours before either dosing or induction could be done. [0603] Twitch response was measured as described by Lisse TS et al (1). Fish was placed intact on a moist sponge foil until calm, and the distal tail fin was stimulated with an insect pin until twitching of the fish was observed. The number of stimuli until twitch was recorded. Twitch responses were recorded once every day in the evening spaced about 4-6 hours since dosing in the morning for eight days. [0604] Brain Pathology: Study fish samples were collected on day 8, zebrafish from respective groups were euthanized by rapid cooling just before the sample collection. Whole fish was used intact and were fixed using 10% NBF solution as primary fixative for 24 hrs followed by Davidson’s fixative for 12 hrs. Further, tissues were processed as per protocol mentioned in ZIRC (Zebrafish International Resource Center) manual. Trichloro acetic acid (TCA) at 5% was used as decalcifying agent followed be tissue dehydration using increasing concentration of ethanol and cleared using chloroform and xylene. Paraffin wax with ceresin was used to infiltrate tissues post clearing and the tissues were embedded in to histology mold. [0605] The MPTP treated animals exhibited significant (p<0.0001) reduction in locomotion activity that progressively worsened over duration of the treatment in comparison to the control group animals (FIG.9). The groups treated with sEH inhibitor (Compound A of Formula I) exhibited a rescue of MPTP-induced abnormal locomotion exhibiting behaviour similar to the control animal group. Treatment of Compound A 5-days after daily MPTP- challenge rescued the locomotor dysfunction of the animals as shown in FIG 9. [0606] Nerve damage (Twitch response) - Selective loss of fine cutaneous fibers and axons projecting along the bony rays within the distal fin regions resulted in a notable temporal change in touch response, which was demonstrated by the MPTP-treated animals requiring significantly more stimulations with an insect pin at the distal fin before a twitching response was evoked. Compound A treatment (given as preventive or therapeutic model) demonstrated a substantial rescue of the peripheral nerve damage induced by MPTP treatment (FIG.10). [0607] Dopamine Neuron -. MPTP-treated groups showed significant reduction in the number of TH+ cells. Compound A treated groups exhibited a dose dependent rescue activity by preventing the loss of TH+ cells (FIG 11). Compound A treated in therapeutic mode (day 5-treatment) also exhibited protection from neuronal degeneration. [0608] The therapeutically effective amount of a compound of Formula I may be administered in a single dose or a dose repeated one or several times after a certain time interval. As described earlier, a number of factors may be considered in dose selection by the attending medical practitioner, including but not limited to - the potency and duration of action of the compounds used; the nature and severity of the illness to be treated as well as the sex, age, weight, general health and individual responsiveness of the subject to be treated, and other relevant circumstances. Therapeutic dose for the human subject can be estimated based on the data from animal studies and considering factors such as body surface area, pharmacokinetics profle and related parameters. Allometric scaling and related methodologies can be used as described in literature [e.g.; J Basic Clin Pharm. March 2016-May 2016; 7(2): 27–31] to estimate human equivalent dose. [0609] The therapeutically effective compositions of the present disclosure for treatment of a neurodegenerative disease in a subject may include compounds of the Formula I administered at doses of about 0.5 mg/day to about 3000 mg/day. [0610] Mutations in GBA are associated with Parkinson’s disease and Gaucher’s disease as reported in the literature – The link between the GBA gene and parkinsonism. Lancet Neurol. 2012 Nov;11(11):986-98. doi: 10.1016/S1474-4422(12)70190-4. PMID: 23079555; PMCID: PMC4141416; GBA gene: Parkinson's disease and the Gaucher Disease, https://www.gaucherdisease.org/blog/gaucher-disease-and-park insons-what-to-know-now/; GBA Variants in Parkinson’s disease, Movement disorders, 35 (12), 2201- 2210, https://doi.org/10.1002/mds.28225 [0611] Mutations in human kinase PINK1 are associated with Parkinson's disease (PD) see e.;g; Structure of PINK1 and mechanisms of Parkinson's disease-associated mutations, https://doi.org/10.7554/eLife.29985; Parkinson's disease mutations in PINK1 result in decreased Complex I activity and deficient synaptic function. EMBO Mol Med. 2009 May;1(2):99-111. doi: 10.1002/emmm.200900006. PMID: 20049710; PMCID: PMC3378121. [0612] SEQUENCE LISTING XML INCORPORATION STATEMENT [0613] The herewith provided Sequence Listing XML is hereby incorporated by reference. The name of the XML file is NeuroPn 1.xml with a date of creation of 09/29/2023 and a size of 6 kilobytes. [0614] <110> NeuroPn Therapeutics Inc. [0615] <120> PIPERIDINE UREA DERIVATIVES FOR TREATMENT OF NEURODEGENERATIVE DISEASES [0616] <130> 0039486.0000020 [0617] <140> Unknown [0618] <141> *** [0619] <150> U.S. Application No. [0620] <151> 09/29/2023 [0621] <160> 1 [0622] <170> WIPO Sequence [0623] <210> 1 [0624] <211> 536 [0625] <212> PRT [0626] <213> Homo sapiens [0627] <221> CDS [0628] <222> 1-586 [0629] <400> 1 [0630] MEFSSPSREECPKPLSRVSIMAGSLTGLLLLQAVSWASGARPCIPKSFGYSS VVCVCNATYCDSFDPPTFPALGTFSRYESTRSGRRMELSMGPIQANHTGT GLLLTLQPEQKFQKVKGFGGAMTDAAALNILALSPPAQNLLLKSYFSEEG IGYNIIRVPMASCDFSIRTYTYADTPDDFQLHNFSLPEEDTKLKIPLIHRAL QLAQRPVSLLASPWTSPTWLKTNGAVNGKGSLKGQPGDIYHQTWARYF VKFLDAYAEHKLQFWAVTAENEPSAGLLSGYPFQCLGFTPEHQRDFIAR DLGPTLANSTHHNVRLLMLDDQRLLLPHWAKVVLTDPEAAKYVHGIAV HWYLDFLAPAKATLGETHRLFPNTMLFASEACVGSKFWEQSVRLGSWD RGMQYSHSIITNLLYHVVGWTDWNLALNPEGGPNWVRNFVDSPIIVDIT KDTFYKQPMFYHLGHFSKFIPEGSQRVGLVASQKNDLDAVALMHPDGS AVVVVLNRSSKDVPLTIKDPAVGFLETISPGYSIHTYLWRRQ [0631] <210> 2 [0632] <211> 581 [0633] <212> PRT [0634] <213> Homo sapiens [0635] <221> CDS [0636] <222> 1-581 [0637] <400> 2 [0638] MAVRQALGRGLQLGRALLLRFTGKPGRAYGLGRPGPAAGCVRGERPGW AAGPGAEPRRVGLGLPNRLRFFRQSVAGLAARLQRQFVVRAWGCAGPCGRAVFLA FGLGLGLIEEKQAESRRAVSACQEIQAIFTQKSKPGPDPLDTRRLQGFRLEEYLIGQSI GKGCSAAVYEATMPTLPQNLEVTKSTGLLPGRGPGTSAPGEGQERAPGAPAFPLAIK MMWNISAGSSSEAILNTMSQELVPASRVALAGEYGAVTYRKSKRGPKQLAPHPNIIR VLRAFTSSVPLLPGALVDYPDVLPSRLHPEGLGHGRTLFLVMKNYPCTLRQYLCVNT PSPRLAAMMLLQLLEGVDHLVQQGIAHRDLKSDNILVELDPDGCPWLVIADFGCCL ADESIGLQLPFSSWYVDRGGNGCLMAPEVSTARPGPRAVIDYSKADAWAVGAIAYEI FGLVNPFYGQGKAHLESRSYQEAQLPALPESVPPDVRQLVRALLQREASKRPSARVA ANVLHLSLWGEHILALKNLKLDKMVGWLLQQSAATLLANRLTEKCCVETKMKMLF LANLECETLCQAALLLCSWRAAL [0639] <210> 3 [0640] <211> 579 [0641] <212> PRT [0642] <213> Homo sapiens [0643] <221> CDS [0644] <222> 1-579 [0645] <400> 3 [0646] MVQYELWAALPGASGVALACCFVAAAVALRWSGRRTARGAVVRARQR QRAGLENMDRAAQRFRLQNPDLDSEALLALPLPQLVQKLHSRELAPEAVLFTYVGK AWEVNKGTNCVTSYLADCETQLSQAPRQGLLYGVPVSLKECFTYKGQDSTLGLSLN EGVPAECDSVVVHVLKLQGAVPFVHTNVPQSMFSYDCSNPLFGQTVNPWKSSKSPG GSSGGEGALIGSGGSPLGLGTDIGGSIRFPSSFCGICGLKPTGNRLSKSGLKGCVYGQE AVRLSVGPMARDVESLALCLRALLCEDMFRLDPTVPPLPFREEVYTSSQPLRVGYYE TDNYTMPSPAMRRAVLETKQSLEAAGHTLVPFLPSNIPHALETLSTGGLFSDGGHTFL QNFKGDFVDPCLGDLVSILKLPQWLKGLLAFLVKPLLPRLSAFLSNMKSRSAGKLWE LQHEIEVYRKTVIAQWRALDLDVVLTPMLAPALDLNAPGRATGAVSYTMLYNCLDF PAGVVPVTTVTAEDEAQMEHYRGYFGDIWDKMLQKGMKKSVGLPVAVQCVALPW QEELCLRFMREVERLMTPEKQSS