Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CALPAIN MODULATORS AND METHODS OF PRODUCTION AND USE THEREOF
Document Type and Number:
WIPO Patent Application WO/2017/156074
Kind Code:
A1
Abstract:
The present technology relates to compounds, kits, compositions, and methods useful for the treatment of fibrotic disease. In some aspects, the present technology provides for treatment of various diseases or disorders associated or mediated, at least in part, by calpains, such as CAPN1, CAPN2, and/or CAPN9. The present technology is generally applicable to compounds which inhibit myofibroblast differentiation.

Inventors:
BUCKMAN BRAD OWEN (US)
YUAN SHENDONG (US)
NICHOLAS JOHN BEAMOND
Application Number:
PCT/US2017/021288
Publication Date:
September 14, 2017
Filing Date:
March 08, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BLADE THERAPEUTICS INC (US)
International Classes:
C07D245/02; A61K38/12; A61P29/00; A61P35/00; A61P43/00; C07D273/08
Domestic Patent References:
WO2015179441A22015-11-26
WO2013166319A12013-11-07
WO2008048121A22008-04-24
Foreign References:
US20110021434A12011-01-27
US20150133368A12015-05-14
Other References:
DAMALANKA, V. C. ET AL.: "Oxadiazole-Based Cell Permeable Macrocyclic Transition State Inhibitors of Norovirus 3CL Protease", JOURNAL OF MEDICINAL CHEMISTRY, vol. 59, no. 5, 2016, pages 1899 - 1913, XP055415445
JONES, S. A. ET AL.: "The Preparation of Macrocyclic Calpain Inhibitors by Ring Closing Metathesis and Cross Metathesis", AUSTRALIAN JOURNAL OF CHEMISTRY, vol. 67, no. 8 - 9, 2014, pages 1257 - 1263, XP055415448
JONES, S. A. ET AL.: "A Template-Based Approach to Inhibitors of Calpain 2, 20S Proteasome, and HIV-1 Protease", CHEMMEDCHEM, vol. 8, no. 12, 2013, pages 1918 - 1921, XP055415449
MANDADAPU, S. R. ET AL.: "Macrocyclic inhibitors of 3C and 3C-like proteases of picornavirus, norovirus, and coronavirus", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 23, no. 13, 2013, pages 3709 - 3712, XP028564927
PEHERE, A.D. ET AL.: "Synthesis and Extended Activity of Triazole-Containing Macrocyclic Protease Inhibitors", CHEMISTRY - A EUROPEAN JOURNAL, vol. 19, no. 24, 2013, pages 7975 - 7981, XP055415454
MORTON, J. D. ET AL.: "A macrocyclic calpain inhibitor slows the development of inherited cortical cataracts in a sheep model", INVESTIGATIVE OPHTHALMOLOGY & VISUAL SCIENCE, vol. 54, no. 1, 2013, pages 389 - 395, XP055415459
PEHERE, A D. ET AL.: "New cylindrical peptide assemblies defined by extended parallel beta- sheets", ORGANIC & BIOMOLECULAR CHEMISTRY, vol. 11, no. 3, 2013, pages 425 - 429
CHEN, H. ET AL.: "New Tripeptide-Based Macrocyclic Calpain Inhibitors Formed by N- Alkylation of Histidine", CHEMISTRY & BIODIVERSITY, vol. 9, no. 11, 2012, pages 2473 - 2484, XP055415468
PEHERE, A. D. ET AL.: "New beta-strand templates constrained by Huisgen cycloaddition", ORGANIC LETTERS, vol. 14, no. 5, 2012, pages 1330 - 1333, XP055415470
STUART, B. G. ET AL.: "Molecular Modeling: A Search for a Calpain Inhibitor as a New Treatment for Cataractogenesis", JOURNAL OF MEDICINAL CHEMISTRY, vol. 54, no. 21, 2011, pages 7503 - 7522, XP055261342
JONES, M. A. ET AL.: "Efficient large-scale synthesis of CAT811, a potent calpain inhibitor of interest in the treatment of cataracts", AUSTRALIAN JOURNAL OF CHEMISTRY, vol. 62, no. 7, 2009, pages 671 - 675, XP055415502
ABELL, A. D. ET AL.: "Molecular modeling, synthesis, and biological evaluation of macrocyclic calpain inhibitors", ANGEWANDTE CHEMIE , INTERNATIONAL EDITION, vol. 48, no. 8, 2009, pages 1455 - 1458, XP055415505
WALKER, M. A. ET AL.: "General method for the synthesis of cyclic peptidomimetic compounds", TETRAHEDRON LETTERS, vol. 42, no. 34, 2001, pages 5801 - 5804, XP004295928
Attorney, Agent or Firm:
MALLON, Joseph J. (US)
Download PDF:
Claims:
WE CLAIM

1. A compound comprising Formula I:

Formula I where: n is from 1-12; each A is independently selected from the group consisting of: O and S;

each R4 is independently selected from the group consisting of: H and ; each R3and R3 is independently selected from the group consisting of: hydrogen, C Cg alkyl, substituted Ci-Cg alkyl, Cj-Cg alkenyl, substituted Ci-Cg alkenyl, Ci-Cg alkynyl, substituted Cj-Cg alkynyl, C3-C7 aiyl, substituted C3-C7 ai l, cyano, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3- C9 heteroaryl, substituted C3-C9 heteroaryl, Ci-Cg alkoxv, substituted Cj-Cg alkoxv, amino, substituted amino, alkyithio, substituted aikylthio, OH, OR2, Ni l -. NHR2, SR2, substituted suifonyi, and substituted sulfmyl, wherein any R1 substituent may be covalentiy bonded to or share another Rl substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system, each R2 is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-Cg alkyl, Cj -Cg alkenyl, substituted Cj-Cg alkenyl, Cj-Cg alkynyl, substituted Ci-Cg alkynyl, C3-C7 aiyl, substituted C3-C7 aryl, cyano, OH, Ci -Cg alkoxy, substituted Ci -Cg alkoxv, amino, substituted amino, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl , and any naturally or n on -naturally occurring amino acid side chain, wherein any R2 substituent may be covalentlv bonded to or share another R2 substituent to form a C3-C12 cyclic, heterocyclic, aiyl, heteroaryl, spirocyclic, or bieyclie ring system; each W is independently selected from the group consisting of: CH2, C(R2)2, CHR2, O, NH, NR2, C(=0), C(=S), S, S(=0), S(=0)2, -C-, -CH-, a C3-C12 spirocyclic group

R2— (R2)m ot ¾ >¾ where m is 1.-19, sulfonyl, substituted sulfonyl, sulfinyl, and substituted sulfinyl, wherein any two adjacent W atoms may form CR^CR1 or C≡C;

L4 is selected from the group consisting of: R^, acyl, acylamino, aminothiocarbonyl, aminoacyl carbonvloxv, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethyiene glycol, substituted sulfonyl, substituted sulfinyl, thioacyi, aminoacyl, phthalimido, and formyl; and wherein any two or more W groups can be bonded together to form a C3-C12 cycloalkyl ring structure, a C3-Ci2 fused cycloalkyl ring structure, a C3-C12 aryl ring structure, a C3-C 12 fused aryl ring structure, a C3-C12 heteroaryl ring structure, a C3-C 12 fused heteroaryl ring structure, a iVC V> heterocyclic ring structure, and a C3-C12 fused heterocyclic ring structure; and wherein W„ does not form an (S) ^ εΗ2εΗ2ε(°}Ν Η group in the ring starting from the carbon attached to R2 which is adjacent to the keto-amide or thioketo-amide functional group;

or a tautomer and/or a pharmaceutically acceptable salt thereof.

2. The compound of claim 1 , wherein R4 is

3. The compound of claim 1 or 2, wherein n is 1, Wi is CHR2 where R2 i s a Ci-Cs alkyi or Ci-Cg alkenyl.

4. The compound of any one of claims 1-3, wherein L4 is -S02-Ar.

5. The compound of any one of claims 1-3, wherein L4 is -CO-O-substituted alkyl.

6. The compound of any one of claims 1-3, wherein L4 is -CO-0-ίί Ί ί φ or - CO-0-CH2Ar.

7. The compound of any one of claims 1-3, wherein L' is -CO-alkyl or -CO-substituted alkyl.

8. The compound of any one of claims 1-3, wherein L4 is -CO-CH or - CO-Ar.

9. The compound of any one of claims 1-8, wherein one R' on the carbon atom is a substituted Ci~C8 alkyl .

10. The compound of any one of claims 1-8, wherein one R" on the carbon atom is - CH-(CH3)2 or -€H2-CH-(CH3)2.

11. The compound of any one of claims 1-8, wherein one R" on the carbon atom is a C3-C9 heteroaryi or a substituted C3-C9 heteroaryl.

12. The compound of any one of claims 1-8, wherein one R' on the carbon atom is a C3-C9 heteroaryl or a naturally or unnaturally occurring amino acid side chain.

13. The compound of claim 1 or 2, wherein n is 4, Wi is CUR , and one R" on the carbon atom is -CH-(CH3)2, and W2 is CHR2 where R2 is a C]-C4 alkyl covalently bonded to the R group on Wj and forms a bicyclic ring structure.

14. A compound comprising Formula II:

Formula II where: n is from 1-1 1 ; each R5and R3 is independently selected from the group consisting of: hydrogen, Cj - Cg alkyl, substituted Ci-Cg aikyl, Ci-Cg alkenyl, substituted Ci-Cg alkenyl, Ci-Cg aikynyl, substituted Ci-Cg aikynyl, C3-C7 aryl, substituted C3-C7 aryi, cyano, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3- C9 heteroaryl, substituted C3-C9 heteroaryl , Ci-Cg alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR2, NH2, NHR2, SR2, substituted sulfonyl, and substituted sulfinyl, wherein any R1 substituent may be covalently bonded to or share another R1 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaiyl, spirocyclic, or bicyclic ring system; each R ' is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-Cg al kyl, Ci-Cg alkenyl, substituted Ci-Cg alkenyl, Ci-Cg aikynyl, substituted C Cg aikynyl, C3-C7 aryl , substituted C3-C7 aryl, cyano, OH, Ci-Cg alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R2 substituent may be covalently bonded to or share another R2 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each W is independently selected from the group consisting of: CH2, C(R2)2, CHR2, O, NH, NR2, C( ()), C( S ). S, Si O), S(=0)2, -C-, -CH-, a C3-C12 spirocyclic group of where m is 1-19, sulfonyl, substituted sulfonyl, sulfinyl, and substituted sulfinyl, wherein any two adjacent W atoms may form CR1=CR5 or C≡C;

L4 is selected from the group consisting of: R2, acyl, acylamino, arninothiocarbonyl , aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfinyl, thioacyl, aminoacyl,

phthalimido, and formyl; and wherein any two or more W groups can be bonded together to form a C3-C12 cycloalkyl ring structure, a C3-C12 fused cycloalkyl ring structure, a C3-C12 aryl ring structure, a C3-C12 fused aryl ring structure, a C3-C12 heteroaryl ring structure, a C3-C12 fused heteroaryl ring structure, a C3-C12 heterocyclic ring structure, and a C3-C12 fused heterocyclic ring structure; and wherein W„ does not form an (S)

from the carbon attached to R" which is adjacent to the keto-amide functional group, or a tautomer and/or a pharmaceutically acceptable salt thereof.

15. The compound of claim 14, wherein L4 is selected from the group consisting of acyl, aminoacyl, carboxy ester, -CO-ethylene glycol, -CO-polyethylene glycol, substituted sulfonyl, and formyl.

16. The compound of claim 14, wherein L4 is -CO-0-CH2-Ph or -S02-Ar.

17. The compound of claim 14, wherein L4 is a carboxy ester or substituted sulfonyl and LJ is C3 or C4 alkyl ,

18. A compound comprising Formula III:

Formula where:

Z1 is selected from the group consisting of: CH2, C(R!)2, CHR1, O, H, NR!, C(=0),

C(=S), S, S(=0), and S(=0)2, a C3-C12 spirocyclic group of where m is 1 -19; each Rxand R3 is independently selected from the group consisting of: hydrogen, Ci- Cg alkyl, substituted Ci-Cg alkyl, Ci~Cg alkenyl, substituted Ci-Cg alkenyl, Ci-Cg alkynyl, substituted C Cg alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, C3-C10 cycloalkyl, substituted C3-Cio cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3 C9 heteroaryl, substituted C3-C9 heteroaryl, C3-Cg alkoxy, substituted C-.-Cg alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR2, NH2, NHR2, SR , substituted sulfonyl, and substituted sulfinyl, wherein any R1 substituent may be covalently bonded to or share another R1 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R2 is independently selected from the group consisting of: hydrogen, Ci-C8 alkyl, substituted Ci-Cs alkyl, Ci-Cg alkenyl, substituted Ci-C& alkenyl , Ci-Cg alkynyl, substituted Ci-C8 alkynyl, C3-C7 aryl, substituted C3-C7 aryl , cyano, OH, Ci -Cg alkoxy, substituted Ci -Cg alkoxy, amino, substituted amino, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R2 substituent may be covalently bonded to or share another R2 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system;

L4 is selected from the group consisting of: R2, acyl, acylamino, arninothiocarbonyl , aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfinyl, thioacyl, aminoacyl, phthalimido, and formyl; or a tautomer and/or a pharmaceutically acceptable salt thereof.

19. A compound comprising Formula IV:

Formula IV where:

Z1 and Z2 are each independently selected from the group consisting of: CH2, C(R2)2, CHR2, O, NH, NR2, C(=0), C(=S), S, S(=0), S(=0)2, -C-, -CH-, a C3-C12 spirocyclic group

of - « » where m is 1-19, wherein any two adjacent Z atoms may form CR^ R

C C; each R and R is independently selected from the group consisting of: hydrogen, Ci

C8 alkyl, substituted Ci~C8 alkyl, Ci-Cg alkenyl, substituted Ci-C8 alkenyl, Ci-Cg alkynyl, substituted Ci-C alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3- C9 heteroaryl, substituted C3-C9 heteroaryl, Ci~C8 alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, alkyl thio, substituted alkylthio, OH, OR2, NH2, IR2, SR2, substituted suifonyi, and substituted sulfinyl, wherein any R1 substituent may be covalently bonded to or share another Rl substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyciic ring system; each R is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-Cg alkyl, Ci-Cg alkenyl, substituted Ci-Cg alkenyl, Ci-C8 alkynyl, substituted Ci-Cg alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, OH, Cj-Cg alkoxy, substituted Cj-Cg alkoxy, amino, substituted amino, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R2 substituent may be covalently bonded to or share another R2 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyciic ring system;

L4 is selected from the group consisting of: R% acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethyiene glycol, substituted suifonyi, substituted sulfinyl, thioacyi, aminoacyl, phthalimido, and formyi; and wherein any two or more Z groups can be bonded together to form a C3-C12 cycloalkyl ring structure, a C3-C12 fused cycloalkyl ring structure, a C3-C12 aryl ring structure, a C3-C12 fused aryl ring structure, a C3-C12 heteroaryl ring structure, a C3-C12 fused heteroaryl ring structure, a C3-C12 heterocyclic ring structure, and a C3-C12 fused heterocyclic ring structure; or a tautomer and/or a pharmaceutically acceptable salt thereof

20. A compound comprising Formula V

Formula V where:

Z1, Z2, and Z3 are each independently selected from the group consisting of: CH?, C(R2)2, CUR2, O, NH, R2, C(=0), C(=S), S, S(=0), S(=0)2, -C-, -CH-, a C3-C12 spirocyclic group of

or C C. each Rxand R3 is independently selected from the group consisting of: hydrogen, Cj - Cg alkyi, substituted Ct-Cg alkyl, C i-Cg alkenyl, substituted Ct-Cg alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C3-C7 and, substituted C3-C7 aryi, cyano, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3- C9 heteroaryl, substituted C3-C9 heteroaryl, Ci-Cg alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR2, NH2, NHR2, SR2, substituted sulfonyl, and substituted sulfinyl, wherein any R1 substituent may be covalently bonded to or share another R1 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaiyl, spirocyclic, or bicyclic ring system; each R ' is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-Cg alkyl, Ci-Cg alkenyl, substituted Ci-Cg alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C3-C7 aryl , substituted C3-C7 aryl, cyano, OH, Ci-Cg alkoxy, substituted Ci-Cg al koxy, amino, substituted amino, C3-C10 cycloal kyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R2 substituent may be covalently bonded to or share another R2 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; L4 is selected from the group consisting of: R2, acyl, acylamino, aminothiocarbonyj , aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfinyl, thioacyl, aminoacyl, phthalimido, and formyi; group in the ring starting from the carbon attached to R which is adjacent to the keto-amide functional group; and wherein any two or more Z groups can be bonded together to form a C3-C12 cycloalkyl ring structure, a C3-C12 fused cycloalkyl ring structure, a C3-C12 aryl ring structure, a C3-C12 fused aryl ring structure, a C3-C12 heteroaryl ring structure, a C3-C12 fused heteroaryl ring structure, a C3-C12 heterocyclic ring structure, and a C3-C12 fused heterocyclic ring structure; or a tautomer and/or a pharmaceutically acceptable salt thereof. 21. A compound comprising Formula VI:

Formula VI where:

Z1, 1 , Z3, and Z4 are each independently selected from the group consisting of: CH2, C(R2)2, CUR2, O, NH, NR2, C(=0), C(=S), S, S(=0), S(=0)2, -C-, -CH-, a C3-C12 spirocyclic group of

or C≡C; each Rxand R3 is independently selected from the group consisting of: hydrogen, Ci- Cg alkyl, substituted Ci-Cg alkyl, C3-Cg alkenyl, substituted Ci-Cg alkenyl, C Cg aikynyl, substituted Ci-Cg aikynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-

C9 heteroaryl, substituted CVC9 heteroaryl, Ci-Cg alkoxy, substituted Cj-Cg alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR2, NH?, HR2, SR , substituted sulfonyl, and substituted sulfinvl, wherein any R1 substituent may be covalently bonded to or share another R1 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyciic ring system; each R2 is independently selected from the group consisting of; hydrogen, Ci-Cg alky] , substituted Ci-Cg alkyl, Ci~C8 alkenyl, substituted Cj-Cg alkenyl, Ci~Cg alkynyl, substituted Ci-Cg alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, OH, Ci-C alkoxy, substituted Ci-C alkoxy, amino, substituted amino, CV-CJO cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R2 substituent may be covalently bonded to or share another R2 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyciic ring system;

L4 is selected from the group consisting of: R2, acyl, acylamino, aminothiocarbonyl, aminoacyi carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethyiene glycol, substituted sulfonyl, substituted sulfmyl, thioacyi, aminoacyi, phthalimido, and formyl; and wherein any and all Z groups do not form an (S) ^ CH2CH2C(°)NH group in the ring starting from the carbon attached to R2 which is adjacent to the keto-amide functional group; and wherein any two or more Z groups can be bonded together to form a C3-C12 cycloalkyl ring structure, a C3-C12 fused cycloalkyl ring structure, a C3-C12 aryl ring structure, a C3-C12 fused aryl ring structure, a C3-C12 heteroaryl ring structure, a C3-C12 fused heteroaryl ring structure, a C3-C12 heterocyclic ring structure, and a C3-C12 fused heterocyclic ring structure; or a tautomer and/or a pharmaceutical ly acceptable salt thereof. 22. A compound comprising Formula VII:

Formula VH where:

Z1, Z2, Z Z\ and Z5 are each independently selected from the group consisting of:

Ci k C(R2)2, CHR2, NR2, C( O { S ). S, S( ()}, S( ()),.. -C-, -CH-, a C3-C12 spirocyclic group of where m is 1-19, wherein any two adjacent Z atoms may form

CR =CR! or C≡C; each Rxand R3 is independently selected from the group consisting of: hydrogen, Ci-

Cg alkyl, substituted Ci-Cg alkyl, C Cg alkenyl, substituted Ci-Cg alkenyl, Ci-Cg alkynyl, substituted CVCg alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3- C9 heteroaryl, substituted C3-C9 heteroaryl, C3-Cg alkoxy, substituted Ci -Cg alkoxy, amino, substituted amino, alkyithio, substituted aikylthio, OH, OR2, NH2, NHR"', SR2, substituted suifonyi, and substituted sulfmyl, wherein any R! substituent may be covalently bonded to or share another Rf substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R' is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted C3-Cg alkyl, Ci-Cg alkenyl, substituted C-. -Cg alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, OH, C . -Cg alkoxy, substituted C . -Cg alkoxy, amino, substituted amino, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any natural ly or non-naturally occurring amino acid side chain, wherein any R2 substituent may be covalently bonded to or share another R2 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; L4 is selected from the group consisting of: R2, acyl, acylamino, aminothiocarbonyj , aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfinyl, thioacyl, aminoacyl, phthalimido, and formyi; and wherein any and all Z groups do not form an (S) ^ CH2CH2C(°)NH group in the ring starting from the carbon attached to R2 which is adjacent to the keto-amide functional group; and wherein any two or more Z groups can be bonded together to form a C3-C12 cycloalkyl ring structure, a C3-C12 fused cycloalkyl ring structure, a C3-C12 aryl ring structure, a C3-C12 fused aryl ring structure, a C3-C12 heteroaryl ring structure, a C3-C12 fused heteroaryl ring structure, a C3-C12 heterocyclic ring structure, and a C3-C12 fused heterocyclic ring structure: or a tautomer and/or a pharmaceutically acceptable salt thereof. 23. A compound comprising Formula VIH:

Formula VIII where:

Z1, /. '. Z3, Z4, Z5, and Z6 are each independently selected from the group consisting of: Ci k C(R2)2, CHR2, O, NH, NR2, C( ()}, Π S), S, S(=0), S(=0)2, -C-, -CH-, a C3-C12 spirocyclic group of where m is 1-19, wherein any two adjacent Z atoms may torm

C\V C\V or ( C. each Rxand R3 is independently selected from the group consisting of: hydrogen, Ci- Cg alkyl, substituted Ci-Cg alkyl, C Cg alkenyl, substituted Ci-Cg alkenyl, Ci-Cg alkynyl, substituted CVCg alkynyl, C3-C7 aryl, substituted C3-C7 aryl, c ano, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3- C9 heteroaryl, substituted C3-C9 heteroaryl, C3-Cg alkoxy, substituted Ci -Cg alkoxy, amino, substituted amino, alkyfthio, substituted alkylthio, OH, OR2, NH2, NHR2, SR2, substituted suifonyi, and substituted sulfmyl, wherein any R! substituent may be eovalentfy bonded to or share another R1 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R~ is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted C3-Cg alkyl, Ci-Cg alkenyl, substituted Ci -Cg alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, OH, C . -Cg alkoxy, substituted C . -Cg alkoxy, amino, substituted amino, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any natural ly or non-naturally occurring amino acid side chain, wherein any R2 substituent may be covalentiv bonded to or share another R2 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system;

L4 is selected from the group consisting of: R2, acyl, acyiamino, aminothiocarbonyl, aminoacyi carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted suifonyi, substituted sulfmyl, thioacyl, aminoacyi, phthalimido, and formy ; and wherein any and all Z groups do not form an (S) ^ ci ½c^2c(°)N group in the ring starting from the carbon attached to R2 which is adjacent to the keto-amide functional group; and wherein any two or more Z groups can be bonded together to form a C3-C12 cycloalkyl ring structure, a C3-C12 fused cycloalkyl ring structure, a C3-C12 aryl ring structure, a C3-C12 fused aryl ring structure, a C3-C12 heteroaryl ring structure, a C3-C12 fused heteroaryl ring structure, a C3-Q2 heterocyclic ring structure, and a C3-C12 fused heterocyclic ring structure; or a tautomer and/or a pharmaceutically acceptable salt thereof.

24. A compound comprising Formula IX:

Formula IX where:

Z1, Z2, Z3, Z Z5, Z6, and Z 7 are each independently selected from the group consisting of: C S k C(R2)2, C IR2, O, NFL NR.2, C( < )), Π S ), S, S{ ( )), S(=0)2, -C-, -CH-, a C3-C12 spirocyclic group of ' - where m is 1-19, wherein any two adjacent Z atoms may form CR1==CR1 or C C: each Rland R3 is independently selected from the group consisting of: hydrogen, C\- C8 alkyl, substituted Ci-C8 aikyl, Ci-C8 alkenyl, substituted Ci-C8 alkenyl, C C8 aikynyl, substituted Ci-C8 aikynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3- C9 heteroaryl, substituted (VGs heteroaryl, Ci-C8 alkoxy, substituted C]-C8 alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OFL OR2, NFi?, HR2, SR , substituted sulfonyl, and substituted sulfinvl, wherein any R1 substituent may be covalently bonded to or share another R1 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R2 is independently selected from the group consisting of: hydrogen, Ci-C8 alkyl , substituted Ci-C8 alkyl, Ci~C8 alkenyl, substituted C]-C8 alkenyl, Ci~C8 aikynyl, substituted Ci-Cs aikynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, OH, Ci-C alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, C -Cio cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R2 substituent may be covalentlv bonded to or share another R2 substituent to form a C3-C12 cyclic, heterocyclic, aiyl, heteroaryl, spirocyclic, or bicyclic ring system;

L4 is selected from the group consisting of: R2, acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfinyl, thioacyi, aminoacyl, phthalimido, and formyi; and wherein any and all Z groups do not form an (S) ^ ci ½c^2c(°)N group in the ring starting from the carbon attached to R2 which is adjacent to the keto-amide functional group; and wherein any two or more Z groups can be bonded together to form a C3-C12 cycloalkyl ring structure, a (' ;--(>> fused cycloaikyl ring structure, a C3-C12 aryi ring structure, a C3-C12 fused aryi ring structure, a C3-C12 heteroaryl ring structure, a C3-C12 fused heteroaryl ring structure, a C3-Q2 heterocyclic ring structure, and a C3-C12 fused heterocyclic ring structure; or a tautomer and/or a pharmaceutically acceptable salt thereof. 25. A compound comprising Formula X:

Formula X where:

Z1, 1 , Z3, Z'*, Z5, Ζυ, Z ', and Z8 are each independently selected from the group consisting of: CH2, C(R2)2, CHR2, O, H, R2, C(=0), C(=S), S, S(=0), S(=0)2, -C-, -CH- a C3-C 12 spirocyclic group ot where m is 1-19, wherein any two adjacent Z atoms may form C R '! C R '! or C≡C; each R!and R3 is independently selected from the group consisting of: hydrogen, Ci- C8 alkyl, substituted Ci~C8 alkyl, Ci-Cg alkenyl, substituted Ci-Cg alkenyl, Ci-Cg alkynyl, substituted Ci-C alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3- C9 heteroaryl, substituted C3-C9 heteroaryl, Ci~C8 alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, alkyl thio, substituted alkylthio, OH, OR2, NH2, NHR2, SR2, substituted sulfonyl, and substituted sulfmyl, wherein any R1 substituent may be covalently bonded to or share another Rl substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-Cg alkyl, Ci-Cg alkenyl, substituted Ci-Cg alkenyl, C]-Cg alkynyl, substituted Ci-Cg alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, OH, Cj-Cg alkoxy, substituted Cj-Cg alkoxy, amino, substituted amino, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R2 substituent may be covalently bonded to or share another R2 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system ;

L4 is selected from the group consisting of: % acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfmyl, thioacyl, aminoacyl, phthalimido, and formyl; and wherein any and all Z groups do not form an (S) ^ CH2CH2C(°)NH group in the ring starting from the carbon attached to R2 which is adjacent to the keto-amide functional group; and wherein any two or more Z groups can be bonded together to form a C3-C12 cycloalkyl ring structure, a C3-C12 fused cycloalkyl ring structure, a C3-C12 aryl ring structure, a C3-C 12 fused aryl ring structure, a C3-C12 heteroaryl ring structure, a C3-C12 fused heteroaryl ring structure, a C3-C12 heterocyclic ring structure, and a C3-C12 fused heterocyclic ring structure;

or a tautomer and/or a pharmaceutically acceptable salt thereof. 26, A compound comprising Formula XI:

Formula XI where:

Z1, Z2, ZJ, Z4, Z5, Z6, Z 7, Z8, and Z9 are each independently selected from the group consisting of: CI k. C( R . CHR2, O, M l, NR2, C(=0), C( S), S, S( ()}, S{ ( )) ··. -C-, -CI 1-, a C3-C12 spirocyclic group ot where m is 1- 19, wherein any two adjacent Z atoms may form C R '! C R '! or C≡C; each R!and R3 is independently selected from the group consisting of: hydrogen, Ci- Cg alkyl, substituted Ci-C8 alkyl, Ci-Cg alkenyl, substituted Ci-Cg alkenyl, Ci-Cg aikynyl, substituted Ci-C aikynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, C3-C10 cycloalkyl, substituted C3-C10 cycloal kyl, C3-C9 heterocycloalkyl, substituted CVC V. heterocycloalkyl, C3- C9 heteroaryl, substituted C3-C9 heteroaryl, Ci~C8 alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR2, NH2, NHR2, SR2, substituted suifonyi, and substituted sulfmyl, wherein any R1 substituent may be covalently bonded to or share another Rl substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaiyl, spirocyclic, or bicyciic ring system; each R is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-Cg alkyl, Ci-Cg alkenyl, substituted Ci-Cg alkenyl, C]-Cg aikynyl, substituted Ci-Cg aikynyl, C -C7 aryl, substituted C3-C7 aryl, cyano, OH, Cj-Cg alkoxy, substituted Cj-Cg alkoxy, amino, substituted amino, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R2 substituent may be covalently bonded to or share another R2 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system;

L4 is selected from the group consisting of: R2, acyl, acylamino, aminothiocarbonyl, aniinoacy! carbonyloxy, aminosulfonyl, ami din o, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfinyl, thioacyl, aminoacyl, phthalimido, and formyl; and wherein any and all Z groups do not form an (S) ^ CH2CH2C(°)NH group in the ring starting from the carbon attached to R' which is adjacent to the keto-amide functional group; and wherein any two or more Z groups can be bonded together to form a C3-C12 cycloalkyl ring structure, a C3-C12 fused cycloalkyl ring structure, a C3-C12 aryl ring structure, a C3-C12 fused aryl ring structure, a C3-C12 heteroaryl ring structure, a C3-C12 fused heteroaryl ring structure, a C3-C12 heterocyclic ring structure, and a C3-C12 fused heterocyclic ring structure;

or a tautomer and/or a pharmaceutically acceptable salt thereof.

27, A compound compri sing Formula XII:

Formula XII where /Λ Z6, Z7, Z8, Z9 and zi0 are each independently selected from the grou 2)2, CHR2, (). M L NR2, Π O h C(=S), S, S( ()}. S( ()) -C-, -

CH-, where m is 1- 19, wherein any two adjacent Z atoms may form CR =CR or C=C; each R and R3 is independently selected from the group consisting of: hydrogen, Ci-

C8 alkyl, substituted Ci~C8 alkyl, Cj-C8 alkenyl, substituted Ci~C8 alkenyl, Ci-C8 alkynyl, substituted Ci-C8 alkynyl, C3-C7 aryl, substituted C3-C7 ai l, cyano, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3- C9 heteroaryl, substituted C3-C9 heteroaryl, C i-Cg alkoxy, substituted Ci-C8 alkoxy, amino, substituted amino, alkyithio, substituted aikylthio, OH, OR2, H2, HR2, SR2, substituted suifonyi, and substituted sulfmyl, wherein any R1 substituent may be covalently bonded to or share another R! substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system, each R is independently selected from the group consisting of: hydrogen, Ci-C8 alkyl, substituted C i-Cg alkyl, Cj -C8 alkenyl, substituted Ci-C8 alkenyl, C j-C8 alkynyl, substituted Ci-Cg alkynyl, C3-C7 aryl, substitirted C3-C7 aryl, cyano, OH, Ci -C8 alkoxy, substituted C C8 alkoxy, amino, substituted amino, C3-C10 cycloalkyl, substituted C3-C1.0 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl , and any naturally or non-naturally occurring amino acid side chain, wherein any R2 substituent may be covalently bonded to or share another R2 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system;

L4 is selected from the group consisting of: ^, acyl, acyianiino, aminothiocarbonyl, aminoacyi carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethyiene glycol, substituted suifonyi, substituted sulfmyl, thioacyi, aminoacyi, phthalimido, and formyl; and wherein any and all Ζ groups do not form an (S) ^ CH2CH2C(0)NH group jn t|ie ring starting from the carbon attached to R2 which is adjacent to the keto-amide functional group, and wherein any two or more Z groups can be bonded together to form a C3-C12 cycloalkyl ring structure, a C3-C12 fused cycloalkyl ring structure, a C3-C12 aryl ring structure, a C3-C12 fused aryl ring structure, a C3-C12 heteroaryl ring structure, a C3-C12 fused heteroaryl ring structure, a C3-C12 heterocyclic ring structure, and a C3-C12 fused heterocyclic ring structure;

or a tautomer and/or a pharmaceutically acceptable salt thereof. 28. A compound comprising Formula XIH:

Formula XIII where:

Z1, Z"', Z Z4, Z5, Z6, Z 7, Z8, Z9, Zu' and Z are each independently selected from the group consisting of: C¾, C(R2)2, CUR2, O, NH, NR2, C(=0), C(=S), S, S(=0), S(=0)2, -C-, - where m is 1- 19, wherein any two adjacent Z atoms may form CR =CR or C:≡=C; each R and R" is independently selected from the group consisting of: hydrogen, Cj - Cg alkyl, substituted Ci-Cg alkyl, Ci-Cg alkenyl, substituted Ci-Cg alkenyl, Ci-Cg aikynyl, substituted Ci-Cg aikynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, C3-C10 cycloalkyl, substituted C3-C10 cycloal kyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3- C9 heteroaryl, substituted C3-C9 heteroaryl , Ci-Cg alkoxy, substituted Ci -Cg alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR2, NH2, NHR2, SR2, substituted sulfonyl, and substituted sulfinyl, wherein any R1 substituent may be covalently bonded to or share another Rl substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system, each R2 is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-Cg alkyl, Ci-Cg alkenyl, substituted C .-Cg alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C3-C7 aryl, substituted C3-C7 and, cyano, OH, C Cg alkoxy, substituted C Cg alkoxy, amino, substituted amino, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaiyl, and any naturally or non-naturally occurring amino acid side chain, wherein any R2 substituent may be covalently bonded to or share another R2 substituent to form a C3-Ci2 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system;

L4 is selected from the group consisting of: R2, acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethyiene glycol, substituted sulfonyl, substituted sulfmyl, thioacyl, aminoacyl, phthalimido, and formyl; and wherein any and all Z groups do not form an (S) ^ CH2CH2C(°)NH group in the ring starting from the carbon attached to R2 which is adjacent to the keto-amide functional group, and wherein any two or more Z groups can be bonded together to form a C3-C12 cycloalkyl ring structure, a C3-C12 fused cycloalkyl ring structure, a C3-C12 aryl ring structure, a C3-C12 fused aryl ring structure, a C3-C12 heteroaryl ring structure, a C3-C12 fused heteroaryl ring structure, a C3-C12 heterocyclic ring structure, and a C3-C12 fused heterocyclic ring structure;

or a tautomer and/or a pharmaceutically acceptable salt thereo

29. A compound comprising Formula XIV.

Formula X IV where:

Z1, Z2, Z3, Z Z5, Z6, Z ', Zs, Z1 , Z1 ' and Z'2 are each independently selected from the group consisting of: i l k C(R2)2, CHR2, O, NH, NR2, C( ( )), C( S), S, 8(0), S( O. -C-. -

CH-, a C3-C12 spirocyclic group of where m is 1-19, wherein any two adjacent Z atoms may form CR^CR1 or C≡C; each Rland R3 is independently selected from the group consisting of: hydrogen, Ci- C8 alkyl, substituted Ci-C8 aikyl, Ci-Cg alkenyl, substituted Ci-Cg alkenyl, C C8 alkynyl, substituted Ci-C8 alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3- C9 heteroaryl, substituted C -.~ -.. heteroaryl, Ci-C8 alkoxy, substituted C]-C8 alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR2, NH?, HR2, SR , substituted sulfonyl, and substituted sulfinvl, wherein any R1 substituent may be covalently bonded to or share another R1 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R2 is independently selected from the group consisting of: hydrogen, Ci-C8 alkyl , substituted Ci-C8 alkyl, Ci~C8 alkenyl, substituted C]-C8 alkenyl, Ci~C8 alkynyl, substituted Ci-Cg alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, OH, Ci-Cg alkoxy, substituted Ci-C alkoxy, amino, substituted amino, CV-CJO cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R2 substituent may be covalentlv bonded to or share another R2 substituent to form a C3-C12 cyclic, heterocyclic, aiyl, heteroaryl, spirocyclic, or bicyclic ring system;

L4 is selected from the group consisting of: R2, acyl, acylamino, aminothiocarbonyl, aminoacyi carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfmyl, thioacyi, aminoacyi, phthalimido, and formyl; and wherein any and all Z groups do not form an (S) ^ CH2CH2C(°)NH group jn he ring starting from the carbon attached to R2 which is adjacent to the keto-amide functional group; and wherein any two or more Z groups can be bonded together to form a C3-C12 cycloalkyl ring structure, a C3-C12 fused cycloalkyl ring structure, a C3-C12 aryl ring structure, a C3-C32 fused aryl ring structure, a C3-C12 heteroaryl ring structure, a C3-C12 fused heteroaryl ring structure, a C3-C12 heterocyclic ring structure, and a C3-C12 fused heterocyclic ring structure;

or a tautomer and/or a pharmaceutical ly acceptable salt thereof. 30. A compound compri sing Formula XV:

Formula XV where: n is from 1-12; each A is independently selected from the group consisting of: O and S; each Rxand R3 is independently selected from the group consisting of: hydrogen, Ci- Cg alkyl, substituted C\-C% alkyl, C Cg alkenyl, substituted C\-C% alkenyl, Ci-Cg alkynyl, substituted C i-Cg alkynyl, C3-C7 and, substituted C3-C7 aryl, cyano, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3- C9 heteroaryl, substituted C3-C9 heteroaryl, Ci-Cg alkoxy, substituted Ci -Cg alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR2, NH2, NHR2, SR , substituted sulfonyl, and substituted sulfmyl, wherein any R1 substituent may be covalently bonded to or share another R1 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R2 is independently selected from the group consisting of: hydrogen, Ci-C8 alkyl, substituted Ci-Cs alkyl, Ci-Cg alkenyl, substituted Ci-C8 alkenyl, Ci-Cg alkynyl, substituted Ci-C8 alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, OH, Ci -Cg alkoxy, substituted Ci -Cg alkoxy, amino, substituted amino, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R2 substituent may be covalently bonded to or share another R2 substituent to form a C3-C12 cyclic, heterocyclic, and, heteroaryl, spirocyclic, or bicyclic ring system; each W is independently selected from the group consisting of: CH2, C(R2)2, CHR2, 2, C( O). C( S), S, S( O), S( O b, -C-, -CH-, a C3-C12 spirocyclic group of where m is 1-19, sulfonyl, substituted sulfonyl, sulfinyl, and substituted sulfmyl, wherein any two adjacent W atoms may form CR^CR1 or C≡C;

Z is selected from the group consisting of: Ci-Cs alkyl, substituted Ci-Cg alkyl, Ci-C8 alkenyl, substituted Ci~C8 alkenyl, Cj-Q alkynyl, substituted Ci~C8 alkynyl, Cj-C8 alkoxy, substituted Ci-C8 alkoxy, substituted amino, alkylthio, substituted alkylthio, sulfonyl, C(R2)2, CHR2, O, NH, NR2, S, substituted sulfonyl, sulfinyl, substituted sulfinyl .and a covalent bond;

L4 is selected from the group consisting of: R2, acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfinyl, thioacyl, aminoacyl, phthalimido, and formyl; and wherein any two or more W groups can be bonded together to form a C3-C12 cycloalkyl ring structure, a C3-C12 fused cycloalkyl ring structure, a C3-C12 aryl ring structure, a C3-C12 fused aryl ring structure, a C3-C12 heteroand ring structure, a C3-C12 fused heteroaryl ring structure, a C3-C12 heterocyclic ring structure, and a C3-C12 fused heterocyclic ring structure; and wherein Wn does not form an (S) * Λ 2 { ' group in the ring starting from the carbon attached to R2 which i s adjacent to the keto-amide or thioketo-amide functional group; or a tautomer and/or a pharmaceutically acceptable salt thereof.

31. A compound compri sing Formula XVI:

Formula XVI where: n is from 1-12; each R and R3 is independently selected from the group consisting of: hydrogen, C C8 alkyl, substituted Ci-C8 alkyl, Cj-C8 alkenyl, substituted Ci-C8 alkenyl, Ci-C8 alkynyl, substituted C3-C8 alkynyl, C3-C7 aryl, substituted C3-C7 and, cyano, C3-C10 cycioalkyl, substituted C3-C10 cycioalkyl, ( ' ;··("·,, heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3- C9 heteroaryl, substituted C3-C9 heteroaryl, Ci-C8 alkoxy, substituted Ci-C8 alkoxy, amino, substituted amino, alkylthio, substituted alkyl thio, OH, OR2, 'NH2, NHR2, SR , substituted sulfonyl, and substituted sulfmyl , wherein any R1 substituent may be covalently bonded to or share another R1 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system, each R2 is independently selected from the group consisting of: hydrogen, Ci-Cs alkyl, substituted Ci-C8 alkyl, Ci-C8 alkenyl, substituted Ci-C8 alkenyl, Ci-C8 alkynyl, substituted Ci-C8 alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, OH, Ci-C8 alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, C3-C10 cycioalkyl, substituted C3-C10 cycioalkyl , C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R2 substituent may be covalently bonded to or share another R2 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each W is independently selected from the group consisting of: CH2, C(R2)2, CHR2, 2, C( O). C( S), S, S( O), S(=0)2, -C-, -CH-, a C3-C12 spirocyclic group of where m is 1-19, sulfonyl, substituted sulfonyl, sulfmyi, and substituted sulfinyl, wherein any two adjacent W atoms may form CR^CR1 or C=C;

Z is selected from the group consisting of: Ci-Ce alkyl, substituted Ci-Cg alkyl, Ci-Cg alkenyl, substituted Ci-C8 alkenyl, C]-C8 alkynyl, substituted Ci-C8 alkynyl, Cj-Cg alkoxy, substituted Cj-Cg alkoxy, substituted amino, alkylthio, substituted alkylthio, sulfonyl, C(R2)2, CHR2, O, NH, NR2, S, substituted sulfonyl, sulfinyl, substituted sulfinyl .and a covalent bond;

L4 is selected from the group consisting of: R2, acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfinyl, thioacyl, aminoacyl, phthalimido, and formyl; and wherein any two or more W groups can be bonded together to form a C3-C12 cycloaikyl ring structure, a C3-C12 fused cycloaikyl ring structure, a C3-C12 aryi ring structure, a C3-C 12 fused aryi ring structure, a C3-C12 heteroaryl ring structure, a C3-C12 fused heteroaryl ring structure, a C3-C12 heterocyclic ring structure, and a C3-C12 fused heterocyclic ring structure; and wherein Wn does not form an (S) ^ C' ",2CH2C(°)NH group n j-}ie r ng starting from the carbon attached to R2 which is adjacent to the keto-amide functional group; or a tautomer and/or a pharmaceutically acceptable salt thereof.

32, A compound comprising Formula XXIII:

Formula XVH where: each A is independently selected from the group consisting of: O and S;

each R4 is independently selected from the group consisting of: H and ; each R and R3 is independently selected from the group consisting of: hydrogen, C C8 alkyl, substituted Ci-Cg aikyl, Ci-Cg alkenyl, substituted Ci-Cg alkenyl, Ci-Cg aikynyl, substituted Ci-C8 aikynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3- C9 heteroaryl, substituted C3-C9 heteroaryl, Cj-C8 alkoxy, substituted C]-C8 alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR2, NH2, NliR2, SR , substituted sulfonyl, and substituted sulfmyl, wherein any R1 substituent may be covalently bonded to or share another R1 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyciic ring system; each R2 is independently selected from the group consisting of: hydrogen, Ci-C8 alkyl, substituted C]-C8 alkyl, Ci-Cg alkenyl, substituted C C8 alkenyl, Ci~C8 aikynyl, substituted Ci-Cg aikynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, OH, CVCg alkoxy, substituted Ci~C8 alkoxy, amino, substituted amino, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-Cg heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R2 substituent may be covalently bonded to or share another R2 substituent to form a C3-C12 cyclic, heterocyclic, and, heteroaryl, spirocyclic, or bicyciic ring system;

LJ is a group containing 1-8 atoms and is selected from the group consisting of: Ci~C8 alkyl, substituted Ci-Cg alkyl, Ci~C8 alkenyl, substituted Ci-Cg alkenyl, Ci-Cg aikynyl, substituted Ci-Cg aikynyl, Ci-Cg alkoxy, substituted Cj-Cg alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, sulfonyl, C(R2)2, CHR2, O, NH, NR2, S, substituted sulfonyl, sulfmyl, and substituted sulfmyl;

Z is selected from the group consisting of: Cj-Cg alkyl, substituted Ci-Cg alkyl, Ci-Cg alkenyl, substituted Ci-Cg alkenyl, Ci-Cg aikynyl, substituted Ci-Cg aikynyl, Cj-Cg alkoxy, substituted Cj-Cg alkoxy, substituted amino, alkylthio, substituted alkylthio, sulfonyl, C(R2)2, CHR , O, NH, NR2, S, substituted sulfonyl, sulfmyl, substituted sulfmyl ,and a covalent bond,

L4 is selected from the group consisting of: R2, acyl, acyiamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfmyl, thioacyl, aminoacyl, phthalimido, and formyl; wherein each L3 and Z are capable of being covalently bonded to the same or other R2 functional groups such that a bicyclic or spirocyclic ring system is formed; and wherein 1/ and Z do not form an (S) ^ CH2CH2C(0^H group in the ring starting from the carbon atom closest to the keto-amide or thio-keto-amide functional group; or a tautomer and/or a pharmaceutically acceptable salt thereof.

33. The compound of claim 32, wherein R4 is

34. The compound of claim 32 or 33, wherein L is a C3 alkyl or C3 substituted alkyl.

35. The compound of claim 32 or 33, wherein LJ is a C4 alkyl or C4 substituted alkyl.

36. The compound of claim 32 or 33, wherein L3 is a C4 alkoxy or C4 substituted aikoxy.

37. The compound of claim 32 or 33, wherein LJ is a C4 amino or C4 substituted amino.

38. A compound comprising Formula XVIII:

Formula XVHI where: n is from 1-12; each A is independently selected from the group consisting of: O and S;

each R' is independently selected from the group consisting of: H and each Rxand R3 is independently selected from the group consisting of: hydrogen, Ci-

Cg alkyl, substituted Ci-C8 alky] , Ci~C8 alkenyl, substituted Ci-C8 alkenyl, C C8 alkynyl, substituted Ci~C8 alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, C3-C10 cycloalkyl, substituted C Cjo cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3- C9 heteroaryl, substituted C3-C9 heteroaryl, C Cg alkoxy, substituted Ci -C8 alkoxy, amino, substituted amino, alkyithio, substituted aikylthio, OH, OR2, NH2, HR2, SR2, substituted suifonyi, and substituted sulfmyl, wherein any R! substituent may be covalentiy bonded to or share another R1 substituent to form a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R2 is independently selected from the group consisting of: hydrogen, C C8 alkyl, substituted Cj-Cg alkyl, Ci-C8 alkenyl, substituted Ci -C8 alkenyl, C i-Cg alkynyl, substituted Ci-C8 alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, OH, Cj-Cg alkoxy, substituted Cj-C8 alkoxy, amino, substituted amino, C3-C10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any natural ly or non-naturally occurring amino acid side chain, wherein any R2 substituent may be covalentiy bonded to or share another R2 substituent to form a C3-C12 cyclic, heterocyclic, aiyl, heteroaryl, spirocyclic, or bicyclic ring system; each W is independently selected from the group consisting of: CH2, C(R2)2, CHR2, O, NH, R2, C(=0), C(=S), S, S(=0), S(=0)2, -C-, -CH-, a C3-C12 spirocyclic group of where m is 1 -19, suifonyi , substituted suifonyi, sulfmyl, and substituted sulfmyl, wherein any two adjacent W atoms may form CR^CR1 or C≡C;

Z is selected from the group consisting of: Cj-Cg alkyl, substituted Ci-C8 alkyl, Cj -C8 alkenyl, substituted Ci-Cg alkenyl, Ci -Cg alkynyl, substituted Ci-Cg alkynyl, C3-C8 alkoxy, substituted Ci-C8 alkoxy, substituted amino, aikylthio, substituted aikylthio, suifonyi, C(R2)2, CHR , O, NH, NR', S, substituted sulfonyl, sulfmyl, substituted sulfmyl ,and a covalent bond,

L4 is selected from the group consisting of: R2, acyl, acyiamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfmyl, thioacyl, aminoacyl, phthalimido, and formyl; and wherein any two or more W groups can be bonded together to form a C3-C12 cycloaikyl ring structure, a C3-C12 fused cycloaikyl ring structure, a C3-C12 aryi ring structure, a C ;-(' ; ·> fused aryi ring structure, a C3-C12 heteroaryl ring structure, a C3-C12 fused heteroaryl ring structure, a C3-C12 heterocyclic ring structure, and a C3-C12 fused heterocyclic ring structure; and wherein Wn does not form an (S) ^ CH^CH C(0)NH group m fae rjng starting from the carbon attached to R2 which is adjacent to the keto-amide or thioketo-amide functional group; or a tautomer and/or a pharmaceutically acceptable salt thereof,

3 '9. The compound of claim 38, wherein R4 is

344

and pharmaceutically acceptable salts thereof.

41. A compound having the staicture or pharmaceutically acceptable salts thereof.

42. A compound from any one of Tables 1-2.

43. A compound from Table 1.

44. A compound from Table 2.

45. A pharmaceutical composition comprising any one of the compounds of claims 1-44 and a pharmaceutically acceptable excipient.

46. A kit comprising a composition including the compound of any one of claims 1 -44 and instructions for use.

47. A method for treating fibrotic disease or a secondary disease state or condition thereof, comprising administration of an effective amount of a composition comprising a compound of any one of claims 1-44 to a subject in need thereof,

48. The method of claim 47, wherein the disease is selected from the group consisting of liver fibrosis, renal fibrosis, lung fibrosis, hypersensitivity pneumonitis, interstitial fibrosis, systemic scleroderma, macular degeneration, pancreatic fibrosis, fibrosis of the spleen, cardiac fibrosis, mediastinal fibrosis, myelofibrosis, endomyocardial fibrosis, retroperitoneal fibrosis, progressive massive fibrosis, nephrogenic systemic fibrosis, fibrotic complications of surgery, chronic allograft vascuiopathy and/or chronic rejection in transplanted organs, ischemic-reperfusion injury associated fibrosis, injection fibrosis, cirrhosis, diffuse parenchymal lung disease, post-vasectomy pain syndrome, and rheumatoid arthritis.

49. The method of claim 47, wherein the treatment decreases the expression level and/or activity of a calpain.

50. The method of claim 49, wherein the calpain is CAPN1, CAPN2, or CAPN9.

51. The method of claim 47, wherein the treatment inhibits myofibroblast differentiation or treats a disease associated with myofibroblast differentiation.

52. The method of claim 47, wherein the treatment inhibits Fibroblast-to-Myofibrobiast Transition (FMT).

53. The method of claim 47, wherein the treatment inhibits Epithelial to Mesenchymal Transition or Endothelial to Mesenchymal Transition.

54. The method of claim 51 , wherein the myofi broblast differentiation is a TGF -mediated my ofib robl ast different! ati on .

55. The method of claim 47, wherein the fibrotic disease is a cancer.

56. The method of claim 55, wherein the cancer is a cancer of epithelial origin,

57. The method of claim 56, wherein the cancer of epithelial origin is selected from the group consisting of breast cancer, basal cell carcinoma, adenocarcinoma, gastrointestinal cancer, lip cancer, mouth cancer, esophageal cancer, small bowel cancer, stomach cancer, colon cancer, liver cancer, brain, bladder cancer, pancreas cancer, ovary cancer, cervical cancer, lung cancer, skin cancer, prostate cancer, and renal cell carcinoma.

58. The method of claim 47, wherein the fibrotic disease is stiff skin syndrome (S S).

59. The method claim 47, wherein the compound is of Formula IV.

60. The method of claim 47, wherein the subject is a mammal. 61, . The method of claim 47, wherein the subject is a human.

62. The method of claim 47, wherein the route of administration is selected from the group consisting of: enteral, intravenous, oral, intraarticular, intramuscular, subcutaneous, intraperitoneal, epidural, transdermal, and transmucosal.

63. The method of claim 47, wherein the administration is intravenous.

64. A method of inhibiting myofibroblast differentiation comprising contacting a cell with a compound of any one of claims 1-44.

65. The method of claim 64, wherein the cell is in a fibrotic tissue.

66. The method of claim 64, wherein the cell is in a cancerous tissue.

67. The method of claim 64, wherein the cell is in a tissue with high TGF signaling.

68. A method for inhibiting calpain, the method comprising contacting a compound of any one of claims 1 -44 with a CAPN1, CAPN2, and/or CAPN9 enzyme residing inside a subject.

69. A method of competitive binding with calpastatin (CAST), the method comprising contacting a compound of any one of claims 1-44 with CAPN1, CAPN2, and/or CAPN9 enzymes residing inside a subject.

70. The method of claim 69, wherein the compound specifically inhibits one or more of the enzymes selected from the group consisting of: CAPNl, CAPN2, and CAPN9 by at least 4- fold.

71. The method of claim 69, wherein the compound specifically inhibits one or more of the enzymes selected from the groups consisting of: CAPN1, CAPN2, and CAPN9 by at least 10- fold.

72. The method of claim 69, wherein the compound specifically inhibits one or more of the enzymes selected from the groups consisting of: CAPN1, CAPN2, and CAPN9 by at least 100-fold.

73. The method of claim 69, wherein the compound inhibits one or more of the enzymes selected from the group consisting of: CAPN1 , CAPN2, and CAPN9 with at least an IC50 value of 15 μΜ.

74. The use of a compound of any one of claims 1-44 for the manufacture of a medicament for treating fibrotic disease.

75. The use of a compound of any one of claims 1-44 for treating fibrotic disease.

76. A pharmaceutical composition comprising a therapeutic compound for treatment, prevention or reduction of a symptom of a fibrotic disease or a fibrotic condition, wherein the therapeutic compound is a calpain modulator present in the composition in an amount effective to alter the fibrotic state of at least one tissue in a human subject to whom the composition is administered.

77. The pharmaceutical composition of claim 76, wherein the therapeutic compound is a selective calpain modulator.

78. The composition of claim 77, wherein the selective calpain modulator comprises a specific calpain-9 modulator.

79. The composition of claim 77, wherein the selective calpain modulator comprises a selective calpain-9 modulator,

80. The composition of claim 77, wherein the selective calpain modulator comprises a selective calpain-9 and calpain- 1 modulator.

81. The composition of claim 77, wherein the selective calpain modulator comprises a selective calpain-9 and calpain-2 modulator.

82. The composition of claim 77, wherein the selective calpain modulator comprises a selective calpain-9 calpain-2 and calpain- 1 modulator.

83. The composition of claim 76, wherein the calpain modulator decreases the expression level and/or activity of caipain-9 in a fibrotic tissue when administered to the subject.

84. The composition of any one of claims 76-83, wherein an alteration of the fibrotic state of at least one tissue comprises one or more of the following: a reduction or reversal of growth of a fibrous tissue, a reduction in fibrogenesis a reduction or halting of a

fibroproliferative state, a reduction in a migration of a fibroblasts or a fibroblast precursor, a reduction in extracellular matrix deposition, a reduction in fibroblast proliferation, a reduction in fibroblast accumulation, a reduction in mesenchymal transition, a reduction in

myofibroblast accumulation, a reduction in migration of fibroblasts, a reduction in migration of myofibroblasts, a reduction in cytokine production, a reduction in growth factor production, a preservation of organ function from progressive decline associated with the fibrotic disease or fibrotic condition a reversal of the fibrosis, a restoration of physiological organ function to a pre -fibrotic state, a reduction in accumulation of fibrotic matrix in a tumor environment, a reduction in cancer growth or tumor burden, an induction of cancer cell senescence, an induction of apoptosis of cancer cells, an enhancement of cancer cell apoptosis, an induction of cancer cell death, an inhibition of angiogenesis, or an inhibition of metastases.

85. The composition of any one of claims 76-83, wherein the fibrotic disease is a disease selected from the group consisting of or that produces a symptom selected from the group consisting of: liver fibrosis, renal fibrosis, lung fibrosis, hypersensitivity pneumonitis, ischemic-reperfusion injury, interstitial fibrosis, systemic scleroderma, chronic allograft vasculopathy and/or chronic rejection in transplanted organs, ischemic-reperfusion injur}' associated fibrosis, macular degeneration, pancreatic fibrosis, fibrosis of organ transplant recipients, fibrosis of the spleen, cardiac fibrosis, mediastinal fibrosis, myelofibrosis, endomyocardial fibrosis, retroperitoneal fibrosis, progressive massive fibrosis, nephrogenic systemic fibrosis, fibrotic complications of surgery, injection fibrosis, cirrhosis, diffuse parenchymal lung disease, post-vasectomy pain syndrome, stiff skin syndrome, and rheumatoid arthritis.

86. The composition of any one of claims 76-83, wherein the fibrotic disease is selected from the group consisting of sarcoma, adenoma, breast cancer, basal cell carcinoma, leiomyoma, neurofibroma, fibroadenoma, glioma, astrocytoma, ependymoma meningioma, neuroma, islet cell tumor, gastrinoma, glucagonoma, VIPoma, sons atostati noma, plasma cell tumor, transitional cell tumor, intramedullary and extramedullar}' tumor, schwannoma, adenocarcinoma, gastrointestinal cancer, lip cancer, mouth cancer, esophageal cancer, small bowel cancer, stomach cancer, colon cancer, liver cancer, brain, bladder cancer, pancreatic cancer, ovarian cancer, cervical cancer, lung cancer, skin cancer, prostate cancer, and renal ceil carcinoma.

87. A method for the treatment, prevention or reduction of a symptom of a fibrotic, sclerotic or post inflammatory disease or condition comprising the step of administering to a human subject in need thereof a pharmaceutical composition comprising a therapeutic compound, wherein the therapeutic compound is a calpain modulator present in the composition in an amount effective to alter the fibrotic, sclerotic or post-inflammatory state of at least one tissue in the human subject.

88. The method of claim 87, wherein the therapeutic compound is a selective calpain modulator.

89. The composition of claim 88, wherein the selective calpain modulator comprises a specific calpain-9 modulator.

90. The composition of claim 88, wherein the selective calpain modulator comprises a selective calpain-9 modulator.

91. The composition of claim 88, wherein the selective calpain modulator comprises a selective calpain-9 and calpain-1 modulator.

92. The composition of claim 88, wherein the selective calpain modulator comprises a selective calpain-9 and calpain-2 modulator.

93. The composition of claim 88, wherein the selective calpain modulator comprises a selective calpain-9 calpain-2 and calpain-1 modulator.

Description:
BACKGROUND

Field of the Invention

[0001] The present disclosure relates to small molecule calpain modulatory compounds, pharmaceutical compositions comprising the compounds, and methods of using the compounds and compositions to modulate calpain activity, including methods to treat diseases and conditions mediated at least in part by the physiologic effects of CAPN1, CAPN2 or CAPN9, or combinations thereof.

Description of the Related Art

[0002] Fibrotic disease accounts for an estimated 45% of deaths in the developed world but the development of therapies for such diseases is still in its infancy. The current treatments for fibrotic diseases, such as for idiopathic lung fibrosis, renal fibrosis, systemic sclerosis, and liver cirrhosis, are few in number and only alleviate some of the symptoms of fibrosis while failing to treat the underlying cause.

[0003] Despite the current limited understanding of the diverse etiologies responsible for these conditions, similarities in the phenotype of the affected organs, across fibrotic diseases, strongly support the existence of common pathogenic pathways. At present, it is recognized that a primary driver of fibrotic disease is a high transforming growth factor-beta (TGFp) signaling pathway which can promote the transformation of normally functioning cells into fibrosis-promoting cells. Termed "myofibroblasts," these transformed cells can secrete large amounts of extracellular matrix proteins and matrix degrading enzymes, resulting in the formation of scar tissue and eventual organ failure. This cellular process is transformative and termed "myofibroblast differentiation" (which includes Epithelial-to-Mesenchymal Transition (EpMT) and its variations like Endothelial-to-Mesenchymal Transition (EnMT) and

Fibroblast-to-Myofibroblast Transition (FMT)), This process is a major target for the treatment of fibrotic diseases. Myofibroblast differentiation has also been shown to occur within cancer cells that have been chronically exposed to high ΊΧϊΡβ, causing stationary epithelial cells to become motile, invasive, and metastasize. Thus, within the context of cancer, the signaling has been documented to associate with the acquisition of drug resistance, immune system evasion, and development of stem cell properties,

[0004] Despite the tremendous potential of myofibroblast differentiation-inhibiting daigs, and the numerous attempts to develop a working treatment, the data gathered thus far has yet to translate into practical therapy. This is partly due to the lack of an ideal target protein. Initial strategies to target the myofibroblast differe tiation process focused on proximal inhibition of the TGFp signaling pathway by various methods, including targeting ligand activators (e.g., alpha-v integrins), ligand-receptor interactions (e.g., using neutralizing antibodies) or TGFp receptor kinase activity (e.g., small molecule chemical compound drugs to block signal transduction). Unfortunately, ΤΟΡβ is a pleiotropic cytokine with many physiological functions such that global suppression of TGF signaling was also associated with severe side effects. Additionally, current data suggests that such proximal inhibition may be vulnerable to pathologic workaround strategies (i.e., due to redundancy or compensation), that would limit the utility of such drags. Further complicating matters is that, in cancer, ΤίίΡβ signaling early on functions as an anti -turn ori genie growth inhibitor but later becomes tumor promoting and is another reason why selective inhibiti on of pathogenic el ements of signaling is so strongly desired. In light of these inherent limitations, current treatment strategies have refocused on identification and inhibition of critical distal events in ΤΟΡβ signaling, which in theory would preferentially target the pathologic, but not physiological functions of ΊΤτΡβ signaling.

SUMMARY OF THE INVENTION

[0005] The inventors have found a series of keto-amide compounds that inhibit CAPNl, CAPN2, and/or CAPN9 and affect a chain of cellular effects so as to elicit therapeutic benefits. In some embodiments, compounds are inhibitors ciapains. In some embodiments, compounds are selective and/or specific calpain inhibitors. In some embodiments, compounds are specific inhibitors of one of: CAPNl, CAPN2 or CAPN9. In some embodiments, compounds are selective inhibitors of one of: CAPNl , CAPN2 or CAPN9. In some embodiments, compounds are selective inhibitors of: CAPNl and CAPN2, or CAPNl and CAPN9, or CAPN2 and CAPN9. In some embodiments, compounds are effective inhibitors of CAPN1 , CAPN2 and/or CAPN9.

[0006] Combination therapy through one molecule is unique in pharmacology since most drugs have only one or two enzymatic targets. Keto-amide compounds of the present invention are broadly effective in treating a host of conditions arising from fibrosis or inflammation, and specifically including those associated with myofibroblast differentiation. Accordingly, compounds of the invention are active therapeutics for a diverse set of diseases or disorders that include or that produces a symptom which include, but are not limited to; liver fibrosis, renal fibrosis, lung fibrosis, hypersensitivity pneumonitis, interstitial fibrosis, systemi c scleroderma, macular degeneration, pancreatic fibrosis, fibrosis of the spleen, cardiac fibrosis, mediastinal fibrosis, myelofibrosis, endomyocardial fibrosis, retroperitoneal fibrosis, progressive massive fibrosis, nephrogenic systemic fibrosis, fibrotic complications of surgery, chronic allograft vasculopathy and/or chronic rejection in transplanted organs, ischemic- reperfusion injury associated fibrosis, injection fibrosis, cirrhosis, diffuse parenchymal lung disease, post- vasectomy pain syndrome, and rheumatoid arthritis diseases or disorders.

[0007] Certain embodiments of the present invention are directed toward using these keto- amide compounds to treat diseases or conditions or that produces a symptom in a subject which include, but not limited to: liver fibrosis, renal fibrosis, lung fibrosis, hypersensitivity pneumonitis, interstitial fibrosis, systemic scleroderma, macular degeneration, pancreatic fibrosis, fibrosis of the spleen, cardiac fibrosis, mediastinal fibrosis, myelofibrosis, endomyocardial fibrosis, retroperitoneal fibrosis, progressive massive fibrosis, nephrogenic systemic fibrosis, fibrotic complications of surgery, chronic allograft vasculopathy and/or chronic rejection in transplanted organs, ischemic-reperfusion injury associated fibrosis, injection fibrosis, cirrhosis, diffuse parenchymal lung disease, post-vasectomy pain syndrome, and rheumatoid arthritis diseases.

[0008] In certain embodiments methods are provided for alleviating or ameliorating a condition or disorder, affected at least in part by the enzymatic activity of CAPN1 , CAPN2, and/or CAPN9, or mediated at least in part by the enzymatic activity of CAPN1, CAPN2, and/or CAPNl wherein the condition includes or produces a symptom which includes: liver fibrosis, renal fibrosis, lung fibrosis, hypersensitivity pneumonitis, interstitial fibrosis, systemic scleroderma, macular degeneration, pancreatic fibrosis, fibrosis of the spleen, cardiac fibrosis, mediastinal fibrosis, myelofibrosis, endomyocardial fibrosis, retroperitoneal fibrosis, progressive massive fibrosis, nephrogenic systemic fibrosis, fibrotic complications of surgery, chronic allograft vasculopathy and/or chronic rejection in transplanted organs, ischemic- reperfusion injury associated fibrosis, injection fibrosis, cirrhosis, diffuse parenchymal lung disease, post-vasectomy pain syndrome, and/or rheumatoid arthritis.

[0009] In some embodiments, the methods, compounds, and/or compositions of the present invention are used for prophylactic therapy.

[0010] In some embodiments, the CAPNl, CAPN2, and/or CAPN9 inhibiting compounds demonstrate efficacy in animal models of human disease. Specifically, in-vivo treatment of mice, rabbits, and other mammalian subjects with compounds of the present invention establish the utility of these compounds as therapeutic agents to modulate CAPNl, CAPN2, and/or CAPN9 activities in humans and thereby ameliorate corresponding medical conditions.

[0011] Some embodiments provide compounds, pharmaceutical compositions, and methods of use to inhibit myofibroblast differentiation. Some embodiments provide compounds, pharmaceutical compositions, and methods of use for inhibiting CAPNl, CAPN2, and/or CAPN9 or combinations of these enzyme activities such as CAPNl and CAPN2, or CAPNl and CAPN9, or CAPN2 and CAPN9. Some embodiments provide methods for treatment of diseases and disorders by inhibiting CAPNl , CAPN2, and/or CAPN9 or combinations of these enzymatic activities.

[0012] Certain embodiments provide novel small molecules that are specific or selective inhibitors of CAPNl, CAPN2, and/or CAPN9 activity, or selective inhibitors of combinations of CAPNl , CAPN2 and CAPN9 activities, e.g., selective inhibitors of CAPNl and CAPN2, or CAPNl and CAPN9, or CAPN2 and CAPN9.

[0013] In certain embodiments, the compound has a structure represented by Formula I:

Formula I where: n is from 1 -12; each A is independently selected from the group consisting of: O and S;

each R 4 is independently selected from the group consisting of: H and each R x and R J is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-Cg alkyl, Ci-Cg aikenyl, substituted Ci-Cg alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C 3 -C 7 aiyl, substituted C 3 -C 7 aryl, cyano, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloal kyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocvcloalkvl, C 3 - C 9 heteroaryl , substituted C3-C9 heteroaryl, Ci-Cg alkoxy, substituted Ci-Cg al koxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR 2 , NH 2 , NHR 2 , SR 2 , substituted sulfonyl, and substituted sulfmyl, wherein any R 1 substituent may be covalently bonded to or share another R 1 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaiyi, spirocyclic, or bicyclic ring system; each R 2 is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-Cg alkyl, C i -Cg alkenyl, substituted Ci-Cg al kenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, OH, Ci-Cg alkoxy, substituted Cj-Cg alkoxy, amino, substituted amino, C 3 -C 10 cycloalkyl, substituted C3-C 10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any substituent may be covalently bonded to or share another R 2 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each W is independently selected from the group consisting of: CH 2 , C(R') 2 , CHR , O,

NH, R 2 , C(=0), C(=S), S, S(=0), where m is 1-19, sulfonyl, substituted sulfonyl, sulfmyl, and substituted sulfmyl, wherein any two adjacent W atoms may form CR CR or C { ' :

L is selected from the group consisting of: R ? , acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethyiene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfmyl, thioacyl, aminoacyl, phthaiimido, and formyl; and wherein any two or more W groups can be bonded together to form a C 3 -C 12 cycloalkyl ring structure, a C Ci 2 fused cycloalkyl ring structure, a C 3 -C 12 aryl ring structure, a C3-C 12 fused aryl ring structure, a C 3 -C 12 heteroaryi ring structure, a C 3 -C 12 fused heteroaryl ring structure, a C 3 -C 12 heterocyclic ring structure, and a C 3 -C 12 fused heterocyclic ring structure; and wherein W n does not form an (S) ^ CH 2 CH 2 C (°) N H group in the ring starting from the carbon attached to R " which is adjacent to the keto-amide or thioketo-amide functional group;

or a tautomer and/or a pharmaceutically acceptable salt thereof.

[0014] Some embodiments include a compound selected from the group consisting of:

pharmaceutically acceptable salt thereof.

[0015] In a related aspect, pharmaceutical compositions are provided that include an effective amount of one or more compounds of Formula I-XVIII described herein and a

pharmaceutically acceptable excipient.

[0016] In certain embodiments, methods are provided for inhibiting CAPNl, CAPN2 or CAPN9 that includes contacting (in vivo or in vitro) cells (including neurons/microglia /invading macrophages) with an effective amount of one or more compounds of Formula I- XVIII described herein.

[0017] In certain embodiments, methods are provided for selectively inhibiting one of:

CAPNl, CAPN2, or CAPN9 that includes contacting (in vivo or in vitro) ceils (including neurons/microglia /invading macrophages) with an effective amount of one or more compounds of Formula I-X VIII described herein. [0018] In other embodiments, methods are provided for selectively inhibiting two of: CAPNI, CAPN2, or CAPN9 (e.g., CAP I and CAPN2, or CAPNI and CAPN9 or CAPN2 and CAPN9) that includes contacting (in vivo or in vitro) ceils (including neurons/mi croglia /invading macrophages) with an effective amount of one or more compounds of Formula I- XVIII described herein.

[0019] In other embodiments, methods are provided for specifically inhibiting one of:

CAPNI, CAPN2, or CAPN9 that includes contacting (in vivo or in vitro) cells (including neurons/microglia /invading macrophages) with an effective amount of one or more compounds of Formula I-XVIH described herein.

[0020] In other embodiments, methods are provided for specifically inhibiting two of:

CAPNI , CAPN2, or CAPN9 (e.g., CAPNI and CAPN2, or CAPNI and CAPN9 or CAPN2 and CAPN9) that includes contacting (in vivo or in vitro) (including neurons/microglia /invading macrophages) with an effective amount of one or more compounds of Formula I- XVIII described herein.

[0021] In an aspect, a method is provided for treating a disease or disorder affected at least in part by CAPNI, CAPN2, and/or CAPN9, where the method comprises administering to a subject in need of treatment an effective amount of one or more compounds of Formula I- XVIII, or a pharmaceutical composition comprising a phannaceuticaliy acceptable excipient and an effective amount of one or more compounds of Formula I-XVDI described herein.

[0022] In one aspect, an article of manufacture is provided for use in inhibiting CAPNI, CAPN2, and/or CAPN9 and treating a disease or disorder affected at least in part by CAPNI, CAPN2, and/or CAPN9, wherein the article comprises a a compound of Formula I-XVDI as provided herein. The diseases affected at least in part by CAPNI, CAPN2, and/or CAPN9 are as provided herein. In some embodiments, the article of manufacture further includes a label with instructions for using the composition to treat a disease or disorder affected at least in part by C APNI, CAPN2, and/or CAPN9,

[0023] These and other embodiments are described in further detail herein.

Q [0024] Compounds of the present invention inhibit calpain. Specifically, these compounds of Formula I-XVUI inhibit CAPN 1, 2, and/or 9. Such compounds are useful for the treatment or therapy of fibrotic and other diseases.

Definitions

[0025] Terms used in the claims and specification are defined as set forth below unless otherwise specified. Further, if any term or symbol used herein is not defined as set forth below, it shall have its ordinary meaning in the art.

[0026] As used herein and in the appended claims, singular articles such as "a," "an" and "the" and similar referents in the context of describing the elements (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value failing within the range, including the upper and lower bounds of the range, unless otherwise indicated herein, and each separate value is incorporated into the

specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplar)' language (e.g., "such as") provided herein, is intended merely to better illuminate the embodiments and does not pose a limitation on the scope of the claims unless otherwise stated. No language in the specification should be construed as indicating any non-claimed element as essential.

[0027] Generally, reference to a certain element such as hydrogen or H is meant to include all isotopes of that element. For example, if an R group is defined to include hydrogen or H, it also includes deuterium and tritium. Compounds comprising radioisotopes such as tritium, C 14 , P 3 and S 35 are thus within the scope of the present technology. Procedures for inserting such labels into the compounds of the present technology will be readily apparent to those skilled in the art based on the disclosure herein.

[0028] As used herein, "a fibrotic disease" includes, for example, liver fibrosis (alcoholic, viral, autoimmune, metabolic and hereditary chronic disease), renal fibrosis (e.g., resulting from chronic inflammation, infections or type II diabetes), lung fibrosis (idiopathic or resulting from environmental insults including toxic particles, sarcoidosis, asbestosis, hypersensitivity pneumonitis, bacterial infections including tuberculosis, medicines, etc.), interstitial fibrosis, systemic scleroderma (autoimmune disease in which many organs become fibrotic), macular degeneration (fibrotic disease of the eye), pancreatic fibrosis (resulting from, for example, alcohol abuse and chronic inflammatory disease of the pancreas), fibrosis of the spleen (from sickle cell anemia, other blood disorders) cardiac fibrosis (resulting from infection, inflammation and hypertrophy), mediastinal fibrosis, myelofibrosis, endomyocardial fibrosis, retroperitoneal fibrosis, progressive massive fibrosis, nephrogenic systemic fibrosis, fibrotic complications of surgery, especially surgical implants, injection fibrosis and secondary conditions and disease states of fibrosis. Secondary conditions and disease states which occur as a consequence of or associated with fibrosis include for example, cirrhosis, diffuse parenchymal lung disease, post-vasectomy pain syndrome and rheumatoid arthritis, among others.

[0029] The term "ameliorating" refers to any therapeutically beneficial result in the treatment of a disease state, e.g., an inflammatory disease state, including lessening in the severity or progression, remission, or cure thereof. In some embodiments, "ameliorating" includes prophylaxis of a disease state,

[0030] The term "in vitro refers to processes that occur in a living cell growing separate from a living organism, e.g., growing in tissue culture.

[0031] The term "/ / vivo" refers to processes that occur in a living organism.

[0032] The term "mammal" as used herein includes both humans and non-humans and include but is not limited to humans, non-human primates, canines, felines, murines, bovines, equines, and porcines.

[0033] The term "sufficient amount" means an amount sufficient to produce a desired effect, e.g., an amount sufficient to modulate protein aggregation in a cell,

[0034] The term "therapeutically effective amount" is an amount that is effective to ameliorate a symptom of a disease. A therapeutically effective amount can, in some embodiments, be a "prophylactically effective amount" as prophylaxis can be considered therapy. [0035] Stereoisomers of compounds (also known as optical isomers) include all chiral, diastereomeric, and racemic forms of a structure, unless the specific stereochemistry is expressly indicated. Thus, compounds used in the present technology include enriched or resolved optical isomers at any or all asymmetric atoms as are apparent from the depictions. Both racemic and diastereomeric mixtures, as well as the individual optical isomers can be isolated or synthesized so as to be substantially free of their enantiomeric or diastereomeric partners, and these stereoisomers are all within the scope of the present technology,

[0036] The compounds of the present technology can exist as solvates, especially hydrates. Hydrates may form during manufacture of the compounds or compositions comprising the compounds, or hydrates may form over time due to the hygroscopic nature of the compounds. Compounds of the present technology can exist as organic solvates as well, including DMF, ether, and alcohol solvates among others. The identification and preparation of any particular solvate is within the skill of the ordinary artisan of synthetic organic or medicinal chemistry, [0037] "Subject" refers to a mammalian organism treated using a compound of the present invention. The "subject" can be a human or non-human mammalian organism.

[0038] "Tautomer" refer to alternate forms of a compound that differ in the position of a proton, such as enol-keto and imine-enamine tautomers, or the tautomeric forms of heteroaryl groups containing a ring atom attached to both a ring NH moiety and a ring =N moiety such as pyrazoies, imidazoles, benzimidazoles, triazoles, and tetrazoles.

[0039] "Treating" or "treatment" of a disease or disorder in a subject refers to 1) preventing the disease or disorder from occurring in a subject that is predisposed or does not yet display symptoms of the disease or disorder; 2) inhibiting the disease or disorder or arresting its development; or 3) ameliorating or alleviating the cause of the regression of the disease or disorder.

[0040] Unless indicated otherwise, the nomenclature of substituents that are not explicitly defined herein are arrived at by naming the terminal portion of the functionality followed by the adjacent functionality toward the point of attachment. For example, the substituent "alkoxycarbonylalkyl" refers to the group (alkoxy)-C(0)-(alkyl)-. [0041] As used herein, the following definitions shall apply unless otherwise indicated. Further, if any term or symbol used herein is not defined as set forth below, it shall have its ordinary meaning in the art.

[0042] As used herein, "naturally occurring amino acid side chain" will be understood to be the substituent of a natural amino acid. Naturally occurring amino acids have a substituent attached to the a-carbon. Naturally occurring amino acids include the list shown below. Charged:

• Arginine - Arg ~ R

• Lysine - Lys - K

• Aspartic acid - Asp - D

• Glutamic acid - Glu - E

Polar (may participate in hydrogen bonds):

• Glutamine - Gin - Q

• Asparagine - Asn - N

• Histidine - His - H

¾ Serine - Ser - S

• Threonine - Thr - T

• Tyrosine - Tyr - Y

• Cysteine - Cys - C

• Methionine - Met - M

• Tryptophan - Trp - W

Hydrophobic (normally buried inside the protein core):

• Alanine - Ala - A

• Isoleucine - Be - 1

• Leucine - Leu - L

• Phenylalanine - Phe - F

• Valine - Val - V * Proline - Pro - P

• Glycine - Gly - G.

[0043] As used herein, "non-naturally occurring amino acid side chain" will be understood to be the substituent of a non-naturally occurring amino acid. Non-naturally occurring amino acids have a substituent attached to the a-carbon. Non-naturally occurring amino acids include the list described below.

Non-natural amino acids include β-amino acids (β 3 and β 2 ), Homo-ami no acids, Proline and Pyruvic acid derivatives, 3-substituted Alanine derivatives, Glycine derivatives. Ring-substituted Phenylalanine and Tyrosine Derivatives, Linear core amino acids and N- methyl amino acids. Exemplary non-natural amino acids are available from Sigma-Aldridge, listed under "unnatural amino acids & derivatives." See also, Travis S. Young and Peter G. Schultz, "Beyond the Canonical 20 Amino Acids: Expanding the Genetic Lexicon," J. Biol. Chem. 2010 285: 11039-1 1044.

[0044] As used herein, the phrase "modulating or inhibiting (the activity of) CAPNl, CAPN2, and/or CAPN9" refers to use of any agent capable of altering the cellular expression levels and/or biological activity of the CAPNl, CAPN2, and/or CAPN9 gene, messenger RNA, or protein. In some embodiments, an agent that modulates or inhibits the biological activity of CAPNl, CAPN2, and/or CAPN9 directly interferes with the expression (such as transcription, splicing, transport, etc.) of the gene encoding the CAPNl , CAPN2, and/or CAPN9 mRNA. In other embodiments, an agent that modulates or inhibits the activity of CAPNl, CAPN2, and/or CAPN9 directly interferes with the biological activity or production of the CAPNl , CAPN2, and/or CAPN9 proteins (such as though inhibition of translation, post-translational modifications, intracellular transport, disruption of interactions between one or more proteins, etc.). In yet other embodiments, an agent that modulates or inhibits the activity of CAPNl, CAPN2, and/or CAPN9 does not directly affect the expression level or activity of CAPNl, CAPN2, and/or CAPN9 but, instead, alters the activity or expression levels of a protein whose function directly impacts the expression or activity of CAPNl, CAPN2, and/or CAPN9 (such as, for example, calpastatin). In other embodiments, an agent may specifically inhibit one or more CAPN l, CAPN2, and/or CAPN9 enzymes. In yet other embodiments, an agent may selectively inhibit CAPNl, CAPN2, and/or CAPN9 enzymes. In yet other embodiments, an agent may both specifically and selectively inhibit one or more CAPNl, CAPN2, and/or CAPN9 enzymes. In yet other embodiments, an agent is neither a specific nor selective inhibitor for one or more CAPNl, CAPN2, and/or CAPN9 enzymes, but is still active for the inhibition of one or more of the CAPN l, CAPN2, and/or CAPN9 enzymes. Calpains are also expressed in cells other than neurons, microglia and invading macrophages. In particular, they are important in skeletal muscle and herein inhibition of calpains also refers to inhibition in these cells as well.

[0045] As used herein, an agent is said to be "specific" if it reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity with a particular target (for example, a protein, such as CAPN9, CAPN2, or CAPNl as well as a nucleic acid encoding the same) than it does with alternative substances, especially as compared to substances that are structurally related to the target, e.g., an isoform of the target. For instance, an antibody "specifically binds ' to a target protein (such as CAPN9, CAPN2, or CAPNl if it binds with greater affinity, avidity, more readily, and/or with greater duration than it binds to other substances,

[0046] In some embodiments, an agent is "specifi c" for a target if a concentration of the agent that produces a maximal effect in an in vitro or in vivo target assay (e.g., a binding assay or an enzyme activity assay) produces no measurable effect in a comparable assay carried out using another substance, especially one or more substances that are structurally related to the target.

[0047] As used herein, an agent is said to be a "specific inhibitor" of CAPNl , CAPN2 or CAPN9 if it inhibits the biological activity and/or expression level of CAPNl, CAPN2 or CAPN9 without inhibiting the biological activity and/or expression level of other members of the calpain family of proteases or other members of the TRP family of calcium channels.

[0048] As used herein, an agent is an agent is said to be "selective" if it reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity with a particular target or a small set of structurally-related targets (for example, a protein, such as CAPN9, CAPN2, or CAPNl as well as a nucleic acid encoding the same) than it does with alternative substances, especially as compared to other substances that are structurally related to the target or small set of structurally-related targets, e.g., an isoform of the target. In certain embodiments, a "selective" agent reacts similarly with multiple related targets, whereas a "specific" agent reacts with its target in a manner that is markedly differently from the way it interacts with other biological molecules. For instance, an antibody "selectively binds" to a target protein (such as CAPN9, CAPN2, or CAPNl if it binds with greater affinity, avidity, more readily, and/or with greater duration to two related targets (e.g., CAPN9, CAPN2) than it binds to other substances.

[0049] In some embodiments, an agent is "selective" for a set of targets if a concentration of the agent that produces a maximal effect in an in vitro or in vivo assay (e.g., a binding assay or an enzyme activity assay) with a first target molecule produces a measurable effect in a comparable assay carried out using a second target molecule. In some embodiments, an agent is selective if it binds to two or more targets (especially structurally-related targets) with Kd or IC50 (or other related measures) ratios of first to second target that are within a range of 1 : 1 to about 1 :500.

[0050] As used herein, in some embodiments, an agent is said to be a "selective inhibitor" of CAPNl , CAPN2 or CAPN9 if it can be shown to inhibit the biological activity and/or expression level of two or three of CAPNl, CAPN2 or CAPN9 (e.g., CAPNl and CAPN2, or CAPNl and CAPN9, or CAPN2 and CAPN9 or CAPNl, CAPN2 and CAPN9) without inhibiting the biological activity and/or expression level of level of other members of the calpain family of proteases or other members of the TRP family of calcium channels.

[0051] As used herein, the term "contacting," as used herein, includes both directly contacting cells, for example, in vivo, in vitro, or ex vivo, or indirectly contacting cells, such as, for example, by administering an agent to a subject. Further, "contacting" a cell with an agent includes administering or applying a prodrug version of the agent. In some embodiments, the cell is in a fibrotic tissue, a cancerous tissue, and/or tissue with high TGFP signaling.

[0052] As used herein, the term "fibrosis" refers to the increased extracellular matrix protein synthesis and deposition that results in the accumulation of scar tissue. Similarly, as used herein, the term "fibrotic tissue" refers to tissue that has high levels of extracellular matrix proteins {i.e., collagen), undergone extensive remodeling (though activity of matrix metalloproteinases) and exhibits progressively diminished physiological function, due to the activity of ceils that have undergone myofibroblast differentiation (such as, EMT and/or FMT). In some embodiments, the cell is in a cancerous tissue, such as in tissue that comprises at least one cancer cell. In some embodiments, the cell is in a tissue with high ΤΟΡβ signaling.

[0053] As used herein, the term "inhibit," "decrease" and grammatical derivations thereof, refers to the ability of an agent to block, partially block, interfere, reduce or deactivate a pathway or mechanism of action. Thus, one of ordinary skill in the art would appreciate that the term "inhibit" or "decrease" encompasses a complete and/or partial loss of activity, e.g., a loss in activity by at least 10%, in some embodiments, a loss in activity by at least 20%, 30%, 50%, 75%, 95%, 98%, and up to and including 100%.

[0054] As used herein, the term "expression level and/or activity of a calpain" refers to the amount of a calpain found in a ceil, tissue and/or subject, and/or a function of a calpain. Such functions can include, without limitation, its protease ability, its function in modulating ΤΟΡβ signaling, its function in myofibroblast transition (such as EMT and/or FMT). In some embodiments, the expression level of a calpain refers to mRNA expression level. In other embodiments, the expression level of a calpain refers to protein expression level. In some embodiments, the myofibroblast transition i s EMT (such as a TGFp-mediated EMT). In some embodiments, at least one agent inhibits Fibroblast-to-Myofibroblast Transition (FMT). In some embodiments, the calpain is CAPN9, CAPN1, and/or CAPN2.

[0055] As used herein, the terms "prevent," "preventing," "prevention," "prophylactic treatment" and the like refer to reducing the probability of developing a disease, disorder, or condition in a subject, who does not have, but is at risk of or susceptible to developing a disease, disorder, or condition. Thus, in some embodiments, an agent can be administered prophylactically to prevent the onset of a disease, disorder, or condition, or to prevent the recurrence of a disease, disorder, or condition.

[0056] For the purposes of this specification and appended claims, unless othemise indicated, all numbers expressing amounts, sizes, dimensions, proportions, shapes, formulations, parameters, percentages, parameters, quantities, characteristics, and other numerical values used in the specification and claims, are to be understood as being modified in all instances by the term "about" even though the term "about" may not expressly appear with the value, amount or range. Accordingly, unless indicated to the contrary, the numerical parameters set forth in the following specification and attached claims are not and need not be exact, but may be approxiraate 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 depending on the desired properties sought to be obtained by the presently disclosed subject matter. For example, the term "about," when referring to a value can be meant to encompass variations of, in some aspects, ± 100% in some aspects ± 50%, in some aspects ± 20%, in some aspects ± 10%, in some aspects ± 5%, in some aspects ±1%, in some aspects ± 0.5%, and in some aspects ± 0.1% from the specified amount, as such variations are appropriate to perform the disclosed methods or employ the disclosed compositions.

[0057] Unless defined otherwise, all technical and scientific terms used herein have the meaning commonly understood by a person skilled in the art to which this invention belongs.

[0058] "Alkyi" refers to monovalent saturated aliphatic hydrocarbyl groups having from 1 to 10 carbon atoms and preferably 1 to 6 carbon atoms. This term includes, by way of example, linear and branched hydrocarbyl groups such as methyl (CH 3 ~), ethyl (CH 3 CH 2 -), «-propyl (CH 3 CH 2 CH 2 -), isopropyl ((CH 3 ) 2 CH-), w-butyl (CH 3 CH 2 CH 2 CH 2 ~), isobutyl

((CH 3 ) 2 CHCH 2 -), sec-butyl ((CH 3 )(CH 3 CH 2 )CH-), /-butyl ((CH 3 ) 3 C-), »-pentyl

(CH 3 CH 2 CH 2 CH 2 CH 2 -), and neopentyl ((CH 3 ) 3 CCH 2 -). (\ alkyi refers to an a k ] group having x number of carbon atoms.

[0059] "Alkenyl" refers to straight or branched hydrocarbyl groups having from 1 to 6 carbon atoms and preferably 2 to 4 carbon atoms and having at least 1 and preferably from 1 to 2 sites of unsaturation (>C=C<). Such groups are exemplified, for example, by vinyl, allyl, and but-3-en-l-yl. Included within this term are the cis and trans isomers or mixtures of these isomers. C x alkenyl refers to an alkenyl group having x number of carbon atoms.

[0060] "Alkynyi" refers to straight or branched monovalent hydrocarbyl groups having from 2 to 6 carbon atoms and preferably 2 to 3 carbon atoms and having at least 1 and preferably from 1 to 2 sites of acetyl enic (-C≡ C-) unsaturation. Examples of such alkynyi groups include acetyl enyl (-C≡ CH), and propargyl (-CH 2 C≡ CH). C x alkynyi refers to an alkynyi group having x number of carbon atoms.

[0061] "Substituted alkyi" refers to an alkyi group having from 1 to 5, preferably 1 to 3, or more preferably 1 to 2 substituents selected from the group consisting of alkoxy, substituted alkoxy, acyl, acyiamino, aminocarbonylamino, acyloxy, amino, substituted amino, aminocarbonyl, aminothiocarbonyl , aminocarbonylamino, aminothiocarbonylamino, aminocarbonyloxy, aminosulfonyl, aminosulfonyloxy, aminosulfonylamino, amidino, aryl, substituted aryl, aryloxy, substituted aiyloxy, arylthio, substituted arylthio, carboxyl, carboxyl ester, (carboxyl ester)amino, (carboxyl ester)oxy, cyano, cycloalkyl, substituted cycloaikyl, cycloalkyloxy, substituted cycloalkyloxy, cycloalkylthio, substituted cycloalkylthio, guanidino, substituted guanidino, halo, hydroxy, heteroaryl, substituted heteroaryl, heteroaryloxy, substituted heteroaryl oxy, heteroarylthio, substituted heteroarylthio, heterocyclic, substituted heterocyclic, heterocyciyloxy, substituted heterocyclyloxy, heterocyclylthio, substituted heterocyclylthio, nitro, S0 3 H, substituted sulfonyl, sulfonyloxy, thioacyl, thiol, alkylthio, and substituted alkylthio, wherein said substituents are defined herein,

[0062] In some embodiments the substituted alkyi groups include halogenated alkyl groups and particularly halogenated methyl groups such as trifluoromethyl, difluromethyi, fluorom ethyl and the like.

[0063] "Cycloalkyl" or "Cyclyl alkyl" refers to a saturated or partially saturated, but not aromatic, group having from 1 to 10 ring carbon atoms and no heteroatoms. Cycloalkyl encompasses single ring or multiple condensed rings, including fused bridged and spiro ring systems. In fused ring systems, one or more of the rings can be cycloalkyl, aryl,

heterocycloalkyl, or heteroaryl provided that the point of attachment is through the original non-aromatic cycloalkyl ring.

[0064] "Substituted alkenyl" refers to alkenyl groups having from J to 3 substituents, and preferably 1 to 2 substituents, selected from the group consisting of alkoxy, substituted alkoxy, acyl, acylamino, aminocarbonylamino, acyloxy, amino, substituted amino, aminocarbonyl, aminothiocarbonyl, aminocarbonylamino, aminothiocarbonylamino, aminocarbonyloxy, aminosulfonyl, aminosulfonyloxy, aminosulfonylamino, amidino, aryl, substituted aryl, aiyloxy, substituted aryloxy, arylthio, substituted arylthio, carboxyl, carboxyl ester, (carboxyl ester)amino, (carboxyl ester)oxy, cyano, cycloalkyl, substituted cycloalkyl, cycloalkyloxy, substituted cycloalkyloxy, cycloalkylthio, substituted cycloalkylthio, guanidino, substituted guanidino, halo, hydroxy, heteroaryl, substituted heteroaryl, heteroaryloxy, substituted heteroaryloxy, heteroarylthio, substituted heteroarylthio, heterocyclic, substituted heterocyclic, heterocyclyloxy, substituted heterocyclyloxy, heterocyclvithio, substituted heterocyclvithio, nitro, S0 3 H, substituted sulfonyl, sulfonyl oxy, thioacyl, thiol, alkylthio, and substituted alkylthio, wherein said substituents are defined herein and with the proviso that any hydroxy or thiol substitution is not attached to a vinyl (unsaturated) carbon atom.

[0065] "Substituted alkynyl" refers to aikynyl groups having from 1 to 3 substituents, and preferably 1 to 2 substituents, selected from the group consisting of alkoxy, substituted alkoxy, acyl, acylamino, aminocarbonylamino, acyloxy, amino, substituted amino, aminocarbonyl, aminothiocarbonyl, aminocarbonylamino, aminothiocarbonylamino, aminocarbonyloxy, aminosulfonyl, aminosulfonyloxy, aminosulfonyl amino, amidino, aryl, substituted aryl, aryloxy, substituted aryloxy, arylthio, substituted arylthio, carboxyl, carboxyl ester, (carboxyl ester)amino, (carboxyl esterjoxy, cyano, cycloalkyl, substituted cycloalkyl, cycloalkyl oxy, substituted cycloalkyl oxy, cycloalkylthio, substituted cycloalkylthio, guanidino, substituted guanidino, halo, hydroxy, heteroaryl, substituted heteroaryl, heteroaryloxy, substituted heteroaryl oxy, heteroarylthio, substituted heteroaryl thio, heterocyclic, substituted heterocyclic, heterocyclyloxy, substituted heterocyclyloxy, heterocyclvithio, substituted heterocyclvithio, nitro, S0 3 H, substituted sulfonyl, sulfonyloxy, thioacyl, thiol, alkylthio, and substituted alkylthio, wherein said substituents are defined herein and with the proviso that any hydroxyl or thiol substitution is not attached to an acetyl enic carbon atom.

[0066] "Ar" refers to any group which is aromatic. This group must be cyclic; however, it may contain heteroatoms or may not.

[0067] "Alkoxy" refers to the group -O-alkyl wherein alkyl is defined herein. Alkoxy includes, by way of example, methoxy, ethoxy, «-propoxy, isopropoxy, w-butoxy, t-butoxy, sec-butoxy, and w-pentoxy.

[0068] "Substituted alkoxy" refers to the group -0-(substituted alkyl) wherein substituted alkyl is defined herein. Preferred substituted alkyl groups in -0-(substituted alkyl) include halogenated alkyl groups and particularly halogenated methyl groups such as trifluoromethyl, diflurom ethyl, fluoromethyl and the like. [0069] "Acyl" refers to the groups H-C(O)-, alkyl-C(O)-, substituted alkyl-C(O)-, alkenyl-C(O)-, substituted alkenyl-C(O)-, alkynyl-C(O)-, substituted alkynyl-C(O)-, cycloalkyl-C(O)-, substituted cycloaikyl-C(O)-, aryl-C(O)-, substituted aryl-C(O)-, heteroaryl -C(O)-, substituted heteroaryl-C(O)-, heterocyclic-C(O)-, and substituted heterocyclic-C(O)-, wherein alkyl, substituted alkyl, alkenyi, substituted alkenyi, alkoxy, substituted alkoxy, aikynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein. Acyl includes the "acetyl" group CH 3 C(0)-.

[0070] "Acylamino" refers to the groups -NR 0 C(G)aikyl, - R 30 C(O)substituted alkyl, - R 30 C(O)cycloaikyl, -NR 30 C(O)substituted cycloalkyl -N R 30 C(O)alkenyl,

-NR 30 C(O)substituted alkenyi, alkoxy, substituted alkoxy-NR 30 C(O)alkynyi,

-NR 30 C(O)substituted alkynyl, -NR 30 C(O)aryl, -NR 30 C(O)substituted and,

-NR 30 C(O)heteroaryl, -NR 30 C(O)substituted heteroaryl, -NR 30 C(O)heterocyclic, and -NR 30 C(O)substituted heterocyclic wherein R 3 ° is hydrogen or alkyl and wherein alkyl, substituted alkyl, alkenyi, substituted alkenyi, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaiyl, substituted heteroaiyl, heterocyclic and substituted heterocyclic are as defined herein.

[0071] "Aminoacyi" refers to the groups H-C(N)-, alkyl-C(N)-, substituted alkyl-C(N)-, alkenyl-C( )-, substituted alkenyl-C(N)-, alkynyl -C(N)-, substituted alkynyl-C(N)-, cycloalkyl -C(N)-, substituted cycloalkyl-C(N)-, aryl-C(N)-, substituted aryl-C(N)-, heteroaiyl -C(N)-, substituted heieroaiyl-C(N)-, heterocyclic-C(N)-, and substituted heterocyclic-C(N)-, wherein alkyl, substituted alkyl, alkenyi, substituted alkenyi, alkoxy, substituted alkoxy, aikynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein. Acyl includes the "acetyl" group CH 3 C(N)-.

[0072] "Acyloxy" refers to the groups alkyi-C(())0-, substituted alkyl-C(0)0-, aikenyl-C(0)0-, substituted alkenyl-C(0)0-, alkynyl-C(0)0-, substituted alkynyl -C(0)0-, aryl-C(0)0-, substituted aryl-C(0)0-, cycloalkyl-C(0)0~, substituted cycloalkyl -C(0)0-, heteroaryl -C(0)0-, substituted heteroaryl-C(0)0-, heterocyclic-C(0)0-, and substituted heterocyclic-C(0)0- wherein alkyl, substituted alkyl, alkenyi, substituted alkenyi, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl , substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein,

[0073] "Amino" refers to the group -NH 2 .

[0074] "Substituted amino" refers to the group -NR l R 32 where R 31 and I 1 are

independently selected from the group consisting of hydrogen, alkyl, substituted alkyl , alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, and substituted sulfonyl and wherein R 3 i and R j2 are optionally joined, together with the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic group, provided that R 3 ' and R 3i are both not hydrogen, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein. When R i is hydrogen and R 32 is alkyl, the substituted amino group is sometimes referred to herein as alkylamino. When R 31 and R 3 are alkyl, the substituted amino group is sometimes referred to herein as dialkylamino. When referring to a rnonosubstituted amino, it is meant that either R 31 or R 32 is hydrogen but not both. When referring to a disubstituted amino, it is meant that neither R 31 nor R 32 are hydrogen.

[0075] "Aminocarbonyl" refers to the group -C(0)NR 3 R 3 " where R 33 and R' 4 are

independently selected from the group consisting of hydrogen, alkyl, substituted alkyl , alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic and where R 33 and R 34 are optionally joined together with the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic group, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.

[0076] "Aminoacyl carbonyloxy" refers to the group -C(NR 33 )OR 34 where R 33 and R 4 are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic and where R j3 and R 34 are optionally joined together with the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic group, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.

[0077] "Arninothiocarbonyi" refers to the group -C(S)NR j3 R 34 where R 33 and R 34 are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic and where R' 3 and R 34 are optionally joined together with the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic group, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.

[0078] "Aminocarbonylamino" refers to the group -NR 3J C(0)NR 3 R 34 where R 30 is hydrogen or alkyl and R 33 and R 34 are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic and where R 33 and R 34 are optionally joined together with the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic group, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.

[0079] "Arninothiocarbonyi amino" refers to the group -NR " C(S)NR " R" where R " is hydrogen or alkyl and R 33 and R j4 are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic and where R j3 and R 34 are optionally joined together with the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic group, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.

[0080] "Aminocarbonyloxy" refers to the group -0-C(0)NR 33 R 34 where R 33 and R j4 are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic and where R' 3 and R 34 are optionally joined together with the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic group, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.

[0081] "Aminosuifonyi" refers to the group -S0 2 R 3 R 3it where R 3 and R' 4 are

independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic and where R 33 and R j4 are optionally joined together with the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic group, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.

[0082] " Aminosuifonyi oxy" refers to the group where R 33 and R "' are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic and where R j3 and R 34 are optionally joined together with the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic group, and wherein aikyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.

[0083] "Aminosulfonylamino" refers to the group -NR J J -S0 2 NR :> R ~ " where R ~ is hydrogen or alkyl and R JJ and R 34 are independently selected from the group consisting of hydrogen, alkyl, substituted aikyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic and where R 3 ' and R 34 are optionally joined together with the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic group, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.

[0084] "Amidino" refers to the group -C(=NR 5 )NR 3 R 34 where R 33 , R 4 , and R 33 are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic and where R 33 and R 34 are optionally joined together with the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic group, and wherein aikyl, substituted alkyl, alkenyl, substituted alkenyl, alkoxy, substituted alkoxy, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.

[0085] "Substituted aryl" refers to aryl groups which are substituted with 1 to 5, preferably 1 to 3, or more preferably J to 2 substituents selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, alkoxy, substituted alkoxy, acyl, acylamino, aminocarbonylamino, acyloxy, amino, substituted amino, aminocarbonyl , aminothiocarbonyl, aminocarbonylamino, aminothiocarbonylamino, aminocarbonyloxy, aminosulfonyl, aminosulfonyloxy, aminosulfonylamino, amidino, aryl, substituted aryl, aryloxy, substituted aryloxy, arylthio, substituted arylthio, carboxyl, carboxyl ester, (carboxyl ester)amino, (carboxyl ester)oxy, cyano, cycloalkyi, substituted cycloalkyi, cycloalkyioxy, substituted cycloalkyloxy, cycloalkylthio, substituted cycloalkylthio, guanidino, substituted guanidino, halo, hydroxy, heteroaryl, substituted heteroaryl, heteroaryloxy, substituted heteroaryloxy, heteroarylthio, substituted heteroarylthio, heterocyclic, substituted heterocyclic, heterocyclyloxy, substituted heterocyclyloxy, heteiOcvclylthio, substituted heteiOcvclylthio, nitro, S0 3 H, substituted sulfonvl, sulfonyloxy, thioacyl, thiol, alkylthio, and substituted alkylthio, wherein said substituents are defined herein.

[0086] "Aryloxy" refers to the group -O-aryl, where aryl is as defined herein, that includes, by way of example, phenoxy and naphthoxy.

[0087] "Substituted aryloxy" refers to the group -0-(substituted aryl) where substituted aryl is as defined herein.

[0088] "Arylthio" refers to the group -S-aryl, where aryl is as defined herein.

[0089] "Substituted arylthio" refers to the group -S-(substituted aryl), where substituted aryl is as defined herein.

[0090] "Carbonyi" refers to the divalent group -C(O)- which is equivalent to -C(=0)-.

[0091] "Carboxy" or "carboxyl" refers to -COOH or salts thereof.

[0092] "Carboxyl ester" or "carboxy ester" refers to the groups -C(0)0-alkyl,

-C(0)0-substituted alkyl, -C(0)0-alkenyl, -C(0)0-substituted alkenyl, -C(0)0-alkynyl, -C(0)0-substituted alkynyl, -C(0)0-aryl, -C(0)0-substituted aryl, -C(0)0-cycloalkyl, -C(0)0-substituted cycloalkyi, -C(0)0-heteroaryl, -C(0)0-substituted heteroaryl,

-C(0)0-heterocyclic, and -C(0)0-substituted heterocyclic wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyi, substituted cycloalkyi, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein. [0093] "(Carboxyl ester)amino" refers to the group -NR. ~ -C(0)0-alkyl,

-NR 30 -C(O)O-substituted alkyl, -NR 30 -C(O)O-alkenyl, -NR 30 -C(O)O-substituted alkenyl, -NR 30 -C(Q)O-alkynyl, -NR 30 -C(O)O-substituted alkynyl, -NR 30 -C(O)O-aryl,

-NR 30 -C(O)O-substituted aryl, - R 30 -C(O)O-cycloalkyl, -NR 30 -C(O)O-substituted cvcloalkyl, - R 30 -C(O)O-heteroaryl, - R 30 -C(O)O-substituted heteroaryl, -NR 30 -C(O)O-heterocyclic, and -NR 30 -C(O)O-substituted heterocyclic wherein R 30 is alkyl or hydrogen, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cvcloalkyl, substituted cvcloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein.

[0094] "(Carboxyl ester)oxy" refers to the group -0-C(0)0-alkyl, -0-C(0)0-substituted alkyl, -0-C(0)0-alkenyl, -0-C(0)0-substituted alkenyl, -0-C(0)0-alkynyl,

-0-C(0)0-substituted alkynyl, -0-C(0)0-aryl, -0-C(0)0-substituted aryl,

-0-C(0)0-cycloalkyl, -0-C(0)0-substituted cvcloalkyl, -0-C(0)0-heteroaryl,

-0-C(0)0-substituted heteroaryl, -0-C(0)0-heterocyclic, and -0-C(0)0-substituted heterocyclic wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cvcloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic are as defined herein.

[0095] "Cyano" refers to the group -C≡N.

[0096] "Cycloalkyl" refers to a saturated or unsaturated but nonaromatic cyclic alkyl groups of from 3 to 10 carbon atoms having single or multiple cyclic rings including fused, bridged, and spiro ring systems. C x cvcloalkyl refers to a cycloalkyl group having x number of ring carbon atoms. Examples of suitable cycloalkyl groups include, for instance, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, and cyclooctyl. One or more the rings can be aryl, heteroaryl, or heterocyclic provided that the point of attachment is through the non-aromatic, non-heterocyclic ring saturated carbocyclic ring, "Substituted cycloalkyl" refers to a cycloalkyl group having from 1 to 5 or preferably 1 to 3 substituents selected from the group consisting of oxo, thione, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, alkoxy, substituted alkoxy, acyl, acylamino, aminocarbonyl amino, acyloxy, amino, substituted amino, aminocarbonyl, aminothiocarbonyl, aminocarbonylamino, aminothiocarbonylamino, aminocarbonyloxy, aminosulfonyl, aminosulfonyloxy, aminosulfonylamino, amidino, aryl, substituted aryl, aryloxy, substituted aryloxy, arylthio, substituted arylthio, carboxyl, carboxyl ester, (carboxyl esteiiamino, (carboxyl ester)oxy, cyano, cycloalkyl, substituted cycloalkyl, cycloalkyloxy, substituted cycloalkyloxy, cycloalkylthio, substituted cycloalkylthio, guanidino, substituted guanidino, halo, hydroxy, heteroaryl, substituted heteroaryl, heteroaryl oxy, substituted heteroaryloxy, heteroarylthio, substituted heteroarylthio, heterocyclic, substituted heterocyclic, heterocyclvloxv, substituted heterocyclvloxv, heterocyclylthio, substituted heterocyclylthio, nitro, S0 3 H, substituted sulfonyl, sulfonyloxy, thioacyl, thiol, alkyithio, and substituted alkylthio, wherein said substituents are defined herein.

[0097] "Cycloalkyloxy" refers to -O-cycloalkyl.

[0098] "Substituted cycloalkyloxy ' " refers to -0-(substituted cycloalkyl).

[0099] "Cycloalkylthio" refers to -S-cycloalkyl.

[00100] "Substituted cycloalkylthio" refers to -S-(substituted cycloalkyl).

[00101 ] "Ethylene glycol" refers to the group -0-CH 2 CH 2 -0-E, wherein E is either H or

CH 3 .

[00102] "Guanidino" refers to the group -\ HC( \ ' ! l )N l k

[00103] "Substituted guanidino" refers to - R 36 C(=NR 36 )N(R 36 ) 2 where each R 36 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, and, substituted aryl, heteroaryl , substituted heteroaryl, heterocyclic, and substituted heterocyclic and two R 36 groups attached to a common guanidino nitrogen atom are optionally joined together with the nitrogen bound thereto to form a heterocyclic or substituted heterocyclic group, provided that at least one R 36 is not hydrogen, and wherein said substituents are as defined herein.

[00104] "Halo" or "halogen" refers to fluoro, chloro, bromo and iodo and preferably is iluoro or chloro,

[00105] "Hydroxy" or "hydroxy!" refers to the group -OH.

[00106] "Heteroaryl" refers to an aromatic group of from 1 to 10 carbon atoms and 1 to 4 heteroatoms selected from the group consisting of oxygen, nitrogen and sulfur within the ring. Such heteroaryl groups can have a single ring (e.g., pyridinyl or furyl) or multiple condensed rings (e.g., indolizinyl or benzothienyl) wherein the condensed rings ma or may not be aromatic and/or contain a heteroatom provided that the point of attachment is through an atom of the aromatic heteroaryl group. In one embodiment, the nitrogen and/or the sulfur ring atom(s) of the heteroaryl group are optionally oxidized to provide for the N-oxide (N→0), sulfinyl, or sulfonyl moieties. Preferred heteroaryl s include 5 or 6 membered heteroaryl s such as pyridinyl, pyrrolyl, indolyl, thiophenyl, and furanyl.

[00107] "Substituted heteroaryl" refers to heteroaryl groups that are substituted with from 1 to 5, preferably 1 to 3, or more preferably 1 to 2 substituents selected from the group consisting of the same group of substituents defined for substituted aryl.

[00108] "Heteroaryl oxy" refers to -O-heteroaryl.

[00109] "Substituted heteroaryl oxy" refers to the group -0-(substituted heteroaryl).

[00110] "Heteroarylthio" refers to the group -S-heteroaryl.

[00111] "Substituted heteroarylthio" refers to the group -S-(substituted heteroaryl).

[00112] "Heterocycle" or "heterocyclic" or "heterocycloalkyl" or "heterocyclvl" refers to a saturated or partially saturated, but not aromatic, group having from 1 to 10 ring carbon atoms and from I to 4 ring heteroatoms selected from the group consisting of nitrogen, sulfur, or oxygen. C x cycloalkyl refers to a heterocycloalkyl group having x number of ring atoms including the ring heteroatoms. Heterocycle encompasses single ring or multiple condensed rings, including fused bridged and spiro ring systems. In fused ring systems, one or more the rings can be cycloalkyl, aryl or heteroaryl provided that the point of attachment is through the non-aromatic ring. In one embodiment, the nitrogen and/or sulfur atom(s) of the heterocyclic group are optionally oxidized to provide for the N-oxide, sulfinyl, sulfonyl moieties,

[00113] "Substituted heterocyclic" or "substituted heterocycloalkyl" or "substituted heterocyclvl" refers to heterocyclvl groups that are substitiited with from 1 to 5 or preferably I to 3 of the same substituents as defined for substituted cycloalkyl.

[00114] "Heterocyclyloxy" refers to the group -O-heterocycyl.

[00115] "Substituted heterocyclyloxy" refers to the group -0-(substituted heterocycyl).

[00116] "Heterocyclvl thio" refers to the group -S-heterocycyl.

[00117] "Substituted heterocyclylthio" refers to the group -S-(substituted heterocycyl).

[00118] Examples of heterocycle and heteroaryl include, but are not limited to, azetidine, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, dihydroindole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthylpyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, phenanthroline, isothl zole, phenazine, isoxazole, phenoxazine, phenothl zine, imidazolidine, imidazoline, piperidine, piperazine, indoline, phthalimide, 1,2,3,4-tetrahydroisoquinoline, 4,5,6,7-tetrahydrobenzo[b]thiophene, thiazoie, thiazolidine, thiophene, benzo[b]thiophene, morpholinyl, thiomorpholinyl (also referred to as thl morpholinyl), l ,l-dioxothiomo holinyl, piperidinyl, pyrrolidine, and tetrahydrofuranyl.

[00119] "Nitro" refers to the group -N0 2 .

[00120] "Oxo" refers to the atom (=0) or (-0 " ).

[00121] "Phthalimido" refers to the group . Phthalimide functional groups are well known in the art and can be generated by covalentiy bonding a nitrogen atom to a C 6 H 4 (CO) 2 group,

[00122] "Polyethylene glycol" refers to the group -0-(CH 2 CH 2 -0) n -E, wherein E is either H or CH 3 , where n is between 2-20,000.

[00123] "Spirocyciic ring system" refers to a ring system with two rings that has a single ring carbon atom in common to both rings. Herein used the term bi cyclic can incorporate up to four heteroatoms in either ring.

[00124] "Bicyclic ring system" refers to a ring system with two rings that has two ring carbon atoms in common, and which can located at any position along either ring. Herein used the term bicyclic ring system can incorporate up to four heteroatoms in either ring.

[00125] "Sulfinyl" refers to the divalent group -SO-.

[00126] "Sulfonyl" refers to the divalent group -S(O)?-.

[00127] "Substituted sulfonyl" refers to the group -S0 2 -alkyl, -S0 2 - substituted alkyl, -SO 2 -OH, -SO 2 -alkenyl, -S0 2 -substituted alkenyl, -S0 2 -cycloalkyl, -S0 2 -substituted cvlcoalkyl, -S0 2 -aryl, -S0 2 -substituted aryl, -S0 2 -heteroaryl, -S0 2 -substituted heteroarvl, -S0 2 - eterocycIic, -S0 2 -substituted heterocyclic, wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein. Substituted sulfonyl includes groups such as m ethyl -S0 2 -, phenyl-S0 2 -, and 4-methylphenyl-S0 2 -. Preferred substituted alky] groups on the substituted alkyl-S0 2 - include halogenated alkyl groups and particularly halogenated methyl groups such as tnfluoromethyl, difluromethyl, fluoromethyl and the like.

[00128] "Substituted sulfmyl" refers to the group -SO-alkyl, -SO-substituted alkyl, -SO-alkenyl, -SO-substituted alkenyl, -SO-cycloalkyl, -SO-substituted cyicoalkyl, -SO-aryl, -SO-substituted aryl, -SO-heteroaryl, -SO-substituted heteroaryl, -SO-heterocyclic,

-SO-substituted heterocyclic, wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaivl, heterocyclic and substituted heterocyclic are as defined herein. Substituted sulfmyl includes groups such as methyl-SO-, phenyl-SO-, and

4-methylphenyl-SO-. Preferred substituted alkyl groups on the substituted alkyl-SO- include halogenated alkyl groups and particularly halogenated methyl groups such as trifluoromethyl, difluromethyl, fluoromethyl and the like.

[00129] "Sulfonyloxy" or "substituted sulfonyl oxy" refers to the group -OS0 2 -alkyl, -OS0 2 -substituted alkyl, -OS0 2 -OH, -OS0 2 -alkenyl, -OS0 2 -substituted alkenyl,

-OS0 2 -cycioalkyl, -OS0 2 -substituted cyicoalkyl, -OS0 2 -aryl, -OS0 2 -substituted aryi, -OS0 2 -heteroaryl, -OS0 2 -substituted heteroaryl, -OS0 2 -heterocyclic, -OS0 2 -substituted heterocyclic, wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.

[00130] "Substitution" or "substitution" generally refers groups which are covalently bonded to an atom to replace a hydrogen atom. The atom in this general context can be a carbon atom or a heteroatom, for example a nitrogen atom.

[00131] "Thioacyl" refers to the groups H-C(S)-, alkyl-C(S)-, substituted alkyl-C(S)-, alkenyl -C(S)-, substituted alkenyl-C(S)-, alkynyl-C(S)-, substituted alkynyl-C(S)-, cycloalkyl-C(S)-, substituted cycloalkyl-C(S)-, aryl-C(S)-, substituted aryl-C(S)-,

heteroaryl-C(S)-, substituted heteroaryl-C(S)-, heterocyclic-C(S)-, and substituted

heterocyclic-C(S)-, wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.

[00132] "Mercapto" or "thiol" refers to the group -SH.

[00133] "Formyl" refers to the group -C(0)H.

[00134] "Thiocarbonyl" refers to the divalent group -C(S)~ which is equivalent to -C(=S)-.

[00135] "Thione" refers to the atom (=S).

[00136] "Alkylthio" refers to the group -S-alkyl wherein alkyl is as defined herein.

[00137] "Substituted alkylthio" refers to the group -S~(substituted alkyl) wherein substituted alkyl is as defined herein. Preferred substituted alkyl groups on -S-(substituted alkyl) include halogenated alkyl groups and particularly halogenated methyl groups such as trifluoromethyl, difluromethyl, fluorom ethyl and the like.

[00138] Stereoisomers of compounds (also known as optical isomers) include all chiral, diastereomeric, and raeemic forms of a structure, unless the specific stereochemistry is expressly indicated. Thus, compounds used in the present technology include enriched or resolved optical isomers at any or all asymmetric atoms as are apparent from the depictions. Both raeemic and d or 1 enriched stereomeric mixtures, as well as the individual optical isomers can be isolated or synthesized so as to be substantially free of their enantiomeric or diastereomeric partners, and these stereoisomers are all within the scope of the present technology.

[00139] The compounds of the present technology may exist as solvates, especially hydrates. Hydrates may form during manufacture of the compounds or compositions comprising the compounds, or hydrates may form over time due to the hygroscopic nature of the compounds. Compounds of the present technology may exist as organic solvates as well, including DMF, ether, and alcohol solvates among others. The identification and preparation of any particular solvate is within the skill of the ordinary artisan of synthetic organic or medicinal chemistry.

[00140] "Tautomer" refer to alternate forms of a compound that differ in the position of a proton, such as enol-keto and imine-enamine tautomers, or the tautomeric forms of heteroaryl groups containing a ring atom attached to both a ring -NH- moiety and a ring =N- moiety such as pyrazoles, imidazoles, benzimidazoies, triazoles, and tetrazoles. [00141] Herein any substituted functional group is substituted at from one to three different positions, and those one to three substituting groups are capable of each independently being substituted at one to three positions, wherein any and each substituting group is independently selected from the group consisting of: halogen, hydroxyl, Cj -Cg alkyl, substituted Ci-Cg alkyl, Ci-Cg alkenyl, substituted Ci-Cg alkenyl, Ci-Cg aikynyl, substituted Ci-Cg alkynyl, acyl, acylamino, aminocarbonylamino, aminoacyl, acyloxy, amino, substituted amino,

aminocarbonyl, aminothiocarbonyl, aminoacyl carbonyloxy, aminocarbonylamino, aminothiocarbonylamino, aminocarbonyloxy, aminosulfonyl, aminosulfonyloxy,

aminosulfonylamino, amidino, Ci-Cg alkoxy, substituted Ci-Cg alkoxy, C3-C7 aryl, substituted C 3 -C aryl, C 3 -C 7 aryloxy, substituted C3-C 7 aryloxy, C 3 -C 7 arylthio, substituted C3-C 7 arylthio, carboxyl, carboxyl ester, (carboxyl ester)amino, (carboxyl ester)oxy, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloaikyl, C 3 -C7 heterocycloalkyi, guanidino, substituted guanidino, C3-C7 heteroaryl oxy, C3-C7 substituted heteroaryloxy, C3-C7 heteroarylthio, C3-C7 substituted heteroarylthio, sulfonyl, substituted sulfonyl, sulfinyl, substituted sulfinyl, sulfonyloxy, substituted sulfonyloxy, thioacyl, alkyl thio, substituted alkyl thio, C3-C7 heteroaryl, and substituted C3-C 7 heteroaryl.

[00142] Herein any and ail heteroaryl and heterocycloalkyi substituents may contain up to four heteroatoms selected from the group consisting of: O, N, and S.

[00143] It is understood that in all substituted groups defined above, polymers arrived at by defining substituents with further substituents to themselves (e.g., substituted aryl having a substituted aryl group as a substituent which is itself substituted with a substituted aryl group, etc.) are not intended for inclusion herein. In such cases, the maximum number of such substituents is three. That is to say that each of the above definitions is constrained by a limitation that each functional group is substituted (at from one to three positions) and that any and all of those substituent groups may be substituted one more time (at from one to three positions).

[00144] It is understood that the above definitions are not intended to include

impermissible substitution patterns (e.g., methyl substituted with 5 fiuoro groups). Such impermissible substitution patterns are well known to the skilled artisan. [00145] Throughout this application, the text refers to various embodiments of the present compounds, compositions, and methods. The various embodiments described are meant to provide a variety of illustrative examples and should not be construed as descriptions of alternative species. Rather, it should be noted that the descriptions of various embodiments provided herein may be of overlapping scope. The embodiments discussed herein are merely illustrative and are not meant to limit the scope of the present technology.

[00146] Compound numbering used herein is merely for convenience and meant for the specific section, scheme, or table in which they appear. They should not be construed or confused with the same numberings, if any, used in other sections, schemes, or tables.

[00147] The present technology is directed to compounds, compositions, and methods of using said compounds or compositions to inhibit CAPN1, CAPN2, and/or CAPN9. Also provided are methods useful in order treating diseases or disorders which are affected at least in part by CAPN1, CAPN2, and/or CAPN9.

[00148] In one aspect, the present technology provides for one or more compounds of a core structure of a mono- or di- amino acid or tri -amino acid wherein the compounds may be substituted by one or more organic functional groups at the C-terminus, N-terminus, and/or the side-chain.

[00149] In some embodiments, the invention is a compound comprising Formula II:

Formula II where: n is from 1 -1 1 ; each R ! and R 3 is independently selected from the group consisting of: hydrogen, Ci-C 8 alkyl, substituted Ci-Cg alkyl, Ci-Cg alkenyl, substituted C Cg alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, C3-C 10 cyeloalkyl, substituted C 3 -C 10 cyeloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C 3 - C9 heteroaryl, substituted C3-C9 heteroaryl, Ci-Cg alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR 2 , H 2 , NHR 2 , SR 2 , substituted sulfonyl, and substituted sulfmyl, wherein any R 1 substituent may be covalentlv bonded to or share another R 1 substituent to form a C3-C 12 cyclic, heterocyclic, aryi, heteroaryl, spirocyclic, or bi cyclic ring system; each R 2 is independently selected from the group consisting of: hydrogen, C]-C 8 alkyl, substituted Ci-Cg alkyl, Cj-Cg alkenyl, substituted Ci-Cg a!kenyl, Ci~Cg alkynyl, substituted C Cg alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, OH, Ci-Cg alkoxy, substituted C Cg alkoxy, amino, substituted amino, C3-C 10 cyeloalkyl, substituted C 3 -C 10 cyeloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R 2 substituent may be covalentlv bonded to or share another R 2 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each W is independently selected from the group consisting of: CH 2 , C(R 2 ) 2 , CUR 2 , O, NH, R 2 , C(=0), C(=S), S, S(=0), S(=0) 2 , -C-, -CH-, a C3-C12 spirocyclic group of where m is 1-19, sulfonyl, substituted sulfonyl, sulfmyl, and substituted sulfmyl, wherein any two adjacent W atoms may form CR ! =CR ! or C≡C;

L 4 is selected from the group consisting of: R ^ , acy!, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosu!fony!, amidino, carboxy ester, -CO-ethylene glycol, -C0- polyethylene glycol, substituted sulfonyl, substituted sulfmyl, thioacyl, aminoacyl,

phthaiimido, and formyl; and wherein any two or more W groups can be bonded together to form a C 3 -C 12 cyeloalkyl ring structure, a C3-C 12 fused cyeloalkyl ring structure, a C3-C 12 aryl ring structure, a C3-C 12 fused aryl ring structure, a C 3 -C 12 heteroaryl ring structure, a C3-C 12 fused heteroaryl ring structure, a C3-C 12 heterocyclic ring structure, and a C3-C 12 fused heterocyclic ring structure; and wherein W n does not form an (S) * 2 2 ' group in the ring starting from the carbon attached to R 2 which is adjacent to the keto-amide functional group; or a tautomer and/or a pharmaceutically acceptable salt thereof.

[00150] In some embodiments, the invention is a compound comprising Formula IH:

Formula III where:

Z 1 is selected from the group consisting of: CH 2 , CiR 1 ^, CUR 1 , O, NH, NR 1 , C(=0),

C(=S), S, S(=0), and S(=0) 2 , a C 3 -C 12 spirocyclic group of where m is 1-19; each R x and R 3 is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Cj -Cg alkyl, C \ -C % alkenyl, substituted Cj-C 8 alkenyl, Ci~C 8 alkynyl, substituted Ci-Cg alkynyl, C 3 -C 7 aryl, substituted C 3 -C 7 aryl, cyano, C -C 10 cycloalkyl, substituted C 3 -C 10 cycloalkyl, C3-C 9 heterocycloalkvl, substituted C3-C9 heterocycloalkvl, C 3 - C 9 heteroaryl, substituted C3-C 9 heteroaryl, Ci-Cg alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, alkyl thio, substituted alkyithio, OH, OR 2 , NH 2 , X! Hi *' , SR 2 , substituted sulfonyl, and substituted sulfinyl, wherein any R f substituent may be covalently bonded to or share another R 1 substituent to fonn a C3-C12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R" is independently selected from the group consisting of: hydrogen, Ci -Cg alkyl, substituted Ci-Cg alkyl, Ci -Cg alkenyl, substituted Ci-Cg alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C3-C7 aryl, substituted C 3 -C 7 aryl, cyano, OH, Ci-C 8 alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, C 3 -C 10 cycloal kyl, substituted C3-C10 cycloalkyl, C3-C 9 heterocycloal kyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R 2 substituent may be covalently bonded to or share another R 2 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system;

L 4 is selected from the group consisting of: R 2 , acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol , substituted sulfonyl, substituted sulfinyl, thioacyl, aminoacyl,

phthaiimido, and formyl ; or a tautomer and/or a pharmaceutically acceptable salt thereof.

[00151] In some embodiments, the invention is a compound comprising Formula IV:

Formula IV where:

Z ! and Z 2 are each independently selected from the group consisting of: CH 2 , C(R 2 ) 2 , CHR 2 , O, NH, NR. 2 , C(=0), Π S ), S, S{ ( )), S(=0) 2 , -C-, -CH-, a C3-C12 spirocyclic group of

«* where m is 1-19, wherein any two adjacent Z atoms may form CR =CR or C≡C; each R and R 3 is independently selected from the group consisting of: hydrogen, C C 8 alkyl, substituted Ci-Cg alkyl, Ci-Cs alkenyl, substituted Ci-Cx alkenyl, Ci-Cg alkynyl, substituted Ci-C 8 alkynyl, C 3 -C 7 aryl, substituted C 3 -C 7 aryl, cyano, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C 3 - C9 heteroaryl, substituted C3-C9 heteroaryl, C \ -C & alkoxy, substituted Ci-C 8 alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR 2 , H 2 , NHR. 2 , SR 2 , substituted sulfony], and substituted sulfinvl, wherein any R substituent may be covalently bonded to or share another R 1 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each 2 is independently selected from the group consisting of: hydrogen, Ci-C 3 alkyl, substituted Ci-C% alkyl, C . -Cs aikenyl, substituted Ci-C% alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C 3 -C 7 aryl, substituted C 3 -C 7 aryl, cyano, OH, Ci-C 8 alkoxy, substituted Ci-C 8 alkoxy, amino, substituted amino, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloalkyl, C 3 -C9 heterocycloalkyl, substituted C 3 -C9 heterocycloalkyl, C 3 -C9 heteroaryl , substituted C 3 -C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R 2 substituent may be covalently bonded to or share another R 2 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system;

L 4 is selected from the group consisting of: R 2 , acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl , amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethyiene glycol, substituted sulfonyl, substituted sulfinyl, thioacyl, aminoacyl, phthalimido, and formyl; and wherein any two or more Z groups can be bonded together to form a C 3 -C 12 cycloalkyl ring structure, a C 3 -C 12 fused cycloalkyl ring structure, a C 3 -C 12 aiyl ring structure, a C 3 -C 12 fused aiyl ring staicture, a C 3 -C 12 heteroaryl ring structure, a C 3 -C 12 fused heteroaryl ring structure, a C 3 -C 12 heterocyclic ring structure, and a C 3 -C 12 fused heterocyclic ring structure; or a tautomer and/or a pharmaceutically acceptable salt thereof.

[00152] In some embodiments, the invention is a compound comprising Formula V :

Formula V where:

Z 1 , Z 2 , and Z 3 are each independently selected from the group consisting of: CH 2 ,

C(R 2 ) 2 , CHR 2 , O, H, R 2 , C(=0), C(=S), S, S(=0), S(=0) 2 , -C-, -CH-, a C3-C12 spirocyciic

or C≡C; each R and R; is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci~C 8 alkyl, C C 8 alkenyl, substituted C Cg alkenyl, Cj-C 8 alkynyl, substituted Cj-C 8 alkynyl, C 3 -C7 aryl, substituted C 3 -C7 aryl, cyano, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloalkyl, C 3 -C9 heterocycloalkyl, substituted C 3 -C9 heterocycloaikyl, C 3 - C9 heteroaryl, substituted C 3 -C9 heteroaryl, Ci-C 8 alkoxy, substituted Ci-C 8 alkoxy, amino, substituted amino, alkylthio, substituted alkyithio, OH, OR 2 , NH 2 , NHR 2 , SR 2 , substituted sulfony], and substituted sulfinyl, wherein any R l substituent may be covalently bonded to or share another R 1 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyciic, or bicyclic ring system; each R 2 is independently selected from the group consisting of: hydrogen, Ci-C 8 alkyl, substituted Ci-C 8 alkyl, Cj-C 8 alkenyl, substituted Ci-C 8 alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C 3 -C 7 aryl, substituted C 3 -C 7 aryl, cyano, OH, Ci-C 8 alkoxy, substituted Ci-C 8 alkoxy, amino, substituted amino, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloalkyl, C 3 -C9 heterocycloalkyl, substituted C 3 -C9 heterocycloalkyl, C 3 -C9 heteroaryl, substituted C 3 -C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R 2 substituent may be covalently bonded to or share another R 2 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyciic, or bicyclic ring system;

L 4 is selected from the group consisting of: R 2 , acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfinyl, thioacyl, aminoacyl,

phthalimido, and formyl; and wherein any and all Z groups do not form an (S) ^ ^^ " ^(Ο) 1 ^ group in the ring starting from the carbon attached to R 2 which is adjacent to the keto-amide functional group, and wherein any two or more Z groups can be bonded together to form a C 3 -C 12 cycloalkyl ring structure, a C3-C 12 fused cycloalkyl ring structure, a C3-C12 aryl ring structure, a C3-C12 fused aryl ring structure, a C3-C12 heteroaryl ring structure, a C3-C12 fused heteroaryl ring structure, a C 3 -C 12 heterocyclic ring structure, and a C3-C . fused heterocyclic ring structure; or a tautomer and/or a pharmaceutically acceptable salt thereof,

[00153] In some embodiments, the invention is a compound comprising Formula VI:

Formula VI where:

Z 1 , Z 2 , Z 3 , and Z 4 are each independently selected from the group consisting of: CH 2 , , CHR 2 , O, NH, NR 2 , C(=0), C(=S), S, S(=0), S(=0) 2 , -C-, -CH-, a C 3 -C 12 spirocyclic group of each R x and R 3 is independently selected from the group consisting of: hydrogen, C C s alkyl, substituted Cj -Cg alkyl, C \ -C% alkenyl, substituted C C* alkenyl, Ci-Cg alkynyl, substituted Ci-C 8 alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C 3 - C 9 heteroaryl, substituted C3-C9 heteroaryl, Ci-C 8 alkoxy, substituted Ci-C 8 alkoxy, amino, substituted amino, alkyl thio, substituted alkylthio, OH, OR 2 , NH 2 , NHR 2 , SR 2 , substituted sulfonyl, and substituted sulfinyl, wherein any R f substituent may be covalently bonded to or share another R 1 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R 2 is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-C 8 alkyl, Cj-Cg alkenyl, substituted Ci-C 8 alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C 3 -C 7 aryl, substituted C 3 -C7 aryl, cyano, OH, Ci-Cg alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycioalkyl, C 3 -C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R 2 substituent may be covalently bonded to or share another R 2 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system,

L 4 is selected from the group consisting of: R 2 , acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfinyl, thioacyl, aminoacyl, phthalimido, and formyl; and wherein any and all Z groups do not form an (S) ^ C^ ^^C 0 )^ group in the ring starting from the carbon attached to R 2 which is adjacent to the keto-amide functional group, and wherein any two or more Z groups can be bonded together to form a C 3 -C 12 cycloalkyl ring structure, a C 3 -C 12 fused cycioalkyl ring structure, a C 3 -C . aryl ring structure, a C 3 -C 12 fused aryl ring staicture, a C 3 -C 12 heteroaryl ring structure, a C 3 -C 12 fused heteroaryl ring structure, a C 3 -C 12 heterocyclic ring structure, and a C 3 -C 12 fused heterocyclic ring structure; or a tautomer and/or a pharmaceutically acceptable salt thereof.

[00154] In some embodiments, the invention is a compound comprising Formula VII:

Formula YH where:

Z l , Z 2 , Z 3 , Z 4 , and Z 5 are each independently selected from the group consisting of: CH 2 , C(R 2 ) 2 , CHR 2 , O, Nil NR 2 , C(=0), C( S ). S, S( O), S( OK -C-, -CH-, a C3-C12 spirocyclic group of where m is 1-19, wherein any two adjacent Z atoms may form

CR^CR 1 or C≡C; each R and R 3 is independently selected from the group consisting of: hydrogen, Ci~C 8 alky], substituted Ci-C 8 alkyl, Ci-C 8 alkenyl, substituted C]-C 8 alkenyl, Ci~C 8 al kynyl, substituted Ci~C 8 alkynyl, C 3 -C 7 aryl, substituted C 3 -C7 aryl, cyano, C 3 -C 10 cycloalkyl, substituted C -C 10 cycloalkyl, C 3 -C9 heterocycloalkyl, substituted C 3 -C9 heterocycloalkyl, C - C9 heteroaryl, substituted C 3 -C9 heteroaryl, Cj -C 8 alkoxy, substituted Cj -C 8 alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR 2 , NH 2 , HR 2 , SR 2 , substituted sulfonyl, and substituted sulfinyl, wherein any R 1 substituent may be covalently bonded to or share another R 1 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bi cyclic ring system; each R 2 is independently selected from the group consi sting of: hydrogen, Ci -C 8 alkyl, substituted Cj -C 8 alkyl, Ci -C 8 alkenyl, substituted Cj -C 8 alkenyl, Ci-C 8 alkynyl, substituted Ci-C 8 alkynyl, C 3 -C7 and, substituted C 3 -C 7 aryl, cyano, OH, Ci-C 8 alkoxy, substituted Ci-C 8 alkoxy, amino, substituted amino, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloalkyl, C 3 -C9 heterocycloalkyl, substituted C 3 -C9 heterocycloalkyl, C 3 -C9 heteroaryl, substituted C 3 -C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R 2 substituent may be covalently bonded to or share another R 2 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system;

L 4 is selected from the group consisting of: R 2 , acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethyiene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted suifmyl, thioacyl, aminoacyl,

phthalimido, and formyl; and wherein any and all Z groups do not form an (S) ^ CH2CH2C (°) NH group in the ring starting from the carbon attached to R 2 which is adjacent to the keto-amide functional group; and wherein any two or more Z groups can be bonded together to form a C 3 -C 12 cycloalkyi ring structure, a C 3 -C 12 fused cycioalkyi ring structure, a C 3 -C 12 aryl ring structure, a C 3 -C 12 fused aryl ring structure, a C3-C 12 heteroaryl ring structure, a C3-C 12 fused heteroaryl ring structure, a C3-C 12 heterocyclic ring structure, and a C 3 -C 12 fused heterocyclic ring structure; or a tautomer and/or a pharmaceutically acceptable salt thereof.

[00155] In some embodiments, the invention is a compound comprising Formula VIH:

Formula where:

Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , and Z 6 are each independently selected from the group consisting of: I 2 , C(R 2 ) 2 , CHR 2 , O, NH, NR 2 , C(=0), C(=S), S, S(=0), S(=0) 2 , -C, -Π I-, a C 3 -C 12 spirocyclic group of where m is 1-19, wherein any two adjacent Z atoms may lorm

CR 1 CR 1 or ( C; each R x and R J is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-Cg alkyl, Ci-Cg aikenyl, substituted Ci-Cg alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C 3 -C 7 aryl, substituted C 3 -C 7 aryl, cyano, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloal kyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C 3 - C 9 heteroaryl, substituted C3-C9 heteroaryl, Ci-Cg alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR 2 , NH 2 , NHR 2 , SR 2 , substituted sulfonyl, and substituted sulfmyl, wherein any R 1 substituent may be covalently bonded to or share another R 1 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R 2 is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-Cg alkyl, Ci -Cg alkenyl, substituted Ci-Cg al kenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C 3 -C 7 aryl, substituted C3-C7 aryl, cyano, OH, Ci-Cg alkoxy, substituted Cj-Cg alkoxy, amino, substituted amino, C 3 -C 10 cycloalkyl, substituted C3-C 10 cycloalkyl, C3-C 9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any substituent may be covalently bonded to or share another R 2 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system;

L 4 is selected from the group consisting of: R ^ , acyl, acylamino, aminothiocarbonyl, aminoacyl carbonvloxv, aminosuifonyi, amidino, carboxy ester, -CO-ethylene glycol, ~CO- polyethylene glycol, substituted sulfonyl, substituted sulfmyl, thioacyl, aminoacyl,

phthalimido, and formyl; and wherein any and all Z groups do not form an (S) ^ CH CH2c (°) NH group in the ring starting from the carbon attached to R 2 which is adjacent to the keto-amide functional group; and wherein any two or more Z groups can be bonded together to form a C 3 -C 12 cycloalkyl ring structure, a C3-C 12 fused cycloalkyl ring structure, a C3-C 12 aryl ring structure, a C3-C12 fused aryl ring structure, a C 3 -C 12 heteroaryl ring structure, a C3-C 12 fused heteroaryl ring structure, a C 3 -C 12 heterocyclic ring structure, and a C 3 -C 12 fused heterocyclic ring structure; or a tautomer and/or a pharmaceutically acceptable salt thereof.

[00156] In some embodiments, the invention is a compound comprising Formula IX:

Formula IX where:

7.} .7.} . ΖΛ Z 4 , ΖΛ Z 6 , and Z ' are each independently selected from the group consisting of: Ci k C(R 2 ) 2 , CH 2 , O, Nil, NR 2 , C( O). Π S), S, S( ()). S( ()),, -C-, -CH-, a C 3 ~C !2 spirocyclic group of where m is 1-19, wherein any two adjacent Z atoms may form

CR ! =CR ! or C≡C; each R and R 3 is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-Cg alkyl, Ci-Cg alkenyl, substituted Cj-Cg alkenyl, Ci-Cg alkynyl, substituted Ci-C 8 alkynyl, C 3 -C 7 aryl, substituted C3-C7 aryl, cyano, C 3 -C 10 cycloalkyl, substituted C3-C 10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3- C 9 heteroaryl, substituted C3-C9 heteroaryl, Cj -Cg alkoxy, substituted Cj -Cg alkoxy, amino, substituted amino, alkyl thio, substituted alkyithio, OH, OR 2 , M k NHR 2 , SR 2 , substituted sulfonyl, and substituted sulfinyf, wherein any R 1 substituent may be covalently bonded to or share another R 1 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R 2 is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-C 8 alkyl, Cj-Cg alkenyl, substituted Ci-C 8 alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C 3 -C 7 aryl, substituted C 3 -C7 aryl, cyano, OH, Ci-Cg alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycioalkyl, C 3 -C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R 2 substituent may be covalently bonded to or share another R 2 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system,

L 4 is selected from the group consisting of: R 2 , acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfinyl, thioacyl, aminoacyl, phthalimido, and formyl; and wherein any and all Z groups do not form an (S) ^ C^ ^^C 0 )^ group in the ring starting from the carbon attached to R 2 which is adjacent to the keto-amide functional group, and wherein any two or more Z groups can be bonded together to form a C 3 -C 12 cycloalkyl ring structure, a C 3 -C 12 fused cycioalkyl ring structure, a C 3 -C . aryl ring structure, a C 3 -C 12 fused aryl ring staicture, a C 3 -C 12 heteroaryl ring structure, a C 3 -C 12 fused heteroaryl ring structure, a C 3 -C 12 heterocyclic ring structure, and a C 3 -C 12 fused heterocyclic ring structure; or a tautomer and/or a pharmaceutically acceptable salt thereof.

[00157] In some embodiments, the invention is a compound comprising Formula X:

Formula X where:

Z ! , Z 2 , Z', Z 4 , Z 3 , Z 6 , Z ', and Z 8 are each independently selected from the group consisting of: CH 2 , C(R 2 ) 2 , CHR 2 , O, NH, NR 2 , C(=0), C(=S), S, S(=0), S(=0) 2 , -C-, -CH-, a

C 3 -C 12 spirocyclic group of where m is 1 -19, wherein any two adjacent Z atoms may form C\V C\V or C≡C; each R and R 3 is independently selected from the group consisting of: hydrogen, Ci-C 8 alkyl, substituted Ci-C 8 alkyl, Ci-Cs aikenyl, substituted Ci-Cx alkenyl, Ci-Cg alkynyl, substituted Ci-C 8 alkynyl, C 3 -C 7 aryl, substituted C 3 -C 7 aryl, cyano, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C 3 - C9 heteroaryl, substituted C3-C9 heteroaryl, Ci-C 8 alkoxy, substituted Ci-C 8 alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR 2 , NH 2 , NHR 2 , SR 2 , substituted sulfonyl, and substituted su!finyl, wherein any R 1 substituent may be covalently bonded to or share another R 1 substituent to form a C3-C 12 cyclic, heterocyclic, aryi, heteroaryl, spirocyclic, or bicyclic ring system; each R 2 is independently selected from the group consi sting of: hydrogen, Ci-Cs alkyl, substituted Ci-C 8 alkyl, C]-C 8 alkenyl , substituted Ci-C 8 alkenyl, Ci-C 8 alkynyl , substituted Ci~C 8 alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, OH, Ci-C 8 alkoxy, substituted Ci~C 8 alkoxy, amino, substituted amino, C 3 -C 10 cycloalkyl, substituted C Cjo cycloalkyl, C 3 -C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R 2 substituent may be covalentiy bonded to or share another R 2 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system;

L is selected from the group consisting of: R 2 , acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfmyl, thioacyl, aminoacyl, phthaiimido, and formyl; and wherein any and all Z groups do not form an (S) ^ c ^ 2Ci ½ c (°) N ^ group in the ring starting from the carbon attached to R 2 which is adjacent to the keto-amide functional group; and wherein any two or more Z groups can be bonded together to form a C 3 -C 12 cycloalkyl ring structure, a C3-C 12 fused cycloalkyl ring structure, a C3-C 12 aryl ring structure, a C3-C 12 fused aryl ring structure, a C 3 -C 12 heteroaryl ring structure, a C 3 -C 12 fused heteroaryl ring structure, a C 3 -C 12 heterocyclic ring structure, and a C 3 -C 12 fused heterocyclic ring structure;

or a tautomer and/or a pharmaceutically acceptable salt thereof. [00158] In some embodiments, the invention is a compound comprising Formula XI:

Formula XI where:

Z l , y. ΖΛ / ' }, Z 5 , ' :, z Z s , and Z 9 are each independently selected from the group consisting of: CH 2 , C(R 2 ) 2 , CHR 2 , O, NH, NR 2 , C( O). C( S), S, S( ()), S( ()}>, -C-, -CH- C3-C 1 2 spirocyclic group of where m is 1- 19, wherein any two adjacent Z atoms may form ( \V ( \V or ( C. each R and R J is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-Cg alkyl, Ci-Cg aikenyl, substituted Ci-Cg alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloal kyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C 3 - C 9 heteroaryl , substituted C3-C9 heteroaryl, Ci-Cg alkoxy, substituted Ci-Cg al koxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR 2 , NH 2 , NHR 2 , SR 2 , substituted sulfonyl, and substituted sulfmyl, wherein any R 1 substituent may be covalently bonded to or share another R 1 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R 2 is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-Cg alkyl, C i -Cg alkenyl, substituted Ci-Cg al kenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, OH, Ci-Cg alkoxy, substituted Cj-Cg alkoxy, amino, substituted amino, C 3 -C 10 cycloalkyl, substituted C3-C10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any substituent may be covalently bonded to or share another R 2 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system;

L 4 is selected from the group consisting of: R ^ , acyl, acylamino, aminothiocarbonyl, aminoacyl carbonvloxv, aminosuifonyi, amidino, carboxy ester, -CO-ethylene glycol, ~CO- polyethylene glycol, substituted sulfonyl, substituted sulfmyl, thioacyl, aminoacyl,

phthaiimido, and formyl; and wherein any and all Z groups do not form an (S) ^ CH CH2c (°) NH group in the ring starting from the carbon attached to R 2 which is adjacent to the keto-amide functional group; and wherein any two or more Z groups can be bonded together to form a C 3 -C 12 cycloalkyl ring structure, a C3-C 12 fused cycloalkyl ring structure, a C3-C 12 aryl ring structure, a C3-C12 fused aryl ring structure, a C 3 -C 12 heteroaryl ring structure, a C3-C 12 fused heteroaryl ring structure, a C 3 -C 12 heterocyclic ring structure, and a C 3 -C 12 fused heterocyclic ring structure;

or a tautomer and/or a pharmaceutically acceptable salt thereof. [00159] In some embodiments, the invention is a compound comprising Formula XII:

Formula XH where:

7.} . 7.} . ΖΛ Z 4 , ΖΛ Z 6 , Z ' , 7 . 7? and Z 10 are each independently selected from the group consisting of: il k C(R 2 ) 2 , CHR 2 , O, NH, NR 2 , C( O). C(=S), S, S( ()), S(=0) 2 , -C-, -CH-, a

( ' ;··( ' ; ·■ spirocyciic group of - where m is 1-19, wherein any two adjacent Z atoms may form CR ! =CR ! or C≡C; each R ! and R 3 is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-Cg alkyl, Ci-Cg alkenyl, substituted Cj-Cg alkenyl, Ci-Cg alkynyl, substituted C \ -C» alkynyl, C 3 -C 7 aryl, substituted C3-C7 aryl, cyano, C 3 -C 10 cycloalkyl, substituted C3-C 10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloaikyl, C3- C 9 heteroaryl, substituted C3-C9 heteroaryl, Cj -Cg alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, alkyl thio, substituted alkyithio, OH, OR 2 , M f ·. NHR 2 , SR 2 , substituted sulfonyl, and substituted sulfmyi, wherein any R 1 substituent may be covalently bonded to or share another R 1 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R 2 is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-C 8 alkyl, Cj-Cg alkenyl, substituted Ci-C 8 alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C 3 -C 7 aryl, substituted C 3 -C7 aryl, cyano, OH, Ci-Cg alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycioalkyl, C 3 -C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R 2 substituent may be covalently bonded to or share another R 2 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system,

L 4 is selected from the group consisting of: R 2 , acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfinyl, thioacyl, aminoacyl, phthalimido, and formyl; and wherein any and all Z groups do not form an (S) ^ C^ ^^C 0 )^ group in the ring starting from the carbon attached to R 2 which is adjacent to the keto-amide functional group, and wherein any two or more Z groups can be bonded together to form a C 3 -C 12 cycloalkyl ring structure, a C 3 -C 12 fused cycioalkyl ring structure, a C 3 -C . aryl ring structure, a C 3 -C 12 fused aryl ring staicture, a C 3 -C 12 heteroaryl ring structure, a C 3 -C 12 fused heteroaryl ring structure, a C 3 -C 12 heterocyclic ring structure, and a C 3 -C 12 fused heterocyclic ring structure;

or a tautomer and/or a pharmaceutically acceptable salt thereof [00160] In some embodiments, the invention is a compound comprising Formula XIII:

Formula XIII where:

Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 , Z ' ', Z 8 , Z 9 , Ζ and Z 11 are each independently selected from th consisting of: CH 2 , C(R 2 ) 2 , CHR 2 , O, NH, NR 2 , C(=0), C(=S), S, S(=0), S(=0) 2 , -C-

CH-, a C3-C 12 spirocyclic group of where m is 1-19, wherein any two adjacent Z atoms may form CR^ R 1 or C≡C; each R x and R 3 is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Cj -Cg alkyl, Ci-Cg alkenyl, substituted Cj-Cg alkenyl, Ci-Cg alkynyl, substituted Ci-C 8 alkynyl, C 3 -C7 aryl, substituted C 3 -C7 aryl, cyano, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloalkyl, C 3 -C heterocycloalkyl, substituted C 3 -C9 heterocycloaikyl, C 3 - C 9 heteroaryl, substituted C 3 -C9 heteroaryl, Ci-C 8 alkoxy, substituted Ci-C 8 alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR 2 , NH 2 , NHR 2 , SR 2 , substituted sulfonyl, and substituted sulfinyl, wherein any R f substituent may be covalently bonded to or share another R 1 substituent to form a C 3 -C 12 cyclic, heterocyclic, aiyl, heteroaryl, spirocyclic, or bicyclic ring system; each R 2 is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-C 8 alkyl, C Cg alkenyl, substituted Ci-C 8 alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C 3 -C 7 aryl, substituted C 3 -C7 aryl, cyano, OH, Ci-Cg alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, C 3 -C 10 cycloal kyl, substituted C 3 -C 10 cycloalkyl, C 3 -C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R 2 substituent may be covalently bonded to or share another R 2 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system;

L 4 is selected from the group consisting of: R 2 , acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfinyl, thioacyl, aminoacyl, phthaiimido, and formyl; and wherein any and all Z groups do not form an (S)

ring starting from the carbon attached to R 2 which is adjacent to the keto-amide functional group; and wherein any two or more Z groups can be bonded together to form a C 3 -C 12 cycloalkyl ring structure, a C 3 -C 12 fused cycloalkyi ring structure, a C 3 -C 12 aryl ring structure, a C3-C 12 fused aryl ring structure, a C 3 -C 12 heteroaryl ring structure, a C 3 -C 12 fused heteroaryl ring structure, a C 3 -C 12 heterocyclic ring structure, and a C 3 -C 12 fused heterocyclic ring structure;

or a tautomer and/or a pharmaceutically acceptable salt thereof. [00161] In some embodiments, the invention is a compound comprising Formula XIV:

Formul where: Z 1 , Z 2 , Z 3 , Z 4 , Z 5 , Z 6 , Z 7 , Z 8 , Z f 0 , Z 11 and Z 12 are each independently selected from the group consisting of: i l k C(R 2 ) 2 , CHR 2 , (). NH, NR 2 , C( ( )). C{ S ). S, Si O h S(=0) 2 , -C-, -

CH-, a C3-C 12 spirocyclic group of where m is 1-19, wherein any two adjacent Z atoms may form CR 1= CR 5 or C≡C: each R ! and R 3 is independently selected from the group consisting of: hydrogen, Ci-C 8 alkyl, substituted Ci~C 8 alkyl, Ci~C 8 alkenyl, substituted Ci-Cg alkenyl, Ci-C 8 alkynyl, substituted Ci-C 8 alkynyl, C3-C 7 aryl, substituted C3-C 7 aryl, cyano, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloalkyl, C3-C 9 heterocycloalkyl, substituted C 3 -C 9 heterocycloalkyl, C 3 - C 9 heteroaryl, substituted C3-C9 heteroaryl, Ci~C 8 alkoxy, substituted Ci~C 8 alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR 2 , NH 2 , NHR 2 , SR 2 , substituted sulfonyl, and substituted sulfmyl, wherein any R l substituent may be covalently bonded to or share another R' substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R 2 is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted C C 8 alkyl, Ci~C 8 alkenyl, substituted Ci-C 8 alkenyl, Cj -C 8 alkynyl, substituted Ci-Cg alkynyl, C3-C 7 aryl, substituted C3-C7 aryl, cyano, OH, Cj -C 8 alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, C3-C 10 cycloalkyl, substituted C 3 -C 10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C 3 -C9 heterocycloalkyl, C3-C9 heteroaiyd, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R 2 substituent may be covalently bonded to or share another R 2 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaiyl, spirocyclic, or bicyclic ring system,

L 4 is selected from the group consisting of: R 2 , acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, ~CO- polyethylene glycol, substituted sulfonyl, substituted sulfmyl, thioacyl, aminoacyl,

phthalimido, and formyl; and wherein any and all Z groups do not form an (S) ^ CH ^ CH2C (°) NH group in the ring starting from the carbon attached to R 2 which is adjacent to the keto-amide functional group, and wherein any two or more Z groups can be bonded together to form a C 3 -C 12 cycloalkyl ring structure, a C3-C 12 fused cycloalkyl ring structure, a C3-C 12 aryl ring structure, a C3-C 12 fused aryl ring structure, a C 3 -C 12 heteroaryl ring structure, a C3-C 12 fused heteroaryl ring structure, a C 3 -C 12 heterocyclic ring structure, and a C 3 -C 12 fused heterocyclic ring structure;

or a tautomer and/or a pharmaceutically acceptable salt thereof. [00162] In some embodiments, the invention is a compound comprising Formula XV:

Formula XV where: n is from 1-12; each A is independently selected from the group consisting of: O and S; each R x and R 3 is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Cj -Cg alkyl, C \ -C % alkenyl, substituted Cj-C 8 alkenyl, Ci~C 8 alkynyl, substituted Ci-Cg alkynyl, C3-C7 aryl, substituted C3-C 7 aryl, cyano, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloalkyl, C3-C 9 heterocycloalkyl, substituted C3-C9 heterocycloalkvl, C 3 - C 9 heteroaryl, substituted C3-C 9 heteroaryl, Ci-Cg alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, alkyl thio, substituted alkylthio, OH, OR 2 , NH 2 , NHR , SR 2 , substituted sulfonyl, and substituted sulfinyl, wherein any R 1 substituent may be covalently bonded to or share another R 1 substituent to form a C3-C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R ^ is independently selected from the group consisting of: hydrogen, Ci -Cg alkyl, substituted Ci-Cg alkyl, Ci -Cg alkenyl, substituted Ci-Cg alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, C 3 -C 7 aryl, substituted C 3 -C 7 aryl, cyano, OH, Ci-C 8 alkoxy, substituted Ci-Cg alkoxy, amino, substituted amino, C 3 -C 10 cycloal kyl, substituted C 3 -C 10 cycloalkyl, C3-C 9 heterocycloal kyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R 2 substituent may be covalently bonded to or share another R 2 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each W is independently selected from the group consisting of: (Ί k C(R ) 2 , CHR 2 , O, NH, NR 2 , C(=0), C(=S), S, S(=0), S(=0) 2 , -C-, -CH-, a C 3 -C 12 spirocyclic group of where m is 1.-19, sulfonyl, substituted sulfonyl, suifinyl, and substituted sulfmyl, wherein any two adjacent W atoms may form CR^CR 1 or C≡C;

Z is selected from the group consisting of: C|-C 3 alkyl, substituted Ci-C 8 alkyl, C|-C 3 alkenyl, substituted Ci-Cs alkenyl, Cj-Cg alkynyl, substituted Ci-Cx alkynyl, Ci-Cs alkoxy, substituted Ci-C 8 alkoxy, substituted amino, alkylthio, substituted alkylthio, sulfonyl, C(R ) 2 , CHR 2 , O, NH, NR 2 , S, substituted sulfonyl, suifinyl, substituted sulfmyl ,and a covalent bond,

L 4 is selected from the group consisting of: R 2 , acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfmyl, thioacyl, aminoacyl,

phthaiimido, and formyl; and wherein any two or more W groups can be bonded together to form a C 3 -C 12 cycloalkyl ring structure, a C3-C 12 fused cycloalkyl ring structure, a C3-C 12 aryl ring structure, a C3-C 12 fused aryl ring structure, a C 3 -C 12 heteroaryl ring structure, a C 3 -Ci 2 fused heteroaryl ring structure, a C 3 -C 12 heterocyclic ring structure, and a C 3 -C 12 fused heterocyclic ring structure; and wherein W n does not form an (S) ^ CH2CH2C (°) NH group in the ring starting from the carbon attached to R 2 which is adjacent to the keto-amide or thioketo-amide functional group; or a tautomer and/or a pharmaceutically acceptable salt thereof.

[00163] In some embodiments, the invention is a compound comprising Formula XVI:

Formula XVI where: n is from 1-12; each R x and R 3 is independently selected from the group consisting of: hydrogen, Ci-C 8 alkyl, substituted Ci-C 8 alkyl, Ci-Cs aikenyl, substituted Ci-C 8 alkenyl, Ci-Cg alkynyl, substituted Ci-C 8 alkynyl, C 3 -C 7 aryl, substituted C 3 -C 7 aryl, cyano, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C 3 - C 9 heteroaryl, substituted C3-C9 heteroaryl, Ci-C 8 alkoxy, substituted Ci-C 8 alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR 2 , NH 2 , NHR 2 , SR 2 , substituted sulfonyl, and substituted sulfinyl, wherein any R 1 substituent may be covalently bonded to or share another R 1 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R 2 is independently selected from the group consi sting of: hydrogen, Ci-C 8 alkyl, substituted Ci-C 8 alkyl, C C 8 alkenyl , substituted Ci-C 8 alkenyl, Ci-Cg alkynyl , substituted Ci~C 8 alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, OH, Ci~C 8 alkoxy, substituted Ci~C 8 alkoxy, amino, substituted amino, C 3 -C 10 cycloalkyl, substituted C Cjo cycloalkyl, C 3 -C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl, substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R 2 substituent may be covalently bonded to or share another R 2 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each W is independently selected from the group consisting of: CH 2 , C(R 2 ) 2 , CUR 2 , O, NH, NR 2 , Π ()}, C(=S), S, S( O h S( ()) . , -C-, -CH-, a C 3 -C 12 spirocyclic group of where m is 1-19, sulfonyl, substituted sulfonyl, sulfinyl, and substituted sulfinyl, wherein any two adjacent W atoms may form CR 1 ==CR 1 or C≡C;

Z is selected from the group consisting of: Ci-Cg alkyl, substituted Cj-Cg alkyl, Ci-Cg alkenyl, substituted Ci-Cg alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl, Ci-Cg alkoxy, substituted Ci-Cg alkoxy, substituted amino, alkylthio, substituted alkylthio, sulfonyl, C(R 2 ) 2 , CHR 2 , O, NH, NR. 2 , S, substituted sulfonyl, sulfinyl, substituted sulfinyl ,and a covalent bond;

L 4 is selected from the group consisting of: R ^ , acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyioxy, aminosulfonyl, amidino, carboxy ester, -CO-ethyiene glycol, -CG- polyethylene glycol, substituted sulfonyl, substituted sulfinyl, thioacyl, aminoacyl,

phthalimido, and formyl; and wherein any two or more W groups can be bonded together to form a C 3 -C 12 cycloalkyl ring structure, a C3-C12 fused cycloalkyl ring structure, a C3-C12 aryl ring structure, a C3-C12 fused aryl ring structure, a C3-C12 heteroaryl ring structure, a C3-C12 fused heteroaryl ring structure, a C3-C12 heterocyclic ring structure, and a C 3 -C 12 fused heterocyclic ring structure; and wherein W„ does not form an (S) ^ C H _: C H 2 C ( 0 ) N H group in the ring starting from the carbon attached to R ^ which is adjacent to the keto-amide functional group; or a tautomer and/or a pharmaceutically acceptable salt thereof

[00164] In some embodiments, the invention is a compound comprising Formula XVII:

^ L 3

Formula XVH where: each A is independently selected from the group consisting of: O and S, each R 4 is independently selected from the group consisting of: H and ; each R ! and R is independently selected from the group consisting of; hydrogen, Ci-C 8 alkyl, substituted Ci-Cg alky], Ci-C 8 alkenyl, substituted C Cg alkenyl, Ci-Cg alkynyl, substituted Cj -Cg alkynyl, C3-C7 aryl, substituted C 3 -C 7 aryl, cyano, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloaikyl, C 3 - C 9 heteroaryl, substituted C3-C9 heteroaryi, Ci-C 8 alkoxy, substituted Ci-C 8 alkoxy, amino, substituted amino, alkylthio, substituted alkyithio, OH, OR "' , NH 2 , NHR 2 , SR 2 , substituted sulfonyl, and substituted sulfmyi, wherein any R ! substituent may be covalently bonded to or share another R 1 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R 2 is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-C 8 alkyl, Cj-Cg alkenyl, substituted Ci-C 8 alkenyl, Cj -Cg alkynyl, substituted Ci-Cg alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, OH, Ci-Cg alkoxy, substituted Cj-Cg alkoxy, amino, substituted amino, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloalkyl, C3-C9 heteroaryl , substituted C3-C9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R 2 substituent may be covalently bonded to or share another R 2 substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system,

L 3 is a group containing 1-8 atoms and is selected from the group consisting of: Ci-Cg alkyl, substituted Ci-C 8 alkyl, Ci-Cg alkenyl, substituted Ci-C 8 alkenyl, Ci-Cg alkynyl, substituted Ci-Cg alkynyl , Ci~C 8 alkoxy, substituted Ci-C 8 alkoxy, amino, substituted amino, alkylthio, substituted al kylthio, sulfonyl , C(R 2 ) 2 , CHR 2 , O, NFL NR 2 , S, substituted sulfonyl, sulfinyl, and substituted sulfmyi;

Z is selected from the group consisting of: C C 8 alkyl, substituted Cj -Cg alkyl, C C 8 alkenyl, substituted Ci-Cg alkenyl, Ci-C 8 alkynyl, substituted Cj-Cg alkynyl, Ci-C 8 alkoxy, substituted Ci-C 8 alkoxy, substituted amino, alkylthio, substituted alkylthio, sulfonyl, C(R 2 ) 2 , CHR 2 , O, NH, NR. 2 , S, substituted sulfonyl, sulfinyl, substituted sulfinyl ,and a covalent bond; L 4 is selected from the group consisting of: R 2 , acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethylene glycol, substituted sulfonyl, substituted sulfinyl, thioacyl, aminoacyl,

phthalimido, and formyl; wherein each L 3 and Z are capable of being covalently bonded to the same or other R 2 functional groups such that a bicyclic or spirocyclic ring system is formed; and wherein L 3 and Z do not form an (S) ^ CH 2 CH 2 C(0)NH g rou p j n† h e r j n g starting from the carbon atom closest to the keto-amide or thio-keto-amide functional group; or a tautomer and/or a pharmaceutically acceptable salt thereof,

[00165] In some embodiments, the invention is a compound comprising Formula Formula XVf l l:

Formula XVIII where: n is from 1-12; each A is independently selected from the group consisting of: O and S;

each R 4 is independently selected from the group consisting of: H and ; each R and R J is independently selected from the group consisting of: hydrogen, Ci-Cg alkyl, substituted Ci-C 8 alkyl, Ci-Cg alkenyl, substituted Ci-Cg alkenyl, Cj-Cg alkynyl, substituted Cj-Cg alkynyl, C 3 -C 7 aryl, substituted C3-C7 aryl, cyano, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloalkyl, C3-C9 heterocycloalkyl, substituted C3-C9 heterocycloaikyl, C 3 - C 9 heteroaryl, substituted C3-C 9 heteroaryl, Ci-C 8 alkoxy, substituted Ci-C 8 alkoxy, amino, substituted amino, alkylthio, substituted alkylthio, OH, OR 2 , NH 2 , NHR 2 , SR 2 , substituted sulfonyl, and substituted sulfinyl, wherein any R 1 substituent may be covalently bonded to or share another R 1 substituent to form a C3-C 1? . cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each R 2 is independently selected from the group consisting of: hydrogen, Ci-C 8 alkyl, substituted d-Cg alkyl, Ci-C 8 alkenyl , substituted d-Cg alkenyl, d-Cg alkyny] , substituted Ci-Cg alkynyl, C3-C7 aryl, substituted C3-C7 aryl, cyano, OH, Ci-Cg al koxy, substituted d-Cg alkoxy, amino, substituted amino, C 3 -C 10 cycloalkyl, substituted C 3 -C 10 cycloalkyl, C3-C 9 heterocyeloalkyl, substituted C3-C9 heterocycloalkyl, C 3 -C 9 heteroaryl, substituted C 3 -C 9 heteroaryl, and any naturally or non-naturally occurring amino acid side chain, wherein any R 2 substituent may be covalently bonded to or share another R ^ substituent to form a C 3 -C 12 cyclic, heterocyclic, aryl, heteroaryl, spirocyclic, or bicyclic ring system; each W is independently selected from the group consisting of: CH 2 , C( ' CHR 2 , O, 2, Π ( )}, C(=S), S, S( O h S( () ) ! : -C-, -CH-, a C3-C spirocyclic group of where m is 1-19, sulfonyl, substituted sulfonyl, sulfinyl, and substituted sulfinyl, wherein any two adjacent W atoms may form CR l =CR l or C≡C;

Z is selected from the group consisting of: d-Cg alkyl, substituted d-Cg al kyl, d-Cg alkenyl, substituted d-Cg alkenyl, d-Cg alkynyl, substituted d-Cg alkynyl, Ci-Cg alkoxy, substituted Ci-C 8 alkoxy, substituted amino, alkylthio, substituted alkylthio, sulfonyl, C(R 2 ) 2 , CHR 2 , O, NH, NR 2 , S, substituted sulfonyl, sulfinyl, substituted sulfinyl ,and a covalent bond;

L 4 is selected from the group consisting of: R 2 , acyl, acylamino, aminothiocarbonyl, aminoacyl carbonyloxy, aminosulfonyl, amidino, carboxy ester, -CO-ethylene glycol, -CO- polyethyiene glycol, substituted sulfonyl, substituted sulfinyl, thioacyl, aminoacyl,

phthalimido, and formyl; and wherein any two or more W groups can be bonded together to form a C 3 -C 12 cycloalkyl ring structure, a C 3 -C 12 fused cycloalkyl ring structure, a C3-C 12 aryl ring structure, a C3-C 12 fused aryl ring staicture, a C 3 -C 12 heteroaryl ring structure, a C 3 -C 12 fused heteroatyl ring structure, a C 3 -C 12 heterocyclic ring structure, and a C 3 -C 12 fused heterocyclic ring structure; and wherein W n does not form an (S)

from the carbon attached to R 2 which is adjacent to the keto-amide or thioketo-amide functional group,

or a tautomer and/or a pharmaceutically acceptable salt thereof.

[00166] In one aspect, the present technology provides two or more compounds of Formula I-XVIII described herein.

[00167] In some embodiments, the compound is of Formula If-XVI!I, wherein all R x and R 3 are I f

[00168] In some embodiments, the compound is of Formula II-XVIII, wherein R 1 is H and all N-R 2 are \-H.

[00169] In some embodiments, the compound is of Fonnula II, wherein W contains a heteroatom .

[00170] In some embodiments, the compound is of Formula Π, wherein W contains an oxygen heteroatom. In some embodiments, the compound is of Formula II, wherein W contains a sulfur heteroatom. In some embodiments, the compound is of Formula II, wherein W contains a nitrogen heteroatom,

[00171] In some embodiments, the compound is of Formula II, wherein W contains sulfonyl functional group.

[00172] In some embodiments, the compound is of Formula II, wherein one R l is an alkyl functional group.

[00173] In some embodiments, the compound is of Formula Π, wherein one R 1 is an aromatic functional group.

[00174] In some embodiments, the compound is of Formula IE, wherein one R f is an alkyl functional group.

[00175] In some embodiments, the compound is of Formula IH, wherein Z l is nitrogen.

[00176] In some embodiments, the compound is of Formula III, wherein Z ! is CH ? . 77] In some embodiments, the compound is of Formula III, wherein one R on the carbon atom is H and the other is CH(CH 3 ) 2 .

] In some embodiments, the compound is of Formula III, wherein one R 2 on the carbon atom is H and the other is CH 2 -CH(CH 3 ) 2 .

] In some embodiments, the compound is of Formula III, wherein one R 2 on the carbon atom is H and the other is Ar.

[00180] In some embodiments, the compound is of Formula III, wherein one L 4 forms a carbamate functional group.

[00181] In some embodiments, the compound is of Formula III, wherein one L 4 forms a sulfamate functional group.

[00182] In some embodiments, the compound is of Formula III, wherein one L 4 is - C(0)C(CH 3 ) 3 .

[00183] In some embodiments, the compound is of Formula ΙΠ, wherein one L 4 is - C(0)C(CH 3 ) 3 .

[00184] In some embodiments, the compound is of Formula IV, wherein Z 1 is CH 2 and R' is H.

85] In some embodiments, the compound is of Formula IV, wherein Z 1 is CH 2 , Z is 2, and one R 1 is H.

186] In some embodiments, the compound is of Formula IV, wherein Z ' is CH 2 , Z is O, and one R 1 is H.

[00187] In some embodiments, the compound is of Formula V, wherein Z 1 is CH 2 , Z 2 is NH, and both R 1 are H.

[00188] In some embodiments, the compound is of Formula VI, wherein Z 1 is CH 2 , ΐ ' is

NH, and both R 1 are H.

89] In some embodiments, the compound is of Formula III-XIV, wherein R 1 is H, both

O

N- are N-H, and L 4 is [00190] In some embodiments, the compound is of Formula III-XIV, wherein both R 1 are

H, both N-R 2 are N-H, and L 4 is

[00191 j In some embodiments, the compound is of Formula III-XIV, wherein both R 1 are

O

H, both N-R 2 are N-H, and L 4 is Ph^O-¾

[00192] In some embodiments, the compound is of Formula III-XIV, wherein both IV are,

O both N-R 2 are N-H, and L 4 is .

[00193] In some embodiments, the compound is of Formula IH-XIV, wherein both R 1 are

H, both N-R 2 are N-H, one R 2 of R2 is H and the other is H ° CH' , and L 4 is

O

O"

[00194] In some embodiments, the compound is of Formula III-XIV, wherein both R x are, both N-R' are N-H, one R 2 of K is H and the other is HsC CHs , and L 4 is

[00195] In some embodiments, the compound is of Formula III-XIV, wherein both R 1 are, both N-R 2 are N-H, one R 2 of R2 r2 is H and the other is H3 ° Ch3 , and L 4 I S

[00196] In some embodiments, the compound is of Formula III, wherein both IV are, both

r ·. 1 O

N-R 2 are N-H, one R 2 of R2 ^ 2 is H and the other is H 3 C ^ C H 3 ; a „d L 4 is Ar-^O

[00197] In some embodiments, the compound is of Formula IH, wherein both R 1 are, both

N-R 2 are N-H, one R 2 of R2 is H and the other is 98] In some embodiments, the compound is of Formula III, wherein one R 1 is H, both

O

N-R 2 are N-H, one R 2 of R2 ^ 2 is H and the other is H i ° CHa , and L is ^ .

[00199] In some embodiments, the compound is of Formula IV, wherein one R 1 is H, both

N-R 2 are N-H, one R 2 of R2 ^ 2 is H and the other is H3C CH3 , and L 4 is Ar/ O '

[00200] In some embodiments, the compound is of Formula TV, wherein both R ! are H, both N-R 2 are N-H, one R 2 of

[00201] In some embodiments, the compound is of Formula IV, wherein both R 1 are H,

O both N-R 2 are N-H, one R 2 of R2 2 is H and the other is H 3 C CH 3 , and L 4 is ^ .

[00202] In some embodiments, the compound is of Formula V, wherein both R 1 are H, 1

both N-R ^ are N-H, one R " of K is H and the other is 3 3 , and L is

! 3] In some embodiments, the compound is of Formula V, wherein one R 1 is H, both

are N-H, one R 2 of

] In some embodiments, the compound is of Formula V, wherein both R 1 are H,

both N-R 2 are N-H, one R 2 of

[00205] In some embodiments, the compound is of Formula VI, wherein both R 1 are H, both N-R 2 are N-H, one R 2 of R * 2 is H and the other is H' ° CHa , and L 4 is

O [00206] In some embodiments, the compound i s of Formula VI, wherein both R 1 are H, both N-R 2 are N-H, one R 2 of

[00207] In some embodiments, the compound is of Formula VI, wherein both R ! are H,

both N-R 2 are N-H, one R 2 of .

[00208] In some embodiments, the compound is of Formula VII, wherein both R : are H, both N-R 2 are N-H, one R 2 of R " is H and the other is CHz , and L 4 is

[00209] In some embodiments, the compound is of Formula VII, wherein both R 1 are H, both N-R 2 are N-H, one R 2 of R " r2 is H and the other is CH 3 , and L 4 is

[00210] In some embodiments, the compound is of Formula VII, wherein both R 1 are H,

O

^λ' Af

both N-R 2 are N-H, one R 2 of R ^ ^ 2 is H and the other is H 3 C CH3 , and L 4 is ^ .

[00211] In some embodiments, the compound is of Formula ΪΠ-XIV, wherein one I 1 is H,

both N-R 2 are N-H, one R 2 of .

[00212] In some embodiments, the compound is of Formula ΙΠ-XIV, wherein one R f is H,

both N-R 2 are N-H, one R 2 of R " 2 is H and the other is oh3 , and L 4 is Ph °' f " some embodiments, the compound is of Formula III-XIY, wherein one R 1 is H,

-H, one R ' of R r2 is H and the other is , and L 4 is

[00214] In some embodiments, the compound is of Formula lil-XIV, wherein one R is H, of is H and the other is selected from the group consisting

[00215] Ei some embodiments, the compound is of Formula ΙΠ-Χΐν, wherein both R ! are

H, both N-R z are N-H, one R l of ^ K' is H and the other is selected from the group

HQ

[00216] In some embodiments, the compound is of Formula Ili-XIV, wherein both R 1 are

H, both N-R' r -H n R of K is H and the other is selected from the consisting j o off: [00217] In some embodiments, the compound is of Formula V-XIV, wherein both R 1 are

! °

H, all \ -R : are N-H, and L 4 is ' ° ^ .

[00218] In some embodiments, the compound is of Formula V-XIV, wherein one iV is H, all N-R 2 are N-H, and L 4 is

[00219] In some embodiments, the compound is of Formula V-XIV, wherein one R 1 is H,

O

all N-R 2 are N-H, and L 4 is Ph '^ ' °

[00220] In some embodiments, the compound is of Formula V-XIV, wherein one R 1 is H,

O all N-R 2 are N-H, and L 4 i s & .

[00221] In some embodiments, the compound is of Formula V-XIV, wherein one R 1 is H,

X

7 R 2 p2

all N-R" are N-H, two R ' of the two K groups on separate carbon atoms are H and one of the other two R groups is

[00222] In some embodiments, the compound is of Formula V-XIV, wherein one R 1 is H, all N-R 2 are N-H, two R of the two R K ' groups on separate carbon atoms are H and one

O

H C"^ CH Λ Ph'^^-d"'^^

the other two R groups is 3 3 , and L * is η υ .

[00223] In some embodiments, the compound is of Formula V-XIV, wherein both R ! are

H, all -R " are N-H, two " of the two K groups on separate carbon atoms are H and

one o . [00224] In some embodiments, th ound is of Formula ΧΙΠ, wherein both R x are H, all N-R" are N-H, two R ' of the two groups on separate carbon atoms are H and one of the other two R groups is

[00225] In some embodiments, the compound is of Formula XIH, wherein both R 1 are H, all N-R 2 are N-H, two R of the two R K ' groups on separate carbon atoms are H and one of the other two R groups is

[00226] In some embodiments, the compound is of Formula XIII, wherein one R ! is H, all

N-R are N-H, two R of the two K groups on separate carbon atom s are H and one of

the other two R z groups is .

[00227] In some embodiments, the compound is of Formula XIV, wherein one R 1 is H, all -R " are N-H, two R of the two groups on separate carbon atoms are H and one of

i O

the other two R z groups is 3 3 , and L7 is A u

[00228] In some embodiments, the compound is of Formula XIV, wherein both R 1 are H, both N-R" are N-H, two R" of the two K groups on separate carbon atoms are H and one or the other two R groups i s .

[00229] In some embodiments, the compound is of Formula XIV, wherein both R 1 are H, all N-R 2 are N-H, two R of the two K K ' groups on separate carbon atoms are H and one of

o the other two R 2 groups is HaC CH3 , and L 4 is [00230] In some embodiments, the compound is of Formula XV, wherein both R 1 are H, all

N-R ^ are N-H, two R 2 of the two R K' groups on separate carbon atoms are H and one of the other two R groups is

[00231] In some embodiments, the compound is of Formula XV, wherein both R 1 are H, all

N-R are N-H, two R " of the two groups on separate carbon atoms are H and one of the other two R groups is

[00232] In some embodiments, the compound is of Formula XV, wherein both R 1 are H, all

N-R are N-H, two R of the two K groups on separate carbon atom s are H and one of

the other two R z groups is .

[00233] In some embodiments, the compound is of Formula XVI, wherein both R 1 are H, all N-R" are N-H, two R z of the two K ' groups on separate carbon atoms are H and one of

i O

the other two R z groups is 3 3 , and L7 is A u

[00234] In some embodiments, the compound is of Formula XVI, wherein both R 1 are H, all N-R 2 are N-H, one R 2 of

5] In some embodiments, the compound is of Formula XVI, wherein R l is H, all N-R 2 are N-H, two R of the two K ' groups on separate carbon atoms are H and one of the

O ΑΓ-¾- -

., |-| C'^^CH 4

other two R" groups is 3 3 , and L is υ . [00236] In some embodiments, th ound is of Formula XVII, wherein both R 1 are H, all N-R" are N-H, two R ' of the two groups on separate carbon atoms are H and one of the other two R groups is

[00237] In some embodiments, the compound is of Formula XVII, wherein both R 1 are H, all N-R 2 are N-H, two R of the two R K ' groups on separate carbon atoms are H and one of the other two R groups is

[00238] In some embodiments, the compound is of Formula XVII, wherein one R 1 is H, all

N-R are N-H, two R of the two K groups on separate carbon atom s are H and one of

the other two R z groups is .

[00239] In some embodiments, the compound is of Formula V-XIV, wherein one R 1 is H, all N-R " are N-H, two R z of the two K ' groups on separate carbon atoms are H and one of

the other two R 2 groups is Hs , and L 4 is

[00240] In some embodiments, the compound is of Formula V-XIV, wherein both R 1 are

H, all N-R are N-H, two R " of the two K groups on separate carbon atoms are H and

one of the other two R 2 groups is Hs , and L 4 is some embodiments, the compound is of Formula V-XVUI, wherein both R x are all N-R are N-H, two R " of th groups on separate carbon atoms are H and

one of the other two R 2 groups is and L 4 is .

[00242] In some embodiments, the compound is of Formula V-XV III, wherein one R 1 is H, all N-R" are N-H, two R" of the two K groups on separate carbon atoms are H and one of

the other two R groups is selected from the group consisting of:

¾j and L 4 1S / -(X

[00243] In some embodiments, the compound is of Formula V-XVIII, wherein one R ! is H, all N-R are N-H, two R of the two groups on separate carbon atoms are H and one of

the other two R" groups is selected from the group consisting of: ?

[00244] In some embodiments, the compound is of Formula V-XVUI, wherein both R 1 are

H, all N-R are N-H, two R " of the two K groups on separate carbon atoms are H and

one of the other two R ? groups is selected from the group consisting of: ^ ,

[00245] In some embodiments, the present technology includes a compound which is selected from the compound of the tables below:

TABLE 1

Macrocycles



80

81

82

83

85

88

91

93

96

98

99

100

101

or a tautomer and/or a pharmaceutically acceptable salt thereof.

108

109

110

112

113

114

115

116

130

131

ı33







ı44

150

151

ı52

ı53

ı54

ı55





160

161

ı62

ı64

ı66

 267 268

211

275

280

Ϊ84

288

292

296 299 500

503 304

307 308

312

ı73 331 (32

335 336

339 340

343 344 347 548

351 35

355 356

359 360

ı76 379

384

391 392





180 447 448

451 452

455 456

59 460 462 464

467 468

471 472

475 476 479 480

484

487 488

491 492

ı84

684

692

696

ı96 7

748

ı98

ı99

200

201

202

autorner and/or a pharmaceutically acceptable salt thereof.

[00246] in some embodiments, the present technology is a pharmaceutical composition comprising one or more compounds disclosed herein and a pharmaceutically acceptable excipient.

[00247] In one embodiment, the compounds of the present invention or a pharmaceutically acceptable salt thereof!, or a prodrug or metabolite thereof, can be administered in

combination with any other pharmaceutical compound approved for treating fibrotie or myofibroblast differentiation associated diseases or disorders.

[00248] The compounds represented by Formula I-XVIH or their tautomers and/or pharmaceutically acceptable salts thereof can effectively act as CAPNL CAPN2, and/or CAPN9 inhibitors and treat conditions affected at least in part by CAPNl, CAPN2, and/or CAPN9. In one aspect of the present technology, the present technology provides pharmaceutical compositions comprising one or more compounds of Formula I-XVIII and a pharmaceutically acceptable excipient. In another aspect of the present technology, the present technology provides a method for treating a fibrotie disease with an effective amount of one or more compound of Formula I-XVIII as provided herein.

[00249] In another aspect of the present technology, the present technology provides a method for inhibiting CAPNl, CAPN2, and/or CAPN9 and/or a method for treating a disease affected at least in part by CAPNl, CAPN2, and/or CAPN9 with an effective amount of one or more compound of Formula I-XVIII as provided herein. [00250] The compounds of the present technology are useful in inhibiting CAPN l, CAPN2, and/or CAPN9 enzymes and/or treating disorders relating to fibrosis or

my ofib robl ast differ enti ation .

[00251] In one of its method aspects, the present technology is directed to a method for inhibiting CAPNl, CAPN2, and/or CAPN9 which method comprises contacting cells (including neurons/microglia /invading macrophages) with an effective amount of one or more compound of Formula I-XVIII as described herein,

[00252] In another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds of Formula I-XVDI or a pharmaceutical composition comprising a pharmaceutically acceptable excipient.

[00253] In another aspect, the present technology is directed to a method for treating a disease affected at least in part by CAPNL CAPN2, and/or CAPN9, which method comprises administering to a subject an effective amount of one or more compounds of Formula I-XVIII or a pharmaceutical composition comprising a pharmaceutically acceptable excipient.

[00254] In an aspect, a method for inhibiting CAPNl, CAPN2, and/or CAPN9 is provided wherein the method comprises contacting cells with an effective amount of one or more compounds of Formula I-XVIH. In one aspect, the method for inhibiting CAPNl, CAPN2, and/or CAPN9 is performed in-vitro or in-vivo.

[00255] Calpains are also expressed in cells other than neurons, microglia and invading macrophages. In particular, they are important in skeletal muscle and herein inhibition of calpains also refers to inhibition in these cells as well.

Selective inhibition

[00256] In another aspect, a method is provided for competitive binding with caipastatin (CAST), the method comprising contacting a compound of any one of claims 1-56 with CAPNl, CAPN2, and/or CAPN9 enzymes residing inside a subject. In such a method, the compound specifically inhibits one or more of the enzymes selected from the group consisting of: CAPNl, CAPN2, and CAPN9 by at least 2-fold, by at least 3-fold, by at least 4-fold, by at least 5-fold, by at least 10-fold, by at least 15-fold, by at least 20-fold, by at least 50-fold, by at least 100-fold, by at least 150-fold, by at least 200-fold, by at least 400-fold, or by at least 500-fold. [00257] In another aspect, a method is provided for selectively inhibiting CAPN1 in the presence of CAPN2 and CAPN9, which includes contacting cells (including

neurons/microglia /invading macrophages) with an effective amount of one or more compounds of Formula I-XVIII described herein.

[00258] In another aspect, a method is provided for selectively inhibiting CAPN2 in the presence of CAPNl and CAPN9, which includes contacting cells (including

neurons/microglia /invading macrophages) with an effective amount of one or more compounds of Formula I-XVUI described herein.

[00259] In another aspect, a method is provided for selectively inhibiting CAPN9 in the presence of CAPN2 and CAPNl , which includes contacting cells (including

neurons/microglia /invading macrophages) with an effective amount of one or more compounds of Formula I-XVUI described herein.

[00260] In another aspect, a method is provided for selectively inhibiting CAPNl and CAPN2 in the presence of CAPN9, which includes contacting cells (including

neurons/microglia /invading macrophages) with an effective amount of one or more compounds of Formula I-XVIII described herein.

[00261] In another aspect, a method is provided for selectively inhibiting CAPNl and CAPN9 in the presence of CAPN2, which includes contacting cells (including

neurons/microglia /invading macrophages) with an effective amount of one or more compounds of Formula I-XVIII described herein.

[00262] In another aspect, a method is provided for selectively inhibiting CAPN2 and CAPN9 in the presence of CAPNl , which includes contacting cells (including

neurons/microglia /invading macrophages) with an effective amount of one or more compounds of Formula I-XVUI described herein.

[00263] In another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds which specifically inhibits CAPNl , CAPN2, and/or CAPN9, said compounds being selected from Formula I-XVIII or a pharmaceutical composition comprising one or more compounds from Formula I-XVIII and a pharmaceutically acceptable excipient.

[00264] In another aspect, the present technology is directed to a method for treating a disease affected at least in part by CAPNl, CAPN2, and/or CAPN9, which method comprises administering to a subject an effective amount of one or more compounds which specifically inhibits CAPN1, CAPN2, and/or CAPN9, said compounds being selected from Formula I- XVIII or a pharmaceutical composition comprising one or more compounds from Formula I- XVIH and a pharmaceutically acceptable excipient.

[00265] in another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds which selectively inhibits CAPN1 , CAPN2, and/or CAPN9, said compounds being selected from Formula I-XVIQ or a pharmaceutical composition comprising one or more compounds from Formula I-XVDI and a pharmaceutically acceptable excipient.

[00266] In another aspect, the present technology is directed to a method for treating a disease affected at least in part by CAPN1, CAFN2, and/or CAP 9, which method comprises administering to a subject an effective amount of one or more compounds which selectively inhibits CAPN1, CAPN2, and/or CAPN9, said compounds being selected from Formula I- XVIII or a pharmaceutical composition comprising one or more compounds from Formula I- XVIH and a pharmaceutically acceptable excipient.

[00267] in another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds which specifically inhibits two or more enzymes selected from the group consisting of CAPN1, CAPN2, and CAPN9 in a ratio of at least 1 : 1 :5.

[00268] In another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds which specifically inhibits two or more enzymes selected from the group consisting of CAPN1, CAPN2, and CAPN9 in a ratio of at least 1 : 1 : 10.

[00269] In another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds which specifically inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least : 1 :20.

[00270] In another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds which specifically inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least 1 : 1 :50. [00271] In another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds which specifically inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least : 1 : 100.

[00272] In another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds which specifically inhibits two or more enzymes selected from the group consisting of CAPN1, CAPN2, and CAPN9 in a ratio of at least 1 : 1 :200.

[00273] In another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds which specifically inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least 1 : 1 :250,

[00274] In another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds which specifically inhibits two or more enzymes selected from the group consisting of CAPNl , CAPN2, and CAPN9 in a ratio of at least 1 : 1 :500.

[00275] In another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds which selectively inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least 1 : 1 :5.

[00276] In another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds which selectively inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least 1 : 1 : 10.

[00277] In another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds which selectively inhibits two or more enzymes selected from the group consisting of C APNl, C APN2, and CAPN9 in a ratio of at least 1 : 1 :20.

[00278] In another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds which selectively inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least 1 : 1 :50. [00279] In another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds which selectively inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least : 1 : 100.

[00280] In another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds which selectively inhibits two or more enzymes selected from the group consisting of CAPN1, CAPN2, and CAPN9 in a ratio of at least 1 : 1 :200.

[00281] In another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds which selectively inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least 1 : :250,

[00282] In another aspect, the present technology is directed to a method for treating a fibrotic disease, which method comprises administering to a subject an effective amount of one or more compounds which selectively inhibits two or more enzymes selected from the group consisting of CAPNl , CAPN2, and CAPN9 in a ratio of at least 1 : 1 :500.

[00283] In another aspect, the present technology is directed to a method for treating a disease affected at least in part by CAPNl, CAPN2, and/or CAPN9, which method comprises administering to a subject an effective amount of one or more compounds which specifically inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and

CAPN9 in a ratio of at least 1 : 1 :5.

[00284] In another aspect, the present technology is directed to a method for treating a disease affected at least in part by CAPNl, CAPN2, and/or CAPN9, which method comprises administering to a subject an effective amount of one or more compounds which specifically inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least 1 : 1 : 10.

[00285] In another aspect, the present technology is directed to a method for treating a disease affected at least in part by CAPNl , CAPN2, and/or CAPN9, which method comprises administering to a subject an effective amount of one or more compounds which specifically inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least 1 : 1 :20. [00286] In another aspect, the present technology is directed to a method for treating a disease affected at least in part by CAPNl, CAPN2, and/or CAPN9, which method comprises administering to a subject an effective amount of one or more compounds which specifically inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least 1 : 1 :50.

[00287] In another aspect, the present technology is directed to a method for treating a disease affected at least in part by C APNl , CAPN2, and/or CAPN9, which method comprises administering to a subject an effective amount of one or more compounds which specifically inhibits two or more enzymes selected from the group consisting of CAP l, CAPN2, and CAPN9 in a ratio of at least 1 : 1 : 100.

[00288] In another aspect, the present technology is directed to a method for treating a disease affected at least in part by CAPNl, CAPN2, and/or CAPN9, which method comprises administering to a subject an effective amount of one or more compounds which specifically inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least 1 : 1 :200.

[00289] In another aspect, the present technology is directed to a method for treating a disease affected at least in part by CAPNl , CAPN2, and/or CAPN9, which method comprises administering to a subject an effective amount of one or more compounds which specifically inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least 1 : 1 :250.

[00290] In another aspect, the present technology is directed to a method for treating a disease affected at least in part by CAPNl, CAPN2, and/or CAPN9, which method comprises administering to a subject an effective amount of one or more compounds which specifically inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least I ; 1 :500.

[00291] In another aspect, the present technology is directed to a method for treating a disease affected at least in part by CAPNl , CAPN2, and/or CAPN9, which method comprises administering to a subject an effective amount of one or more compounds which selectively inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least 1 : 1 :5.

[00292] In another aspect, the present technology is directed to a method for treating a disease affected at least in part by CAPNl, CAPN2, and/or CAPN9, which method comprises administering to a subject an effective amount of one or more compounds which selectively inhibits two or more enzymes selected from the group consisting of CAPN1, CAPN2, and CAPN9 in a ratio of at least 1 : 1 : 10.

[00293] in another aspect, the present technology is directed to a method for treating a disease affected at least in part by CAPNl , CAPN2, and/or CAPN9, which method comprises administering to a subject an effective amount of one or more compounds which selectively inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least 1 : 1 :20.

[00294] In another aspect, the present technology is directed to a method for treating a disease affected at least in part by CAPNl, CAPN2, and/or CAPN9, which method comprises administering to a subject an effective amount of one or more compounds which selectively inhibits two or more enzymes selected from the group consisting of CAPNl , CAPN2, and CAPN9 in a ratio of at least 1 : 1 :50.

[00295] In another aspect, the present technology is directed to a method for treating a disease affected at least in part by CAPNL CAPN2, and/or CAPN9, which method comprises administering to a subject an effective amount of one or more compounds which selectively inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least 1 : 1 : 100.

[00296] In another aspect, the present technology is directed to a method for treating a disease affected at least in part by CAPNl, CAPN2, and/or CAPN9, which method comprises administering to a subject an effective amount of one or more compounds which selectively inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least 1 : 1 :200.

[00297] In another aspect, the present technology is directed to a method for treating a disease affected at least in part by CAPNl, CAPN2, and/or CAPN9, which method comprises administering to a subject an effective amount of one or more compounds which selectively inhibits two or more enzymes selected from the group consisting of CAPNl, CAPN2, and CAPN9 in a ratio of at least 1 : 1 :250.

[00298] In another aspect, the present technology is directed to a method for treating a disease affected at least in part by CAPNl , CAPN2, and/or CAPN9, which method comprises administering to a subject an effective amount of one or more compounds which selectively inhibits two or more enzymes selected from the group consisting of CAPN1 , CAPN2, and CAPN9 in a ratio of at least I : 1 :500.

[00299] In another aspect, a method is provided for prophylactic therapy or treatment of a subject having a fibrotic disorder wherein said method comprising administering an effective amount of one or more compounds of Formula I-XVIII to the subject in need thereof.

[00300] In another aspect, a method is provided for prophylactic therapy or treatment of a subject having a disorder affected by CAPN!, CAPN2, and/or CAPN9 wherein said method comprising administering an effective amount of one or more compounds of Formula I-XVIII to the subject in need thereof.

[00301] In another aspect, a method for inhibiting myofibroblast differe tiation (e.g., Epithelial/Endothelial-to-Mesenchymal Transition (EpMT/EnMT)) is provided wherein the method comprises contacting cells with an effective amount of one or more compounds of Formula I-XVIII disclosed herein. In one aspect, the method for inhibiting myofibroblast differentiation (e.g., Epithelial/Endothelial-to-Mesenchymal Transition (EpMT/EnMT)) is performed in-vitro or in-vivo.

[00302] In some embodiments, the present technology is a method for treating a disease or condition selected from the group consisting of or that produces a symptom selected from the group consisting of: liver fibrosis, renal fibrosis, lung fibrosis, hypersensitivity pneumonitis, interstitial fibrosis, systemic scleroderma, macular degeneration, pancreatic fibrosis, fibrosis of the spleen, cardiac fibrosis, mediastinal fibrosis, myelofibrosis, endomyocardial fibrosis, retroperitoneal fibrosis, progressive massive fibrosis, nephrogenic systemic fibrosis, fibrotic complications of surgery, chronic allograft vascuiopathy and/or chronic rejection in transplanted organs, ischemic-reperfusion injury associated fibrosis, injection fibrosis, cirrhosis, diffuse parenchymal lung disease, post-vasectomy pain syndrome, and rheumatoid arthritis diseases, wherein which method comprises administering to a subject an effective amount of one or more compounds Formula I-XVIII to a subject in need thereof.

[00303] In some embodiments, the present technology is a method for treating liver fibrosis.

[00304] In some embodiments, the present technology is a method for treating cardiac fibrosis,

[00305] In some embodiments, the present technology is a method for treating fibrosis in rheumatoid arthritis diseases. [00306] In some embodiments, the present technology is a method for treating a condition affected by CAPNl, CAPN2, and/or CAPN9, which is in both a therapeutic and prophylactic setting for subjects. Both methods comprise administering of one or more compounds of Formula ί-ΧΥίίί to a subject in need thereof.

[00307] In some embodiments, the present technology is a method for treating stiff skin syndrome.

[00308] In another aspect, the present technology is directed to a method wherein one or more compounds of Formula I-XVIil may be administered with other CAPNl , CAPN2, and/or CAPN9 inhibitor agents, such as anti-CAPNl, CAPN2, AND/OR CAPN9 antibodies or antibody fragments, CAPNl , CAPN2, and/or CAPN9 antisense, iRNA, or other small molecule CAPNl, CAPN2, and/or CAPN9 inhibitors, or in combination with other agents as described in detail herein.

[00309] Diseases and/or disorders or produced symptoms associated or affected at least in part by CAPNl, CAPN2, and/or CAPN9 include those selected from the group consisting of: liver fibrosis, renal fibrosis, lung fibrosis, hypersensitivity pneumonitis, interstitial fibrosis, systemic scleroderma, macular degeneration, pancreatic fibrosis, fibrosis of the spleen, cardiac fibrosis, mediastinal fibrosis, myelofibrosis, endomyocardial fibrosis, retroperitoneal fibrosis, progressive massive fibrosis, nephrogenic systemic fibrosis, fibrotic complications of surgery, chronic allograft vasculopathy and/or chronic rejection in transplanted organs, ischemic-reperfusion injury associated fibrosis, injection fibrosis, cirrhosis, diffuse parenchymal lung disease, post-vasectomy pain syndrome, and rheumatoid arthritis.

[00310] In one embodiment, therapeutically effective amount is a specific amount which causes a specific physiological effect which results in the amelioration of the disorder being treated or protects against a risk associated with the disorder.

[00311] The compounds of the present technology are useful in the diagnosis and treatment of a variety of human diseases selected from the group consisting of or that produces a symptom selected from the group consisting of: liver fibrosis, renal fibrosis, lung fibrosis, hypersensitivity pneumonitis, interstitial fibrosis, systemic scleroderma, macular

degeneration, pancreatic fibrosis, fibrosis of the spleen, cardiac fibrosis, mediastinal fibrosis, myelofibrosis, endomyocardial fibrosis, retroperitoneal fibrosis, progressive massive fibrosis, nephrogenic systemic fibrosis, fibrotic complications of surgery, chronic allograft

vasculopathy and/or chronic rejection in transplanted organs, ischemic-reperfusion injury associated fibrosis, injection fibrosis, cirrhosis, diffuse parenchymal lung disease, post- vasectomy pain syndrome, and rheumatoid arthriti s. The compounds of the present technology are particularly useful in treating disorders arising from fibrosis and complications thereof.

[00312] in another aspect, the present technology is directed to a method wherein one or more compounds of Formula I-XVIII which are used as a means to inhibit myofibroblast differentiation (e.g., Epithelial/Endothelial-to-Mesenchymal Transition (EpMT/EnMT)). In particular, one or more of these compounds which are inhibitors of one or more (or all three) CAPNI, CAFN2, and/or CAPN9, alone or in combination with other TGFp signaling inhibitors, could be used to treat or protect againstor reduce a symptom of a fibrotic, sclerotic or post inflammatory disease or condition including: liver fibrosis, renal fibrosis, lung fibrosis, hypersensitivity pneumonitis, interstitial fibrosis, systemic scleroderma, macular degeneration, pancreatic fibrosis, fibrosis of the spleen, cardiac fibrosis, mediastinal fibrosis, myelofibrosis, endomyocardial fibrosis, retroperitoneal fibrosis, progressive massive fibrosis, nephrogenic systemic fibrosis, fibrotic complications of surgery, chronic allograft

vasculopathy and/or chronic rejection in transplanted organs, ischemic-reperfusion injur}' associated fibrosis, injection fibrosis, cirrhosis, diffuse parenchymal lung disease, post- vasectomy pain syndrome, and rheumatoid arthritis.

[00313] Compounds of the present technology are shown to have improved safety and potency, such as the potency of inhibiting CAPNI , CAPN2, and/or CAPN9 at low

micromolar concentratio s and possessing low relative cytotoxicity. In general, compounds of the present technology are shown to have potency, ameliorate, and/or possess efficacy in treating diseases or disorders which include, as a component, some form of fibrosis or inflammation.

[00314] The amount of active compound administered will vary depending upon the disease treated, the mammalian species, and the particular mode of administration, etc.

Suitable doses for the compounds of the present technology can be, for example, between 0.1 mg to about 1000 mg, between 1 mg to about 500 mg, between I mg to about 300 mg, or between 1 mg to about 100 mg per day. Such doses can be administered once a day or more than once a day, for example 2, 3, 4, 5 or 6 times a day, but preferably 1 or 2 times per day. In some embodiments, the total dosage for a 70 kg adult is in the range of 0.001 to about 15 mg per kg weight of subject per administration or 0.01 to about 1.5 mg per kg weight of subject per administration, and such therapy can extend for a number of days, a number of weeks or months, and in some cases, years. It will be understood, however, that the specific dose level for any particular subject will depend on a variety of factors including the activity of the specific compound employed: the age, body weight, general health, sex and diet of the individual being treated; the time and route of administration; the rate of excretion; other drugs that have previously been administered; and the severity of the particular disease undergoing therapy, as is well understood by those of skill in the area.

[00315] The compounds of the present technology can be prepared from readily available starting materials using the following general methods and procedures. It will be appreciated that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.

[00316] Additionally, as will be apparent to those skilled in the art, conventional protecting groups may be necessary to prevent certain functional groups from undergoing undesired reactions. Suitable protecting groups for various functional groups as well as suitable conditions for protecting and deprotecting particular functional groups are well known in the art. For example, numerous protecting groups are described in T. W. Greene and P. G M. Wuts, Protecting Groups in Organic Synthesis, Third Edition, Wiley, New York, 1999, and references cited therein.

[00317] if the compounds of the present technology contain one or more chiral centers, such compounds can be prepared or isolated as pure stereoisomers, i.e., as individual enantiomers or dfl ) stereoisomers, or as stereos sorrier-enriched mixtures. All such

stereoisomers (and enriched mixtures) are included within the scope of the present technology, unless otherwise indicated. Pure stereoisomers (or enriched mixtures) may be prepared using, for example, optically active starting materials or stereoselective reagents well-known in the art. Alternatively, racemic mixtures of such compounds can be separated using, for example, chiral column chromatography, chiral resolving agents and the like.

[00318] The starting materials for the following reactions are generally known compounds or can be prepared by known procedures or obvious modifications thereof. For example, many of the starting materials are available from commercial suppliers such as Aldrich Chemical Co. (Milwaukee, Wisconsin, USA), Bach em (Torrance, California , USA), Emka- Chemce or Sigma (St. Louis, Missouri, US A). Others may be prepared by procedures, or obvious modifications thereof, described in standard reference texts such as Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-15 (John Wiley, and Sons, 1991), Rodd's Chemistry of Carbon Compounds, Volumes 1-5, and Supplemental (Elsevier Science Publishers, 1989), Organic Reactions, Volumes 1-40 (John Wiley, and Sons, 1991 ), March's Advanced Organic Chemistry, (John Wiley, and Sons, 5th Edition, 2001), and Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989).

Synthesis Routes to Compounds of the Present Technology

[00319] In one general embodiment, the method involves starting with an appropriately N- protected amino acid compound with an alkene functionalized side-chain, and coupling it with an appropriately C-protected amino acid compound with an alkene functionalized side- chain as a coupling partner to give the di-functionalized di-amino acid derivative. It is appreciated that other suitable coupling conditions and reagents, such as HOBt and/or DMAP, may be used to form a requisite di-amino acid derivative, di-amino acid derivative is then cyclized upon with a metathesis reaction using, for example, the Grubbs catalyst. The skilled artisan will appreciate that there are many synthetic conditions and catalysts which are capable of metathesizing the olefin groups. Such synthetic methods and catalysts are well within in the scope of the present technology disclosed. The C -protection is removed by reduction with DIBAL to directly give the aldehyde. Alternatively, the C-protection can be reduced with LAH (other reduction conditions such as birch reduction and so forth will work as well) to afford the corresponding alcohol, which is then oxidized with PCC (or DMP conditions or like) to the aldehyde. The aldehyde then undergoes a cyanohydrin reaction upon treatment with KCN, which is then hydrolyzed under acidic conditions in alcohol to the corresponding a-hydroxy ester. At this point, any desired groups may be placed onto the nitrogen atom of the keto-amide by a substitution reaction, such as with an aikyl iodide, or perhaps with reductive animation. The skilled artisan will note that if the desired compound has functionality on the nitrogen of the amide, the synthesis can use NaNH 2 or a strong base of the like in the step before the cyanohydrin reaction, in order to selectively place a differing group onto those nitrogen atoms as opposed to the nitrogen atom of the keto-amide. Finally, the a-hydroxyl group is oxidized under conditions for Dess-Martin Periodinane oxidation (with hvpervaleiit iodine) or by an oxidizing agent such as PCC (pyridinium chlorochromate) or the like. The skilled artisan will once again appreciate that there are many other oxidizing conditions and agents which are within the scope of this disclosure to oxidize the hydroxy! group. Of course it is recognized that at any point during this synthesis, the keto compounds may be interchanged with thio-keto compounds as treatment with Lawesson's reagent or the like will give the corresponding thio-carbonyl compound. This synthesis route is generally shown in Scheme 1.

[00320] In one example, the compounds of general Formula I-XXIV can be generally prepared according to representative Scheme I :

Scheme 1

[00321] As an alternative, the skilled artisan will appreciate that these keto-amide compounds can al so be prepared by converting the amino acid side-chain of the starting material (N-terminus protected) into side-chain with heteroatom functionality. In this way, the heteroatom and substituent bond connection on the ring can be affected, optionally stereoseiectively. Such functional group conversions are well known in the art and also allow: for complex substitution at the ring juncture(s) and opens pathways for bi cyclic ring systems. Typical substrates for these kind of intramolecular reactions are usually setup with functional groups, one electrophilic, one nucleophilic in nature and may include, but are not limited to; aldehydes, halides, alkenes, and ketones, and the like. Of course, either way the cyclization is affected, the skilled artisan will recognize that the requisite functional groups may be selected so that heteroatom s and multiple substituents can exist at any position of the ring. An ex

[00322] In one example, the compounds of general Formula I-XXIV can also be generally prepared according to representative Scheme 2: eh erne 2

Herein it is understood that amino, keto, thio, hydroxy!, and any other necessary protecting groups and their methods of deprotection are known in the art, such as those described in T. W. Greene and P. G. M. Wuts, Protecting Groups in Organic Synthesis, Third Edition, Wiley, New York, 1999.

[00323] Alternatively, the skilled artisan will recognize that there is additional synthetic functional group modifications that can use to prepare spirocyclic and other bicyclic compounds onto the cyclic keto-amide ring.

[00324] in one example, the compounds of general Formula I-XXIV can also be generally prepared according to representative Scheme 3 :

Scheme 3

[00325] Yet in another alternative aspect, the skilled artisan will recognize that other bicyclic systems can be prepared thru similar functional group transformations with groups on the amino acid side chain or on the nitrogen atoms outside the keto-amide ring.

[00326] The present technology provides novel compounds possessing CAPNl, CAPN2, and/or CAPN9 inhibitory activity and, accordingly, are useful in treating conditions and/or disorders affected by (or at least in part by) CAPNl, CAPN2, and/or CAPN9. Such conditions include fibrosis and/or complications thereof. [00327] Methods for treatment of fibrotic diseases are also encompassed by the present invention. Said methods of the invention include administering a therapeutically effective amount of any one of compounds of Formula I-XVIH. The compounds and solvates of the invention can be formulated in pharmaceutical compositions. These compositions can comprise a pharmaceutically acceptable excipient, carrier, buffer, stabilizer or other materials well known to those skilled in the art. The precise nature of the carrier or other material can depend on the route of administration, e.g. oral, intravenous, cutaneous or subcutaneous, nasal, intramuscular, intraperitoneal routes.

[00328] Pharmaceutical compositions for oral administration can be in tablet, capsule, powder or liquid form. A tablet can include a solid carrier such as gelatin or an adjuvant. Liquid pharmaceutical compositions generally include a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil. Physiological saline solution, dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol can be included,

[00329] For intravenous, cutaneous or subcutaneous injection, or injection at the site of affliction, the active ingredient will be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability. Those of relevant skill in the art are well able to prepare suitable solutions using, for example, isotonic vehicles such as Sodium Chloride Injection, Ringer's Injection, Lactated Ringer's Injection.

Preservatives, stabilizers, buffers, antioxidants and/or other additives can be included, as required.

[00330] A composition can be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated.

[00331] In general, the compounds of the present technology will be administered in a therapeutically effective amount by any of the accepted modes of administration for agents that serve similar utilities. The actual amount of the compound of the present technology, i.e., the active ingredient, will depend upon numerous factors such as the severity of the disease to be treated, the age and relative health of the subject, the potency of the compound used, the route and form of administration, and other factors well known to the skilled artisan. The drug can be administered at least once a day, preferably once or twice a day.

[00332] An effective amount of such agents can readily be determined by routine experimentation, as can the most effective and convenient route of administration and the most appropriate formulation. Various formulations and drug delivery systems are available in the art. See, e.g., Gennaro, A.R., ed. (1995) Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing Co.

[00333] A therapeutically effective dose can be estimated initially using a variety of techniques well-known in the art. Initial doses used in animal studies may be based on effective concentrations established in cell culture assays. Dosage ranges appropriate for human subjects can be determined, for example, using data obtained from animal studies and cell culture assays.

[00334] An effective amount or a therapeutically effective amount or dose of an agent, e.g., a compound of the present technology, refers to that amount of the agent or compound that results in amelioration of symptoms or a prolongation of survival in a subject. Toxicity and therapeutic efficacy of such molecules can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., by determining the LD 50 (the dose lethal to 50 % of the population) and the ED 50 (the dose therapeutically effective in 50 % of the population). The dose ratio of toxic to therapeutic effects is therapeutic index, which can be expressed as the ratio LD 50 / ED 50 . Agents that exhibit high therapeutic indices are preferred.

[00335] The effective amount or therapeutically effective amount is the amount of the compound or pharmaceutical composition that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinari an, medical doctor or other clinician. Dosages particularly fall within a range of circulating concentrations that includes the ED 50 with little or no toxicity. Dosages may vary within this range depending upon the dosage form employed and/or the route of administration utilized. The exact formulation, route of administration, dosage, and dosage interval should be chosen according to methods known in the art, in view of the specifics of a subject's condition.

[00336] Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety that are suffi cient to achieve the desired effects, i.e., the minimal effective concentration (MEC). The MEC will vary for each compound but can be estimated from, for example, in vitro data and animal experiments. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration. [00337] The amount of agent or composition administered may be dependent on a variety of factors, including the sex, age, and weight of the subject being treated, the severity of the affliction, the manner of administration, and the judgment of the prescribing physician.

[00338] The present technology is not limited to any particular composition or

pharmaceutical carrier, as such may vary. In general, compounds of the present technology will be administered as pharmaceutical compositions by any one of the foll owing routes: oral, systemic (e.g., transdermal, intranasal or by suppository), or parenteral (e.g., intramuscular, intravenous or subcutaneous) administration. The preferred manner of administration is oral using a convenient daily dosage regimen that can be adjusted according to the degree of affliction. Compositions can take the form of tablets, pills, capsules, semisolids, powders, sustained release formulations, solutions, suspensions, elixirs, aerosols, or any other appropriate compositions. Another preferred manner for administering compounds of the present technology is inhalation.

[00339] The choice of formulation depends on various factors such as the mode of drug administration and bioavailability of the drug substance. For delivery via inhalation the compound can be formulated as liquid solution, suspensions, aerosol propeilants or dry powder and loaded into a suitable dispenser for administration. There are several types of pharmaceutical inhalation devices-nebulizer inhalers, metered dose inhalers (MDI) and dry- powder inhalers (DPI). Nebulizer devices produce a stream of high velocity air that causes therapeutic agents (which are formulated in a liquid form) to spray as a mist that is carried into the subject's respirator}' tract, MDFs typically are formulation packaged with a compressed gas. Upon actuation, the device discharges a measured amount of therapeutic agent by compressed gas, thus affording a reliable method of administering a set amount of agent. DPI dispenses therapeutic agents in the form of a free flowing powder that can be dispersed in the subject's inspiratory air-stream during breathing by the device. In order to achieve a free flowing powder, therapeutic agent is formulated with an excipient such as lactose, A measured amount of therapeutic agent is stored in a capsule form and is dispensed with each actuation.

[00340] Pharmaceutical dosage forms of a compound of the present technology may be manufactured by any of the methods well-known in the art, such as, for example, by conventional mixing, sieving, dissolving, melting, granulating, dragee-making, tabletting, suspending, extruding, spray-drying, levigating, emulsifying, (nano/micro-) encapsulating, entrapping, or lyophilization processes. As noted above, the compositions of the present technology can include one or more physiologically acceptable inactive ingredients that facilitate processing of active molecules into preparations for pharmaceutical use.

[00341] Recently, pharmaceutical formulations have been developed especially for drugs that show poor bioavailability based upon the principle that bioavailability can be increased by increasing the surface area i.e., decreasing particle size. For example, U.S. Pat. No.

4, 107,288 describes a pharmaceutical formulation having particles in the size range from 10 to 1,000 rim in which the active material is supported on a crosslinked matrix of

macromolecules. U.S. Patent No. 5, 145,684 describes the production of a pharmaceutical formulation in which the drug substance is pulverized to nanoparticles (average particle size of 400 nm) in the presence of a surface modifier and then dispersed in a liquid medium to give a pharmaceutical formulation that exhibits remarkably high bioavailability.

[00342] The compositions are comprised of in general, a compound of the present technology in combination with at least one pharmaceutically acceptable excipient.

Acceptable excipients are non-toxic, aid administration, and do not adversely affect therapeutic benefit of the claimed compounds. Such excipient may be any solid, liquid, semisolid or, in the case of an aerosol composition, gaseous excipient that is generally available to one of skill in the art.

[00343] Solid pharmaceutical excipients include starch, cellulose, talc, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, magnesium stearate, sodium stearate, glycerol monostearate, sodium chloride, dried skim milk and the like. Liquid and semisolid excipients may be selected from glycerol, propylene glycol, water, ethanol and various oils, including those of petroleum, animal, vegetable or synthetic origin, e.g., peanut oil, soybean oil, mineral oil, sesame oil, etc. Preferred liquid carriers, particularly for injectable solutions, include water, saline, aqueous dextrose, and glycols.

[00344] Compressed gases may be used to disperse a compound of the present technology in aerosol form. Inert gases suitable for this purpose are nitrogen, carbon dioxide, etc. Other suitable pharmaceutical excipients and their formulations are described in Remington's Pharmaceutical Sciences, edited by E. W. Martin (Mack Publishing Company, 18th ed., 1990).

[00345] The present compositions may, if desired, be presented in a pack or dispenser device containing one or more unit dosage forms containing the active ingredient. Such a pack or device may, for example, comprise metal or plastic foil, such as a blister pack, or glass, and rubber stoppers such as in vials. The pack or dispenser device may be

accompanied by instructions for administration. Compositions comprising a compound of the present technology formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.

[00346] The amount of the compound in a formulation can vary within the full range employed by those skilled in the art. Typically, the formulation will contain, on a weight percent (wt %) basis, from about 0.01 99.99 wt % of a compound of the present technology based on the total formulation, with the balance being one or more suitable pharmaceutical excipients. Preferably, the compound is present at a level of about 1 80 wt %. Representative pharmaceutical formulations are described below.

Formulation Examples

The following are representative pharmaceutical formulations containing a compound of Formula I-XVUI.

Formulation Example 1 ~~ Tablet formulation

The following ingredients are mixed intimately and pressed into single scored tablets.

Quantity per

Ingredient tablet, mg compound of this the present technology 400

cornstarch 50

croscarmellose sodium 25

lactose 120

magnesium stearate 5

Formulation Example 2— Capsule formulation

The following ingredients are mixed intimately and loaded into a hard-shell gelatin capsule.

Quantity per

Ingredient capsule, mg compound of this the present technology

lactose, spray-dried

magnesium stearate

Formulation Example 3— Suspension formulation

The following ingredients are mixed to form a suspension for oral administration. Ingredient Amount compound of this the present technology 1.

fumaric acid 0.5 g

sodium chloride 2.0 g

methyl paraben 0.15 g

propyl paraben 0.05 g

granulated sugar 25.0 g

sorbitol (70% solution) 13.00 g

Veegum K (Vanderbilt Co.) 1.0 g

flavoring 0.035 mL

colorings 0.5 mg

distilled water q.s. to 100 mL

Formulation Example 4 -- Injectable formulation

The following ingredients are mixed to form an injectable formulation.

Ingredient Amount compound of this the present technology 0.2 mg-20 mg

sodium acetate buffer solution, 0.4 M 2,0 mL

HC1 (IN) or NaOH (IN) q.s. to suitable pH

water (distilled, sterile) q.s. to 20 mL

Formulation Example 5 — Suppository Formulation

A suppository of total weight 2.5 g is prepared by mixing the compound of the present technology with Witepsol® H-15 (triglycerides of saturated vegetable fatty acid;

Riches-Nelson, Inc., New York), and has the following composition:

Ingredient Amount

Compound of the present technology 500 mg

Witepsol® Ft- 15 balance

[00347]

[00348] The following synthetic and biological examples are offered to illustrate this the present technology and are not to be construed in any way as limiting the scope of this the present technology. Unless otherwise stated, ail temperatures are in degrees Celsius.

EXAMPLES

[00349] Below are examples of specific embodiments for carrying out the present invention. The examples are offered for illustrative purposes only, and are not intended to limit the scope of the present invention in any way. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperatures, etc.), but some experimental error and deviation should, of course, be allowed for,

[00350] The practice of the present invention will employ, unless otherwise indicated, conventional methods of protein chemistr) ' ', biochemistry, recombinant DNA techniques and pharmacology, within the skill of the art. Such techniques are explained fully in the literature. See, e.g., T.E. Creighton, Proteins: Structures and Molecular Properties {W . I i. Freeman and Company, 1993); A.L. Lehninger, Biochemistry (Worth Publishers, Inc., current addition), Sarnbrook, et a!., Molecular Cloning: A Laboratory Manual (2nd Edition, 1989); Methods in Enzymology (S. Colowick and N. Kaplan eds., Academic Press, Inc.); Remington's

Pharmaceutical Sciences, 18th Edition (Easton, Pennsylvania: Mack Publishing Company, 1990); Carey and Sundberg Advanced Organic Chemistry 3 ru Ed. (Plenum Press) Vols A and B(1992).

[00351] The present technology is further understood by reference to the following examples, which are intended to be purely exemplary of the present technology. The present technology is not limited in scope by the exemplified embodiments, which are intended as illustrations of single aspects of the present technology only. Any methods that are functionally equivalent are within the scope of the present technology. Various modifications of the present technology in addition to those described herein will become apparent to those skilled in the art from the foregoing description and accompanying figures. Such

modifications fall within the scope of the appended claims.

[00352] In the examples below, the following abbreviations have the following meanings. If an abbreviation is not defined, it has its generally accepted meaning.

aq. = aqueous

LC-MS liquid chromatography-mass spectrometry

MS mass spectrometry

THF = tetrahydrofuran

NaHCOs = sodium bicarbonate

DIE A diisopropylethylamine

MS mass spectrometry

NaH = sodium hydride

o/n = overnight

HATU l-[Bis(dimethylamino)methylene]-lH- 1,2,3- trl zolo[4,5-b]pyridinium 3-oxid hexafluorophosphate r.t. = room temperature

LAH = lithium aluminum hydride

DCM: di chl orom ethane

DMF dimethylformamide dimethyl sulfoxide

Ethyl Acetate

equivalent

ethyl acetate

ethanol

gram

hours

hours

hydrochloric acid

high-performance liquid chromatography

acetic acid

molar

methanol

milligrams

milliliters

millimols

melting point

mass to charge ratio

sodium chloride

sodium carbonate

nuclear magnetic resonance

sodium hydroxide

sodium sulfate

Petroleum Ether

saturated

thin layer chromatography

ultraviolet

weight percent

micromolar

General experimental details:

[00353] Final compounds were confirmed by HPLC/MS analysis and determined to be > 90%. Ή and 13 C MR spectra were recorded in CDCW (residual internal standard CHCW = δ 7.26), DMSQ-i¾ (residual internal standard CD 3 SOCD 2 H = δ 2.50), methanol-i¾ (residual internal standard CD 3 OD), or acetone-t& (residual internal standard CD 3 COCD 2 H = δ 2.05). The chemical shifts (δ) reported are given in parts per million (ppm) and the coupling constants (J) are in Hertz (Hz). The spin multiplicities are reported as s :: = singlet, bs :: = broad singlet, bm = :: broad multiplet = :: doublet, t = triplet, q = :: quartet, p = pentuplet, dd :: = doublet of doublet, ddd = doublet of doublet of doublet, dt = doublet of triplet, td = triplet of doublet, tt = triplet of triplet, and m = multiplet. [00354] HPLC-MS analysis was carried out with gradient elution. Medium pressure liquid chromatography (MPLC) was performed with silica gel columns in both the normal phase and reverse phase.

EXAMPLE 1 : Com ounds synthesis procedure and data:

To a solution of compound 1A (50 g, 194 mmol) in THF (500 mL) was added 3- butenylmagnesium bromide (0.522 M, 558 mL, 291 mmol) dropwise at -40 °C to -50 °C for 30 min. The mixture was stirred at -50 °C for 1.5 hrs. The mixture was quenched with sat. NH4CI (100 mL) and separated. The aqueous layer was extracted with EA (3 x 50 mL). The combined organic layers were dried over Na 2 S0 4 and concentrated. The residue was purified by column chromatography (Si0 2 , petroleum ether/ethyl acetate :=: 5: 1 to 3 : 1) to afford compound IB (58.8 g, yield 96.5%) as white solid. MS (ESI) m/z (M+Na + ) 335.9. 1H NMR (CDC1 3 , 400MHz) δ 5.90 - 5.71 (m, 1H), 5.19 - 4.89 (m, 3H), 4.31 - 4.15 (m, 3H), 2.65 - 2.42 (m, 4H), 2.38 - 2.26 (m, 2H), 2.20 - 2.08 (m, 1H), 1.95 - 1.82 (m, 1H), 1.50 - 1.38 (m, 9H), 1.30 - 1.26 (m, 3H)..

To a solution of compound IB (58.8 g, 187.6 mmol) in AcOH (500 mL) was added 4- methylbenzenesulfonohydrazide (41 ,6 g, 223.3 mmol) at 10 °C. The mixture was stirred at 10 °C for 1 h. Then NaBH(OAc) 3 (167 g, 788 mmol) was added to the mixture in portions. The mixture was stirred at 37 °C for 17 hrs. The mixture was quenched with water (500 mL) and extracted with EA (3 x 150 mL). The combined organic layers were basified to pH ~ 8 with 10% NaOH aqueous solution. Then the organic layer was separated and dried over anhydrous Na 2 S0 4 , filtered and concentrated. The residue was purified by column chromatography (Si0 2 , petroleum ether/ethyl acetate = 10/1 to 5/1 ) to afford compound 1C (35.0 g, yield 62.3%) as colorless oil. 1H NMR (CDC1 3 , 400MHz) δ 5,90 - 5.71 (m, 1H), 5.19 - 4.89 (m, 3H), 4.31 - 4.15 (m, 3H), 2.15 - 1.95 (m, 2H), 1.90 - 1.56 (m, 1H), 1 ,75 - 1.55 (m, 1H), 1.52 ~ 1.22 (m, 1H).

[00357] Step 3: Synthesis of compound! 1D

To a solution of compound 1C (24.0 g, 80.2 mmol) in DCM (150 mL) was added TFA (70 mL) at 10 °C. The mixture was stirred at 10 °C for 2 h. The mixture was concentrated to give compound ID (25.1 g, crude), which was used directly for next step without further purification. 1H NMR (CDC1 3 , 400MHz) δ 7.97 - 7.50 (m, 3H), 5.89 - 5.67 (m, 1H), 5,07 - 4,85 (m, 2H), 4.38 - 4.22 (m, 2H), 4, 13 - 3.96 (m, 1H), 2.10 - 1 ,82 (m, 4H), 1.53 - 1.20 (m, 9H).

[00358] Step 4: Synthesis of compound! 1E

To a solution of compound I D (40 g, 128 mmol, TFA salt) in DCM^ (60 mL) was added Et 3 N (61.9 mL, 446.9 mmol) and benzyl chloroformate (15.4 mL, 108.5 mmol) drop wise at 0 °C for 30 min. The mixture was stirred at 10 °C for 14 hrs. The mixture was quenched with water (200 mL) and separated. The aqueous layer was extracted with DCM (2 x 100 mL). The combined organic layers were dried over Na 2 S0 4. filtered and concentrated. TThhee rreessiidduuee wwaass ppuurriiffiieedd bbyy ccoolluummnn cchhrroommaattooggrraapphhyy ((SSii00 22 ,, ppeettrroolleeuumm eetthheerr//eetthhyyll aacceettaattee == 55//11)) ttoo aaffffoorrdd ccoommppoouunndd IIEE ((3344 gg,, yyiieelldd 7799..99%%)) aass yyeellllooww ooiill.. ll HH NNMMRR ((CCDDCC11 33 .. 440000MMHHzz)) δδ 77..4455 -- 77..2277 ((mm,, 55HH)),, 55..9900 -- 55..7700 ((mm,, IIHH)),, 55..3399 -- 55..2255 ((mm,, IIHH)),, 55..1122 ((ss,, 22HH)),, 55..0044 -- 44..9900 ((mm,, 22HH)),, 44..4422 -- 44..3311 ((mm,, II HH)),, 44..2288 -- 44..0055 ((mm,, 22HH)),, 22..1122 -- 11 ..9988 ((mm,, 22HH)),, 11..9911 -- 11..7766 ((mm,, 1IHH)),, 11..7744 -- 11..5588 ((mm,, IIHH)),, 11..4466 -- 11 ,, 1166 ((mm,, 99HH))„„

[[0000335599]] SStteepp 55:: SSyynntthheessiiss ooff c coommppoouunndd 11FF

TToo aa ssoolluuttiioonn ooff ccoommppoouunndd IIEE ((3344 gg,, 110022..00 mmmmooll)) iinn 11 {{ ■■ ■■(( )) ((110066 mmLL)) aanndd MMeeOOHH ((332255 m mLL)) wwaass aaddddeedd LLiiOOHH''HHjjOO ((1111..11 gg,, 226655 mmmmooll)) aatt 77 °°CC.. TThhee mmiixxttuurree wwaass ssttiirrrreedd aatt 2288 °°CC ffoorr 1166 hhrrss.. TThheenn MMeeOOHH wwaass rreemmoovveedd uunnddeerr rreedduucceedd pprreessssuurree.. TToo tthhee rreessiidduuee wwaass ddiilluutteedd wwiitthh wwaatteerr ((110000 mmLL)).. TThhee mmiixxttuurree wwaass eexxttrraacctteedd wwiitthh MMTTBBEE ((22 xx 4400 mmLL)).. TThhee aaqquueeoouuss llaayyeerr wwaass aacciiddiififieedd ttoo ppHH ~~ 33 wwiitthh 11MM HHCC11 aanndd tthheenn wwaass eexxttrraacctteedd wwiitthh EEAA ( (33 xx 110000 mmLL)).. TThhee ccoommbbiinneedd oorrggaanniicc llaayyeerrss wweerree ddririeedd oovveerr aannhhyyddrroouuss NNaa 22 SS00 44 ,, fi filltteerreedd aanndd ccoonncceennttrraatteedd ttoo ggiivvee ccoommppoouunndd IIFF ((2288..55 gg,, ccrruuddee,, yyiieelldd:: 9911..55%%)) aass yyeellllooww ooiill,, wwhhiicchh wwaass uusseedd foforr nneexxtt sstteepp ddiirreeccttllyy.. 1H1H NNMMRR ((CCDDCC11 33 ,, 440000MMHHzz)) δδ 77..5500 -- 77..2288 ((mm,, 55HH)),, 55..9911 -- 55..7711 ((mm,, IIHH)),, 55..3344 -- 55..0099 ((mm,, 33HH)),, 55..0088 -- 44..8855 ((mm,, 22HH)),, 44..5500 -- 44..3344 ((mm,, IIHH)),, 22..0066 · -· 11..9999 ((mm,, 22HH)),, 11..9966 -- 11..8811 ((mm,, IIHH)),, 11..7777 -- 11..6633 ((mm,, IIHH)),, 11..4499 -- 11..2299 ((mm,, 66HH))..

To a solution of compound IF (2 g, 6.55 mmol) and compound 1G (2 g, 6.55 mmol, HC1 salt) in DCM (50 mL) was added DIPEA (5.7 mL, 32.75 mmol, 5 eq), EDCI (1.88 g, 9.82 mmol) and HOBt (884.9 nig, 6.55 mmol) at 5 °C. The mixture was stirred at 5 °C for 15 hrs. The mixture was concentrated. To the mixture was added EA (150 mL). The mixture was washed with 1M HC1 (20 mL), sat. NaHC0 3 (4 x 20 mL) and brine (20 mL), dried over Na 2 S0 4 and concentrated to give compound IH (3 g, crude), which was used in next step without further purification. l H NMR (DMSO-fife, 400MHz) δ 8.73 - 8.63 (m, IH), 7.85 - 7.74 (m, IH), 7.40 - 7.20 (m, 5H), 5.86 - 5.69 (m, IH), 5.67 - 5.51 (m, IH), 5.32 - 5.16 (m, IH), 5.12 - 4.84 (m, 5H), 4,33 - 4.17 (m, IH), 4.1 1 - 3 ,90 (m, 4H), 2.15 - 2,02 (m, I H), 2.00 - 1.95 (m, 2H), 1.69 - 1.36 (m, 6H), 1.35 - 1 ,05 (m, 10H), 0.93 - 0.68 (m, 6H).

[00361] Step 7: Synthesis of compound 1 J

To a solution of compound IH (1 g, 1.80 mmol) in DCM (650 mL) was added Hoveyda-Grubbs' 1 st Generation catalyst (108 mg, 179.95 umol) under N 2 atmosphere . The mixture was stirred at 50 °C under N 2 for 15 hrs. The mixture was concentrated and the residue was purified by column chromatography (SiO ? ., Petroleum ether/Ethyl acetate = 2: 1 to 1 : 1) to afford compound 1J (500 mg, yield 52.6%) as white solid. MS (ESI) m/z (M ! f ) 528.1.

To a solution of compound 1J (1 10 mg, 0.21 mmol) in THF ( 10 raL) was added LiAlH,| (24 mg, 0.63 mmol) at 0°C. The mixture was stirred at 0°C for 1 h. The mixture was diluted with EtOAc (20 mL), quenched with H 2 0 (0.03 mL), 5% aq, NaOH (0.03 mL), H 2 () (0,09 mL) at 0°C. The mixture was warmed up to 25°C and stirred for 15 min, MgS0 4 was added and the mixture was stirred for 15 min. After filtered, the filtrate was separated, and the organics were collected and concentrated. The residue was purified by column chromatography (petroleum ether/ethyl acetate = 1/1) to give compound IK (65 mg, yield 63.2%) as white solid. MS (ESI) m/z (M+H) + 486.3. ! H NMR (DMSO-i¾ , 400 MHz): δ 8.31 (br. s, I H), 8.14 (d, J 7.6 Hz, 1 1 1 ). 7.38 - 7.31 (m, 51 ! }. 7, 12 (d, ,/ 7.6 Hz, 1 H), 5.49 - 5.43 (m, 1H), 5.12 - 5.05 (m, 1H), 4.99 (s, 2H), 4.26 - 4,05 (m, 2H), 3.74 - 3,60 (m, I H), 3.32 - 3.25 (m, H), 2,34 - 2, 18 (m, IH), 1.97 - 1.82 (m, IH), 1 ,71 - 1.33 (m, 7H), 1.32 - 1.05 (m, 6H), 0.97 - 0.67 (m, 7H).

[00363] Step 9: Synthesis of compound! 1

To a solution of compound I K (65 mg, 0, 13 mmol) in DCM (20 mL) was added Dess-Martin periodinane (86 mg, 0.20 mmol). The mixture was stirred at 25°C for 2 hrs. TLC (PE: EA = 2: 1) showed the reaction was completed. The mixture was quenched with a solution of 10% aq. Na 2 S 2 0 3 and 10% aq. NaHC0 3 (v/v = 1/1, 20 mL). The organics were collected, washed with brine. The organics were collected, dried with Na 2 S0 4 , filtered and concentrated to afford compound 1 (21.2 mg, yield: 32.7%) as white solid. MS (ESI) m/z CM 1 1 } 484.3 l H NMR (DMSO-de , 400 MHz): δ 9.18 (s, IH), 8,77 (s, I H), 8,35 - 8.20 (m, IH), 7.45 - 7.25 (m, 5H), 7.21 - 7, 10 (m, IH), 5,63 - 5.50 (m, IH), 5.39 - 5.29 (m, IH), 5.00 (s, 2H), 4.30 - 4, 19 (m, IH), 4, 13 - 4,03 (m, IH), 2,31 - 2, 17 (m, IH), 2, 10 - 1.99 (m, IH), 1.98 - 1.85 (m, IH), 1.76 - 1.42 (m, 7H), 1.38 - 1.09 (m, 6H), 0.99 - 0.77 (m, 6H).

EX. AM: 'LE 2

Benzyl ((lR,4>y,7 t y. > 1.5 ?)-l-formy -4-isobutyl-3. > 6-dioxo-2,5- diazabicyclo| " 13.1.01he¾adecan-7-Y )carbamate (2)

[00364] Step 1: Synthesis of compounds 2A and 2B

To a solution of compound IK (86 mg, 177.1 umol) in MeOH (20 niL) was added Pd/C (25 mg, 10% purity) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H 2 (15 psi) at 5°C for 2 hours. The mixture was filtered. The filtrate was concentrated to afford the mixture of 2A and 2B (crude) (60 mg) was obtained as yellow solid. MS (ESI) m/z (M ! f ) 354.2.

To a solution of compound 2A and compound 2B (60 mg) in DCM (10.00 mL) was added CbzCi (19.3 mg, 1 13.15 umol) and Et 3 N (12.6 mg, 124.47 umol) dropwise at 0 °C. The mixture was stirred at 5 °C for Ih. The mixture was quenched with water (20 mL) and separated. The aquous layer was extracted with DCM (2 x 15 mL). The combined organic layers were dried and concentrated. The residue was purified by preparatory-HPLC (HCl condition) to afford compound 2C (16 mg, 14.50% yield) and 2D (38 mg, 34.30% yield) as a white solid.

[00366] Step 3: Synthesis of compound 2 To a solution of compound 2D (14 rng, 28.71 umo!) in DCM (15 mL) and DMSO (1.5 niL) was added DMP (24.35 mg, 57.42 umol) at 0 °C under N 2 . The mixture was stirred at 5 °C for 16 hrs under N 2 . The reaction mixture was quenched by addition sat. Na 2 S 2 0 3 (10 mL), sat. NaHC0 3 (10 mL) and stirred for 30 min. The mixture was separated. The organic layers were washed with water (10 mL x 3), brine (10 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. The residue was triturated in (i-Pr) 2 0 (3 mL) and filtered to afford compound 2 (12 mg, 23.72 umol, 41.31% yield, 95.97% purity) as a white solid. MS (ESI) m/z (M+H) + 486.2. 1H NMR (DMSO-i/ 6 , 400MHz) S 9.36 (s, 1H), 8.69 - 8.53 (m, IH), 8.28 - 8.06 (m, IH), 7.45 - 7.23 (m, 5H), 7.11 - 6.94 (m, IH), 5.00 (s, 2H), 4.32 - 4.04 m, 2H), 1.78 - 1.07 (m, 20H), 0.99 - 0.76 (m, 6H).

3

[00367] ¾ nthesisaf∞mpoundl 3

Compound 3 was prepared from compound 2D following the procedure of compound 2. Compound 3 (13.3 mg, yield 47,7 %) was obtained as a white solid. 1H NMR (400MHz, DMSC ft ) δ 9.24 (s, 1H), 8.67 - 8.58 (m, IH), 8.24 ·· 8.13 (m, 1H), 7.38 - 7.23 (m, 5H), 7.17 - 7.09 (m, 1H), 5.04 - 4.95 (m, 2H), 4.58 - 4.46 (m, 1H), 4.14 - 4.02 (m, IH), 1.96 - 1.84 (m, IH), 1.69 - 1.32 (m, 8H), 1.30 - 1.10 (m, 10H), 1.01 (s, 3H), 0.93 - 0.72 (m, 6H). MS (ESI) m/∑(M+H) + 488.3.

To a solution of (S)-2-(((benzyloxy)carbonyl)amino)non-8-enoic acid (5.22 g, 17,08 mmol), EDCI (4.91 g, 25.62 mrnol), HOBt (2.31 g, 17.08 mmol) and D1EA (6,62 g, 51 ,24 mmol, 9 mL) in DCM (50 mL) was added a solution of ethyl (R)-2-((S)-2-amino-4- methylpentanamido)pent-4-enoate (5 g, 17.08 mmol, HCl salt) in DCM (50 mL) dropwise at 0 °C, and then the reaction was stirred at 15 °C for 26 hrs. The reaction mixture was partitioned between H 2 0 (100 mL) and DCM (50 mL). The organic phase was separated and washed with brine (20 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Si0 2 , Petroleum ether/Ethyl acetate= 13 : l to 6: 1) to give compound 4A (8 g, yield: 86.6%) as a white solid. Ή NMR (400X1 Hz. DMSO-t ) δ 8.25 (d, ,/ 7.3 Hz, 1H), 7.88 (d, ,/ 8.5 Hz, 1H), 7.43 - 7.24 (m, 6H), 5.86 - 5.66 (m, 2H), 5.16 - 4.88 (m, 6H), 4.41 - 4.33 (m, IH), 4.30 - 4.22 (m, 1H), 4.14 - 3.93 (m, 3H), 2.46 - 2.34 (m, 2H), 2.02 - 1.97 (m, 2H), 1.67 - 1.54 (m, 2H), 1.52 - 1.39 (m, 3H), 1.38 - 1.20 (m, 6H), 1.16 (t, J = 7.2 Hz, 3H), 0.92 - 0.77 (m, 6H). MS (ESI) m/z (M+H) + 544, 1 .

To a solution of compound 4A (2 g, 3.68 mmol) in DCM (500 mL) was added Grubbs catalyst 2 aa generation (312 mg, 368 umol). The mixture was stirred at 40 °C for 16 hrs. The mixture was concentrated to give a residue. The residue was purified by column chromatography (SiO?, Petroleum ether/Ethyl acetate=5/l ~ 2/1 - DCM: EtOAc = 3 : 1) compound 4B (1.7 g, yield: 79.7%) was obtained as a grey solid. Ή NMR (400MHz, DMSO-fife) δ 8.16 (d, J = 7.9 Hz, 2H), 7.32 (s, 5H), 7.05 (d, J = 7.5 Hz, IH), 5.44 - 5.23 (m, 2H), 5.04 - 4.91 (m, 2H), 4.48 - 4.24 (m, 2H), 4. 13 - 3.99 (m, 3H), 2,40 (br. s,, IH), 2.28 - 2.13 (m, I H), 1.92 (br. s,, 2H), 1.73 - 1.62 (m, I H), 1.60 - 1.39 (m, 4H), 1.37 - 1.04 (m, 8H), 1.03 - 0.93 (m, IH), 0,90 - 0,74 (m, 6H). MS (ESI) m/z (\ Ι · Η ) 516.1.

[00370] Step 3: Synthesis of compound 4C

To a mixture of LiAlH 4 ( 24 mg, 3.27 mmol) in THE (4 mL) was degassed and purged with N 2 for 3 times at 0 °C, and the solution of compound 4B (560 mg, 1.09 mmol) in THE (4 mL) was added dropwise, and then the mixture was stirred at 15 °C for 2 hrs under i atmosphere at 0 °C. The reaction was quenched with H 2 0 (0.1 mL), then added 15% NaOH (0.1 mL), and then diluted with THF (20 mL). The mixture was dried over and stirred for 30 min, then filtered; the filtrate was concentrated under reduced pressure to give a residue. The residue was stirred in EtOAc (5 mL) for 30 min and filtered to give compound 4C (475 mg, yield: 87.4%) as a white solid. MS (ESI) m/z (M i l ) ' 474. 1 .

[00371] Step 4: Synthesis of compound 4D

To a solution of compound 4C ( 100 mg, 211.14 umol) in MeOH (5 mL) was added Pd/C (30 mg) under N 2 . The mixture was stirred at 15 °C for 2 hrs under H 2 balloon. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO?., DCM: MeOH = 10: 1) to give compound 4D (45 mg, yield: 51.8%) as a white solid. ! H NMR (400MHz, Methanol-<¾) δ 4.56 (t, ,/ 7.5 Hz, I H), 4.04 - 3,93 (m, IH), 3,63 - 3.61 (m, I H), 3.47 - 3.38 (m, 2H), 1.85 - 1.51 (m, 6H), 1 .49 - 1.14 (m, 12H), 1.13 - 1 .03 (m, IH), 0,95 (t, ,/ 6,6 Hz, 6H), MS (ESI) m/z (M+H 342.1.

[00372] Step S: Synthesis of compound 4E

To a solution of benzyl (2,5-dioxopyrrolidin-l-yl) carbonate (36 mg, 144.96 umol) and compound 41) (45 mg, 131.78 umol) in H 2 0 (2 mL) and dioxane (4 mL) was added K 2 CO 3 (18.21 mg, 131 ,78 umol). Then the solution was stirred at 15 °C for 16 hrs. This solution was diluted with DCM (20 mL) and washed with H 2 0 (20 mL), sat, NaHC0 3 (20 mL), dried over Na 2 S0 4 , filtered and the solvent was removed in vacuo. The residue was purified by preparatory-HPLC (HC1 condition) to give compound 4E (40 mg, yield: 63.8%) as a white solid. Ί Ι MR (400MHz, CDC1 3 ) δ 7.47 (d, J ------- 8.4 Hz, I I I ). 7,31 (hr. s., 5H),

7.05 (d. J 7,5 Hz, IH), 5.79 (d, J = 7.1 Hz, H i), 5,05 (br, s., 2H), 4.42 (d, J = 6.2 Hz, 1 1 1), 4.28 (br. s., IH), 3 ,96 (br. s., IH), 3.55 - 3.38 (m, 2H), 1.85 (br. s., i l l ). 1 ,54 (br. s., 5H), 1 ,45 ·· 1.00 (m, 14H), 0.88 (br. s., 6H). MS (ESI) m/z (M+H) + 476.1.

[00373] Step 6: Synthesis of compound 4

To a solution of compound 4E (100 mg, 211.15 umol) in DCM (10 mL) and DMSO (1 mL) was added DMP (269 mg, 633.45 umol). The mixture was stirred at 15 °C for 16 hrs. The reaction mixture was quenched by addition sat. NaHC0 3 (10 mL) and Na 2 S 2 0 3 (10 mL) at 15 °C, and then the mixture was stirred until the solution was clear, and extracted with DCIVi (10 mL x 2). The combined organic layers were washed with H 2 0 (20 mL x 2) and brine (20 mL x 2), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. The residue in DCM: PE = 10: I (20 mL) was stirred, filtered, the cake 4 (53 mg, yield: 50%) was obtained as a white solid. 1H NMR (400X1 ! iz. DMSO-afe) δ 9.42 (s, IH), 8.38 (d, J = 7.5 Hz, IH), 8.20 (d, J = 8.8 Hz, IH), 7.34 (br. s., 5H), 7.06 (d, J = 6.6 Hz, IH), 5.10 - 4.93 (m, 2H), 4.51 (d, J = 1.5 Hz, IH), 4.36 (br. s., IH), 4.15 (br. s., IH), 1.79 (br. s., IH), 1 .70 - 1.39 (m, 6H), 1.38 - 0.98 (m, 12H), 0.89 (dd, J= 5.5, 19.2 Hz, 6H). MS (ESI) }n/z (M+H) + 474.2.

EXAMPLE 5

carbamate

[00374] Step 1 : Synthesis of compounds SE and 6Z

The product 4C was separated by SFC to give two isomers.

SFC method: Column: AD (250mm*30mm,5um), Mobile phase: A: C0 2 B: IPA (0,05% NH 3 .H 2 O); Gradient: 30% of B Flow rate: 70 rnL/min Column temperature: 35 °C)

The product was separated by SFC to give compound SE (R t = 4.465 min, 200 mg) and compound 6Z (R t = ;: 4.608 min, 50 mg) both as white solid.

Compound 5E: 1H NMR (400MHz, Methanol-^) δ 7.33 (br. s., 5H), 5.37 (br. s., 2H), 5.16 - 4.99 (m, 2H), 4.50 (br. s., IH), 4.15 (br. s., i l l ). 3.89 (br. s.. H i). 3.53 - 3.36 (m, 1 1 1 ). 2.31 (d, J = 14.6 Hz, 1 1 1). 2.13 - 1.77 (m, 5H), 1.69 - 1.47 (m, 4H), 1.42 - 1.02 (m, 6H), 0.91 (dd, J=5.7, 12.8 Hz, 6H).

Compound 6Z: 1H NMR (400MHz, Methanol-^) S 7.33 (br. s., 5H), 5,37 (br. s., 2H), 5.15 - 5.00 (m, 2H), 4.48 (br, s,, IH), 4.10 (d, J = 6.2 Hz, IH), 3,93 (br, s,, IH), 3.54 (br. s., I l l ), 2.26 (br. s., IH), 2.04 (br. s., IH), 1.81 (br. s., IH), 1.58 (br. s., 5H), 1.47 - 1.12 (m, 8H), 0.95 - 0.84 (m, 6H).

Compound 5 was prepared from compound 5E following the procedure of compound 4. Compound 5 (100 mg, yield 44.7 %) was obtained as a white solid. 1H NMR (400MHz, DMSO- ) δ 9,40 (s, IH), 8.23 - 8,09 (m, 2H), 7.35 - 7,25 (s, 5H), 7, 1 1 (d, ,/ 7.5 Hz, I H), 5,43 - 5.33 (rn, IH), 5,32 - 5.23 (m, IH), 5,04 - 4.94 (m, 2H), 4,51 - 4,42 (m, 1 1 1), 4.28 (t, J 7,7 Hz, IH), 4, 10 (d, J = 3. 1 Hz, IH), 2.55 - 2.51 (m, IH), 2.12 - 1.91 (rn, 3H), 1 ,74 - 1 ,64 (m, IH), 1.63 - 1.40 (m, 4H), 1.34 - 1.12 (m, 5H), 1.00 (br. s., IH), 0.93 - 0.72 (m, 6H). MS (ESI) m/z (M+H) + 472.2.

Compound 6 was prepared from compound 6Z following the procedure of compound 4. Compound 6 (6 mg, yield 28.6 %) was obtained as a grey solid. ! H NMR (400MHz, DMSO- ) δ 9.42 (s, IH), 8.28 (d, J = 8.6 Hz, IH), 8.03 (d, ./ 7.0 Hz, IH), 7.32 (s, 5H), 7.18 (d, ./ 7.0 Hz, IH), 5.35 - 5,29 ( rn, I H), 5,23 (br. s., I H), 5,08 - 4,91 (m, 2H), 4,56 - 4,41 (m, IH), 4,36 - 4.25 (m, I H), 4.05 (br. s,, IH), 2,33 - 2.22 (m, I H), 2.05 - 1.90 (m, 2H), 1.79 - 1.00 (m, 12H 0.99 - 0.61 (m, 6H), MS (ESI) m/z (M+H) ÷ 472,2.



[00377] Step 1 : Synthesis of compound 7A

To a solution of (S)-2-((tert-butoxycarbonyl)amino)pent-4-enoic acid (5.0 g, 23 ,23 mmol) in DMF (60 mL) was added K 2 C0 3 (6.42 g, 46.46 mmol) and Etl (5.43 g, 34,85 mmol). The mixture was stirred at 20 °C for 16 hrs. The reaction mixture was quenched by addition H?0 (300 mL) at 0 °C, and extracted with EtOAc (50 mL x 2). The combined organic layers were washed with brine (200 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give compound 7A (5.5 g, yield: 97.31%) as a yellow oil. l H NMR. (400MHz, CDC1 3 ) δ 5.81 - 5.60 (m, 1H), 5.19 - 4,98 (m, 31 1 ). 4.35 (d, J = 6.2 Hz, 1 1 1 ), 4.26 - 4.09 (m, 2H), 2.63 - 2.39 (m, 2H), 1.43 (s, 9H), 1 .32 - 1. 19 (m,

[00378] Step 2: Synthesis of compound 7B

To a solution of compound 7A (1 ,0 g, 4.1 1 mmol) in EtOAc (5 ml.) was added HCl/EtOAc (4M, 5.1 mL) at 15 °C. The mixture was stirred at 15 °C for 2 hrs. The reaction was concentrated to give compound 7B (730 nig, yield: 98.9%, HCi salt) as a white solid. 1H NMR (400MHz, CDC1 3 ) δ 8.76 (br. s., 3H), 5.93 - 5.76 (m, l ! f ), 5.38 - 5.18 (m, 2H), 4.35 - 4.13 (m, 3H), 2.89 - 2.78 (m, 2H), 1.38 - 1.22 (m, 3H).

[00379] Step 3: Synthesis of coupled product 7B-1

To a solution of (ter -butoxycarbonyl)-L-leucine (900 mg, 3.89 mmol) in DCM (10 mL) was added EDCI (1.12 g, 5.84 mmol) and HOBT (525.78 mg, 3.89 mmol) and DIEA (1.51 g, 1 1.67 mmol). The mixture was stirred at 20 °C for 15 min, then added compound 7B (699 mg, 3.89 mmol). The mixture was stirred at 20 °C for 16 hrs. H 2 0 (20 mL) was added, and the mixture was extracted with DCM (10 mL x 3), the organic phase was washed with brine (20 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue, which was purified by column chromatography (Si0 2 , Petroleum ether/Ethyl acetate == 55//11 ttoo 22//11)) ttoo aaffffoorrdd tthhee ccoouupplleedd pprroodduucctt 77BB--11 ((775500 mmgg,, yyiieelldd:: 7755,.22%%)) aass aa wwhhiittee ssoolliidd.. 1H1H NNMMRR ((440000MMHHzz,, CCDDCC1I 33 )) δδ 66..5544 ((dd,, JJ== 77..55 HHzz,, 1IHH)),, 55..7744 -- 55..5599 ((mm,, 1IHH)),, 55..1111 ((dd,, JJ== 1122..33 HHzz,, 22HH)),, 44..8844 ((bbrr.. ss..,, 1IHH)),, 44..6699 -- 44..5555 ((mm,, 1IHH)),, 44..2266 -- 44..0022 ((mm,, 33HH)),, 22..6655 -- 22..4400 ((mm,, 22HH)),, 11..7755 -- 11..6633 ((mm,, 22HH)),, 11..5522 -- 11..3377 ((mm,, QQHH)),, 11..2277 ((tt,, JJ== 77..11 HHzz,, 33HH)),, 00..9933 ((tt,, JJ== 55..33 HHzz,, 66H1 f ))..

TThhee mmiixxttuurree ooff ccoouupplleedd pprroodduucctt 77BB--11 ((333300 mmgg,, 992255..7777 uummooll)) iinn EEttOOAAcc ((55 mmLL)) aanndd HHCCll//EEttOOAAcc ((44MM,, 44..66 mmLL)) wwaass ssttiirrrreedd aatt 1188 °°CC ffoorr 22 hhrrss.. TThhee ssoolluuttiioonn wwaass ccoonncceennttrraatteedd ttoo ggiivvee ccoommppoouunndd 77CC ((227700 mmgg,, yyiieelldd:: 9999..66%%o,, HHCC11 ssaalltt)) aass aa wwhhiittee ssoolliidd,, wwhhiicchh wwaass uusseedd ttoo tthhee nneexxtt sstteepp wwiitthhoouutt ppuurriifificcaattiioonn.. 33 HH NNMMRR ((440000MMHHzz,, CCDD CCII 33)) δδ 88..3388 ((bbrr.. ss..,, 33HH)),, 77..9900 ((bbrr.. ss..,, I IHl l)),, 55,,8899 -- 55..7733 ((mm,, 11 11 11 )).,.. 55..2288 -- 55,.0011 ((mm,, ..??..!! !!)),, 44,,5566 ((bbrr.. ss..,, I 1H1 1)).,. 44..3355 -- 44,.0022 ((mm,, 33HH)),, 22..6622 ((bbrr.. ss..,, 22HH)),, 11 ,.8844 ((bbrr.. s s.„, 33HH)),, 11..2255 ((tt,, ..// 66..88 HHzz,, 33HH)),, 00,,9988 ((dd,, JJ== 88..44 HHzz,, 66HH))..

[[0000338811]] SStteepp 55:: SSyynntthheessiiss ooff ccoommppoouunndd 7711))

TToo aa ssoolluuttiioonn ooff ((SS))--22--((((tteerrtt--bbuuttooxxyyccaarrbboonnyyll))aamm iinnoo))nnoonn--88--eennooiicc aacciidd ((11..8855 gg,, 66..8833 mmmmooll)) iinn DDCCMM ((4400 mmLL)) wwaass aaddddeedd EEDDCCII ((11..9966 gg,, 1100..2255 mmmmooll)) aanndd HHOOBBTT ((992222,,8877 mmgg,, 66..8833 mmmmooll)) aanndd DDIIEEAA ((22..6655 gg,, 2200..4499 mmmmooll,, 33..66 mmLL)),, tthheenn aaddddeedd ccoommppoouunndd 77CC ((22,,0000 gg,, 66..8833 mmmmooll,, HHCC11 ssaalltt)).. TThhee mmiixxttuurree wwaass ssttiirrrreedd aatt 2200 °°CC ffoorr 1166 hhrrss.. TThhee rreeaaccttiioonn mmiixxttuurree wwaass aaddddeedd HH 22 00 ((5500 mmLL)),, eexxttrraacctteedd wwiitthh DDCCMM ((2200 mmLL xx 33)),, tthhee oorrggaanniicc pphhaassee wwaass wwaasshheedd wwiitthh bbrriinnee ((5500 mmLL)),, ddrriieedd oovveerr NNaa 22 SS00 44 ,, fifilltteerreedd aanndd ccoonncceennttrraatteedd uunnddeerr rreedduucceedd pprreessssuurree ttoo ggiivvee aa rreessiidduuee.. TThhee rreessiidduuee wwaass ppuurirififieedd bbyy ccoolluummnn cchhrroommaattooggrraapphhyy ((SSii00 22 ,, PPeettrroolleeuumm eetthheerr//EEtthhyyll aacceettaattee == 55//11 ttoo 33//11)).. CCoommppoouunndd 77DD ((22..88 gg,, yyiieelldd:: 8800..44%%)) wwaass oobbttaaiinneedd aass aa wwhhiittee ssoolliidd.. 1H1H NNMMRR ((440000MMHHzz,, CCDDCC11 33 )) ·.-->> 66,,5599 ((dd.. J 66,,22 HHzz,, IIHH)),, 66..4499 ((dd,, JJ == 77..99 HHzz,, I 1H1 1)),,. 55..8866 -- 55..5588 ((mm,, 22HH)),, 55..0099 ((dd,, JJ == 1122..33 HHzz,, 22HH)),, 55..0011 -- 44..8877 ((mm,, 33HH)),, 44..6633 -- 44..5533 ((mm,, IIHH)),, 44..4499 -- 44..3377 ((mm,, IIHH)),, 44..2244 -- 44..1100 ((mm,, 22HH)),, 44..0022 ((dd,, J J = 66..22 HHzz,, IIHH)),, 22..6600 ···· 22..4444 ((mm,, 22HH)),, 22..0055 ···· 11..9977 ((mm,, 22HH)),, 11..8844 -- 11..7766 ((mm,, IIHH)),, 11..6699 -- 11..4488 ((mm,, 44HH)),, 11..4422 ((ss,, 99HH)),, 11..3377 -- 11..1188 ((mm,, 88HH)),, 00..9999 -- 00..8844 ((mm,, 6611 ff ))..

To a solution of compound 7D (700 mg, 1.37 mmol) in DCM (300 mL) was added Grubbs catalyst 2 lid generation (58.2 mg, 68.5 umol). The mixture was stirred at 40 °C for 16 hrs under N 2 . The mixture was concentrated to give a residue. The residue was purified by column chromatography (Si0 2 , Petroleum ether/Ethyl acetate ::: 5/1 to 2/1 ) to give compound 7F (520 mg, yield: 78.8%) as a white solid. 1H NMR (400MHz, CDC1 3 ) δ 6.51 (d, J = 7.5 Hz, IH), 6.24 (d, J = 8.8 Hz, IH), 5.49 - 5.35 (m, IH), 5.28 - 5.18 (m, IH), 5.16 (d, J = 7.9 Hz, ! i i ). 4,65 - 4,46 (m, 2H), 4, 21 (q, ,/ 7.1 Hz, 2H), 4.05 (t, ,/ 7.3 Hz, 1H), 2.71 - 2,57 (m, IH), 2.41 - 2,34 (m, H i ), 2,05 - 1.99 (rn, 3H), 1.74 - 1.35 (m, 1 61 1 ).. 1.33 - 1.09 (rn, 6H), 0.90 (dd, J = 6.2, 10.6 Hz, 6H). MS (ESI) m/z (M i l ) ' 482, 3.

100383! Step 7: Synthesis of compound 7G and separation to afford 7Z and 7E

To a solution of compound 7F (900 mg, 1 .87 mmol) in THF (15 mL) was added L1BH 4 (122.1 mg, 5.61 mmol). The mixture was stirred at 15 °C for 16 hrs. The excess lithium borohydride was quenched by addition of aqueous saturated ammonium chloride solution (1 mL) at 0 °C. The mixture was partitioned between ethyl acetate (20 mL) and aqueous saturated NaHC0 3 (30 mL). The aqueous layer was back extracted with ethyl acetate (10 mL x 2). The combined organic layers were dried over Na >S() ., filtered and concentrated under reduced pressure to give a residue 7G, The residue was purified by prep- HPLC (HCl condition) and then separated by SFC ((Column: IC(250mm*30mm, 10um)), Mobile phase: A: CO . B: MeOH (0.05% H 3 .H 2 0) Gradient: 15% of B Flow rate: 60 mL/min Column temperature: 35°C)) to give compound 7E (400 mg, yield: 48.7%) and compound 7Z (40 mg, yield: 4.9%) both as white solid.

Compound 7E: l H NMR (400MHz, DMSO-£¾) δ 8,07 (d, J = 8,8 Hz, IH), 7.67 (d, J = 8.4 Hz, I H), 6.32 (d, ,/ 7. i Hz, I H), 5.37 - 5.22 (m, 2H), 4,66 (t, .,/ 5,5 Hz, IH), 4.45 - 4,33 (m, I H), 4.00 (br, s„ I H), 3.74 (br, s„ I H), 3 ,30 - 3.24 (m, IH), 3.19 - 3.08 (m, I H), 2,33 - 2,21 (m, IH), 2.01 - 1 ,80 (m, 3H), 1.72 - 1.61 (m, IH), 1.57 - 0,91 (m, 19H), 0.83 (dd, 6,4, 14,3 Hz, 6H). MS (ESI) m/z ( W W ) 440.2.

Compound 7Z: 1H NMR (400MHz, Methanol-^) δ 5.37 (br. s., 2H), 4.53 - 4.43 (m, H), 4.08 - 3.88 (m, 2H), 3.60 - 3.48 (m, 2H), 2.33 - 2.20 (m, 2H), 2.14 - 1.93 (m, 2H), 1.88 - 1.17 (m, 20H), 1.00 - 0.75 (m, 6H).

To a solution of compound 7E ( 1 50 mg, 341 .23 umol) in DMSO (100 uL) and DCM (8 mL) was added DMP (289.5 mg, 682,45 umol). The mixture was stirred at 1 5 °C for 16 hrs. The reaction mixture was quenched by addition sat. NaHC0 3 (10 mL) and sat, ' Na 2 S 2 0 3 (10 mL) at 15 °C, and then the mixture was stirred until the solution was clear, and extracted with DCM (10 mL x 2). The combined organic layers were washed with H 2 0 (20 mL x 2) aanndd bbririnnee ((2200 n miLL xx 22)),, ddririeedd oovveerr NNaa 22 SS00 44 ,, ffiilltteerreedd aanndd ccoonncceennttrraatteedd uunnddeerr rreedduucceedd pprreessssuurree ttoo ggiivvee ccoommppoouunndd 77 ((114400 n migg,, yyiieelldd:: 8899..11%%)) aass aa wwhhiittee ssoolliidd.. 1H1H NNMMRR ( (440000MMHHzz,, DDMMSSOO--44 δδ 99..3399 ((ss,, I IHH)),, 88..1188 -- 88..0022 ((mm,, 22HH)),, 66..5544 ((dd,, JJ == 77..55 HHzz,, IIHH)),, 55..4422 -- 55..3322 ((mm,, IIHH)),, 55..2299 -- 55..2200 ((mm,, IIHH)),, 44..4499 -- 44..3399 ((mm,, I IHH)),, 44..2233 ((bbrr.. ss..,, IIHH)),, 33..9988 ((bbrr.. ss..,, IIHI I))., 22.. 11 77 -- 22..0055 ((mm,, I l H! f)),, 22..0011 -- 11..8855 ((mm,, 22HH)),, 11..7777 -- 11..5544 ((mm,, 33HH)),, 11..4499 -- 11..0088 ((mm,, 11 77HH)),, 00..9999 ((bbrr.. ss..,, IIHH)),, 00..9922 -- 00..7744 ((mm,, 66H1 1)).. MMSS ((EESSII)) mm//zz ί (M\Ι+H ! ί)} +' 443388..33..

To a stirred solution of compound 7 (180 mg, 41 1.36 umol) in DCM (10 mL) was added isocyanoethane (27.2 mg, 493.63 umol) (2 drops) and Py (130.2 mg, 1.65 mmol) at 0 °C, then TFA (93,8 mg, 822.72 umol) was added slowly dropwise. The reaction mixture was stirred for 30 min at 0 °C. Then the reaction temperature was allowed to 16 °C and stirred for 16 hrs. The mixture was diluted with DCM (10 mL), washed with 1 N HCl (20 mL x 2), saturated NaHC0 3 (20 mL x 2), dried over anhydrous Na 2 S0 4 , filtered and concentrated. The residue was purified by preparatory-HPLC (HCl condition) to give compound 8A (100 mg, yield: 47.6%) as a white solid. 1H NMR (400MHz, MeOD) δ 5.42 - 5.3 1 (m, 2H), 4.61 - 4.48 (m, 1 i f ), 4.33 (br. s , IH), 4.24 (d, J = 1 1 .0 Hz, 1 H), 4.11 - 3.94 (m, 2H), 3.26 - 3.08 (m, 21 ! }. 2.23 (br. s,, IH), 2.04 (br, s,, IH), 1 ,96 (br, s., IH), 1 ,87 - 1.73 (m, I H), 1 .71 - 1.50 (m, 5H), 1.42 (br. s., 9H), 1.34 - 1.19 (m, 5H), 1.12 (t, J = 7.1 Hz, 3H), 0,98 - 0.83 (m, 6H). MS (ESI) ra/z (M+H) + 51 I ,4.

[00386] Step 10: Synthesis of compound 8:

Tert-hutyl ((3S',6 t y,15»S l . E)-15-(2- ethylamino)-2-oxoacetyl)-3-isobutyl-2,5-dioxo-l,4-

Compound 8 was prepared from compound 8 A following the procedure of compound 7. Compound 8 (80 mg, yield 75.5 %) was obtained as a white solid. 1H NMR (400MHz, ]. ) \ !S(. „} δ 8.71 (br. s., IH), 8.18 - 8,02 (m, 2H), 6.52 (d, J = 7, 1 Hz, 1 1 1 ), 5.47 - 5.37 (m, I H), 5.30 - 5.24 (m, I H), 5,00 (br. s., I H), 4.50 - 4.40 (m, I H), 4,00 (br, s„ I H), 3,20 - 3.05 (m, 2H), 2,44 - 2.38 (m, I H), 2.13 (br. s., I H), 1.95 (br. s,, 2H), 1.69 (br, s,, IH), 1 ,62 - 1 .09 (m, 19H), 1.03 (t, J= 7.1 Hz, 3H), 0,91 - 0,67 (m, 6H), MS (ESI) m/z (M+H) + 509,4, 3 ,

[00387] Step 11: Synthesis of compound 9:

Tert-butyl ((3Ly,6»S',15S , -15-(2-(ethylamino)-2-oxoacetyl)-3-isobutyl-2,5-dioxo-l,4- diazacyclopentadecan-6-y )carbamate To a solution of compound 8 (50 mg, 98,30 umol) in THF (5 mL) was added Pd/C (10 mg) under N 2 . The mixture was stirred at 15 °C for 2 hrs under H 2 balloon. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by preparatory-HPLC (HCl condition) to afford compound 9 (15.5 mg, yield: 30.9%) as a white solid. Ή NMR (400MHz, DMSO- ) δ 8.39 (br. s,, ill), 8.05 (d, ./ 7.5 Hz, IH), 7.87 (d../ 8.5 Hz, 1H), 6.03 (br. s., HI).5.17 - 5,08 (m, III), 4.57 - 4.49 (m, IH), 4.11 - 4.03 (m, IH), 3.21 - 3.15 (m, 2H), 1,85 - 1.75 (m, IH), 1,72 - 1,59 (m, 2H), 1.55 - 1.42 (m, 4H), 1.41 - 1.17 (m, 21H), 1.09 (t, J= 7.3 Hz, 3H), 0.93 - 0.87 (m, 6H). MS (ESI) m/z (W W) 5! 1.2.

Compound 10 was prepared from compound 7Z following the procedure of compound 7. Compound 10 (16.2 mg, yield 27 %) was obtained as a white solid. 1H NMR (400MHz, DMSO-ofe) δ 9,40 (s, IH), 8.24 (d, ,/ 8.8 Hz, IH), 7.91 (d, J= 7.1 Hz, IH), 6.62 (d, J= 7.5 Hz, IH), 5.39 - 5.26 (m, IH), 5,24 - 5.12 (m, IH), 4,52 - 4.41 (m, H), 4.26 (d, J = 3.5 Hz, IH), 3.94 (br. s.. IH), 2,41 (br. s., IH), 2.31 - 2.20 (m, IH), 2.03 - 1.88 (m, 2H), 1.70 -- 1.50 (m, 2H), 1.51 - 1.08 (m, 18H), 0.91 - 0.73 (m, 6H). MS (ESI) m/z { W) 438.3.

[00389] Step 13: Synthesis of compound 11 A

To a solution of compound 7G (100 mg, 227.48 umol) in MeOH (5 mL) was added Pd/C (30 mg) under N 2 . The mixture was stirred at 15 °C for 16 hrs under H 2 balloon. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. Compound 11A (70 mg, yield: 69.7%) was obtained as a w hite solid. 1H NMR (400MHz, CDCI 3 ) > 7.77 (d, ,/ 8.8 Hz, IH), 7.12 (d, ,/ 7.9 Hz, IH), 5.54 (d, ,/ 7.9 Hz, IH), 4.48 - 4.40 (m, IH), 4,30 (br. s., H), 3.97 (br. s., IH), 3.55 - 3.42 (m, 2H), 1,85 - 1.75 (m, IH), 1.69 - 1.50 (m, 5H), 1.45 - 1.02 (m, 22H), 0,89 (I../ 6.2 Hz, 6H). MS (ESI) m/z (M il) 442.2

Tert-butyl ((3^6A15>y-15-formyl-3-isobiityI-2,S-dioso-l.,4-di¾z¾cy cIopentadec¾n-6- ylkarbamate

Compound 11 was prepared from compound 1.1 A following the procedure of compound 7. Compound 11 (29 mg, yield 38.3 %) was obtained as a white solid. L H NMR (400MHz, DMSO-de) δ 9.39 (s, IH), 8.36 (d, 7.5 Hz, 1 H), 8.13 (d, ,/ 8.8 Hz, 1H), 6,41 (d, J = 7.1 Hz, IH), 4.48 (d, J = 7,5 Hz, 1 1 1), 4.30 (br. s., I l l ), 4,03 (br. s., I l l ), 1.76 (br, s., I l l ), 1.65 - 1.55 (rn, 2H), 1.48 - 1.05 (m, 25H), 0.89 -0.82 (m, 6H). MS (ESI) m/z (M I S) 440.3.

[00391] Step 15: Synthesis of compound 12A

To a solution of compound 7F (800 mg, 1.66 mmol) in EtOAc (10 mL) was added dropwise HCl EtOAc (4M, 10 mL) at 0°C. Then the mixture was stirred at 15 °C for 4 hrs. The reaction mixture was concentrated under reduced pressure to give compound 12A (700 mg, cmde, HCl) was obtained as a white solid. The crude product was directly used without further purification. 1H NMR (400MHz, CDC1 3 ) δ 8,66 (d, J = 9.3 Hz, IH), 8,52 (d, J = 8.8 Hz, I H), 8,24 (br, s , 3H), 5.45 - 5,24 (m, 2H), 4.54 id. ./ 7.9 Hz, I H), 4.36 (t, ./ 9.3 Hz, IH), 4,07 (d,■/ 5.7 Hz, 2H), 3.80 (br. s., IH), 2.26 - 2. 1 1 (m, IH), 1 ,96 (br, s,, 2H), 1.76 - 1.43 (m, 4H), 1.41 - 0.99 (m, 101 1 ), 0,90 (dd, ./ 6,0, 16.5 Hz, 6H). MS (ESI) m/z (M I S) 382.0.

[00392] Step 16: Synthesis of compound 12 B

To a solution of 2,5-dioxopyrrolidin-l-yl (2-(2-methoxyethoxy)ethyl) carbonate (750 mg, 2.87 mmol), compound 12A (600 mg, 1.44 mmol, HCl) in dioxane (10 mL) and H 2 0 (5 mL) was added K <C() ; (298 mg, 2.15 mmol) at 0 °C. The mixture was stirred at 20 °C for 3 hrs. This solution was added with H 2 0 (100 mL), filtered and the cake was dissolved with DCM (10 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. The crude product compound 12B (450 mg, yield: 42%) as a grey solid was used into the next step without further purification. 1H NMR (400MHz, CDC1 3 ) δ 6.51 ~ 6.37 (m, IH), 6,24 (d. J 8.8 Hz, IH), 5,63 - 5.51 (m, IH), 5,44 - 5.38 (m, IH), 5,30 - 5. 17 (m, IH), 4.64 - 4.45 (m, 2H), 4.27 - 4.12 (m, 4H), 3.74 - 3.59 (m, 4H), 3.58 - 3.50 (m, 2H), 3.38 (s, 3H), 2.73 - 2.60 (m, IH), 2.33 - 2.25 (m, IH), 2.11 - 1.90 (m, 3H), 1.71 - 1.46 (m, 5H), 1.42 - 1.14 (m, 9H), 0.98 - 0.78 (m, 61 f ). MS (ESI) m/z (M+H) + 528.2.

[00393] Step 17: Synthesis of compound 12C

To a mixture of LiAlH f (108 mg, 2.84 mmol) in THF (10 mL) was degassed and purged with N 2 for 3 times at 0 °C, and the solution of compound 12B (500 mg, 947.60 umol) in THF (10 mL) was added dropwise, and then the mixture was stirred at 0 °C for 2 hrs under N 2 atmosphere. The reaction was quenched with H 2 0 (0.1 mL), 15% NaOH (0.1 mL), then added H 2 0 (0.3 mL) and then diluted with THF (20 nil.). The mixture was dried over Na 2 S0 4 and stirred for 30 min, then filtered the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by preparatory-HPLC (HC1 condition) to give compound 12C (150 mg, yield: 32%) as a white solid. 1H NMR (400MHz, CDC1 3 ) δ 7.59 (d, J = 9.0 Hz, 1H), 6.42 (d, J = 8.2 Hz, I I I). 6.01 (d, J = 7.8 Hz, 1H), 5.44 - 5.32 (m, 2H), 4.68 - 4.56 (m, 1 H), 4.50 (br. s., 1 H), 4.26 - 4, 13 (m, 2H), 4,04 (br, s., 1H), 3,68 - 3.62 (m, 4H), 3.58 - 3,46 (m, 3H), 3.38 (s, 3H), 2,52 -2,42 (m, I H), 2, 12 - 1.91 (m, 4H), 1.76 - 1.49 (m, 6H), 1.40 - 1.10 (rn, 61 1 ), 0.98 - 0.86 (m, 6H). MS (ESI) m/z (M+H) + 486.2.

[00394] Step 18; Synthesis of compound 12:

2-(2-methoxyethoxy)ethyl ((3^6Α^5 ,£ ·-15-!θΓΜν1-3-ί§οΙηιΐνΙ-2,5·-άίο¾ -1,4- diazacyclopentadec-12-en-6-y )carbamate

C Coommppoouunndd 1122 wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 1122CC f foolllloowwiinngg tthhee pprroocceedduurree ooff ccoommppoouunndd 77.. CCoommppoouunndd 1122 ((113300 n migg,, yyiieelldd 7777..55 %%)) wwaass oobbttaaiinneedd aass aa wwhhiittee ssoolliidd.. 1H1H N NMMRR ((440000MMHHzz,, CCDDCC1I 3 3)) δδ 99..5566 ((ss,, 1IHH)),, 66,,6655 ((dd,, JJ== 77..00 HHzz,, IIHH)),, 66..5533 ((dd,, JJ== 77..88 HHzz,, 1IHH)),, 55,,5588 ((dd,, JJ == 77,.44 HHzz,, 1IHH)),, 55..4455 -- 55..4400 ((mm,, 1IHH)),, 55..2288 -- 55..1166 ((mm,, 1IHH)),, 44,,6622 -- 44..5500 ((mm,, 22HH)),, 44..2277 -- 44..1100 ((mm,, 33HH)),, 33..7755 -- 33..5599 ((mm,, 44HH)),, 33..5555 ((dd,, JJ -------------- 44..33 HHzz,, 22HH)),, 33..3388 ((ss,, 33HH)),, 22..7755 -- 22,,6655 ((mm,, II HH)),, 22..3333 --

22..2200 (( rrnn.. IIHH)),, 22..0099 -- 11..9911 ( (mrn,, 33HH)),, 11..7700 -- 11..4477 ((rrnn,, 44HH)),, 11..4411 -- 11 .. 1100 ((mm,, 66HH)),, 00..9922 ((dddd,, ..// 66..33,, 1133..77 HHzz,, 6611 11 )) MMSS ((EESSII)) mm//zz ((MM II ff }} '' 448844 22..

[[0000339955]] SStteepp 1199;; SSyynntthheessiiss ooff ccoommppoouunndd 1133AA

CCoommppoouunndd 1133AA wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 1122 aanndd iissooccyyaannooccyycclloopprrooppaannee fofolllloowwiinngg tthhee pprroocceedduurree ooff ccoommppoouunndd 88AA.. CCoommppoouunndd 1133 AA ((4400 mmgg,, yyiieelldd 4400..99 %%)) wwaass oobbttaaiinneedd aass aa wwhhiittee ssoolliidd.. 1H x \ \ NNMMRR ((440000MMHHzz,, CCDDCCII33)) δδ 88..5500 ((bbrr.. ss..,, IIHH)),, 77..1199 ((bbrr.. ss..,, IIHH)),, 66..8877 -- 66..5599 ((mm,, IIHH)),, 66..1133 ((dd,, JJ 77..44 HHzz,, IIHH)),, 55..4477 -- 55..2211 ((mm,, 22HH)),, 55..0011 -- 44..5500 ((mm,, 33HH)),, 44..3388 -- 44..0088 ((mm,, 44HH)),, 33..7766 -- 33,,4455 ((mm,, 66HH)),, 33..3388 ((ss,, 33HH)),, 22..7766 -- 22..6600 ((mm,, IIHH)),, 22..3399 ((dd,, JJ == 1133..33 HHzz,, HH)),, 22..1166 -- 11..7777 ((mm,, 44HH)),, 11..7766 -- 11..4455 ((mm,, 66HH)),, 11 ,,3388 --11 ,,2200 ((mm,, 44HH)),, 11 ,, 1155 --11 ..0066 ((mm,, IIHH)),, 00,.9999 -- 00..8888 ((mm,, 55HH)),, 00,,8822 -- 00,,6622 ((mm,, 22HH)),, 00..5555 -- 00..3355 ((mm,, 22HH)).. MMSS ((EESSII)) mm//zz ((MM++HH)) ++ 556699..11..

2-(2-methoxyethoxy)ethyl ((3A ' ,6>y,15^,E -15-(2-(cycIopropyIamino)-2-oxoacetyI)-3- isobutyi-2,5-dioxo-l,4-diazacyclopentadec-12-en-6-yi ' )carbamate

Compound 13 was prepared from compound 13A following the procedure of compound 7. Compound 13 (15 mg, yield 28.6 %) was obtained as a white solid. 1H NMR (400MHz, DMSO-i¾) δ 8.73 (br. s., 1H), 8.13 (br. s., 2H), 6.94 (br, s., H), 5,40 (br, s., 1H), 5.30 (br. s., IH), 5,00 (br. s., IH), 4.45 (br. s., IH), 4.00 (br. s., 3H), 3.60 - 3.34 (m, 6H), 3,21 (br, s., 3H), 2.72 (br. s., !!!)..2.42 - 2.29 (m, 111), 2.08 (br. s., IH), 1.95 (br, s., 2H), 1.67 (br. s., IH), 1.57 - 1.09 (m, 9H), 1.01 (br. s., IH), 0.95 - 0.71 (m, 6H), 0.70 - 0.45 (m, 4H). MS (ESI) m/z (M+H + 567.3.

To a solution of ethyl (lR,2S)-l-((tert-butoxycarbonyl)amino)-2- vinylcyclopropane-l-carboxylatecompound (7.0 g, 27.42 mmol) in EtOAc (50 mL) was added HCi/dioxane (4M, 40 mL). Then the reaction was stirred at 15 °C for 3 hours. Then PE (200 mL) was slowly added to the reaction. The white solid was filtered, collected and dried in vacuum to give compound 14A (5.26 g, crude, HCl salt), which was used directly for the next step without further purification, ! 1 1 NMR (DMSO-i&, 400MHz) δ 9.21 (br. s., 3H), 5.71 - 5.55 (m, H I ), 5.35 (d, J = 17,2 Hz, IH), 5, 18 (d, J= 10.6 Hz, 1H), 4.27 - 4.10 (m, 2H), 2.58 - 2.51 (m, IH), 1.93 - 1.82 (m, IH), 1.67 - 1.57 (m, IH), 1.21 (t, J= 7.1 Hz, 3H).

[00398] Step 2: Synthesis of compound 14B

To a solution of (fert-butoxycarbonyl)-L-leucine (6.3 g, 27.4 mmol), compound 14A (5.3 g, 27.4 mmol) and HOBt (3.7 g, 27.4 mmol) in DCM (150 mL) was added ! · !)( ! (7.9 g, 41.1 mmol) and DIPEA (10.6 g, 82.2 mmol, 14.4 mL). The reaction was stirred at 15 °C for 16 hours. The solvent was removed under reduced pressure. The residue was dissolved in EtOAc (200 mL) and then washed with HCl (80 mL, O.SM), sat. NaHC0 3 (70 mL x 2) and brine (80 mL). The organics layers were dried over Na>S() ., filtered and concentrated to afford compound 14B (10 g, yield: 85.7%) as off white solid, which was used directly for next step without further purification. 1H NMR (DMSO-£¾, 400MHz) δ 8.64 (s, IH), 6.71 (d, J = 8.4 Hz, IH), 5.70 - 5.53 (m, IH), 5.33 - 5.20 (m, IH), 5.15 - 5.00 (m, IH), 4.04 - 4.00 (m, 2H), 3.97 - 3.88 (m, IH), 2.12 (q, J = 8.7 Hz, IH), 1.65 - 1.50 (m, 2H), 1.46 - 1.29 (m, 11H), 1.23 - 1.20 (m, IH), 1.13 (t, J = 7.3 Hz, 3H), 0.91 - 0.79 (m, 6H). MS (ESI) m/z ' (M+Na) ÷ 391.1. To a solution of compound 14B (9.9 g, 26.9 mmol) in EtOAc (50 mL) was added HCl/dioxane (4M, 75 mL). The reaction was stirred at 15°C for 6 hours. The reaction mixture was concentrated and then PE (700 mL) was added. The solid was collected by filtered and dried in vacuum to afford compound 1.4C (7.95 g, yield: 97, 1%, HC1 salt) as white solid, which was used directly for the next step without further purification. 1H NMR (DM SO-i/r.. 400MHz) .-> 9,35 (s, H I ), 8.32 (br. s., 3H), 5.81 - 5.62 (m, IH), 5,27 (dd. ./ 1.3, 17.2 Hz, IH), 5.1 1 (dd, J = 1.3, 10.1 Hz, IH), 4.13 - 3.99 (m, 2H), 3.68 (br. s., IH), 2.21 (q, J = 8.8 Hz, IH), 1.75 - 1.48 (m, 4H), 1.43 - 1.31 (m, IH), 1.16 (t, J = 7.1 Hz, 3H), 0.99 - 0.81 (m, 6H).

[00400] Step 4: Synthesis of eomponnd 14D

To a solution of compound 14C (2.0 g, 6.56 mmol), (S)-2-((tert- butoxycarbonyl)amino)non-8-enoic acid (1.78 g, 6.56 mmol) and HOBt (887 mg, 6.56 mmol) in DCM (80 mL) was added EDCI (1.90 g, 9.91 mmol) and DIPEA (2.54 g, 19.68 mmol, 3.44 mL). Then the reaction was stirred at 15°C for 18 hrs. The solvent was removed under reduced pressure. The residue was dissolved in EtOAc (100 mL), and the solution was washed with HC1 (0.5M, 40 mL), sat, NaHC0 3 (40 mL x 3) and brine (50 mL). The organics were collected, dried over Na 2 S0 4 , filtered and concentrated to afford compound 14D (3.1 g, yield: 90.6%) as off-white solid, which was used directly for next step without further purification. l H NMR (DMSO-£¾, 400MHz) δ 8.69 (s, IH), 7.70 (d, J= 8.4 Hz, IH), 6.92 (d, J = 7.9 Hz, IH), 5.84 - 5.72 (m, IH), 5.67 - 5.55 (m, IH), 5.24 (dd, J = 1.5, 17.0 Hz, IH), 5.12 - 5.05 (m, IH), 5,03 - 4.89 (m, 2H), 4.32 - 4.22 (m, IH), 4,06 - 3.93 (m, 2H), 3.91 - 3 ,79 (m, I H), 2.12 - 2.04 (m, I H), 2,03 - 1.95 (m, 2H), 1.65 - 1.48 (m, 4H), 1 .47 - 1 .39 (m, 3H), 1.39 - 1.32 (m, 9H), 1.31 - 1.17 (m, 61 1 ), 1.12 (t, ./ 7, 1 Hz, 3H), 0.92 - 0.78 (m, 6H). MS (ESI) m/z (M+H) + 522.2.

[00401] Step 5: Synthesis of compound 14E

To a solution of compound 14D (500 mg, 0,96 mmol) in DCM (150 mL) was added Hoveyda-Grubbs 1 st Generation catalyst (75 mg, 0.12 mmol). The reaction was stirred at 51 °C for 48 hours. The solvent was removed in vacuum. The residue was purified by column chromatography (SiO?, Petroleum ether/Ethyl acetate = 4/1 to 3/1) to afford compound 14E (340 mg, yield: 69.0%) as off-white solid. 1H NMR (DMSO-£¾, 400MHz) δ 88..5555 ((bbrr.. ss..,, 1I HH)),, 88..1166 ((dd,, JJ == 77..11 HHzz,, II HH)),, 66..4444 ((dd,, ..// 77..11 HHzz,, I I HI I))., 55..5599 -- 55..4444 ( (mm,, 1IHH)),, 55,,3333 ((tt,, ..// 99..55 HHzz,, I HHi)),, 44,,2200 -- 44..0099 ((mm,, IIHH)),, 44,,0077 -- 33..9911 ((mm,, 33HH)),, 22..2277 ((bbrr,, ss..,, IIHH)),, 11..9977 -- 11 ,.8844 ((rmn,, 22HH)),, 11..7777 -- 11..6655 ((mm,, IIHH)),, 11 ,,6633 -- 11..2299 ((mm,, I166HH)),, 11..2288 -- 11..0066 ((mm,, 88HH)),, 00..9988 -- 00..7799 ((rmn,, ( 6:»H! !)}., MMSS ((EESSII)) mm//zz ((MM++HH)) ++ 449944..44..

[[0000440022]] SStteepp 66:: SSyynntthheessiiss ooff ccoommppoouunndd 1144FF

TToo aa mmiixxttuurree ooff L LiiAAllHH 44 ((5533 mmgg,, 11 ..4400 mmmmooll)) iinn TTHHFF ((44 mmLL)) wwaass aaddddeedd aa ssoolluuttiioonn ooff ccoommppoouunndd 1144EE ((223300 mmgg,, 00,,4477 mmmmooll)) iinn TTHHFF ((88 mmLL)) aatt 00°°CC.. TThhee rreessuullttiinngg mmiixxttuurree wwaass ssttiirrrreedd aatt 00°°CC ffoorr 11..55 hhrrss.. TThheenn tthhee mmiixxttuurree wwaass ddiilluutteedd wwiitthh TTHHFF ((3300 mmLL)) aanndd tthheenn q quueenncchheedd wwiitthh HH 22 00 ((00..11 mmLL)),, NNaaOOHH ((1155%%,, 00..11 mmLL)) aanndd HH 22 00 ((00..33 mmLL)).. AAfftteerr bbeeiinngg ssttiirrrreedd aatt rroooomm tteemmppeerraattuurree ffoorr 4400 mmiinnss,, tthhee mmiixxttuurree wwaass ddrriieedd oovveerr NNaa 22 SS00 44 .. TThhee ssoolliidd wwaass rreemmoovveedd bbyy f fiillttrraattiioonn,, aanndd tthhee fifillttrraattee wwaass ccoonncceennttrraatteedd ttoo ggiivvee aa rreessiidduuee,, wwhhiicchh wwaass ppuurriififieedd bbyy rree-- ccrryyssttaalllliizzaattiioonn ffrroomm EEttOOAAcc ((66 mmLL)) ttoo aaffffoorrdd ccoommppoouunndd 1144FF ((113300 mmgg,, yyiieelldd:: 5599..33%%)) aass wwhhiittee ssoolliidd.. HHii NNMMRR. ((DDMMSSOO--ddfefe,, 440000MMHHzz)) δδ 88,,2299 ((bbrr.. ss..,, IIHH)),, 88..1100 ((dd,, JJ== 77..55 HHzz,, IIHH)),, 66,,4499 -- 66,,3344 ((mm,, IIHH)),, 55..5522 -- 55..3399 ((mm,, IIHH)),, 55..0044 ((tt,, JJ == 99..11 HHzz,, I IHH)),, 44..6677 ((bbrr.. ss..,, IIHH)),, 44..2233 -- 44..1111 ((mm,, IIHH)),, 44..0077 -- 33..9955 ((mm,, IIHH)),, 33..6688 -- 33..5588 ((mm,, IIHH)),, 33..2288 ((bbrr.. ss..,, IIHH)),, 22..3300 -- 22..1199 ((mm,, IIHH)),, 11..9988 -- 11..8877 ((mm,, IIHH)),, 11..7722 -- 11..4411 ((mm,, 77HH)),, 11..3355 ((ss,, 99HH)),, 11..3300 -- 11..0066 ((mm,, 66HH)),, 00..9977 -- 00..7799 ((mm,, 66HH)),, 00..7788 -- 00..7722 ((mm,, IIHH)).. MMSS ((EESSII)) mm//zz ((MM++NNaa)) ++ 447744..22..

Tert-butyl ((lR,4A 7A\153^-l-formyl-4-isobutyl-3,6-dioxo-2,5- diazabicyclo[13.1.0]hexadec-13-en-7-yl)carbamate

Compound 14 was prepared from compound 14F following the procedure compound 7. Compound 14 (63 mg, yield 61.4 %>) was obtained as a white solid. H NMR

((DD SSOO--fififcfc,, 440000MMHHzz)) <<)) 99 !1 77 ((ss,, I IHH)),, 88,,7766 ((bbrr,, ss,,,, IIHH)),, 88..2233 ((dd,, ..// 77..11 HHzz,, IIHH)),, 66..4477 ((dd,, ..// 77..11 HHzz,, IIHH)),, 55..5599 -- 55..4488 ((mm,, IIHH)),, 55..3333 ((tt,, JJ == 99..55 HHzz,, IIHH)),, 44..2299 -- 44..1188 ((mm,, IIHH)),, 44..0055 -- 33..9944 ((mm,, I IHH)),, 22..2222 ((bbrr.. ss..,, I IHH)),, 22..0088 · -· 11..9999 ((mm,, IIHH)),, 11..9977 ···· 11..8866 ((mm,, IIHH)),, 11..7799 -- 11..4411 ((mm,, 88HH)),, 11..3355 ((ss,, 99HH)),, 11..2277 -- 11..0077 ((mm,, 55HH)),, 11..0033 -- 00..7799 ((mm,, 66HH)).. MMSS ((EESSII)) mm//zz ((MM++HH)) ++ 445500..33..

Compound ISA was prepared from compound 14 and isocyanoethane following the procedure of compound 8A. Compound ISA (55 mg, yield 19.5 %) was obtained as a white solid, MS (ESI) m/z (M+H) + 523,2, Tert-butyl ((lR,4^,7»S 5»S,Z)-l-(2-(ethyIaniino)-2-oxoacetyl)-4-isobutyl-3,6-dioxo -2,5- diazabicyclo[13.1. (

Compound 15 was prepared from compound ISA following the procedure of compound 7. Compound 15 ( 16 mg, yield 35 ,7 %) was obtained as a white solid. 1H NMR ( DM SO-i/r,, 400MHz) .·) 8.74 (br. s„ 1H), 8.31 (hr. s.. 1 H), 8. 19 (br. s„ 1H), 6,41 (d, 6.6 Hz, IH), 5.54 - 5.45 (m, 1H), 5.43 - 5.36 (m, IH), 4, 16 (br, s,, IH), 4.04 (br. s., H I ). 3.09 - 3.00 (m, 2H), 2.17 (br. s., IH), 2.07 - 2,00 (m, IH), 1 ,95 (br, s., H), 1 ,84 (br. s., IH), 1.70 - 1.51 (m, 5H), 1.45 - 1.39 (m, 2H), 1.35 (br. s., 9H), 1.28 - 1. 13 (m, 5H), 1.00 (t, J = 7.1 Hz, 3H), 0.93 - 0.79 (m, 6H). MS (ESI) m/z (M+H) + 521.5.

To a solution of compound 14F (103 mg, 0,23 mmol) in THF (15 mL) was added Wilkinson's cat. (100 mg, 0.1 1 mmol). The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H 2 (15 psi) at 15°C for 72 hours. The reaction mixture was filtered and the filtrate was concentrated. The residue was purified by column chromatography (Si0 2 , Petroleum ether/Ethyl acetate = 4/1 to 3/1) to afford crude product (85 mg), which was further purified with preparatory-HPLC (HC1) to afford compound 16A (50 mg, yield: 48,3%) as white solid. 1H NMR ( DMSO-i/r,. 400M Hz) δ 8.26 (br. s., IH), 8.02 (d, J = 7.9 Hz, IH), 6.28 (d, J = 7.5 Hz, IH), 4.88 - 4.79 (m, IH), 4.21 - 4. 1 1 (m, IH), 4.07 (br. s., IH), 3.67 - 3.59 (m, IH), 3.45 - 3.38 (m, IH), 1.74 - 1.02 (m, 26H), 1.00 - 0.72 (m, 8H), 0.45 - 0.35 (m, IH). MS (ESI) m/z (M+H) + 454.2.

[00407] Step 11; Synthesis of compound 16:

Compound 16 was prepared from compound 16A following the procedure of compound 7. Compound 16 (21 mg, yield 42.7 %) was obtained as a white solid. l H NMR

I S< )-</,·., 400MHz) δ 9.35 (s, IH), 8.58 (br. s., I H), 8.17 (d, ,/ 7.9 Hz, IH), 6,32 (d, ./ 7.1 Hz, I H), 4.27 - 4, 17 (m, IH), 4.08 (br. s.. IH), 1.80 - 1 .42 (m, 10H), 1 .35 (s, 13H), 1.24 - 1.05 (m, 6H), 0.98 - 0,80 (m, 6H), MS (ESI) m/z (M+Na) ÷ 474.2.

)8] Step 12: Synthesis of compound 17A

To a solution of compound 14F (200 mg, 442.87 umol) in MeOH (20 mL) was added Pd/C (40 mg, 10%wt). The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H 2 (15 psi) at 15 °C for 16 hours. The reaction mixture was filtered through celite and the filtrate was concentrated. The crude product was purified with preparatory-HPLC (HCl condition) to give compound 17A (85 nig, yield

4422..1122%%)) aass wwhhiittee ssoolliidd.. !! HH NNMMRR ((44Q0G0MMHHzz,, DDMMSSOC ^4)) δδ 77..9911 ((dd,, JJ== 99..33 HHzz,, 1IHH)),, 77..3333 ((bbrr.. ss..,, 1HH)),, 66..6644 ((dd,, 77..11 HHzz,, 1IHH)),, 44..4411 ((bbrr.. ss..,, 1IHH)),, 33..9988 ((bbrr.. ss..,, 1IHH)),, 33..4499 -- 33..4400 ((mm,, 22HH)),, 11..9977 ((bbrr.. ss,,,, HH)),, 11 ..7733 -- 11..4499 ((mm,, 33HH)),, 11..4488 -- 11..0088 ((mm,, 2244HH)),, 11 ..0055 ((bbrr.. ss,,,, 33HH)),, 00..9944 -- 00..7766 ((mm,, 66HH)).. MMSS ((EESSII)) mm//zz ((MM++HH)) ++ 445566,,33..

diazacvclopentadecan-6-yl)carbamate

Compound 17 was prepared from compound 17A following the procedure of compound 7. Compound 17 (45 mg, yield 52.4 %) was obtained as a white solid. 1H NMR (400MHz, DMSO-6¾) δ 9.26 (s, 1 1 1), 8,60 (br, s,, IH), 8.09 (d, ./ 8.8 Hz, 1 1 1 ).. 6.58 (d, ./ 7.5 Hz, IH), 4.60 - 4.49 (m, IH), 4.00 (d, J = 3.1 Hz, IH), 1.95 - 1.84 (m, IH), 1.64 - 1.53 (rn. 2H), 1.52 - 1.32 (m, 13H), 1.32 - 1.10 (m, 12H), 1.03 (s, 3H), 0.93 - 0.80 (m, 6H). MS (ESI) m/z (M-55) ÷ 398.3.

EXAMPLE 8

Step 1 : £

To a solution of (2S)-5-oxopyrrolidine-2-carboxylic acid (40 g, 309 mmol) in propan-2-ol (200 mL) was added SOCl 2 (129 g, 1.08 mol) slowly. The mixture was stirred at 15 °C for 2 hrs. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product 18A (52 g, yield: 98.1%) was obtained as red oil, which was used into the next step without further purification. 1H NMR (400MHz, CDC1 3 ) «5 6.89 (br. s., 1H), 5.08 - 4.95 (m, IH), 4.21 - 4.1 1 (m, I I I). 2.49 - 2.22 (m, 3H), 2.19 - 2.06 (m, H i), 1.27 - 1.17 (m. 6H). MS (ESI) m/z (M+H) + 171.8.

[00411] Step 2: Synthesis of compound 18B

To a solution of compound 18A (79.5 g, 364.5 mmol) in CH 3 CN (300 mL) was added DMAP (1.86 g, 15.19 mmol). The mixture was stirred at 10 °C for 1 h. The reaction mixture was concentrated and diluted with MTBE (200 mL), the organic layer was washed with I N HC1 (150 mL) and brine (200 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give compound 18B (80 g, yield: 97%) as a light yellow oil, which was used in the next step without further purification. l H NMR (400MHz, CDCI 3 ) δ 5.16 - 4.99 (m, 1H), 4.59 - 4.49 (m, IH), 2.65 - 2.25 (m, 3H), 2.06 - 1.92 (m, IH), 1.61 ·· 1.34 (m, 9H), 1.31 - 1.19 (m, 6H).

[00412] Step 3: Synthesis of compound 18C

To a solution of compound 18B (40 g, 147.4 mmol) in MeOH (300 mL) was added NaBH 4 (8.37 g, 221.1 mmol) slowly. The mixture was stirred at 18 °C for 2 h. The reaction mixture was diluted with water (500 mL) and extracted with MTBE (500 mL). The organic layers were washed with brine (500 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Si0 2 , Petroleum ether/Ethyl acetate=3/l to 2: 1) to afford compound 18C (26.8 g, yield 52.8%) as a light yellow oil. 1H NMR (400MHz, CDC1 3 ) δ 5.25 - 5.17 (m, IH), 5.08 - 4.99 (m, IH), 4.31 - 4.21 (m, IH), 3.66 (t, ./ 6.2 Hz, 2H), 1.98 - 1.79 (m, 2H), 1.76 - 1.68 (m, IH), 1.65 - 1.57 (m, 2H), 1.43 (s, 9H), 1.28 - 1.21 (m, 6H).

[00413] Step 4: Synthesis of compound 18D

A mixture of compound 18C (10 g, 36.3 mmol), ally! methyl carbonate (14.04 g, 120.95 mmol), Pd 2 (dba) 3 (997.7 mg, 1.09 mmol), [l-(2-diphenylphosphanyl-l-naphthyl)-2- naphthyl] -diphenyl-phosphane (2.71 g, 4.36 mmol) in THE (150 mL) was degassed and purged with N 2 for 3 times. And then the mixture was stirred at 80 °C for 10 hrs under N 2 atmosphere. After cooling to room temperature, the reaction mixture was diluted with MTBE (200 mL), filtered through celite and evaporated to give a dark brown syrup. The residue was ppuurriififieedd bbyy ccoolluummnn cchhrroommaattooggrraapphhyy ((SSii00 22 ,, PPeettrroolleeuumm eetthheerr//EEtthhyyll aacceettaattee==99//ll ttoo 55 :: 11 )) ttoo aaffffoorrdd ccoommppoouunndd 1188DD ((77..8800 gg,, yyiieelldd 6688..11%%)) aass aa lliigghhtt yyeellllooww ooiill.. XX HH NNMMRR ((440000MMHHzz,, CCDDCC1I 33 )) δδ 55..9944 -- 55..7799 ((mm,, II HH)),, 55..2288 -- 55..1199 ((mm,, 1I H1 1))., 55..1188 -- 55..0099 ((mm,, 22HH)),, 55..0066 -- 44..9977 ((mm,, I HHi)),, 44..2266 -- 44..1155 ((mm,, IHH)),, 33..9933 ((dd,, ..// 55 ,,77 HHzz,, 22HH)),, 33 ,,4466 -- 33 ,,3377 ((mm,, 22HH)),, 11 ..9911 -- 11 ..7788 ((mm,, 1IHH)),, 11..7733 -- 11..5555 ((mm,, 33HH)),, 11..4411 ((ss,, 99HH)),, 11..2277 -- 11..1166 ((mm,, 66HH))..

[[0000441144]] SStteepp SS:: SSyynntthheessiiss ooff ccoommppoouunndd 1188EE

TToo aa mmiixxttuurree ooff ccoommppoouunndd 1188DD iinn TTHHFF ((5500 mmLL)) aanndd wwaatteerr ((2200 mmLL)) wwaass aaddddeedd LLii OOHH ((11 ..3333 gg,, 5555..55 mmmmooll ,,)),, aanndd tthheenn tthhee mmiixxttuurree wwaass ssttiirrrreedd aatt 1100 °°CC ffoorr 1166 hhrrss uunnddeerr ' NN 22 aattmmoosspphheerree.. TThhee rreeaaccttiioonn mmiixxttuurree wwaass ddiilluutteedd wwiitthh wwaatteerr ((5500 mmLL)) aanndd eexxttrraacctteedd wwiitthh MMTTBBEE ((7700 mmLL xx 33)).. TThhee aaqquueeoouuss llaayyeerr wwaass aaddjjuusstteedd ttoo ppHH ~~ 44 wwiitthh IINN HHCC1I aanndd eexxttrraacctteedd wwiitthh EEAA ((8800 m mLL xx 33)).. TThhee ccoommbbiinneedd oorrggaanniicc llaayyeerrss wweerree wwaasshheedd wwiitthh bbrriinnee ((220000 mmLL)),, ddrriieedd oovveerr NNaa 22 SS00 44 ,, fifilltteerreedd aanndd ccoonncceennttrraatteedd uunnddeerr rreedduucceedd pprreessssuurree ttoo ggiivvee ccoommppoouunndd 1188EE ((33..44 gg,, yyiieelldd 7788..55%%>)) wwaass oobbttaaiinneedd aass aa ccoolloorrlleessss ooiill.. TThhee ccrruuddee pprroodduucctt wwaass uusseedd iinn tthhee nneexxtt sstteepp wwiitthhoouutt ffuurrtthheerr ppuurriifificcaattiioonn ,, ll HH NNMMRR ( (440000M1H! !/z,, CCDDCC11 33 )) δδ 1100,,2277 -- 99..6644 ((mm,, 22HH)),, 55..9955 -- 55..8833 ( (mm,, 1I HH)),, 55..2299 -- 55..2222 ((mm,, I I HH)),, 55..2200 -- 55..1144 ((mm,, 1IHH)),, 44,,3355 -- 44..2266 ((mm,, 1IHH)),, 33..9966 ((dd,, 55..33 HHzz,, 22HH)),, 33..5500 -- 33..4433 ((mm,, 22H1 i)),, 11..9977 -- 11..8888 ((mm,, I 1 H1 1)),, 11..8866 -- 11..7755 ( (mm., I HHi)),, 11..7744 -- 11..6644 ((mm,, 22H1 1)),, 11..4433 ((ss,, 9911 !! )) ..

[[0000441155]] SStteepp 66:: SSyynntthheessiiss ooff ccoommppoouunndd 1188GG

AA mmiixxttuurree ooff ccoommppoouunndd 1188EE ((33.,44 gg,, 1122,,4444 mmmmooll)),, ccoommppoouunndd 77CC ((33 ,,6644 gg,, 1122,,4444 mmmmooll ,, HHOCI)),, EEDDCCII ((33..5588 gg,, 1188..66 mmmmooll)),, HHOOBBtt ( ( 11..6688 gg,, 1122..4444 mmmmooll)) aanndd DDIIEEAA ((44,,8822 gg,, 3377.,3322 mmmmooll)) iinn DDCCMM ((110000 mmLL)) wwaass ssttiirrrreedd aatt 1100 °°CC ffoorr 4488 hh uunnddeerr NN 22 aattmmoosspphheerree.. TThhee rreeaaccttiioonn mmiixxttuurree wwaass ccoonncceennttrraatteedd uunnddeerr rreedduucceedd pprreessssuurree.. TThhee rreessiidduuee wwaass ddiilluutteedd wwiitthh EEAA ((110000 mmLL)) aanndd wwaasshheedd wwiitthh IINN HHCCii ((110000 mmLL)),, ssaatt.. NNaaHHCC00 33 ((7700 mmLL xx 33)),, ddrriieedd oovveerr NNaa 22 SS00 44 ,, ffiilltteerreedd aanndd ccoonncceennttrraatteedd uunnddeerr rreedduucceedd pprreessssuurree ttoo ggiivvee ccoommppoouunndd 1188GG ((44..11 gg,, yyiieelldd 6644..44%%)) aass aa l liigghhtt yyeellllooww ssoolliidd.. l1 HH NNMMRR ((440000XX 11 !! iizz,, CCDDCC11 3 )) SS 66..8833 -- 66..7766 ((mm,, I 1 H1 1)).,. 66..7722 -- 66,,6622 ((mm,, I 1H1 1)).,. 55..9966 -- 55..8844 ((mm,, II HH)),, 55..7722 -- 55..6600 ((mm,, I 1H1 1)).,. 55..4411 -- 55..3333 ((mm,, IIHH)),, 55 ,,3300 -- 55..2222 ((mm,, I MHi)),, 55,,2200 -- 55..1144 ((mm,, IIHH)),, 55..1133 -- 55..0055 ((mm,, 22HH)),, 44..6622 -- 44..5544 ((mm,, IIHH)),, 44..4466 -- 44..3388 ((mm,, IIHH)),, 33..9999 -- 33..9933 ((mm,, 22HH)),, 33..5533 -- 33..4422 ((mm,, 22HH)),, 22..6622 -- 22..4433 ((mm,, 33HH)),, 11..9922 -- 11..5577 ((mm,, 88HH)),, 11..5566 -- 11..4499 ((mm,, IIHH)),, 11..4422 ((ss,, 99HH)),, 11..2288 -- 11..2233 ((mm,, 33HH)),, 00..9944 -- 00..8866 ((mm,, 66HH))..

To a solution of compound 18G (1.9 g, 3.71 mmol) in DCM (600 mL) was added Grubbs 2nd cat. (157.6 mg, 185.67 umol), the mixture was degassed and purged with N? for 3 t tiimmeess,, t thheenn ssttiirrrreedd aatt 4400 °°CC ffoorr 1166 hh uunnddeerr NN 22 aattmmoosspphheerree.. TThhee rreeaaccttiioonn mmiixxttuurree wwaass ccoonncceennttrraatteedd uunnddeerr rreedduucceedd pprreessssuurree ttoo ggiivvee aa rreessiidduuee.. TThhee rreessiidduuee wwaass ppuurriiffiieedd bbyy ccoolluummnn cchhrroommaattooggrraapphhyy ((SSii00 22 ,, PPeettrroolleeuumm eetthheerr//EEtthhyyll aacceettaattee==44//ll ttoo 11 :: 11)) ttoo aaffffoorrdd ccoommppoouunndd 1188HH ((22..88 gg,, yyiieelldd:: 7788%%)) aass aann ooffff--wwhhiittee ssoolliidd.. ll HH NNMMRR ((440000MMHHzz,,CCDDCCll 33 )) δδ 66..8800 -- 66..6699 ((mm,, IIHH)),, 66..5544 -- 66..4444 ((mm,, I IHH)),, 55..6655 -- 55..4455 ((mm,, 22HH)),, 55..3344 -- 55..2255 ((mm,, I IHH)),, 44..6699 -- 44..6600 ((mm,, 1HH)),, 44..5599 -- 44..5511 ((mm,, 1IHH)),, 33 ,,9922 -- 33,,8833 ((mm,, 22HH)),, 33..5511 -- 33..3366 ((mm,, II HH)),, 33..3355 -- 33..2244 ((mm,, 22HH)),, 22..7733 -- 22,,6611 ((mm,, 1IHH)),, 22..5588 -- 22..4477 ((mm,, IIHH)),, 22..4433 -- 22..3300 ((mm,, IIHH)),, 22..1155 ((ss,, II HH)),, 11..9911 -- 11..7799 ((mm,, II HH)),, 11 ,,7755 -- 11 ,,4488 ((mm,, 6 (:»H] [)},.. 11..4411 ((ss,, 99HH)),, 11..2277 -- 11..2233 ((mm,, 33HH)),, 00..9944 -- 00,,8833 ((mm,, 66H1 1)), MMSS ( (EESSII)) mm//zz ((MM II ff }} '' 448844 33..

[[0000441177]] SStteepp 88:: SSyynntthheessiiss ooff ccoommppoouunndd 1188 JJ

CCoommppoouunndd 11..88 JJ wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 1188HH ffoolllloowwiinngg tthhee pprroocceedduurree ooff ccoommppoouunndd 1144FF,, CCoommppoouunndd 1188 JJ ((330000 mmgg,, yyiieelldd 3322,,88 %%)) wwaass oobbttaaiinneedd aass aa wwhhiittee ssoolliidd.. MMSS ((EESSII)) 77??//zz ((MM++HH)) ++ 444422..33..

[[0000441188]] SStteepp 99:: SSyynntthheessiiss ooff ccoommppoouunndd 1188::

TTeerrtt--bbuuttyyll

13-en-5-yl)carbamate

Compound 18 was prepared from compound 18J following the procedure of compound 7. Compound 18 (25 mg, yield 12.7 %) was obtained as a white solid. ! H NMR (400MHz, DMSO-O δ 9.45 - 9.37 (m, I H), 8.33 - 8.22 (m, I H), 8.22 - 8, 12 (m, IH), 6.77 - 6.56 (m, I H), 5.62 - 5.41 (m, 2H), 4,53 - 4,32 (m, 2H), 4.06 - 3.96 (m, I H), 3.96 - 3.86 (m, IH), 3,82 - 3.72 (m, IH), 3,25 - 3.14 (m, IH), 3, 13 - 3 ,02 (m, IH), 2.69 - 2,54 (m, 2H), 2.20 - 2,06 (m, IH), 1.78 - 1.55 (m, 3H), 1.54 - 1 ,31 (m, 1 21 1 ),. 0.98 - 0.74 (m, (:»! ! }, MS (ESI) m/z (M+H) + 440.5.

[00419] Step 10: Synthesis of compounds 19Z and 19E

Compound 18H (1.0 g) was purified by prep-HPLC (HCl condition) to give two isomers: compound 19Z (200 mg, yield 20%) and compound 19E (480 mg, yield 48%o) was obtained both as white solid. Compound 19Z: 1H NMR (400MHz, CDC1 3 ) δ 6,76 - 6,67 (m, IH), 6,48 - 6.39 (m, I H), 5.67 - 5.57 (m, IH), 5.56 - 5.45 (m, I H), 5.32 - 5.23 (m, IH), 4.71 - 4,61 (m, IH), 4.61 - 4.50 (m, IH), 4,27 - 4, 13 (m, 3H), 3.94 - 3.83 (m, 2H), 3,38 - 3,25 (m, 2H), 2.74 - 2,62 (m, IH), 2.45 - 2.32 (m, IH), 2.09 - 1.97 (m, IH), 1.75 - 1.53 (m, 6H), 1.42 (s, 9H), 1.31 ·· 1.22 (m, 3H), 0.98 - 0.82 (m, 6H). MS (ESI) m/z (M+H) + 484.2. Compound 19E: ! l i NMR CDC1 3 ) δ 6.07 - 5.97 (m, IH), 5,94 - 5.83 (m, IH), 5.66 - 5.55 (m, IH), 5.03 ·· 4.95 (m. IH), 4.71 - 4.61 (m, 1 1 1 ). 4.43 ·· 4.33 (m. I H). 4.25 4.10 (m, 31 ! ). 3.96 ·· 3.84 (m, 2H), 3.72 - 3.64 (m, IH), 3.48 ·· 3.37 (m, IH), 2.60 - 2.48 (m, 2H), 2.16 - 2.03 (m, IH), 1 .92 - 1.82 (m, H), 1.79 - 1.47 (m, 6H), 1.45 - 1.38 (m, 9H), 1.29 1.23 (m, 3H), 0.92 - 0.84 (m, 6H). MS (ESI) m/z (M+H) + 484.2.

Compound 19F was prepared from compound 19E following the procedure of compound 14F. Compound 19F (390 mg, yield 69,41 %) was obtained as a white solid, 1H NMR (400MHz, CDCI 3 ) δ 8.17 - 8.05 (m, I H), 7.87 - 7.67 (m, I H), 6.49 - 6.34 (m, IH), 5,65 - 5.33 (m, 2H), 4,80 - 4.65 (m, IH), 4.48 - 4,3 (m, IH), 4,09 - 3.97 (m, IH), 3.96 - 3.68 (m, 3H), 3.65 - 3.55 (m, IH), 3,23 - 2.96 (m, 3H), 2.39 - 2,23 (m, IH), 2.02 - 1.85 (m, IH), 1.81 - 1.13 (m, 16H), 0.97 - 0.67 (m, 6H). MS (ESI) m/z (M+H) + 442.1..

[00421] Step 12: Synthesis of compound 19:

Tert-butyl ((5 * S 8^,ll»5»£)-ll-formyl-8-isobutyl-6,9-dioxo-l-oxa-7,10-

Compound 19 was prepared from compound 19F following the procedure of compound 7. Compound 19 (45 mg, yield 31 %) was obtained as a white solid. ! H NMR (400MHz, DMSO-de) δ 9.43 (s, IH), 8.27 (d, .1 7.9 Hz, I H), 8.18 (d, ,/ 9.3 Hz, IH), 6,60 (d, ./ 7.5 Hz, I H), 5.60 - 5.43 (m, 21 ! }. 4.53 - 4.42 (m, IH), 4,36 (t, J ------ 8.2 Hz, IH), 4,00

(br, s., IH), 3.96 - 3.86 (m, IH), 3 ,82 - 3.71 (m, IH), 3.24 - 3. 14 (m, IH), 3 , 13 - 3.02 (m, IH), 2.68 - 2,53 (m, 2H), 2.19 - 2.05 (m, IH), 1.80 - 1.54 (m, 3H), 1.49 - 1.33 (m, 12H), 0,97 - 0.77 (m, 6H). MS (ESI) m/z (M+H) + 440.5.

Tert-butyl (3>S l ,6 t y,9>S ' ,£)-9-formyl-6-isobutyl-4,7-dioxo-l-oxa-5,8-diazacvc opentadec-

1 l-ei¾-3-y1)carbamate (20)

To a solution of N-(tert-butoxycarbonyl)-0-(pent-4-en-l -yl)-L-serine (960 mg, 3.51 mmol) in THF (10 mL) was added N-methylmorpholine (533 mg, 5.27 mmol, 580 uL), isobutyl chloroformate (480 mg, 3.51 mmol, 460 uL) at -40 °C. The mixture was stirred at - 40 °C for 30 min. A solution of compound 7C (900 mg, 3.51 mmol) in THF (3 mL) and DMF (3 mL) was added at -40 °C. The mixture was stirred at -40 °C for another 1 .5 hrs. The reaction mixture was quenched with H 2 0 (1 mL) and partitioned between DCM (SO mL) and H?0 (50 mL). The organic phase was separated, washed with sat. NaHC0 3 (50 mL), dried over a 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Si0 2 , Petroleum ether/Ethyl acetate=5/l) to give compound 20A (1.7 g, yield: 77%) as a white semisolid. l H NMR (400MHz, CDC1 3 ) δ 6.82 id. ./ 7.9 Hz, I I I ), 6.58 (br. s., 1H), 5.84 - 5.73 (m, i l l ). 5.71 - 5,55 (m, 1H), 5.43 - 5.27 (m, i l l ). 5.19 - 4.91 (m, 4H), 4.65 - 4.52 (m, 1H), 4.46 - 4.42 (m, H I ), 4.28 - 4.07 (m, 3H), 3.82 (dd, J = 4.0, 9.3 Hz, 1H), 3.56 - 3.38 (m, 3H), 2.63 - 2.42 (m, 2H), 2.14 - 2.01 (m, 2H), 1.73 - 1.58 (m, 4H), 1.56 - 1.48 (m, i l l ). 1.45 (s, 9H), 1.26 (t, ./ 7.1 Hz, 3H), 0.92 (dd, ./ 6.2, 8.8 Hz, 6H). MS (ESI) m/z (M i l ) 512.1. The product was further separated by SFC to give pure compound 20A. SFC method: Column: AD (250mm*30mm,5um), Mobile phase: A: C0 2 B: EtOH (0.05% H 3 .H 2 O) Gradient: 20% of B Flow rate: 65 mL min Column temperature: 35°C). The was separated by SFC to give compound 20A (R t = 2.191 min, 400 mg) as a yellow oil. Compound 20A: ! II NMR (400MHz, Methanol-^) ·' 5.89 - 5.70 (m, .?.!]}, 5.16 - 4,93 (m, 4H), 4.55 - 4.39 (m, 2H), 4.26 - 4.09 (m, 3H), 3.62 - 3.58 (m, 2H), 3.54 - 3.41 (m, 2H), 2.60 - 2.42 (m, 2H), 2.13 - 2.08 (m, 2H), 1.74 - 1.57 (m, 5H), 1.45 (s, 9H), 1.18 (t, J= 7.1 Hz, 3H), 0.94 (dd, J= 6.6, 10.6 Hz, 6H).

[00423] Step 2: Synthesis of compound 20B

Compound 20B was prepared from compound 20A following the procedure of compound 18H. Compound 20B (250 mg, yield 66 %) was obtained as a grey solid. 1H NMR (400MHz, CDC! 3) δ 6.64 (d, 7.9 Hz, 1H), 6.30 (d, 8.8 Hz, IH), 5.53 (d, J = 6.6 Hz, 1H), 5,43 - 5.39 (m, IH), 5.24 - 5.18 (m, IH), 4.75 - 4.65 (m, 1H), 4.58 - 4.50 (m, 1H), 4.22 - 4.17 (m, 3H), 3,75 - 3.58 (m, 3H), 3.48 - 3.40 (m, IH), 2,52 - 2.36 (m, 2H), 2,23 - 2,09 (m, IH), 2.05 - 1.92 (m, IH), 1.84 - 1.80 (m, 2H), 1.71 - 1.60 (m, 211).1.54 - 1.40 (m, 10H), 1.28 (t, J= 7.1 Hz, 3H), 0.94 (dd, J= 6.6, 9.7 Hz, 6H). MS (ESI) m/z ( +H) + 484.1.

[00424] Step 3: Synthesis of compound 20C

Compound 20C was prepared from compound 20B following the procedure of compound 14F. Compound 20C (80 mg, yield 32 %) was obtained as a white solid. 1H NMR (400MHz, DMSO-i4) δ 8.22 (d, J= 9.0 Hz, 111).7.72 (d, J= 9.0 Hz, IH), 5.85 (d, J = 8.2 Hz, IH), 5.36 - 5.20 (m, 2H), 4.65 (t, J= 5.5 Hz, 111).4.47 - 4.31 (m, IH), 4.09 (q, J= 5.5 Hz, 2H), 3.77 (br. s,, IH), 3,68 - 3.60 (m, IH), 3.53 - 3,48 (m, IH), 3,36 - 3.19 (m, 5H), 2,25 (d, J ------- 13.7 Hz, IH), 2,07 - 1.96 (m, IH), 1.90 - 1,75 (m, 2H), 1.60 - 1.26 (m, 12H), 0.86 (dd,

J--- 6.3, 12.9 Hz, 6H). MS (ESI) m/z (M II) ' 442.1.

[00425] Step 4: Synthesis of compound 20

Compound 20 was prepared from compound 20C following the procedure of compound 7. Compound 20 (24.4 mg, yield 30 %) was obtained as a white solid. 1H NMR (400MHz, DMSG-t4) δ 9.43 (s, IH), 8.35 - 8.18 (m, 2H), 6.00 (d, J= 7.8 Hz, IH), 5.42 - 5.21 (m, 2H), 4.51 - 4.42 (m, IH), 4.38 - 4.33 (m, IH), 4.11 (br. s., IH), 3.68 - 3.64 (m, IH), 3.53 - 3.45 (m, IH), 3.27 - 3.13 (m, 211).2.48 - 2.45 (m, IH), 2.08 - 2.03 (m, 2H), 1.89 (br. s., IH), 1.74 - 1.20 (m. 14! f ), 0.89 (dd, ./ 6.5, 19.8 Hz, 6H). MS (ESI) m/z (M+H) + 440.2.

EXAMPLE 10

[00426] Step 1: Synthesis of compound 21 A

Compound 21A was prepared from compound 18H following the procedure of compound 12A. Compound 21A (420 mg, yield 98.4 ¾) was obtained as a brown solid. 1H NMR (400MHz, DMSO-fife) δ 8.73 - 8.64 (m, IH), 8.57 (d, J = 8.8 Hz, 1H), 8.31 - 8.20 (m, 3H), 5.61 - 5.43 (m, 2H), 4.54 - 4.30 (m, 2H), 4.10 - 3.71 (m, 5H), 3.16 - 2.97 (m, 2H), 2.32 - 2.18 (m, IH), 1.86 - 1.44 (m, 6H), 1.38 - 1.25 (m, I I I). 1.20 - 1.12 (m, 3H), 0.94 - 0.80 (m, 6H).

[00427] Step 2: Synthesis of compound 21B

Compound 21B was prepared from compound 21A following the procedure of compound 4E. Compound 21 B (500 mg, yield 96 %) was obtained as a brown solid, 1H NMR (400MHz, DMSO-tg) δ 8.31 - 8.23 (m, I H), 8.23 - 8.15 (m, IH), 7,37 - 7.21 (m, 5H), 7.18 - 7.08 (m, IH), 5,64 - 5.42 (m, 2H), 5.05 - 4.93 (m, 2H), 4,54 - 4.31 (m, 2H), 4.18 - 3 ,73 (m, 51 1 ). 3.23 - 2.99 (m, 2H), 1 ,77 - 1.61 (m, IH), 1.76 - 1.59 (m, IH), 1.58 - 1.38 (m, 5H), 1.37 - 1.24 (m, IH), 1.21 - 1.10 (m, 3H), 0.93 - 0.73 (m, 6H). MS (ESI) m/z (M+H) + 518.1.

[00428] Step 3: Synthesis of compound 21C

Compound 21C was prepared from compound 21B following the procedure of compound 14F. Compound 21C (125 mg, yield 27.2 %) was obtained as a white solid. 1H NMR (400MHz, DMSC 4) δ 8. 5 - 8.09 (m, IH), 7.80 - 7.74 (m, IH), 7.35 - 7.27 (m, 5H),

7.08 - 7.02 (m, IH), 5.63 - 5.32 (m, 2H), 4.97 (s, 2H), 4.69 (br s, IH), 4.41 - 4.34 (m, IH),

4.09 (br s, I H), 3.91 - 3.69 (m, 3H), 3.22 - 3.09 (m, 21 ! }. 3.09 - 2,96 (m, I H), 2.40 - 2,27 (m, IH), 1 ,97 - 1.86 (m, IH), 1.71 - 1 .60 (m, I H), 1 .53 - 1.31 (m, 7H), 0.91 - 0.78 (m, 6H). MS (ESI) m/z (M+H) + 476.3. Compound 21 was prepared from compound 21C following the procedure of compound 7. Compound 21 (20 mg, yield 14.14%) was obtained as a white solid. l H NMR (400MHz,CDCl 3 ) δ 9.55 (s, 1H), 7.34 (br s, 5H), 7.08 - 6.85 (m, 2H), 5.94 - 5.84 (m, 1H), 5.67 - 5.38 (m, 2H), 5.22 - 4.96 (m, 2H), 4.63 (br s, 2H), 4.39 (br s, 1 H), 3.88 (br s, 2H), 3.28 (br s, 21 1 ). 2.69 (br s, H I ), 2.30 - 1.98 (m, 211), 1.72 - 1.47 (m, 6H), 0.98 - 0,86 (m, 6H). MS m/z CM+Ή 474,2.

[00430] Step 1: Synthesis of compound 22A

To a solution of ethyl (S)-2-((tert-butoxycarbonyl)amino)non-8-enoate (4 g, 13.36 mmol) and K 2 0s0 4 .2H 2 0 (246 mg, 668.00 umol) in THF (100 ml.) and H 2 0 (50 mL) was added NaI0 4 (5.72 g, 26.72 mmol) at 0 °C. After addition, the reaction mixture was stirred at 10 °C for 1 h. The reaction mixture was filtered and the filtrate was diluted with 200 mL of sat. Na 2 S 2 0 3 , then the mixture was extracted with EtOAc (100 mL x 2) and the organic layers were washed with 50 mL of sat. Na 2 S 2 0 3 , and brine (50 mL x 2). The organic layer was dried over Na 2 S0 4 and concentrated to give compound 22A (3.9 g, yield: 96.9%) as colorless oil, which was used to next step without further purification. MS (ESI) m/z (M-Boc+H) " ' " 201 .9.

To a solution of compound 22A (2.9 g, 9.62 mmol) in MeOH (50 mL) was added NaBH 4 (473 mg, 12.51 mmol) as portions. After addition, the reaction mixture was stirred at 10 °C for 2 hrs. 50 mL of water was added into the reaction mixture and concentrated to remove MeOH, then the mixture was extracted with EtOAc (50 mL x 2), the combined extracts were washed with brine (40 mL x 2). The mixture was dried over Na 2 SC>4 and concentrated in vacuo to afford crude compound 22B (2.5 g, yield: 85.7%) as colorless oil, which was used to next step without further purification. 1H NMR (400MHz, CDC1 3 ) δ 5.04 - 4.97 (m, 1H), 4.30 - 4.23 (m, 1H), 4.22 - 4.15 (m, 2H), 3.64 (t, J = 6.5 Hz, 2H), 1.47 - 1.28 (m, 22H).

[00432] Step 3: Synthesis of compound 22C

To a solution of PPh 3 (2.59 g, 9.89 mmol) in DCM (30 mL) was added I 2 (2.51 g, 9.89 mmol), then the mixture was stirred for 6 min and DIEA (1.28 g, 9.89 mmol, 1.73 mL), compound 22B (2.5 g, 8.24 mmol) in DCM ( 10 mL) was added. After addition, the reaction mixture was stirred at 10 °C for 15 hrs. 200 mL of Petroleum ether was added into the reaction mixture and stirred for 5 min. The mixture was filtered and the filtrate was washed with Na 2 S 2 0 3 (100 mL x 2) and brine (100 mL). Then the mixture was dried over Na 2 S0 4 and concentrated to afford compound 22C (1.2g, yield: 35.2%>) as colorless oil. 1H NMR (400MHz, CDC1 3 ) S 5.03 - 4.95 (m, 1H), 4.31 - 4.24 (m, IH), 4.23 - 4.17 (m, 2H), 3.18 (t, J =

7.0 Hz, 2H), 1.86 - 1.77 (ni, 3H), 1.66 - 1 .59 (m, IH), 1 .45 (s, 9H), 1.42 - 1 .32 (m, 6H), 1.29 (I, J = 7.2 Hz, 3H).

[00433] Step 4: Synthesis of compound 22D

To a solution of compound 22C in THF (40 mL) was added t-BuOK (1 M, 6.5 mL), then the reaction mixture was stirred at 10 °C for 12 hrs. The reaction mixture was poured into 50 mL of water and the mixture was extracted with MTBE (50 mL x 2), then the aqueous layer was acidified by I N HC1 to pH ~ 3, then extracted with EtOAc (50 mL x 2) and the combined extracts were washed with brine (50 mL), the organic layer was dried over ' Na 2 S0 4 and concentrated in vacuo to afford compound 22D (450 mg, yield: 80.2%) as colorless oil. 1H NMR (DMSO- , 400 MHz): δ 12.37 (br. s, IH), 7.04 - 6.94 (m, IH), 5.86 - 5.73 (m, IH), 5.05 - 4.90 (m, 2H), 3.90 - 3.80 (m, IH), 2.05 - 1.98 (m, 2H), 1.73 - 1.20 (m, 15H).

[00434] Step 5: Synthesis of compound 22E

Compound 22E was prepared from compound 22D following the procedure of compound 7D. Compound 22E (1.2 g, yield 44.34%) was obtained as an off-white solid. ! H NMR (400MHz, CDCI 3 ): δ 6,56 - 6.34 (m, IH), 5.81 - 5.62 (m, 21 1 ). 5.12 (d, J = 13.2 Hz, 2H), 5.01 - 4.92 (m, 2H), 4,61 - 4.42 (m, 2H), 4.22 - 4.1 1 (m, 2H), 2.59 - 2.48 (m, 2H), 2.05 -

2.01 (m, 2H), 1 ,66 (br s, 3H), 1.57 (s, 3H), 1.44 (s, 9H), 1.29 - 1.25 (m, 3H), 0.93 (d, J = 5.3 Hz, 61 1 ) MS (ESI) m/z (M-56) + 440.1 , [00435] Step 6: Synthesis of compound 22F and separation to obtain 22G- and 22H

Compound 22F was prepared from compound 22E following the procedure of compound 7F. Compound 22F was purified by preparatory-HPLC (HCl condition) to give compound 22G (120 mg, yield: 21.2%) as white solid and compound 22H (100 mg, yield: 13.5%) as white solid.

CCoommppoouunndd 2222GG:: 1H1H NNMMRR ( (DDM 8S0O--4,.,. 440000MMHHzz)):: δδ 88., 1199 ((dd,, .,/ 55..77 HHzz,, I IHH)),, 77..2277 ( (bbrr.. ss..,, IIHH)),, 77..0044 ( (bbrr.. ss..,, IIHH)),, 55..2233 -- 55..3388 ((mm,, IIHH)),, 55..1122 -- 55..2211 ((mm,, I HHi)),. 44..4477 -- 44..2255 ((mm,, 22HH)),, 44..1155-- 33.,9988 ((mm,, 22HH)),, 33..8877--33..7744 ( (mrn,, II HH)),, 22,,2277 -- 22., 1144 ((mm,, IIHH)),, 22..0077 ((dd,, ..// 1100.,66 HHzz,, IIHH)),, 11..8888 -- 11 ,,7711 ((mm,, IIHH)),, 11 ,,5599 -- 1I ..440() ((mm,, 55HH)),, 11..3366 ((ss,, 1100HH)),, 11 ..2233 ((dd,, JJ -- 77,,55 HHzz,, 22HH)),, 11..1155 ((tt,, JJ== 77.,33 HHzz,, 44HH)),, 00..8844 ((dddd,, JJ== 1177..00,, 66..44 HHzz,, 77HH)).. MMSS ((EESSII)) mm//zz ((MM--5566)) ++ 441122..11..

CCoommppoouunndd 2222HH:: 1H1H N NMMRR ((440000MMHHzz,, DDMMSSOO--fififefe)):: Sδ 88..4411 -- 88..1133 ((mm,, IIHH)),, 77..9900 ((dd,, JJ == 77..99 HHzz,, I IHH)),, 66..7766 -- 66..5511 ((mm,, IHH)),, 55..3344 ((dddd,, ../ 66.,88,, 1144..33 HHzz,, IIHH)),, 55..2211 -- 55..0066 ((mm,, I IHH)),, 44..5555 -- 44..2299 ((mm,, 22HH)),, 44..0055 ((qq,, JJ== 77..11 HHzz,, 22HH)),, 33..9922 ((bbrr.. ss..,, IHH)),, 11..9955 ((bbrr.. ss..,, 22HH)),, 11..6688 -- 11..4433 ((mm,, 55HH)),, 11..3344 ((ss,, 1133HH)),, 11 .. 1155 ((i1,, JJ 77.. 11 HHzz,, 44HH)),, 00,,8888 -- 00,,7799 ((mm,, 77H1 !)).. MMSS ((EESSII)) mm//zz ((MM--5566)) ++ 441122,, 11 ..

CCoommppoouunndd 2222..ΪΪ wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 2222GG ffoolllloowwiinngg tthhee pprroocceedduurree ooff c coommppoouunndd 1144FF.. CCoommppoouunndd 2222,,11 ((112200 mmgg,, yyiieelldd 2211 ,,22%%)) wwaass oobbttaaiinneedd aass aa wwhhiittee ssoolliidd.. 1H1H NNMMRR ((440000MMHHzz,, DDMMSSOO--ii¾¾)):: <<>> '' 77..7744 ((dd,, JJ == 77,,88 HHzz,, IIHH)),, 77..1133 ((bbrr.. ss..,, IIHH)),, 77..0011 ((bbrr.. ss..,, IIHH)),, 55..1166 ((bbrr,, ss.,.. 22HH)),, 44..6633 ((iL, 55..11 HHzz,, IIHH)),, 44..3344 ((dd,, ..// 77..88 HHzz,, IIHH)),, 33..7777 ((bbrr.. ss..,, ..??..!! ]])),, 33 ,,2222 -- 33..1100 ((mm,, IIHH)),, 22..3300 ((dd,, JJ == 1133..77 HHzz,, IIHH)),, 11..9944 -- 11..6655 ((mm,, 22HH)),, 11..4488 ((dd,, JJ == 55..11 HHzz,, 44HH)),, 11..3355 ((bbrr.. ss..,, 11 11HH)),, 11..2233 -- 11..0033 ((mm,, 44HH)),, 00..8844 -- 00..8800 ((mm,, IIHH)),, 00..8822 ((bbrr.. ss..,, 66HH)).. MMSS ((EESSII)) mm//zz ( (M-- 5566)) '' 337700..00..

Compound 22 was prepared from compound 22 J following the procedure of compound 7. Compound 22 (25 mg, yield 25.1 %) was obtained as a white solid. 3 H NMR (400MHz, CDCI 3 ): δ 9.54 (s, IH), 6.84 (br. s., IH), 6.61 (d, ./ 8.2 Hz, IH), 5.41 - 5.22 (rn, 2H), 4,83 (br. s., IH), 4.66 - 4.38 (m, 2H), 4.15 (br. s., IH), 2,71 (d, J = 14,9 Hz, IH), 2.25 - 2.06 (m, 2H), 1.95 (d, J = 7.4 Hz, 2H), 1.72 - 1.56 (m, 5H), 1.45 (s, 9H), 1.34 - 1.23 (m, IH), 1.22 - 1.09 (m, 2H), 0.96 - 0.90 (m, 6H). MS (ESI) m/z (M+H) + 424.2. Compound 23A was prepared from compound 22H following the procedure of compound 14F. Compound 23A (120 mg, yield 21.2 %) was obtained as a white solid. ! H NMR (400MHz, DMSO-<&): δ 8.09 (d, ./ 9,0 Hz, ill), 7,57 (d, ,/ 8.6 Hz, III).6.47 (d, ./ 7.0 Hz, 1H), 5.20 (br. s,, 211).4.63 (t, ./ 5.3 Hz, 111), 4.35 (q, ,/ 7.8 Hz, III).3.97 - 3.78 (rn, 2H), 3,30 - 3.23 (rn, 2H), 3,17 - 3.07 (rn, III), 2,27 (d. 14.1 Hz, 1H), 2.05 - 1.77 (m, 3H), 1.57 - 1.38 (m, 4H), 1.33 (s, 10H), 1.23 - 1,09 (m, 2H), 0.94 (d../ 4.3 Hz, 1H), 0.86 - 0.79 (m.6H). MS (ESI) m/z (M-56) + 370.1.

[00439] Step 10; Synthesis of compound 23:

Tert-XmXx\ ((3 t S'J4^£ -14-forim -3"¾sobutyl-2,5-dioxo-l,4-di¾¾acyclotetr¾dec-ri-en-6-- vDcarbamate

Compound 23 was prepared from compound 23A following the procedure of compound 7. Compound 23 (10 mg, yield 11.2 %) was obtained as a white solid. 1H NMR (400MHz, CDC!:.): S 9.69 - 9.48 (m, IH), 6.90 (br. s., 1H), 6.74 (d, ,/ 7.0 Hz, 111), 5.44 - 5.23 (m, 3H), 4.67 (br. s., 2H), 4.16 (br. s., IH), 2.72 (d, J 13.3 Hz, IH), 2.21 (br. s., IH), 2.02 (br. s,, 2H), 1.82 (br, s, 2H), 1,73 - 1.62 (m, 4H), 1.42 (br. s., 9H), 1.28 - 1.10 (rn.3H), 0.97 - 0.90 (rn.6H . MS (ESI) m/z (M-56) + 368.2.

Compound 24A was prepared from compound 22F following the procedure of compound 14F. Compound 24A (260 mg, yield 77.21 %) was obtained as a white solid. 1H NMR (400MHz, Methanol-^) : S 5.44 - 5.19 (m, 2H), 4.53 (q, J=7.1 Hz, 1H), 4.18 - 3.78 :(m, 21 IK 3.51 - 3.38 (m, 2H), 2.44 - 2.27 (m, III).2.21 - 1.89 (m, 3H), 1.70 - 1.50 (ni, 5H), 1.44 - 1.39 (ni, 9H), 1.29 - 1.11 (m, 3H), 0.96 - 0.89 (m, 6H). MS (ESI) m/z (M-56) + 370.1.

To a solution of compound 24A (260 mg, 610.96 umol) in MeOH (30 nL) was added Pd/C (80 mg, 10% under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H 2 (15 psi) at 10 °C for 2 hrs. The mixture was filtered. The filtrate was concentrated. The residue was purified by preparatory-HPLC (HC1 condition) to give compound 24B (80 mg, yield: 18.7%) as a white solid and compound 24C (80 mg, yield: 18.1%) as a white solid.

Compound 24B: Ίί NMR (400MHz, DMSO-i¾): δ Ί ,19 (d, 8,8 Hz, ill), 7,19 (d, ./ 7.9 Hz, IH), 7.05 (br. s., HI).4.53 (t, ./ 5,7 Hz, Hi), 4.45 - 4.30 (m, III), 3,78 (d. 5.7 Hz, 2H), 3.27 - 3,09 (m, 2H), 1,60 - 1.46 (m, 4H), 1.41 - 1.32 (m, I Hi), 1.24 (br. s., 5H), 1.17 - 1.01 (m, 6H), 0.83 (dd, J= 6.4, 10.8 Hz, 6H). MS (ESI) m/z (M-56) + 372.1.

Compound 24C: 1H NMR (400MHz, DMSO- ,): δ 8.14 (d, J= 9.3 Hz, 111).7.75 (d, ./ 9,3 Hz, 111), 6.41 (d, ./ 7,5 Hz, 111), 4.52 (t, J= 5.7 Hz, H), 4,43 (q, J= 7.9 Hz, III). 3,96 (br, s,, IH), 3.79 (br. s., H), 3.27 - 3.09 (m, 2H), 1.62 - 1.47 (m, 3H), 1.43 - 1.37 (m, 3H), 1.33 (s, 9H), 1 .28 - 1 .17 (m, 6H), 1.13 - 1.03 (m, 4H), 0,88 - 0,80 (m, 6H), MS (ESI) m/z (Μ-56Ϋ 372,0.

Tert-butyl ((3 ^ V,14iy)-14-formyl-3-isobutyl-2,5-dioxo-l,4-diazacvclot etradecan-6-

Compound 24 was prepared from compound 24B following the procedure of compound 7. Compound 24 (11 mg, yield 13.8 %) was obtained as a white solid. 1H NMR (400MHz, DMSO-fife): <5 9.39 (s, 1H), 8.40 (d, J= 7.4 Hz, IH), 7.40 (d, J = 8.2 Hz, 1H), 6.98 (br. s., IH), 4.44 (d, J= 6,3 Hz, 1H), 4,26 (br. s., IH), 3.81 (br, s., 1H), 1.55 (br. s,, 3H), 1.44 (br. s., 3H), 1 ,36 (br. s., 10! 1). 1.25 (br. s., 5H), 1.12 (br. s., 5H), 0,89 - 0.81 (m, 6H). MS (ESI) m/z (M+H) + 426.3.

ip 4: Synthesis of compound 25:

Hazacvclotetradecan-6-

Compound 25 was prepared from compound 24C following the procedure of compound 7. Compound 25 ( 10 mg, yield 12,6 %) was obtained as a white solid. 1H NMR (400M:HZ, CDCI 3 ): δ 9,56 (s, I H), 6.89 (d, J = 7.0 Hz, I H), 6.64 (d, .1 7.4 Hz, IH), 5.33 (d, J ----- 8,2 Hz, IH), 4,65 (br, s., 2H), 4.12 (br, s,, IH), 2.04 (br. s.. IH), 1.79 (br. s., IH), 1.66 (d, J ----- 6,7 Hz, 6H), 1.43 (br, s., 12H), 1.26 (br. s,, 6H), 0,93 (d, J = 14.5 Hz, 6H). MS (ESI) m/z

Benzyl C(3^ t S',16 t SV16"forniyI"3-isobutyj-2.,5,9J3 etrao¾o-l,4,8.,12- tetr¾az¾€vdohe¾¾decan-6-yl)£arb¾mate (26)

To a solution of TMSC1 (200 mL, 1 .60 mo!) in MeOH (500 mL) was added L- glutamic acid (47 g, 319 mmol) as portions at 0 °C. After addition, the reaction mixture was stirred at 10 °C for 15 hrs. The reaction mixture was concentrated to afford dimethyl (2S)~2~ aminopentanedioate (67 g, yield 99.1%) as colorless oil, which was used to next directly without further purification.

To a solution of dimethyl (2S)-2-aminopentanedioate (67 g, 316.58 mmol) in MeOH (500 mL) was added TEA ( 132 mL, 949.74 mmol) and (Boc) 2 0 (90 g, 41 .55 mmol) at 0 °C slowly. After addition, the reaction mixture was stirred at 10 °C for 14 hrs. The mixture was concentrated and the residue was dissolved in EtOAc (500 mL), the mixture was washed with 0.5 N HC1 (300 ml,), sat. NaHC0 3 (300 mL), and brine (300 mL). The organic layers was dried over Na 2 S0 4 and concentrated in vacuo to afford compound 26A (75 g, yield 86.05%) as colorless oil. ' i f NMR (400 MHz, CDC1 3 ): S 5.17 - 5.05 (m, 1H), 4.40 - 4.28 (m, 1H), 3.75 (s, 3H), 3.69 (s, 3H), 2.49 - 2.33 (m, 2H), 2.25 - 2.13 (m, 1H), 2.02 - 1.89 (m, 1H), 1.44 (s. 9H), MS (ESI) m/z (M+23) + 297.9. TToo aa ssoolluuttiioonn ooff LLiiBBHH 44 ((3300 gg,, 11..3366 mmooll)) iinn TTHi I VF ((550000 mmLL)) wwaass aaddddeedd ddrrooppwwiissee aa ssoolluuttiioonn ooff ccoommppoouunndd 2266AA ((7755 gg,, 227722..4433 mmmmooll)) iinn TTHHFF ((115500 mmLL)) aatt 00 °°CC.. AAfftteerr aaddddiittiioonn,, tthhee rreeaaccttiioonn mmiixxttuurree wwaass ssttiirrrreedd aatt 00 °°CC ffoorr 11 hh,, tthheenn wwaarrmmeedd ttoo 1100 °°CC aanndd ssttiirrrreedd ffoorr 1166 hhrrss.. TThheenn MMeeOOHH ((330000 mmLL)) wwaass aaddddeedd iinnttoo tthhee rreeaaccttiioonn mmiixxttuurree sslloowwllyy aanndd ssttiirrrreedd ffoorr 3300 mmiinn ffoolllloowweedd bbyy aaddddeedd NHH 44 CC11 ((3300 mmLL)) ddrrooppwwiissee iinnttoo tthhee mmiixxttuurree sslloowwllyy.. TThhee mmiixxttuurree wwaass ffiilltteerreedd,, aanndd tthhee ssoolliidd ccoolllleecctteedd wwaass wwaasshheedd wwiitthh EEttOOAAcc ((11 LL)).. TThhee fifillttrraattee wwaass ccoonncceennttrraatteedd aanndd tthhee rreessiidduuee wwaass ddiissssoollvveedd iinn wwaatteerr ((880000 mmLL)) aanndd EEttOOAAcc ((880000 mmLL)),, tthhee mmiixxttuurree wwaass sseeppaarraatteedd aanndd tthhee aaqquueeoouuss wwaass eexxttrraacctteedd wwiitthh EEttOOAAcc ((880000 m mLL xx 22)).. TThhee ccoommbbiinneedd eexxttrraaccttss wweerree ddririeedd oovveerr NNaa 22 SS00 44 aanndd ccoonncceennttrraatteedd iinn vvaaccuuoo.. TThhee rreessiidduuee wwaass ddiilluutteedd wwiitthh MMTTBBEE aanndd fifilltteerreedd ttoo aaffffoorrdd ccoommppoouunndd 2266BB ((3355 gg,, yyiieelldd 5588..66%%)) aass wwhhiittee ssoolliidd.. !! HH NNMMRR ( (440000MMHHzz,, CCDDCC1I 33 )):: δδ 44..9955 -- 44..8844 ((mm,, 11 11 11 )).. 33..7755 -- 33..5500 ((mm,, 55HH)),, 33..0011 ((bbrr.. ss..,, 11HH)),, 22..4499 ( (bbrr.. ss..,, 11HH)),, 11..6677 -- 11..4411 ( (mm,, 1133HH)),,

TToo aa ssoolluuttiioonn ooff ccoommppoouunndd 2266BB ((1155 gg,, 6688..4411 mmmmooll)) iinn DDCCMM ((6600 mmLL)) wwaass aaddddeedd 22,,22-- ddiimmeetthhooxxyypprrooppaannee ((112266 mmLL,, 11..0033 mmooll)) aanndd PPTTSSAA ((11..22 gg,, 66..8844 mmmmooll)).. AAfftteerr aaddddiittiioonn,, tthhee rreeaaccttiioonn mmiixxttuurree wwaass ssttiirrrreedd aatt 11 55 °°CC ffoorr 22 hhrrss.. TThheenn tthhee rreeaaccttiioonn mmiixxttuurree wwaass qquueenncchheedd wwiitthh ssaatt..NNaa..HHCCOO 33 ((110000 mmLL)) aanndd eexxttrraacctteedd wwiitthh EEttOOAAcc ((220000 mmLL xx 22)),, tthhee ccoommbbiinneedd eexxttrraaccttss wweerree wwaasshheedd wwiitthh bbrriinnee ((110000 mmLL)) aanndd ddrriieedd oovveerr NNaa 22 SS00 44 ,, tthheenn ccoonncceennttrraatteedd iinn vvaaccuuoo.. TThhee rreessiidduuee wwaass ppuurriififieedd bbyy ccoolluummnn cchhrroommaattooggrraapphhyy ((SSii00 22 ,, P Peettrroolleeuumm eetthheerr//EEtthhyyll aacceettaattee == 33//11 ttoo 22//11)) ttoo aaffffoorrdd ccoommppoouunndd 2266CC ((99 gg,, yyiieelldd 5500..77%%)) aass ccoolloorrlleessss ooiill.. 11 HH NNMMRR ((440000MMHHzz,, CCDDCCII 33 )):: δδ 44..0022 -- 33..6644 ((mm,, 55HH)),, 11..9911 -- 11..6688 ((mm,, 11HH)),, 11..6666 -- 11..5522 ((mm,, 66HH)),, 11..5522 -- 11..4433 ((mm,, 1133HH))..

To a solution of compound 26C (9 g, 34.70 mmol) in ACN (100 mL) and H 2 0 (100 mL) was added PhI(OAc) 2 (25 g, 76.34 mmol), TEMPO (1.1 g, 6.94 mmol) at 0 °C under N 2 atmosphere. After that, the reaction mixture was stirred at 15 °C for 2 hrs. Then Na 2 S 2 0 3 (200 mL) was added into the reaction mixture and the mixture was stirred for 5 min. and extracted with EtOAc (300 mL x 2), the combined extracts were di luted with sat. NaHC0 3 (300 mL) and separated, the aqueous layer was acidified by IN HC1 to pH ~ 3, then the mixture was extracted with EtOAc (300 mL x 2), the combined extracts were washed with brine (150 mL), dried over Na 2 S0 4 and concentrated in vacuo to afford compound 5 (4 g, yyiieelldd 3388,,33%%)) aass ccoolloorrlleessss ooiill.. 1H1H NNMMRR ((440000ΜMΉHζz,, O DMMSSQO~-dt 6 g)):: SS 1122,,0022 ((bbrr., ss..,, 11HH)),, 33 ,,9900 -- 33 ,,6677 ((mm,, 33HH)),, 22,.2288 -- 22,, 1122 ((rmn,, 22HH)),, 11..8877 -- 11..7777 ((mm,, 1 HHi)),, 11..7744 -- 11..6633 ((rmn,, 11 H1 1)),, 11..5588 -- 11..2277 ((mm,, 1155HH))..

[[0000444488]] SStteepp 55:: SSyynntthheessiiss ooff ccoommppoouunndd 2266EE

CCoommppoouunndd 2266EE wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 2266DD aanndd eetthhyyll 33--aammiinnoopprrooppaannooaattee ffoolllloowwiinngg tthhee pprroocceedduurree ooff ccoommppoouunndd 77DD.. CCoommppoouunndd 2266EE ((55 gg,, yyiieelldd 4455,.22 %%)) wwaass oobbttaaiinneedd aass aa ccoolloorrlleessss ooiill.. 1H1H NNMMRR ((440000MMHHzz,, CCDDCC11 33 )):: δδ 44..2200 -- 44..11 11 ((mm,, 22HH)),, 33..9999 -- 33..8833 ((mm,, 22HH)),, 33..7777 -- 33..6677 ((mm,, 11HH)),, 33..5577 -- 33..4477 ((mm,, 22HH)),, 22..6600 -- 22..4499 ((mm,, 22HH)),, 22..2288 -- 22..1122 ((mm,, 22HH)),, 22..0033 -- 11..8833 ((mm,, 22HH)),, 11..6622 -- 11..4400 ((mm,, 1155HH)),, 11..2277 ((tt,, JJ==66..66 HHzz,, 33HH..

TToo aa ssoolluuttiioonn ooff ccoommppoouunndd 2266EE ((55..00 gg,, 1133..4422 mmmmooll)) iinn TTHHFF ((110000 mmLL)) wwaass aaddddeedd aa ssoolluuttiioonn ooff LLiiOOHH..HH 22 00 ((11..77 gg,, 4400..2266 mmmmooll)) iinn HH 22 00 ((110000 m mLL)) aatt 00 °°CC.. AAfftteerr aaddddiittiioonn,, tthhee rreeaaccttiioonn mmiixxttuurree wwaass ssttiirrrreedd aatt 1100 °°CC ffoorr 22 hhrrss.. TThhee rreeaaccttiioonn mmiixxttuurree wwaass aacciiddiififieedd ttoo ppHH ~~ 88 aanndd eexxttrraacctteedd wwiitthh EEttOOAAcc ((220000 mmLL)),, tthhee aaqquueeoouuss llaayyeerr wwaass aacciiddiififieedd bbyy IINN HHCCll ttoo ppHH ~~ 33 aanndd eexxttrraacctteedd wwiitthh E EttOOAAcc ((220000 mmLL xx 22)),, tthhee ccoommbbiinneedd eexxttrraaccttss wweerree wwaasshheedd wwiitthh bbririnnee ((110000 m mLL xx 22)) aanndd ddrriieedd oovveerr NNaa 22 SSC0>4 4 ,, tthheenn ccoonncceennttrraatteedd iinn vvaaccuuoo ttoo aaffffoorrdd ccoommppoouunndd 2266FF ((44..55 gg,, yyiieelldd 7777..99%%)) aass ccoolloorrlleessss ooiill.. ΉΉ NN ..MM RR ((440000MMHHzz,, DDMMSSOO-- ,,)):: δδ 1122.. 1155 ((bbrr.. ss..,, 11HH)),, 33..8888 -- 33..8800 ((mm,, 11HH)),, 33..7788 -- 33..6655 ((mm,, 22HH)),, 33..2244 -- 33..1155 ((mm,, 22HH)),, 22..3388 ···· 22..33 11 ((mm,, 22HH)),, 22..0066 -- 11..9988 ((mm,, 22HH)),, 11..8844 -- 11..7744 ((mm,, 1l HH)),, 11..6688 -- 11 ..5577 ((mm,, 11HH)),, 11..5522 -- 11..3322 ((mm,, 1155HH)).. MMSS ((EESSII)) mm//zz ((MM++2233)) ++ 336666..99..

Compound 26G was prepared from (S)-2-(((benzyloxy)carbonyl)amino)-3-((tert- butoxycarbonyl)amino)propanoic acid and methyl -leucinate following the procedure of compound 71). Compound 26G (9 g, yield 84.1 %) was obtained as a white solid. 1H NMR (400MHz, (Ί)Π :. ): δ 7.41 - 7.29 (m, 5H), 7,05 (br, s., 1H), 6.25 - 6, 1 5 (m, 1 H), 5.22 (br. s., H I }.. 5. 16 - 5. 10 (m, 2H), 4.60 - 4.52 (m, 1 1 1 }.. 4.34 - 4.27 (m, 1 1 1 }.. 3.72 (br. s., 31 1 ). 3.58 - 3.44 (m, .?.! ! ), 1.67 - 1.56 (m, 3H), 1.43 (s, 9H), 0.96 - 0.90 (m, 6H). MS (ESI) m/z (M- Boc+H) + 365.9.

[00451] Step 8: Synthesis of compound 26H

To a solution of compound 261» (9.0 g, 19.33 mmol) in EtOAc (20 mL) was added HCl/EtOAc (4M, 128 mL) at 0 °C. After addition, the reaction mixture was stirred at 15 °C for 2 hrs. The reaction mixture was concentrated in vacuo to afford compound 26H (7.0 g, yield 90.1%, HCl salt) as white solid, which was used to next without further purification. ! H NNMMRR ((440000MMHHzz,, DDMMSSOO--ii¾¾));; δδ 88..6622 -- 88,,5522 ((mm,, lIHH)),, 88..2266 -- 88..0099 ((mm,, 33HH)),, 77..7711 -- 77..6633 ((mm,, 1IHH)),, 77..4433 -- 77,,2255 ((mm,, 55HH)),, 55..0055 ((ss,, 22HH)),, 44..4400 -- 44..2222 ((mm,, 22HH)),, 33 ,,6600 ((ss,, 33HH)),, 33 ,,2244 -- 33..1100 ((mm,, 1IHH)),, 33..0022 -- 22..8888 ((rmn,, H 1 1 I1 )),, 11..6655 -- 11..5555 ((mm,, 22HH)),, 11..5533 -- 11..4455 ((rmn,, H 1 1 I1 )),, 00..9900 -- 00..7799 ((mm,, 66HH))..

[[0000445522]] SStteepp 99:: SSyynntthheessiiss ooff ccoommppoouunndd 2266JJ

CCoommppoouunndd 2266 JJ wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 2266HH aanndd 2266FF ffoolllloowwiinngg tthhee pprroocceedduurree ooff ccoommppoouunndd 77DD.. CCoommppoouunndd 2266JJ ((77..00 gg,, yyiieelldd 6611..99 %%)) wwaass oobbttaaiinneedd aass aa wwhhiittee ssoolliidd.. 1H1H NNMMRR ((440000MMHHzz,, DDMMSSOO-- ,,)):: δδ 88..3344 -- 88..2277 ((mm,, 1IHH)),, 77..8855 -- 77..7733 ((mm,, 22HH)),, 77..3388 -- 77..2299 ((mm,, 66HH)),, 55..0033 ((ss,, 22HH)),, 44..3355 -- 44..2266 ((mm,, 1IHH)),, 44..2200 -- 44..1111 ((mm,, 1IHH)),, 33..8899 -- 33..8800 ((mm,, 1IHH)),, 33..7788 -- 33..6677 ((mm,, 22HH)),, 33..6655 -- 33..6600 ((mm,, 33HH)),, 33..3322 -- 33..1155 ((mm,, 44HH)),, 22..2277 -- 22..1188 ((mm,, 22Ή1 1)),. 22..1100 -- 11..9944 ((mm,, 22HH)),, 11..8866 -- 11..8800 ((mm,, 1HH)),, 11 ,,7700 -- 11 ..5533 ((mm,, 44HH)),, 11..5522 -- 11..3322 ((mm,, 1166HH)),, 00..9911 -- 00..8877 ((mm,, 33HH)),, 00..8855 -- 00..8822 ((mm,, 33HH))..

[[0000445533]] SStteepp 1100:: SSyynntthheessiiss ooff ccoommppoouunndd 2266KK

CCoommppoouunndd 2266 wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 2266 JJ ffoolllloowwiinngg tthhee pprroocceedduurree ooff ccoommppoouunndd 2266FF.. CCoommppoouunndd 2266JJ ((66..00 gg,, yyiieelldd 7700..00 %%)) wwaass oobbttaaiinneedd aass aa ccoolloorrlleessss ooiill.. 1H1H NNMMRR ((440000MMHHzz,, DDMMSSOO--ddfefe)):: δδ 1122..5522 ((ss,, IIHH)),, 88..1166 -- 88..1100 ((mm,, 1IHH)),, 77..8855 -- 77,,7733 ((mm,, 22HH)),, 77..3377 -- 77..2277 ((mm,, 66HH)),, 55..0011 ((ss,, 22HH)),, 44..2244 -- 44..1177 ((mm,, IIHH)),, 44..1155 -- 44..0088 ((mm,, IIHH)),, 33..8888 -- 33..7799 ((mm,, IIHH)),, 33..7777 -- 33..6644 {{rnrn.. 22HH)),, 33..2266 -- 33..1122 ((mm,, 44HH)),, 22..2255 -- 22..1166 ((mm,, 22HH)),, 22..0088 -- 11..9988 ((mm,, 22HH)),, 11..6688 -- 11..4488 ((mm,, 55HH)),, 11 ..4466 -- 11..3333 ((mm,, 1122HH)),, 00..9922 -- 00..7711 ((mm,, 99HH))..

Compound 26L was prepared from compound 26K following the procedure of compound 26K. Compound 26L (4.5 g, yield 75.3 %, HCl) was obtained as a light yellow solid. 1H NMR (400MHz, DMSO-tg): δ 12.52 (s, I H), 8, 16 - 8.10 (m, IH), 7.85 - 7.73 (m, 2H), 7.37 - 7.27 (m, 6H), 5,01 (s, 2H), 4,24 - 4, 17 (m, IH), 4.15 - 4.08 (m, I H), 3.88 - 3,79 (m, IH), 3 ,77 - 3,64 (m, 21 1 ). 3.26 - 3.12 (m, 4H), 2.25 - 2.16 (m, 2H), 2.08 - 1 ,98 (m, 2H), 1.68 - 1.48 (m, 5H), 1.46 - 1 ,33 (m, 12H), 0.92 - 0.71 (m, 91 1 )

[00455] Step 12: Synthesis of compound 26M

To a solution of compound 26L (1.0 g, 1 ,74 mmol, HCl) and DIEA (0.9 mL, 5,23 mmol,) in dry DMF (250 mL) was added HATU (728 mg, 1.92 mmol). After addition, the reaction mixture was stirred at 10 °C for 1 h. The reaction mixture was concentrated in vacuo to remove DMF. The residue was purified by preparatory-HPLC (HCl) to afford compound 26M (180 mg, yield 19.5%) as white solid. 1H NMR (400MHz, L>\f S0- ,}: δ 8.20 - 8.14 (m, IH), 7,90 - 7.82 (m, 1H), 7.62 - 7.53 (m, 1H), 7,40 - 7.25 (m, 6H), 7.00 - 6,93 (m, !!!),.5.08 - 4.98 (m, 2H), 4,37 - 4,28 (m, IH), 4.08 - 4.00 (m, 1H), 3.81 - 3.73 (rn, 2H), 3.72 - 3.62 (m, IH), 3,54 - 3.44 (m, 111), 3.30 - 3.23 (m, 2H), 2,91 - 2.81 (m, 1H), 2.39 - 2,28 (m, 1H), 2.11 - 1.99 (m, 3H), 1.97 - 1.87 (m, 1H), 1.79 - 1.68 (m, 1H), 1.56 - 1.46 (m, IH), 1.42 - 1.24 (m, 2H), 0.97 - 0.79 (m, 6H). MS (ESI) m/z (M 11} 520.1.).

Compound 26 was prepared from compound 26M following the procedure of compound 7. Compound 26 (18 mg, yield 10.5 %, HCl) was obtained as a white solid. ! H MR (400MHz, DMSO- ,): δ 9.49 (s, IH), 8.34 - 8.20 (m, 2H), 7.54 - 7.28 (m, 7H), 7.23 - 7.17 (m, IH), 5.10 - 4.99 (m, 2H), 4,43 - 4.28 (m, 2H), 4.15 - 4.04 (ni, IH), 3.73 - 3.46 (m, 3H), 2.98 - 2.83 (m, IH), 2,35 - 2.04 (m, 5H), 1.85 - 1,68 (m, IH), 1.61 - 1.40 (m.31 i), 0.98 - 0.91 (m, 3H), 0.91 - 0.86 (m, 3H). MS (ESI) m/z (M ·!!) 518,1

Compounds 27-29

N-((3.y,6^ l J5^'^-3-((^-sec-butyl)-15-formyl-2,5-dioxo-l,4-diazacv clopentadec-12-en-6- -fluorobenzenesufonamide (27)

AA ssoolluuttiioonn ooff DDCCCC ((11..7788 gg,, 88..6655 mmmmooll)) iinn aannhhyyddrroouuss TTHHFF ((2200 mmLL)) wwaass aaddddeedd ttoo aa ssoolluuttiioonn ooff ((tteerrt-~bbuuttooxxyyccaarrbboonnyyll))-~ZL--iissoolleeuuc ciinnee ((22..0000 gg,, 88..6655 mmmmooll)) aanndd ..vv--hhyyddrrooxxyyssuucccciinniimmiiddee

((11..00 gg,, 88..6655 mmmmooll)) iinn aannhhyyddrroouuss TTHHFF ((2200 mmLL)).. TThhee rreessuullttaanntt mmiixxttuurree wwaass ssttiirrrreedd aatt 1155 °°CC ffoorr 1122 hhrrss.. TThhee iinnssoolluubbllee ssuubbssttaannccee wwaass fifilltteerreedd ooffff.. TThhee fifillttrraattee wwaass ccoonncceennttrraatteedd ttoo aaffffoorrdd ccoommppoouunndd 2277AA ((22..9900 gg,, ccrruuddee)) aass ccoolloorrlleessss gguumm,, wwhhiicchh wwaass uusseedd ffoorr nneexxtt sstteepp wwiitthhoouutt ffuurrtthheerr ppuurriifificcaattiioonn,,

[[0000445588]] SStteepp 22:: SSyynntthheessiiss ooff ccoommppoouunndd 2277BB

DDIIEEAA ((00..9977 mmLL,, 55..5577 mmmmooll)) wwaass aaddddeedd ttoo aa mmiixxttuurree ooff ccoommppoouunndd 77BB ((11..00 gg,, 55..5577 mmmmooll)) aanndd ccoommppoouunndd 2277AA ((1.1..8833 gg,, 55..5577 mmmmooll)) iinn DDMMEE ((2200..0000 mmLL)).. TThhee rreessuullttaanntt mmiixxttuurree wwaass ssttiirrrreedd aatt 1155 °°CC ffoorr 1122 hhrrss.. TThhee mmiixxttuurree wwaass ccoonncceennttrraatteedd aanndd tthhee rreessiidduuee wwaass ttrreeaatteedd wwiitthh DDCCMM ((5500 mmLL)) aanndd HH 22 00 ((3300 mmLL)).. TThhee oorrggaanniicc llaayyeerr wwaass sseeppaarraatteedd aanndd tthhee aaqquueeoouuss llaayyeerr wwaass eexxttrraacctteedd wwiitthh DDCCMM ((22 xx 1155 mmLL)).. TThhee ccoommbbiinneedd oorrggaanniicc llaayyeerr wwaass wwaasshheedd wwiitthh ssaattuurraatteedd NNaaHHCC00 33 ((1155 mmLL)),, IINN HHCC11 ((1155 mmLL)),, bbrriinnee ((1155 mmLL)),, ddrriieedd oovveerr aannhhyyddrroouuss MMggSS00 44 ,, fifilltteerreedd aanndd ccoonncceennttrraatteedd.. TThhee rreessiidduuee wwaass ttrriittuurraatteedd wwiitthh PPEE//// '' --PPrr 22 00 ((vv//vv == 55//11,, 1100 mmLL)) ttoo aaffffoorrdd ccoommppoouunndd 2277BB ((11..1155 gg,, yyiieelldd 5577..99%%)) aass wwhhiittee ssoolliidd,, MMSS ((EESSII)) mm//zz ((MM++HH)) ++ 335577..11.. ll HH N NMMRR ((440000MMHHzz,, DDMMSSOO--aafefe)) δδ 88..2200 ((dd,, JJ == 77..66 HHzz,, I 1 H1 1)),, 66..6699 ((dd.. JJ 88,,88 HHzz,, I !H! !)}.,. 55..8844 -- 55,,6699 ((mm,, 1IHH)),, 55..1155 -- 55..0077 ((mm,, 1IHH)),, 55..0055 -- 55..0033 ((mm,, IIHH)),, 44..3366 -- 44..2255 ((mm,, 1IHH)),, 44..1100 -- 44..0066 ((mm,, 22HH)),, 33..8855 -- 33..8833 ((mm,, IIHH)),, 22..4477 -- 22..3311 ((mm,, 22HH)),, 11..6688 -- 11..6666 ((mm,, 1IHH)),, 11..4400 -- 11..3388 ((mm,, llHH)),, 11..3388 ((ss,, 99HH)),, 11..1177 ((tt,, JJ== 77..22 HHzz,, 33HH)),, 11..1111 -- 11..0066 ((mm,, IIHH)),, 00..8899 -- 00..7755 ((mm,, 66HH))..

Compound 27C was prepared from compound 27B following the procedure of compound 12A. Compound 27C (0.8 g, yield 97.2 %) was obtained as a white solid. ! H NNMMRR ((440000MMHHzz,, DDMMSSOO--ii¾¾)) δδ 88..9955 ((dd,, 66..88 HHzz,, IIHH)),, 88..3322 ((bbrr..ss.,,, 33HH)),, 55..8899 -- 55..7777 ((mm,, II HH)),, 55.. 1177 -- 55..1122 ((mm,, IIHH)),, 55 ,,0099 -- 55..0088 ((mm,, IIHH)),, 44..3344 -- 44..3322 ((mm,, IIHH)),, 44,,0099 -- 44..0066 ((mm,, 22HH)),, 33..7733 -- 33 ,,7722 ((mm,, IIHH)),, 22..4477 -- 22..4466 ((mm,, 2.?.H! ])),, 11..8877 -- 11..8855 ((mm,, IIHH)),, 11..5522 -- 11..4488 ((mm,, II HH)),, 11..2200 -- 11 .. 1155 ((rmn,, 44HH)),, 00..9922 ((dd,, JJ == 66..88 HHzz,, 33HH)),, 00..8888 -- 00..8844 ((mm,, 33HH))..

[[0000446600]] SStteepp 44:: SSyynntthheessiiss ooff ccoommppoouunndd 2277PP

CCoommppoouunndd 2277DD wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 2277CC aanndd ((SS))--22--((((tteerrll-- bbuuttooxxyyccaarrbboonnyyll))aammiinnoo))nnoonn--88--eennoo iicc aacciidd ffoolllloowwiinngg tthhee pprroocceedduurree ooff ccoommppoouunndd 77DD.. CCoommppoouunndd 227711)) ((11 ..44 gg,, ccrruuddee)) wwaass oobbttaaiinneedd aass aa ppaallee yyeellllooww ssoolliidd aanndd uusseedd ffoorr nneexxtt sstteepp ddiirreeccttllyy.. MMSS ((EESSII)) mm//zz ((MM++NNaa)) ++ 553322..22.. ΉΉ NNMMRR ((440000MMHHzz,, DDMMSSOO-- )) δδ 88,,4400 ((dd,, ..// 66..88 HHzz,, IIHH)),, 77,,5599 ((dd,, JJ == 88..88 HHzz,, IIHH)),, 66..9988 ((dd,, JJ == 88..00 HHzz,, IIHH)),, 55..7799 -- 55..7711 ((mm,, 22HH)),, 55.. 1122 -- 44,,9922 ((mm,, 44HH)),, 44..2277 -- 44..2233 ((mm,, ..??..!! ]] )),, 44,,0088 -- 44..0055 ((mm,, 22HH)),, 33..9922 -- 33..8899 ((mm,, II HH)),, 22..4433 -- 22..3399 ((mm,, ..??..!! ]] }},, 22..0011 -- 11..9999 ((mm,, 22HH)),, 11..5599 -- 11..1144 ((mm,, 2233HH)),, 00..8855 -- 00..7799 ((rrnn,, 66HH))..

[[0000446611]] SStteepp 55:: SSyynntthheessiiss ooff ccoommppoouunndd 2277EE

CCoommppoouunndd 2277EE wwaass pprreeppaarreedd frfroomm ccoommppoouunndd 2277DD aanndd eetthhyyll 33--aammiinnoopprrooppaannooaattee ffoolllloowwiinngg tthhee pprroocceedduurree ooff ccoommppoouunndd 1188HH.. CCoommppoouunndd 2277EE ((00..88 gg,, yyiieelldd 8844..77 %%)) wwaass oobbttaaiinneedd aass aa ppaallee yyeellllooww ssoolliidd.. MMSS ((EESSII)) mm//zz ((MM++NNaa)) ++ 550044..22.. ]] HH NNMMRR ((440000MMHHzz,, CCDDCC11 33 )) SS 66..4400 -- 66..2200 ((mm,, 22HH)),, 55..4411 -- 55..3300 ((mm,, IIHH)),, 55..2233 -- 55..1155 ((mm,, IIHH)),, 44..5566 -- 44..5544 ((mm,, IIHH)),, 44..2288 -- 44..1100 ((mm,, 33HH)),, 44..0088 -- 33..9977 ((mm,, II HH)),, 22,,6677 -- 22,,4444 ((mm,, IIHH)),, 22..3377 -- 22..1122 ((mm,, II HH)),, 22..0022 -- 11 ..7788 ((mm,, 44HH)),, 11..6600 ((ss,, 22HH)),, 11 ,,4477 -- 11..4400 ((mm,, 22HH)),, 11..4400 -- 11..1199 ((mm,, 1166HH)),, 11..11 88 -- 11 ..0022 ((mm,, 33HH)),, 00,,9900 -- 00,, 7766 ((mm,, 66HH))..

[[0000446622]] SStteepp 66:: SSyynntthheessiiss ooff ccoommppoouunndd 2277FF

CCoommppoouunndd 2277FF wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 2277EE ffoolllloowwiinngg tthhee pprroocceedduurree ooff ccoommppoouunndd 1122AA.. CCoommppoouunndd 2277FF ((00,,7733 gg,, yyiieelldd 9999..22 %%)) wwaass oobbttaaiinneedd aass aann ooffff--wwhhiittee ssoolliidd wwhhiicchh wwaass uusseedd ddiirreeccttllyy.. 1H1H NNMMRR ((440000MMHHzz,, DDMMSSOO--ddfefe)) δδ 88..5588 -- 88,,5566 ((mm,, 22HH)),, 88,, 1188 ((bbrr., ss..,, 33HH)),, 55..4466 -- 55..2299 ((mm,, 22HH)),, 44..4400 -- 44..3366 ((mm,, IIHH)),, 44..3311 -- 44..2277 ((mm,, IIHH)),, 44..11 11 --44..0077 ((mm,, 22HH)),, 33..8899 -- 33..8888 ((mm,, IIHH)),, 22..2233 -- 22..1122 ((mm,, IIHH)),, 22..0033 -- 11 ..9933 ((mm,, 22HH)),, 11 ..8844 --11..8822 ((mm,, II HH)),, 11..6688 --11..6644 ((mm,, 33HH)),, 11..5577 -- 11 ..4444 ((mm,, IIHH)),, 11..2277 --11..2222 ((mm,, 33HH)),, 11..2211 --11..1188 ((mm,, 44HH)),, 11..1144 -- 00..9977 ((mm,, 33HH)),, 00..8899 ((dd,, ..// 66..44 HHzz,, 33HH)),, 00..8877 -- 00..8811 (( rrnn.. 33HH))..

DIEA (0.92 mL, 5.25 mmol) was added to a mixture of compound 27F (730.00 mg, 1.75 mmol) in DCM (30 mL). The suspension turned clear solution. Then 4- flfluuoorroobbeennzzeenneessuullffoonnyyll cchhllooririddee ((337744..6622 raragg,, 11..9933 mmmmooll)) wwaass aaddddeedd.. TThhee rreessuullttaanntt mmiixxttuurree wwaass ssttiirrrreedd aatt 1100 °°CC ffoorr 1122 hhrrss.. TThhee mmiixxttuurree wwaass ttuurrnneedd tthhiicckk sslluurrrryy.. TThhee mmiixxttuurree wwaass ddiilluutteedd wwiitthh DDCCMM ((110000 mmll,,)) aanndd TTHHFF ((3300 mmLL)),, wwaasshheedd wwiitthh HH 22 00 ((2200 mmLL)),, ssaattuurraatteedd N NaaHHCC00 33 ((2200 mmLL)),, IINN HHCC11 ((2200 mmLL)),, bbrriinnee ((1155 mmLL xx 22)),, ddrriieedd oovveerr aannhhyyddrroouuss MMggSS00 44 ,, fifilltteerreedd aanndd ccoonncceennttrraatteedd.. TThhee rreessiidduuee wwaass ttrriittuurraatteedd wwiitthh EEAA ((1155 mmLL)) aass ooffff--wwhhiittee ssoolliidd,, wwhhiicchh wwaass rree-- ppuurriififieedd bbyy ttrriittuurraattiinngg wwiitthh EEAA//MMeeOOHH ((vv//vv ::;; == 1100//11,, 1155 mmLL)) ttoo aaffffoorrdd ttoo aaffffoorrdd ccoommppoouunndd 2277GG ((449900 mmgg,, yyiieelldd 5511..99%%)) aass wwhhiittee ssoolliidd.. MMSS ((EESSII)) mm//zz ((MM++NNaa)) ++ 556622,, 11....

[[0000446644]] SStteepp 88:: SSyynntthheessiiss ooff ccoommppoouunndd 2277HH

CCoommppoouunndd 2277HH wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 2277GG ffoolllloowwiinngg tthhee pprroocceedduurree ooff ccoommppoouunndd 1144FF., CCoommppoouunndd 2277HH ((111100 mmgg,, yyiieelldd 2266..55 %%)) wwaass oobbttaaiinneedd aass aa wwhhiittee ssoolliidd.. MMSS ((EESSII)) mm//zz ((MM++NNaaff 552200..00.. ΊΊ ίί NNMMRR ((440000MMHHzz,, DDMMSSOO--ii¾¾)) δδ 88..0033 ((dd,, ..// 99..66 HHzz,, I I HI I))., 77..8866 -- 77..8833 ((mm,, 22HH)),, 77..7766 ((dd,, JJ== 99..22 HHzz,, IIHH)),, 77..6688 ((dd,, JJ== 88..88 HHzz,, IIHH)),, 77..3388 -- 77..3333 ((mm,, 1IHH)),, 55..2277 ((bbrr.. ss..,, 22HH)),, 44..6688 --44..6655 ((mm,, IIHH)),, 44..0011 -- 33..9933 ((mm,, 22HH)),, 33..7799 -- 33..6688 ((mm,, IIHH)),, 33..3322 -- 33..2255 ((mm,, IIHH)),, 33..1144 -- 33..0066 OOnn,, IIHH)),, 22..3333 -- 22..2255 ( Omn,, IIHH)),, 22..0011 -- 11..7766 ((mm,, 33HH)),, 11..6655 -- 11..5544 ((mm,, IIHH)),, 11..4433 ((dd,, JJ==66..88 HHzz,, 22HH)),, 11..2233 -- 11..0044 ((mm,, 77HH)),, 00..9966 -- 00..8855 ((mm,, IIHH)),, 00..7711 --00..6688 ((mm,, 77HH))..

Compound 27 was prepared from compound 27H following the procedure of compound 7. Compound 27 (25 rag, yield 50.2 %) was obtained as a white solid. MS (ESI) m/z (\l - \a ] [ ;()) ' 536.1. Ί ί NMR (400MHz, DMSO-ofe) S 9.41 (s, IH), 8.27 (d, ./ 8.4 Hz, IH), 8. 10 (d, J --- 9.2 Hz, IH), 7,86 - 7,83 (m, 2H), 7.75 (d, ./ 8,4 Hz, IH), 7.39 - 7.34 (m, 2H), 5.37 - 5,24 (m, 2H), 4.35 - 4.08 (m, IH), 4.08 - 3.97 (m, 2H), 2.02 -1 ,94 (m, 3H), 1.61 - 0.71 (m, 1 8! !}.

4-fluoro-N-((3.y,65',15i.S'^)-15-formvi-3-isopropyl-2,5-diox o-l,4-diazacvcIopentadec-12- e8¾-6- (28)

Compound 28 was prepared using the same synthetic steps as for compound 27, except that in Step 1 (fert-butoxycarbonyl)-L-valine was used as the starting amino acid. Compound 28 (15 rng, yield 37.7 %) was obtained as a white solid. MS (ESI) m/z (M+H) ' 482.1.

N-((3£6^ 5 £ )-3-benzv.-15-formyl-2,5^

flHoroberczercesuIfonamide (29)

Compound 29 was prepared using the same synthetic steps as for compound 27, except that in Step 2, compound 7B was coupled with (tert-butoxycarbonyl)-Z -phenylalanine. Compound 29 (40 mg, yield 57 %) was obtained as a white solid. 1 H NMR (400MHz, DMSO-flfe) δ 9.13 - 8.93 (m, I H), 8,44 (d, ./ 9,0 Hz 1 1 1 ), 8.19 (d, ./ 8.3 Hz, I H), 7,87 - 7.66 (m, 3H), 7.38 - 7.08 fm, 7H), 5.37 - 5.26 (m, 1H), 5.26 - 5.14 (m, IH), 4.48 - 4.36 (m, 1H), 4.30 - 4.16 (m, IH), 3.99 - 3.86 (m, IH), 2.87 - 2.76 (m, IH), 2.62 - 2.52 (m, IH), 2.43 - 2.36 (m, IH), 2.02 - 1.81 (m, 3H), 1.65 - 1.52 (m, IH), 1.47 - 1.34 (m, IH), 1.31 - 1.15 (m, 3H), 1.09 - 0.97 (m, 2H), 0.95 - 0.78 (m, H). MS (ESI) m/z (M+Na + ) 530.1.

To a solution of PPh 3 (4.53 g, 17.28 mmol) and imidazole (1.18 g, 17.28 mmol) in DCM (150 mL) at 0 C 'C was added I 2 (4.38 g, 17.28 mmol) in three portions. The solution was warmed to 10 °C and stirred for 10 min, and re-cooled to 0 °C. Methyl ((benzyloxy)carbonyl)- -serinate (3.50 g, 13.82 mmol) in DCM (25 mL) was then added dropwise. The solution was stirred at 0 °C for 1 hr and 10 °C for 1.5 hrs. The reaction mixture was then filtered through silica gel using PE/EA = 50/50 as eluent, concentrated. The residue was purified by column chromatography (Si0 2 , petroleum ether : ethyl acetate = 1 :0 to 10: 1) to afford the desired compound 30A (4.00 g, yield: 79.70%) as colorless oil. ! H MR (400MHz, CDC1 3 ) δ 7.42 - 7.27 (m, 5H), 5.63 (m, 1 i I ). 5.22 - 5.05 (m, 2U 4.60 (m, 1H), 3.82 (s, 3H), 3.69 - 3.49 (m, 2 H).

To a solution of TMSCl (11.76 mL, 93.1 mmol) in MeOH (25 mL) was added (2S)-2- aminohexanedioic acid (3 g, 18.62 mmol) at 0 °C. The mixture was stirred at 10 °C for 16 hhrrss.. TThhee mmiixxttuurree wwaass ccoonncceennttrraatteedd ttoo aaffffoorrdd ccoommppoouunndd SSOOBB ((66 gg,, ccrruuddee)) aass ccoolloorrlleessss ooiill,, wwhhiicchh wwaass uusseedd ttoo nneexxtt sstteepp wwiitthhoouutt ffuurrtthheerr ppuurriiffiiccaattiioonn..

[[0000447700]] SStteepp 33:: SSyynntthheessiiss ooff ccoommppoouunndd 3300CC

TToo aa ssoolluuttiioonn ooff ddiimmeetthhyyll ((22SS))--22--aammiinnoohheexxaanneeddiiooaattee SSOOBB ((66 gg,, 3311..7711 mmmmoo!i)) iinn MMeeOOHH ((110000 mmLL)) wwaass aaddddeedd TTEEAA ((1133..1199 mmll,,,, 9955..1133 mmmmooii)) aanndd ((BBoocc)) 22 00 ((99 gg,, 4411..2222 mmmmooii)) aatt 00 °°CC.. TThhee mmiixxttuurree wwaass ssttiirrrreedd aatt 1100 °°CC ffoorr 1166 hhrrss.. TThhee mmiixxttuurree wwaass ccoonncceennttrraatteedd aanndd tthhee rreessiidduuee wwaass ddiissssoollvveedd iinnttoo EEttOOAAcc ((110000 mmLL)),, tthhee mmiixxttuurree wwaass wwaasshheedd wwiitthh HHCCll ((00..55 NN,, 2200 mmLL)),, ssaattuurraatteedd \\aall IICCOO ;; ((2200 mmLL xx 22)),, aanndd bbririnnee ((3300 mmLL)).. TThhee oorrggaanniicc llaayyeerrss wwaass ddrriieedd oovveerr NNaa 22 SS00 44 aanndd ccoonncceennttrraatteedd iinn v vaaccuuoo ttoo aafffoforrdd ccoommppoouunndd 3300CC ((88..77 gg,, ccrruuddee)) aass ccoolloorrlleessss ooiill.. 1H1H NNMMRR. ((440000MMHHzz,, CCDDCC11 33 )) δδ 55..1177 -- 44..9955 ((mm,, I !H! !)).,. 44..3399 -- 44..2222 ((mm,, IIHH)),, 33..7755 ((ss,, 33HH)),, 33..6677 ((ss,, 33H1 1)),, 22..4411 -- 22..2277 ((mm,, 22HH)),, 11..9900 -- 11..7799 ((mm,, I 1H1 1)),, 11..7777 -- 11..5577 ((mm,, 33HH)),, 11..4477 -- 11..3377 ((mm,, 9911 11 ))..

[[0000447711]] SStteepp 44:: SSyynntthheessiiss ooff ccoommppoouunndd 3S0OPP

CCoommppoouunndd 3S0O1D) wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 3300CC ffoolllloowwiinngg tthhee pprroocceedduurree ooff ccoommppoouunndd 2266BB.. CCoommppoouunndd SSOODD ((77..99 gg,, yyiieelldd 6633..22 %%)) wwaass oobbttaaiinneedd aass aa ccoolloorrlleessss ooiill.. 1H1H NNMMRR ((440000 MMHHzz,, CCDDCC1i 33 )):: δδ 55..0088 ···· 44..4444 ((mm,, 22HH)),, 33..7755 -- 33..5522 ((mm,, 55HH)),, 33..1155 -- 22..6622 ((mm,, 1IHH)),, 22..4411 -- 11..7788 ((mm,, IIHH)),, 11..6655 -- 11..4400 ( (mni,, 1144HH))..

Compound 30E was prepared from compound SOD following the procedure of compound 26C. Compound 30E (3 10 mg, yield 39.51 %) was obtained as a colorless oil. ! H NMR (400MHz, CDC1 3 ): δ 3,95 - 3.89 (m, I H), 3,81 - 3.69 (m, 2H), 3.69 - 3.61 (m, 2H), 1.64 - 1.53 (m, 6H), 1.52 - 1 ,41 (m, 13H), 1 .41 - 1 .29 (m, 2H).

[00473] Step 6: Synthesis of compound 30F

To a solution of compound 30E (3 0 mg, 1. 13 mmoi) and 3-bromoprop-l-yne (161.31 mg, 1.36 mmoi) in DMF (5 mL) was added NaH (54.24 mg, 1 .36 mmoi, 60% purity) at 0 °C. After addition the mixture was stirred at 0 °C for 2 hrs, then stirred at 5 °C for 14 hrs. To the reaction mixture was added water (15 mL) and extracted with EA (30 mL x 2), the combined extracts were washed with IN HCl (10 mL), sat. NaHC0 3 (10 mL), dried over Ν¾8θ 4 , and then concentrated in vacuo. The residue was purified by column chromatography (Si0 2 , Petroleum ether/Ethyl acetate = 3/1 to 2/1) to afford compound 30F (164 mg, yield 46,6%) as colorless oil. 1H NMR (400MHz, CDC1 3 ): δ 4.14 - 4.09 (m, 2H), 33..9933 -- 33..8866 ((mm,, 11HH)),, 33..8800 -- 33..6677 ((mm,, 22HH)),, 33..5555 -- 33..4466 ((mm,, 22HH)),, 22,,4411 fftt,, 22..33 HHzz,, 11HH)),, 11..6655 -- 11..5511 ((rmn,, 6611 11 )),, 11..5500 -- 11..4400 ((mm,, 113H! !))., 11..3399 -- 11..2299 ((rmn,, 221H1))..

[[0000447744]] SStteepp 11%% SSyynntthheessiiss ooff ccoommppoouunndd 3300GG

TToo aa ssoolluuttiioonn ooff ccoommppoouunndd 3300FF ((225500 mmgg,, 880022..7777 uummooll) ) iinn D T)CCMM ( ( 1100 mmLL)) wwaass aaddddeedd NNIISS ((119988..66 mmgg,, 888833..0055 uummooll)) aanndd AAggNN00 33 ((116633..6644 mmgg,, 996633..3333 uummooll)).. TThhee mmiixxttuurree wwaass ssttiirrrreedd aatt 88 °°CC ffoorr 44 hhrrss.. TThhee mmiixxttuurree wwaass ccoonncceennttrraatteedd.. TThhee rreessiidduuee wwaass ppuurriififieedd bbyy ccoolluummnn cchhrroommaattooggrraapphhyy ((SSii00 22 ,, PPeettrroolleeuumm eetthheerr//EEtthhyyll aacceettaattee==33//ll ttoo 22//11)) ttoo aaffffoorrdd ccoommppoouunndd 3300GG ((229933 mmgg,, yyiieelldd 8833..4466%%)) aass yyeellllooww ooiill.. 1H1H NNMMRR ((440000MMHHzz,, CCDDCC11 33 )):: δδ 44..2288 ((ss,, 22HH)),, 33..9966 -- 33..8855 ((mm,, 11HH)),, 33..8811 -- 33..6699 ((mm,, 22HH)),, 33..5577 -- 33..4444 ((mm,, 22HH)),, 11..6677 -- 11..5511 ((mm,, 77HH)),, 11..5500 -- 11..4433 ((mm,, 112211 II 11..4400 -- 11.,2266 ((mm,, 22HH)),,

To a solution of pre-active Zn powder (154.49 mg, 2.36 mmol) in THF (6 mL, fresh distilled) was added 1,2-dibromoethane (22.9 mg, 121.88 umol). The reaction mixture was stirred at 70 °C then cooled back to 8 °C, this cycle was repeated three times. Freshly distilled TMSC1 (8. 5 mg, 75.00 umol) was added and the reaction was stirred at 8 °C for 0.1 h. A solution of compound 30A (425.59 mg, 1.17 mmol) in anhydrous TFEF (5 mL) was added at 8 °C. The reaction was stirred at 8°C for 0.75 h. Meanwhile, a round bottomed flask, equipped with a stirrer bar, was charged with LiCl (101.34 mg, 2.39 mmol), which was subsequently dried under vacuum at high timeperature with hot gun. To this flask was added CuCN (104.96 mg, 1.17 mmol) and anhydrous THF (5 mL), the mixture was stirred at 8 °C until a yellow solution was obtained that was then cooled to -10 °C. The previously prepared solution of the organozinc iodide was decanted from the excess zinc via syringe and added dropwise to the solution of the copper complex. The reaction mixture was stirred at -10 °C for 0.2 h and then cooled to - 78 °C and a solution of compound 30G (410 mg, 937.55 umol) in anhydrous THF (5 mL) was added. The reaction was stirred at -20 °C for 4 h and then at 8 °C for 16 hrs. The mixture was quenched with 10% NH 4 G (40 mL) and extracted with EA (3 x 50 mL). The combined organic layers were dried over Na 2 S0 4 and concentrated. The residue was purified by column chromatography (Si0 2 , Petroleum ether/Ethyl acetate=T0: l to 5: 1) to afford compound 30H (260 mg, yield 23.08%) as yellow oil. 1H NMR (400MHz, CDCI 3 ): δ 7.38 - 7.35 (m, 5H), 5.70 - 5.51 (m, 1H), 5.13 - 5.10 (m, 2H), 4.10 - 4.05 (m, 1H), 33..9966 -- 33..8844 ((mm,, 22HH)),, 33 ,,8833 -- 33..6688 ((mm,, 66HH)),, 33..4488 -- 33..4422 ((mm,, 1IHH)),, 22,,8877 -- 22..7722 ((mm,, 22HH)),, 11..6611 -- 11 ,,3322 ((mm,, 22 I1 H! !))..

[004 17766]] SStteepp 99:: SSyynntthheessiiss ooff ccoommppoouunndd 3300JJ

CCoommppoouunndd 3300,,11 wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 3300 HH ffoolllloowwiinngg tthhee pprroocceedduurree ooff ccoommppoouunndd 2266KK.. CCoommppoouunndd 3300JJ ((222200 mmgg,, ccrruuddee)) wwaass oobbttaaiinneedd aass aa yyeellllooww ooiill aanndd uusseedd ffoorr nneexxtt sstteepp ddiirreeccttllyy.. MMSS ((EESSII)) mm//zz ((MM++NNaa)) ++ 555555,.00..

[[0000447777]] SStteepp 1100:: SSyynntthheessiiss ooff ccoommppoouunndd 3300

CCoommppoouunndd 3300KK wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 3300 JJ aanndd t feernt~-bbuuttyyil LL--lleeuucciinnaattee ffoolllloowwiinngg tthhee pprroocceedduurree ooff ccoommppoouunndd 226 JJ.. CCoommppoouunndd 3300 ((6600 mmgg,, yyiieelldd 2200..77 %%)) wwaass oobbttaaiinneedd aass aa ccoolloorrlleessss ooiill.. MMSS ((EESSII)) mm//zz ((MM++NNaa)) ++ 772244..44..

[00478] Step 11; Synthesis of compound 30L

CCoommppoouunndd SSOOLL wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 3300 ffoolllloowwiinngg tthhee pprroocceedduurree ooff ccoommppoouunndd 2266LL.. CCoommppoouunndd SSOOLL ((6600 mmgg,, ccrruuddee,, HHCCll)) wwaass oobbttaaiinneedd aass aa yyeellllooww ooiill aanndd uusseedd ffoorr nneexxtt sstteepp ddiirreeccttllyy.. MMSS ((EESSII)) mm//zz ((MM++NNaa)) ÷÷ 556622..11 ,,

To a solution of compound 30L (60 mg, 100.31 umol, HCl) in DCM (3 mL) was added TEA (3 mL, 40.52 mmol). The mixture was stirred at 1 5 °C for 30 min. The reaction mixture was concentrated to afford compound 30M (60 mg, crude, TFA) as yellow oil, which was used to next step without further purification. MS (ESI) m/z (M I f } ' 506, 1 .

[00480] Step 13: Synthesis of compound 30N

Compound 30N was prepared from compound 30M following the procedure of compound 26M. Compound SON (13 mg, yield 22.44 %) was obtained as a white solid. MS (ESI) m/z (M +H) + 488. 1H NMR (400MHz, DMSO-fife) «58.19 - 8.05 (m, 1H), 8. 18 - 8.03 (m, 1 1 I L 7.80 - 7.73 (m, I H), 7.40 - 7.29 (m, 5H), 7.18 - 7.04 (m, 1H), 5.09 - 4.98 (m, 1H), 5.09 - 4.96 (m, 1 1 1 ). 5.10 - 4.96 (m, IH), 5.08 - 4.94 (m, 1 1 1 ). 4.64 - 4.56 (m, IH), 4.66 - 4.52 (m, I H), 4.43 - 4.34 (m, I H), 4.31 - 4. 15 (m, I H), 4.04 - 3.93 (m, 2H), 3 ,88 - 3.73 (m, IH), 3 ,30 - 3.15 (m, 3H), 2.70 - 2,59 (m, IH), 2.56 - 2.45 (m, 43H), 1 ,63 - 1 .47 (m, 3H), 1.46 - 1.33 (m, 3H), 1.33 - 1.15 (m, 5H), 0.93 - 0,77 (m, 61 1 )

[00481] Step 14; Synthesis of compound 30 Compound 30 was prepared from compound SON following the procedure of compound 26. Compound 30 (3.2 mg, yield 21.42 %) was obtained as a white solid. MS (ESI) m/z (M 1 1 } 486.2. ! H NMR (400MHz, D\ISO- i/y.): δ 9.43 (s, 1 i I ). 8.38 - 8.30 (ni, 1 H), 8.25 - 8. 15 (m, 1 H), 7.39 - 7.27 (m, 5H), 7.19 - 7.07 (m, 1H), 5.06 - 4.96 (m, 2H), 4,46 - 4.35 (m, 2H), 4,27 - 4, 18 (m, 1 H), 4.02 - 3.90 (m, 2H), 2,74 - 2.55 (m, 2H), 1.64 - 1.25 (m, 9H), 0.96 - 0.86 (m, 3H), 0.85 - 0.75 (m, 31 1 )

(4R,7^ f -4-isobutyl-l-methyl-2,5-dioxo-l,6-diazacvclotetradec-9-ene- 7-carbaldehvde (31)

(S)-4-benzyloxazolidin-2-one (70 g, 395.03 mmol) was dissolved in THF (700 mL) and cooled to -65 °C under N 2 . n-BuLi (190 mL, 474.04 mmol, 2.5 M in hexane) was added during 1 h and the resultant mixture was stirred for I h at -65 °C to -60°C. Then 4~ methylpentanovl chloride (66.2 mL, 493.79 mmol) was added and the resulting mixture was stirred at -60 C 'C to -65°C for Ih before warming to 0 - 5 °C and stirred at 0 - 5 C 'C for 16 hours. The reaction was quenched with saturated NH 4 C1 (500 mL), the THF was removed under vacuum, and the aqueous phase was washed with EtOAc (500 mL x 2). The organic phase was then washed with IN NaOH (200 mL) and brine (200 rnL), after which it was dried over anhydrous \a : S() : The product was isolated following filtration and removal of the solvent under vacuum to give compound 31 A (1 15.20 g, 79% yield) as a yellow solid.

NMR (CDC! :, 400 MHz) δ 7.43 - 7.21 (m, 5H), 4.76 - 4.68 (m, 1 1 1 ), 4.28 - 4.18 (m, 2H), 3.35 (dd, J =3.3, 13.3 Hz, 1 H), 3.09 - 2.91 (m, 2H), 2.82 (dd, J=9.7, 13.4 Hz, 1H), 1.76 - 1.58 (m, i l 1.02 - 0.90 (m, 71 i ). MS (ESI) m/z (M+Na + ) 412.2,

TThhee ssoolluuttiioonn ccoonnssiissttiinngg ooff ccoommppoouunndd 3311AA ((8800 gg,, 229900..5555 mmmmooll)) aanndd TTHHFF ((880000 mmLL)) wwaass aaddddeedd ttoo LLii HHMMDDSS ((332233 mmLL,, 332222..8833 mmmmooll,, 11MM iinn TTHHFF)) aatt -- 6600°°CC ttoo --6655°°CC uunnddeerr NN 22 .. TThhee mmiixxttuurree wwaass ssttiirrrreedd aatt --6655°°CC ffoorr 11 hhoouurr aanndd tthheenn feferr//--bbuuttyyll 22--bbrroommooaacceettaattee ((11 1199..33 mmLL,, 880077..0088 mmmmooll ,,)) wwaass aaddddeedd aatt -- 6600°°CC ttoo --6655°°CC ddrrooppwwiissee.. TThhee mmiixxttuurree wwaass ssttiirrrreedd aatt --6655°°CC ffoorr II hh aanndd tthheenn wwaarrmmeedd ttoo 55°°CC -- 1100 °°CC sslloowwllyy,, aanndd ssttiirrrreedd ffoorr 1166 hhoouurrss bbeeffoorree qquueenncchhiinngg wwiitthh ssaattuurraatteedd NNHIT 44 CC1I ((550000 mmLL)).. TTHHFF wwaass rreemmoovveedd uunnddeerr vvaaccuuuumm,, aanndd t thhee aaqquueeoouuss pphhaassee wwaass wwaasshheedd wwiitthh EEttOOAAcc ((550000 mmLL xx 22)).. TThhee ccoommbbiinneedd oorrggaanniicc pphhaassee wwaass tthheenn wwaasshheedd wwiitthh 11 NN NNaaOOHH ((220000 mmLL)) aanndd bbrriinnee ((220000 mmLL)),, tthhee oorrggaanniicc pphhaassee wwaass ddrriieedd oovveerr aannhhyyddrroouuss NNaa 22 SS00 44 aanndd ccoonncceennttrraatteedd.. MMuucchh ssoolliidd wwaass pprreecciippiittaatteedd oouutt,, fifilltteerreedd aanndd tthhee fi filltteerr ccaakkee wwaass wwaasshheedd wwiitthh PPEE:: EEAA ((1100:: 11,, 1155 mmLL xx 22)),, tthheenn tthhee fifilltteerr ccaakkee wwaass ddrriieedd uunnddeerr rreedduucceedd pprreessssuurree ttoo ggiivvee tthhee ppuurree ccoommppoouunndd 3311AA ((3300 gg)) aass aa wwhhiittee ssoolliidd.. TThhee fifillttrraattee wwaass ccoonncceennttrraatteedd aanndd ppuurriiffiieedd bbyy FFCCCC ((PPEE:: EEAA==11 ::00 ttoo 11 :: 11)) ttoo aaffffoorrdd ccoommppoouunndd 3311BB ((1144 gg)) aass aa wwhhiittee ssoolliidd.. 1H1H NNMMRR ((CCDDCC1I 33,, 440000 MMHHzz)) δδ 77..3399 -- 77..1199 ((mm,, 55HH)),, 44..7700 -- 44..5599 ((mm,, II HH)),, 44..3311 -- 44..2200 ((mm,, 11HH)),, 44..1199 -- 44..1100 ((mm,, 22HH)),, 33..3388 -- 33..3300 ((mm,, 1IHH)),, 22..8811 -- 22..6677 ((mm,, 22HH)),, 22..5533 -- 22..4433 ((mm,, 11HH)),, 11..6688 -- 11..4499 ((mm,, 33HH)),, 11..4422 (( ss.. 99HH)),, 11..3388 -- 11 ..2288 ((mm,, 1IHH)),, 00..9922 ((tt,, ..!! 66..88 H Hzz,, 66HH)).. MMSS ((EESSII)) mm/zz ((MM++NNaa ++ )) 441122..22,,

BnOH (16.66 g, 154.06 mmol, 16 mL) was dissolved in THF (300 mL) and cooled to 0°C. /z-BuLi (2.5M, 37 mL) was added dropwise and the reaction mixture was stirred for 0.5 h under N 2 atmosphere. A solution of 31B (30.0 g, 77.03 mmol) in THF ( 100 mL) was added and the reaction mixture was stirred at 0 °C for 4 hrs. The reaction mixture was quenched with sat. NH 4 CI (200 mL), and then extracted with EA (200 mL x 3). The combined organic layers were dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Si0 2 , Petroleum ether/Ethyl acetate = 20/1 to 10: 1). Compound 31C (23.00 g, yield: 93.19%) was obtained as a colorless ooiill.. 1H1H NNMMRR ((440000MMHHzz,, CCDDCC1! 3 3)) δδ 77,,4433 -- 77,,2288 ((mm,, 55HH)),, 55..2200 -- 55..0088 ((mm,, 22HH)),, 22,,9966 -- 22..8844 ((mm,, 1IHH)),, 22,,6677 -- 22,,5577 ((rrnn.. IIHH)),, 22..4411 -- 22..3322 ((mm,, 1IHH)),, 11..6644 -- 11..5511 ((mm,, 22HH)),, 11..4411 ((ss,, O9i1 l1 )).. 11..3333 -- 11..2299 ((mm,, 1IHH)),, 00..9933 -- 00..8855 ((rmn,, ((::»»!! !! }}..

[[0000448855]] SStteepp 44:: SSyynntthheessiiss ooff ccoommppoouunndd 3311PP

TToo aa ssoolluuttiioonn ooff 3311CC ((2233..00 gg,, 7711 ..7788 n mimmooll)) iinn DDCCMM ((110000 mmLL)) wwaass aaddddeedd T TFFAA. ((115544 gg,, 11..3355 mmooll,, 110000 mmLL)) sslloowwllyy.. TThhee mmiixxttuurree wwaass ssttiirrrreedd aatt 1188 °°CC ffoorr 22 hhoouurrss.. TThhee rreeaaccttiioonn mmiixxttuurree wwaass ccoonncceennttrraatteedd uunnddeerr rreedduucceedd pprreessssuurree ttoo ggiivvee aa rreessiidduuee.. CCoommppoouunndd 3311DD ((1188..5500 gg,, y yiieelldd:: 9977..5511%%)) wwaass oobbttaaiinneedd aass aa yyeellllooww ooiill.. TThhee pprroodduucctt wwaass uusseedd iinn tthhee nneexxtt sstteepp wwiitthhoouutt ffuurrtthheerr ppuurriiffiiccaattiioonn.. 1H1H NNMMRR ((440000MMHHzz,, CCDDCC11 33 )) δδ 1100..3399 ((bbrr ss,, 1HH)),, 77..4455 -- 77..2299 ((mm,, 55HH)),, 55..2222 -- 55.. 11 11 ((mm,, 22HH)),, 33,,0033 -- 22..9900 ((mm,, 1I HH)),, 22..8855 -- 22,,7733 ((mm,, 1IHH)),, 22,,5599 -- 22..4499 ((mm,, 1I HH)),, 11..6699 -- 11..5522 ((mm,, 22HH)),, 11 ..4433 -- 11..2277 ((mm,, 1I HH)),, 00..9999 -- 00..8833 ((mm,, 66HH))..

[[0000448866]] SStteepp 55:: SSyynntthheessiiss ooff ccoommppoouunndd 3311EE

CCoommppoouunndd 3311EE wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 3311DD aanndd NN--mmeetthhyyllhheexx--55--eenn--ll--aammiinnee ffoolllloowwiinngg tthhee pprroocceedduurree ooff ccoommppoouunndd 77DD.. CCoommppoouunndd 3311EE ((88,,8800 gg,, yyiieelldd 8822..2277 %%)) wwaass oobbttaaiinneedd aass aa ccoolloorrlleessss ooiill.. MMSS ((EESSII)) mm//zz ((MM++HH)) ++ 336600..11.. 1H1H NNMMRR ((440000MMHHzz,, CCDDCC!! 33)) 00 77..4455 -- 77..2233 ( {mrn,. 55HH)),, 55..8855 -- 55..7700 ((mm,, IIHH)),, 55..2222 · -· 55..1144 ((mm,, 1IHH)),, 55..1122 -- 55..0066 ((mm,, 1IHH)),, 55..0055 -- 44..9911 ((mm,, 22HH)),, 33..4466 - - 33..3344 ((mm,, 1IHH)),, 33..3322 -- 33..2200 ((mm,, 1IHH)),, 33..1133 - - 33..0022 ((mm,, IIHH)),, 22..9977 - - 22..8855 ((mm,, 33HH)),, 22..7788 -- 22..6677 ((mm,, 1IHH)),, 22..4411 -- 22..3300 ((mm,, IIHH)),, 22.. 11 11 -- 22..0055 ((mm,, 22HH)),, 11..6633 -- 11..4444 ((mm,, 44HH)),, 11..4433 -- 11..2299 ((mm,, 33HH)),, 00..9977 -- 00..8811 ((mm,, 66HH))..

To a solution of 31E (8.80 g, 24.48 mmol) in MeOH (100 mL) was added NaOH (4.90 g, 122.40 mmol) and H 2 0 (20 mL). The mixture was stirred at 30 °C for 24 hours. The reaction mixture was concentrated under reduced pressure to remove most of the solvent. The residue was diluted with water (100 mL) and extracted with MTBE (100 mL x 3), To the water layer was added IN HC1 until pH ~ 4, then extracted with EA (100 mL x 3). The combined organic layers were washed with brine (200 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. Compound 31F (4.90 g, yield: 61.67%) was obtained as a colorless oil. The product was used in the next step without further purification. 1H NMR (400MHz, CDC1 3 ) δ 10.51 (br s, H), 5.84 - 5.71 (m, IH), 5.07 - 4.90 (m, 2H), 3,46 - 3.19 (m, 2H), 3.03 - 2,90 (m, 4H), 2,75 - 2.61 (m, I H), 2.51 - 2.40 (m, II I), 2.14 - 2.05 (m, 2H), 1.74 - 1.48 (m, 4H), 1.46 - 1 ,33 (m, 2H), 1 .32 - 1.26 (m, I H), 0.97 - 0,86 (rn, ( :»! ! }. MS (ESI) m/z (M+H) + 270, 1).

[00488] Step 7: Synthesis of compound 31G

Compound 31 G was prepared from compound 31F and ethyl (5)-2-aminopent-4- enoate following the procedure of compound 7D. Compound 311» (4.60 g, yield 70,66 %) was obtained as a yellow oil. 1H NMR (400MHz, CDC1 3 ) δ 6.59 - 6.41 (m, IH), 5.82 - 5.59 (m, 2H), 5.16 - 4.85 (m, 4H), 4.63 - 4.50 (m, IH), 4.12 ·· 4.05 (m, IH), 3.41 - 3.10 (m, 3H), 2.98 - 2.79 (m, 4H), 2.73 - 2.60 (m, IH), 2.57 - 2.38 (m, 2H), 2.32 - 2.18 (m, IH), 2.03 - 1.96 (m, IH), 1.70 - 1.42 (m, 5H), 1.41 - 1.28 (m, 2H), 1 .23 (t, J=7.2 Hz, 3H), 1.20 - 1.10 (m, IH), 0.94 - 0,82 (m, 6H). MS (ESI) m/z (M+H) + 396.2.

[00489] Step 8: Synthesis of compound 31H

Compound 31H was prepared from compound 31G following the procedure of compound 18H. Compound 31 H (700 mg, yield 35.7 %) was obtained as brown oil. 1H NMR (400MHz, CDCI 3 ) δ 6.57 (br d, ./ 7.8 Hz, 0.77! ! ). 6.19 (br d, ./ 9 2 Hz, 0.16H), 5.56 - 5.43 (m, IH), 5,37 - 5.23 (m, IH), 4.84 - 4.73 (m, 0.17H), 4,62 - 4,50 (m, I H), 4.29 - 4.09 (m, 3H), 3.67 - 3.54 (m, 0.18H), 3.12 - 3.01 (m, IH), 2.98 (s, 3H), 2.79 - 2.62 (m, 2H), 2.54 - 2.42 (m, IH), 2.27 - 2.09 (m, 2H), 2.08 - 1.85 (m, 2H), 1.84 ·· 1.54 (m, 4H), 1.52 - 1.41 (m, IH), 1.40 - 1.30 On, I H), 1.29 - 1.23 (m, 31 1 ). 1.23 - 1.22 (m, I H), 1.19 - 1.09 (m, IH), 0.93 (d, ./ 0.5 Hz, 3H), 0.89 (d, 6.7 Hz, 3H). MS (ESI) m/z (M+H) ÷ 367.2.

[00490] Step 9; Synthesis of compound 31J

Compound 3U was prepared from compound 31H following the procedure of compound 26K, Compound 3U (330 mg, yield 87.55 %) was obtained as white foam and used for next step directly, MS (ESI) m/z (M+Na) ÷ 339.2,

[00491] Step 10; Synthesis of compound 31

Compound 31 K was prepared from compound 311 and N-methoxymethanamine following the procedure of compound 26J. Compound 31K (150 mg, yield 38.3 1 %) was obtained as a white solid. 1H NMR (400MHz, CDC1 3 ) δ 6.50 (br d, ./ 8.5 Hz, 0.69H), 6,31 (br d, J=9.3 Hz, 0.19H), 5.56 - 5.47 (m, IH), 5.44 - 5.33 (m, IH), 4.97 (br s, IH), 4.32 - 4.19 (m, IH), 3.77 (s, 3H), 3.20 (s, 3H), 3.07 - 2.95 (m, 3H), 2.84 - 2.72 (m, IH), 2.63 - 2.41 (m, 2H), 2.24 - 2.06 (m, 2H), 2.05 - 1.90 (m, 2H), 1.82 - 1 .68 (m, IH), 1.61 - 1.31 (m, 4H), 1.29 - 1.14 (m, 2H), 0.95 - 0.84 (m, 6H). MS (ESI) m/z (M+H) + 382.2. Compound 31 was prepared from compound 31K following the procedure of compound 14F. Compound 31 (90 mg, yield 76.06 %) was obtained as a white solid. ! H NMR (400MHz, CDC1 3 ) δ 9,61 - 9.56 (m, 0.15H), 9.52 (s, 0.7211), 6,75 (br d, 6.4 Hz, !!!}. 5.55 - 5.44 (m, IH), 5.34 - 5,23 (m, IH), 4.54 - 4,45 (m, IH), 4.24 - 4.15 (m, 1H), 3.18 - 3.09 (m, III), 3,02 - 2,96 (m, 3H), 2.77 - 2.64 (m, 2H), 2,55 - 2.46 (m, Hi), 2.25 - 1,94 (m, 411),. 1.85 - 1.74 (m, 2H), 1.57 - 1.42 (m, 2H), 1.39 - 1.28 (m, IH), 1.27 - 1.12 (m, 2H), 0.98 - 0.88 (m, 6H). MS (ESI) m/z (M+H) + 323.2.

7ert-butyl (4»y,7 'J0S',i-^-10-formyl-7-isobutyl-5,8-dio¾o-l-o¾a-6. > 9-diazacvcopentadec- -en-4-yl)carbamate (32)

To a solution of L-homoserine (120 g, 1.01 mol, 1.00 eq) in MeCN (212 mL) was added NaOH (1 M, 1,01 L, 1.00 eq). The mixture was cooled to 0 °C and then B0C2O (233 g, 1,07 mol, 245.32 mL, 1.06 eq) in MeCN (50 mL) was slowly added to the mixture. The rreeaacctitioonn wwaass ssttiirrrreedd aatt 2255 °°CC ffoorr 1122 hhoouurrss.. TThhee oorrggaanniicc ssoollvveenntt wwaass rreemmoovveedd aanndd aaddddeedd tthhee aaqquueeoouuss ttoo ppHH ~~ 55 wwiitthh HHQQ ((22NN,, 220000 mmLL)).. TThheenn NNaaCCll wwaass aaddddeedd uunnttiill tthhee ssoolluuttiioonn rreeaacchheedd ssaattuurraattiioonn.. TThhee mmiixxttuurree wwaass fifilltteerreedd.. TThhee fifilltteerr ccaakkee wwaass ddrriieedd uunnddeerr rreedduucceedd pprreessssuurree ttoo ggiivvee ccoommppoouunndd 3322AA ((448800 gg,, ccrruuddee)) aass aa wwhhiittee ssoolliidd.. !! HH NNMMRR ((DDMMSSOO--ddee,, 440000MMHHzz)) δδ 77..0055 -- 66..9988 ((mm,, IIHH)),, 44..0055 -- 33..9955 ((mm,, 1IHH)),, 33..5500 -- 33..3377 ((mm,, 22HH)),, 11..8833 -- 11..6600 ((mm,, 22HH)),, 11..4455 -- 11..3300 ((mm,, 99HH)),, M MSS ( (EESSII)) mm//zz ((MM++NNaa ++ )) 224411 ..88..

[[0000449944]] SStteepp 22:: SSyynntthheessiiss ooff ccoommppoouunndd 3322BB

TToo aa ssoolluuttiioonn ooff ccoommppoouunndd 3322AA ((224400 gg,, 11..0099 mmooll,, 11..0000 eeqq)) iinn EEttOOHH ((880000 mmLL)) wwaass aaddddeedd tthhee ssoolluuttiioonn ooff N NaaOOHH ((4455..7788 gg,, 11..1144 mmooll,, 11..0055 eeqq)) iinn HH 22 00 ((440000 mmLL)).. TThhee mmiixxttuurree wwaass ssttiirrrreedd aatt 2255 °°CC ffoorr 1166 hhoouurrss.. TThhee ssoollvveenntt wwaass rreemmoovveedd aanndd tthhee rreessiidduuee wwaass ddiissssoollvveedd iinn DDMMFF ((ll OOOOmmLL)).. TThheenn MMeell ((1166 mmLL,, 225555..0044 mmmmooll,, 33..3355 eeqq)) wwaass aaddddeedd ttoo tthhee aabboovvee ssoolluuttiioonn.. TThhee mmiixxttuurree wwaass ssttiirrrreedd aatt 2255 °°CC ffoorr 2244 hhoouurrss.. TThheenn tthhee mmiixxttuurree wwaass eexxttrraacctteedd wwiitthh EEAA ((880000 mmLL xx 33)).. TThhee oorrggaanniicc pphhaassee wweerree ccoolllleecctteedd,, wwaasshheedd wwiitthh bbrriinnee ((11000000 mmLL xx 55)),, ddrriieedd wwiitthh NNaa 22 SS00 44 ,, fifilltteerreedd aanndd ccoonncceennttrraatteedd ttoo aaffffoorrdd ccoommppoouunndd 3322BB ((111166 gg,, yyiieelldd:: 4455..6622%%)) aass yyeellllooww- ooiill.. 1H1H N NMMRR ((DDMMSSC0-i 440000MMHHzz)) δδ 77..2200 -- 77..1155 ((mm,, IIHH)),, 44..1155 -- 44..0055 ((mm,, 1IHH)),, 33..6655 -- 33..6600 ((mm,, 33HH)) 33..5500 -- 33..4400 ((mm,, 22HH)),, 11..8822 -- 11..6633 ((mm,, 22HH)),, 11 ..4455 -- 11..3300 ((mm,, 99HH)),.

TToo aa ssoolluuttiioonn ooff ccoommppoouunndd 3322BB ((116622 gg,, 669944..5500 mmmmooll,, 11..0000 eeqq)) aanndd TTEEAA ((229900 mmLL,, 22..0088 mmooll,, ,, 33..0000 eeqq)) iinn DDCCMM ((550000 mmLL)),, MMssCCll ((9999 mmLL,, 11..2277 mmooll,, 11..8833 eeqq)) wwaass ddrrooppwwiissee aaddddeedd aatt 00 °°CC.. TThhee rreeaaccttiioonn wwaass ssttiirrrreedd aatt 1100 °°CC ffoorr 1122 hhoouurrss b beefoforree ddiilluutteedd wwiitthh wwaatteerr ((110000 mmLL)),, eexxttrraacctteedd wwiitthh DDCCMM ((330000 mmLL xx 22)).. TThhee oorrggaanniicc llaayyeerrss wweerree ccoommbbiinneedd aanndd ddiirreedd oovveerr NNaa 22 SSC0>4 4 .. FFiilltteerreedd aanndd tthhee fifillttrraattee wwaass ccoonncceennttrraatteedd iinn vvaaccccuuoo ttoo ggiivvee ccoommppoouunndd 3322CC ((220000 gg,, ccrruuddee)) aass bbllaacckk bbrroowwnn ooiill.. 1H1H N NMMRR ((DDMMSSO0-- ,, 440000MMHHzz)) δδ 77..4422 -- 77..3322 ((mm,, IIHH)),, 44..3300 -- 44..1100 ((mm,, 22HH)),, 33..6655 ((ss,, 33HH)),, 33..1166 ((ss,, 33HH)),, 22..1155 -- 11..9900 ((mm,, 22HH)),, 11..4455 -- 11..3300 ((mm,, 99HH))..

To a solution of compound 32C (25 g, 80.30 mmol, 1.00 eq) in acetone (400 mL) was added LiBr (21 g, 241 mmol, 3.00 eq). The mixture was stirred at 50 °C for 2 hours. The reaction was concentrated under reduced pressure to give a residue. The residue was purified by FCC (PE/EA = 4: 1 ) to give compound 32D (145 g crude) was obtained as yellow oil. H

NMR (DMSO-i¾, 400MHz) δ 7.40 - 7.32 (m, H), 4,20 - 4, 10 (m, IH), 3.65 (s, 3H), 3 ,60 - 3.45 (s, 2H), 2.20 - 2,05 (m, 2H), 1.50 - 1.30 (m, 9H). 00497]

BBuutt--33--eenn--ll--ooll ((88,,77 mmLL,, 110011..3300 mmmmooll,, 11..5500 eeqq)) wwaass aaddddeedd ttoo aa ssoolluuttiioonn ooff NNaaHH ((66.,7755 gg,, 116688..8833 mmmmooll,, 6600%% ppuuririttyy,, 22.,5500 eeqq)) iinn TTHHFF ((5500 mmLL)) aatt 00 °°CC uunnddeerr NN 22 .. TThhee rreeaaccttiioonn wwaass ssttiirrrreedd aatt 00 °°CC ffoorr 3300 mmiinn uunnddeerr NN 22 .. TThhee ssoolluuttiioonn ooff ccoommppoouunndd 3322DD ((2200 gg,, 6677..5533 mmmmooll,, 11 ..0000 eeqq)) iinn TTHHFF ((5500 mmLL)) wwaass aaddddeedd ddrrooppwwiissee aatt 00 °°CC uunnddeerr NN 22 aanndd ssttiirrrreedd aatt 00 °°CC ffoorr 22 hhoouurrss.. HH 22 00 ((5500 mmLL)) wwaass aaddddeedd aanndd tthhee mmiixxttuurree wwaass ssttiirrrreedd aatt 00 °°CC ffoorr 11 hh .. TThhee mmiixxttuurree wwaass eexxttrraacctteedd wwiitthh eetthhyyll aacceettaattee ((110000 mmLL xx 22)),, TThhee ccoommbbiinneedd aaqquueeoouuss llaayyeerrss wweerree aacciiddiiffiieedd wwiitthh 22NN HHCCll ttoo ppHH ~~ 33 aanndd eexxttrraacctteedd wwiitthh eetthhyyll aacceettaattee ((220000 mmLL xx 22)).. TThhee oorrggaanniicc llaayyeerrss wweerree ccoommbbiinneedd aanndd ddrriieedd oovveerr NNaa 22 SS00 44 ,, fifilltteerreedd aanndd ccoonncceennttrraatteedd uunnddeerr rreedduucceedd pprreessssuurree ttoo ggiivvee ccoommppoouunndd 3322EE ((44.,33 gg,, ccrruuddee)) aass yyeellllooww ssoolliidd..

Compound 32F was prepared from compound 32E and 7C following the procedure of compound 7D. Compound 32F (250 mg, yield 13%) was obtained as a white solid. ! H NMR

I S< )-</,·., 400MHz) δ 8,27(br d, .1 7.3 Hz, I I I ). 7.70(br d, 8,3 Ηζ, Ι Η), 6.93 (br d, .1 8.3 Ηζ, ΙΗ), 5.85 - 5.67 (m, 2H), 5 , 14 - 4,95 (m, 4H) , 4,43 - 4, 20 (m, 2 H), 4.1 1 - 3.90 (m, 3H), 3.45 - 3.33 (m ,4H), 2.47 - 2,31 (m ,3H), 2.23(q. J 6.7 Hz, 2H), 1 ,88 - 1.75 (m, i l l ). 1.73 - I ,55(m, 21 1 ),. 1.43 (br t, J = 7.2 Hz, 2H), 1..39 -1 ,29 (m, 9H), 1.15 (t, J = 7,0 Hz, 31 1 ). 0.85(dd, J=6.5, 14.8 Hz, 6H). MS (ESI) m/z (M+H) + 51 1.6.

[00499] Step 7: Synthesis of compound 32G

Compound 32G was prepared from compound 32F following the procedure of compound 18H. Compound 32G (260 mg, yield 76.33 %) was obtained as a white solid. 1H NMR (DMSO-fife, 400MHz) δ 8.12 (br d, J = 9.3 Hz, 1H), 7.53 (br d, J = 7.5 Hz, 1H), 6.95 - 6.86 (m, 1H), 5.75 - 5.64 (m, 1 1 1 ), 5.37 - 5.24 (m, 1 1 1 ). 4.45 - 4.21 (m, 2H), 4.13 - 3.99 (ni, 3H), 3.34 - 3.19 (m, 4H), 2.45 - 2.30 (m, 2H), 2.12 (br d, J ------- 5.0 Hz, 2H), 1 .93 - 1.81 (m,

1 1 1 ),, 1.73 - 1 .39 (m, 41 1 },, 1.36 (s, 91 1 },, 1 , 16 (t, J 7,0 Hz, 31 ! }. 0,91 - 0,75 (ni, 6H). MS (ESI) m/z (M+H) + 484,2.

[00500] Step 8: Synthesis of compound 32H

Compound 3211 was prepared from compound 32G following the procedure of compound 14F. Compound 32H (100 mg, yield 52 %) was obtained as white solid. 1H NMR (CDC13, 400MHz) δ 6,82 (br d, J = 8.3 Hz, 1H), 6.43 (br d, J = 7.0 Hz, IH), 5.67 - 5.56 (m, IH), 5.55 - 5.44 (m, IH), 5.17 (br d, J = 7.5 Hz, IH), 4.57 - 4.47 (m, IH), 4.38 - 4.29 (m, 1H), 4.06 (br d, ,/ 4.8 Hz, I N ), 3.72 - 3,56 (m, 4H), 3.52 (td, J = 4.9, 9,5 Hz, I I I ). 3 ,44 - 3.35 (rn, 1 1 1), 2.97 (br l. J 5.4 Hz, 1 1 1), 2.47 - 2.37 (m, 1H), 2.34 - 2.18 (m, 3H), 2.10 (dq, J = 4,4, 9,4 Hz, IH), 1.95 - 1.81 (m, 2H), 1.66 - 1.61 (m, IH), 1.53 - 1.38 (m, 10H), 0.92 (dd, J = 3.9, 6.4 Hz, 6H). MS (ESI) m/z {M i l} 442.1.

[00501] Step 9: Synthesis of compound 32

Compound 32 was prepared from compound 32H following the procedure of compound 7. Compound 32 (25 mg, yield 50 %) was obtained as white solid. ¾ NMR (CD CI 3, 400MHz) δ 9.57 (s, IH), 7.02 - 6.90 (m, 1H), 6.74 (br d, J = 7.3 Hz, IH), 5.61 - 5.48 (m, IH), 5.31 (br d, J = 7.3 Hz, IH), 5.24 - 5.17 (m, IH), 4.68 - 4.54 (m, 2H), 4.36 - 4.27 (m, IH), 3.73 - 3.59 (m, 2H), 3,59 - 3.50 (m, IH), 3.48 - 3 ,38 (m, I H), 2.77 - 2.60 (m, I H), 2.40 - 2.50 (m, I H), 2.37 - 2.25 (m, IH), 2,24 - 2.12 (m, 2H), 1.98 - 1.82 (m, 2H), 1 .67 - 1 .61 (m, IH), 1.51 - 1.36 (m, 10H), 0.97 - 0.85 (m, 6H). MS (ESI) m/z (M i l ) ' 440.2.

Tert-butyl ({4A\7^J0^-10-formyI-7-isobut r I-5,8-dioxo-l-os¾-6,9-di¾z¾cycIopentadec¾n-

The mixture of compound 32H (46 mg, 104.18 umol, 1.00 eq) and Pd/C (20 mg) in MeOH (10 mL) was stirred at 20 - 25 °C for 2 hours under H 2 atmospheres. Filtered and the filtration was concentrated under reduced pressure to give compound 33A (40.00 mg, yield: 86,5%) as a white solid. 1H NMR (DMSO- , 400MHz) δ 7.92 (d, . 9.0 Hz, IH), 7,75 (br d, J = 8.8 Hz, IH), 6.60 (br d, J = 7.5 Hz, IH), 4.59 (br s, IH), 4.46 - 4.33 (m, IH), 4.10 - 3.93 (m, IH), 3.72 (br s, IH), 3.29 - 3.20 (m, 3H), 3.19 - 3.10 (m, IH), 1.93 ·· 1.79 (m, IH), 1.72 (qd, ./ 6.7, 1 1 .2 Hz, I H), 1.62 - 1 ,45 (m, 2H), 1.44 - 1 ,08 (m, 15H), 0.85 (br dd, ./ 6.4, 12,9 Hz, 6H). MS (ESI) m/z ( W W ) 444.2.

[00503] Step 2: Synthesis of compound 33

Compound 33 was prepared from compound 33A following the procedure of compound 7. Compound 33 (20 mg, yield 45 %) was obtained as white solid. Ή NMR (CDC13, 400X 1 Hz) δ 9.55 (s, IH), 6.84 (br d, J=7.0 Hz, IH), 6.70 (br d, J=8.5 Hz, IH), 5. 17 (br d, J=8.0 Hz, IH), 4.61 - 4.51 (m, 2H), 4.30 - 4.21 (m, IH), 3.62 - 3.54 (m, IH), 3.53 - 3.43 (m, 2H), 3.41 - 3.34 (m, IH), 2.30 - 2.20 (m, IH), 2.06 - 1.95 (m, I H), 1.88 - 1.79 (m, IH), 1.78 - 1 .68 (m, 2H), 1.67 - 1.65 (m, I H), 1 .63 - 1.58 (m, 2H), 1.55 - 1.49 (m, 2H), 1.45 i s. 9H 1.34 - 1 .30 (m, 2H), 0.92 (dd, ./ 6.3, 10,3 Hz, 6H). MS (ESI) m/z (M+H) + 442.3.

TToo aa mmiixxttuurree ooff ((<<SS))--22--aammiinnoo--22--mmeetthhyyllppeenntt--44 --eennooiicc aacciidd ( (111100 mmgg,, 885511..6666 uummooll)) iinn M MeeOOHH ((1100 mmLL)) wwaass sslloowwllyy aaddddeedd SSOOCCil 22 ((330077 mmgg,, 22..5588 mmrrnnooll)) aatt 00 °°CC,, tthhee mmiixxttuurree wwaass wwaarrmmeedd ttoo 2200 °°CC ((rroooomm tteemmppeerraattuurree)) aanndd ssttiinniinngg ffoorr 3300 mmiinn aatt 2200 °°CC.. TThhee ssoolluuttiioonn wwaass tthheenn rreefflluuxxeedd aatt 6666 °°CC ffoorr 2200hhrrss.. TThhee mmiixxttuurree wwaass ddiirreeccttllyy ccoonncceennttrraatteedd uunnddeerr vvaaccuuuumm ttoo ggiivvee ccoommppoouunndd 3344AA ((115522 mmgg,, 9999..3355%% yyiieelldd,, HHCC11 ssaalltt)) aass ccoolloorrlleessss ooiill.. TThhee ccrruuddee wwaass uusseedd ddiirreeccttllyy ffoorr tthhee nneexxtt sstteepp.. 1H1H NNMMRR ((440000MMHHzz,, CCDD 33 OODD)) δδ 55..8811 -- 55..6688 ((mm,, 11 HH)),, 55..3333 -- 55..2255 ((mm,, 22 HH)),, 33..8833 ((ss,, 33 HH)),, 22..7766 -- 22..6633 ((mm,, 11 HH)),, 22..6622 -- 22..5522 ((mm,, 11 HH)),, 11..5566 ((ss,, 33 HH))..

[[0000550055]] SStteepp 22:: SSyynntthheessiiss ooff ccoommppoouunndd 3344BB

CCoommppoouunndd 3344BB wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 3344AA aanndd ((tteerrtt--bbuuttooxxyyccaarrbboonnyyll))-- -- lleeuucciinnee ffoolllloowwiinngg tthhee pprroocceedduurree ooff ccoommppoouunndd 77DD., CCoommppoouunndd 3344BB ((223300 mmgg,, yyiieelldd 7744..7788%%)) wwaass oobbttaaiinneedd aass aann ooffff--wwhhiittee ssoolliidd., 1H1H NNMMRR ((440000MMHHzz,, CCDDCC11 33 )) δδ 66,,7744 ((bbrr.. ss..,, 11 HH)),, 55,,7711 -- 55..5544 ((mm,, 11 HH)),, 55., 1133 -- 55..0055 ((mm,, 22 HH)),, 44..8822 ((bbrr.. ss..,, 11 HH)),, 44..1111 -- 33..9988 ((mm,, 11 HH)),, 33..7788 -- 33..7700 ((mm,, 33 HH)),, 22..9944 -- 22..8833 ((mm,, 11 HH)),, 22..6622 -- 22..5533 ((mm,, 11 HH)),, 11..7744 -- 11..6600 ((mm,, 33 HH)),, 11..5511 -- 11..3377 ((mm,, 1122 HH)),, 00..9933 ((dddd,, JJ== 44..55,, 66..11 HHzz,, 66 HH))..

CCoommppoouunndd 3344CC wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 3344BB ffoolllloowwiinngg tthhee pprroocceedduurree ooff ccoommppoouunndd 1122AA.. CCoommppoouunndd 3344CC ((118888 mmgg,, yyiieelldd 9999..5511%%,, HHCC11 ssaalltt)) wwaass oobbttaaiinneedd aass aa ccoolloorrlleessss ooiill aannss uusseedd ddiirreeccttllyy ffoorr tthhee nneexxtt sstteepp..

CCoommppoouunndd 3344DD wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 3344CC aanndd ( (SS))--22--((((tteerrtt-- bbuuttooxxyyccaarrbboonnyyll))aammiinnoo))nnoonn--88--eennoo iicc aacciidd ffoolllloowwiinngg tthhee pprroocceedduurree ooff ccoommppoouunndd 7711)).. CCoommppoouunndd 3344DD ((116600 mmgg,, yyiieelldd 3377..00%%)) wwaass oobbttaaiinneedd aass aa ccoolloorrlleessss ooiill.. MMSS ((EESSII)) mm//zz ((MM++NNaa)) ++ 553322..11..

Compound 34E was prepared from compound 34D following the procedure of compound 18H. Compound 34E (250 mg, yield 66.1 %) was obtained as a colorless oil. ! H NMR (400MHz, CDC1 3 ) S 6.80 (s, 1H), 6.33 (br d, ./ 8.4 Hz, 1H), 5.43 - 5.32 (m, 1H), 5.10 (br d, ./ 7 7 Hz, 2H), 4.55 - 4.44 (m, 1 H), 4.05 - 3 ,95 (m, 1 H), 3 ,82 - 3.76 (m, 3H), 3.00 - 2.89 (m, 1H), 2,58 - 2,48 (m, 1 H), 2.12 - 1.90 (m, 4H), 1.71 - 1 .59 (m, 6H), 1.55 - 1 ,48 (m, 2H), 1.46 - 1.31 (m, 101 1 ), 1 ,24 - 1.15 (m, 3H), 0.97 - 0.84 (m, 6H). MS (ESI) m z (M I f } ' 482.2. Compound 34F was prepared from compound 34E following the procedure of compound 14F. Compound 34F (160 mg, yield 59.46 %) was obtained as white solid. ! H NMR (400ΜΉζ, CDC1 3 ) δ 6,54 - 6.42 (m, IH), 6,30 - 6.19 (m, H), 5.47 - 5 ,31 (m, 2H), 5.04 (br d, ./ 7 7 Hz, 1H), 4.53 - 4.42 (m, ! ! ! ), 4.12 - 4,02 (m, I I I ). 3 ,76 - 3.66 (m, 1H), 3.63 - 3.54 (m, 1H), 2,55 - 2,43 (m, 1 1 1 ).. 2.16 - 2.05 (m, 4H), 1.83 - 1.72 (m, IH), 1.56 - 1 ,40 (m, 1 ! f ), 1.31 - 1.19 (m, 7H), 0.95 - 0,85 (m, 6H). MS (ESI) m/z (M+H) + 454,2.

[00510] Step 7: Synthesis of compound 34G

To a solution of compound 34F (90 mg, 198.41 umol) in MeOH (20 mL) was added Pd-C (10%, 20 mg) under N 2 . The suspension was degassed under vacuum and purged with H? several times. The mixture was stirred under H 2 (15 psi) at 20°C for 16 hours. The reaction mixture was filtered through Celite pad and the filtrate was concentrated to give compound 34G (72 mg, 77.23% yield) as white solid. The crude product was used directly for the next step. MS (ESI) m/z (M I I ) 456,3.

Compound 34 was prepared from compound 34G following the procedure of compound 7. Compound 34 (41 mg, yield 65.41 %) was obtained as white solid. Ή NMR (400MHz, CDCI 3 ) .· )' 9. 18 (s, 1 1 1 ), 6,72 (s, IH), 6.32 (br d, ./ 7 3 Hz, IH), 5, 17 (br d, ./ 7.9 Hz, I H), 4.56 - 4.45 (m, I H), 4.07 (br s, IH), 2.48 - 2,38 (m, IH), 1.90 (br s, IH), 1.73 - 1 ,65 (m, 2H), 1 ,55 (br d, J=7.9 Hz, 2H), 1.50 (s, 3H), 1.43 (s, 9H), 1.23 (br s, 1 1H), 0.98 (br s, 2H), 0.94 (br d, J=6.2 Hz, 6H). MS (ESI) m/z ( +H) + 454,3.

Terr-butyl {(3 6 15S)-15-formyl-3-isobutyl-l 5-methyl-2,5-dioxo-l ,4- '■acyclopentadec-12-en-6-yl

Compound 35 was prepared from compound 34F following the procedure compound 7. Compound 35 (55 mg, yield 85.96 %) was obtained as white solid. Ή NF (400MHz, DMSO- ) δ 9.37 - 9.23 (m, H), 8.03 (br d, ,/ 9.5 Hz, 1 H), 7.63 (s, 1H), 7.16 - 7.03 (m, IH), 5.52 - 5.39 (m, 1H), 5.23 (td, J-7.6, 14.9 Hz, H i), 4,38 - 4.24 (m, H i ), 4.01 - 3.89 (m, I I I ), 2.47 - 2.40 (m, 1 1 1), 2.20 (br dd, ./ 8.4.. 13,9 Hz, H i), 1.98 (br d, . 4.4 Hz, 2H), 1.90 - 1.78 (m, IH), 1.65 (br dd, J=6.2, 10.6 Hz, IH), 1.51 (br s, IH), 1.47 - 1.41 (m, 2H), 1.41 - 1.33 (m, 12H), 1.26 - 1.17 (m, 3H), 1.14 (s, 4H), 0.88 - 0.76 (m, 6H). MS (ESI)w/z (M+H) + 452.3.

[00513] Step 1; Synthesis of compound 36A

To a solution of compound 34E (310.0 mg, 643.66 umol) in MeOH (15 mL) was added Pd-C (10%, 30 mg) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H 2 (15 psi) at 20 °C for 45 hours. The reaction mixture was filtered through Celite pad and the filtrate was concentrated. Compound 36A (300.0 mg, yield: 96.4%) was obtained as colorless oil. The crude product was used directly for the next step. TTTTTooooo aaaaa mmmmmiiiiixxxxxtttttuuuuurrrrreeeee ooooofffff cccccooooommmmmpppppooooouuuuunnnnnddddd 3333366666AAAAA (((((333330000000000.....00000 mmmmmggggg,,,,, 666662222200000.....3333300000 uuuuummmmmooooolllll))))) iiiiinnnnn E EEEEtttttOOOOOAAAAAccccc (((((55555 mmmmmLLLLL))))) wwwwwaaaaasssss aaaaaddddddddddeeeeeddddd HHHHHCCCCClllll/////EEEEEtttttOOOOOAAAAAccccc (((((44444MMMMM,,,,, 77777.....0000000000 mmmmmLLLLL,,,,, 4444455555.....1111144444 eeeeeqqqqq))))) aaaaannnnnddddd ttttthhhhheeeee rrrrreeeeeaaaaaccccctttttiiiiiooooonnnnn wwwwwaaaaasssss ssssstttttiiiiirrrrrrrrrreeeeeddddd aaaaattttt 2222200000 °°°°°CCCCC fffffooooorrrrr IIIIIhhhhh..... TTTTThhhhheeeee rrrrreeeeeaaaaaccccctttttiiiiiooooonnnnn mmmmmiiiiixxxxxtttttuuuuurrrrreeeee wwwwwaaaaasssss dddddiiiiirrrrreeeeeccccctttttlllllyyyyy cccccooooonnnnnccccceeeeennnnntttttrrrrraaaaattttteeeeeddddd ..... CCCCCooooommmmmpppppooooouuuuunnnnnddddd 3333366666BBBBB ((( ((222226666600000.....00000 mmmmmggggg,,,,, yyyyyiiiiieeeeelllllddddd::::: 7777799999.....88888%%%%%,,,,, HHHHHCCCCClllll))))) wwwwwaaaaasssss ooooobbbbbtttttaaaaaiiiiinnnnneeeeeddddd aaaaasssss cccccooooolllllooooorrrrrllllleeeeessssssssss oooooiiiiilllll aaaaannnnnddddd uuuuussssseeeeeddddd aaaaasssss sssssuuuuuccccchhhhh wwwwwiiiiittttthhhhhooooouuuuuttttt pppppuuuuurrrrriiiiififififificccccaaaaatttttiiiiiooooonnnnn .....

[[[[[0000000000555551111155555]]]]]

TToo aa mmiixxttuurree ooff ccoommppoouunndd 3366BB ((226600..00 mmgg,, 66 1199..0066 uummooll,, HHCCll)) iinn ddiiooxxaannee ((44 mmLL)) aanndd HH 22 00 ((44 mmLL)) wwaass aaddddeedd KK 22 CC00 33 ((221166..00 mmgg,, 11..5566 mmmmooll)) aanndd CCbbzzOOSSuu ((221155..9999 mmgg,, 886666..6688 uummooll)),, aanndd tthheenn tthhee mmiixxttuurree wwaass ssttiirrrreedd aatt 2255 °°CC ffoorr 22 hhoouurrss uunnddeerr NN 22 aattmmoosspphheerree.. TThhee rreeaaccttiioonn mmiixxttuurree wwaass qquueenncchheedd wwiitthh HH 22 00 ((2200 mmLL)) aanndd eexxttrraacctteedd wwiitthh EEttOOAAcc ((3300 mmLL xx 33)).. TThhee oorrggaanniicc llaayyeerr wwaass wwaasshheedd wwiitthh bbrriinnee ((2200 mmLL)),, ddrriieedd oovveerr N Naa 22 SS00 44 .. TThhee ssoolliidd wwaass rreemmoovveedd bbyy fifillttrraattiioonn aanndd tthhee fifillttrraattee wwaass ccoonncceennttrraatteedd ttoo ggiivvee tthhee rreessiidduuee.. TThhee rreessiidduuee wwaass ppuurriififieedd bbyy ffllaasshh ssiilliiccaa ggeell cchhrroommaattooggrraapphhyy ((EEiiuueenntt ooff 00--2200%% EEAA//''PPEE)).. CCoommppoouunndd 3366CC ((225500 mmgg,, yyiieelldd:: 7788..00%%)) wwaass oobbttaaiinneedd aass wwhhiittee ssoolliidd..

[[0000551166]] SStteepp 44:: SSyynntthheessiiss ooff ccoommppoouunndd 3366DD

CCoommppoouunndd 3366DD wwaass pprreeppaarreedd ffrroomm ccoommppoouunndd 3366CC ffoolllloowwiinngg tthhee pprroocceedduurree ooff c coommppoouunndd 1144FF.. CCoommppoouunndd 336611)) ((110055 mmgg,, yyiieelldd 3377..77%%)) wwaass oobbttaaiinneedd aass aa wwhhiittee ssoolliidd.. 1H1H NNMMRR. ((440000MMHHzz,, DDMMSSOO--aafefe)) δδ 88,,0088 ((bbrr dd,, ,,// 99..00 HHzz,, IIHH)),, 77..4400 -- 77..2266 ((mm,, 55HH)),, 77..2222 ((bbrr dd,, .. 77..77 HHzz,, IIHH)),, 77..1177 ((ss,, II HH)),, 55.. 11 11 -- 44,.8899 ((mm,, 22HH)),, 44..7700 ((bbrr tt,, ..// 55,,88 HHzz,, IIHH)),, 44..4433 -- 44..2277 ((mm,, IIHH)),, 44..11.22 -- 44..0022 ((mm,, IIHH)),, 33..3333 ((bbrr ss,, IIHH)),, 33..1199 ((bbrr dddd,, JJ--66..55,, 1100..77 HHzz,, IIHH)),, 22..0033 -- 11 ..8822 ((mm,, I IHH)),, 11..7700 -- 11..4444 ((mm,, 33HH)),, 11..4433 -- 11..0088 ((mm,, 1188HH)),, 00..9944 ···· 00..7700 ((mm,, 66HH)).. MMSS ((EESSII)) mm//zz {{ WW WW )) 449900..22..

Compound 36 was prepared from compound 36D following the procedure of compound 7. Compound 36 (31 mg, yield 33.3 %) was obtained as a white solid. ! H NMR (400MHz, DMSQ-i¾) δ 9.30 (br s, H), 8.06 (br d, J=9.0 Hz, I H), 7.80 (br s, IH), 7.58 (br d, ./ 7.5 Hz, IH), 7,34 (br s, 5H), 5, 13 - 4,91 (m, 2H), 4.34 (br s, I H), 4.02 (br d, J 6 4 Hz, IH), 1.82 - 1 ,40 (m, 4H), 1 .39 - 1 .08 (m, 1 81 ! }. 0,89 - 0.76 (m, 6H). MS (ESI) m/z ( M i l ) 488.3

EXAMPLE 22

diazabicyclo 3, 0 hexadeo carbamate 1

To a mixture of t-BuOK (6,9 g, 61.72 mmol) in THF (45 mL) at -78°C was added ethyl 2-((diphenylmethylene)amino)acetate (15 g, 56.11 mmol) in THF (30 mL). The reaction mixture was warmed to 0 °C, and strired at this temperature for 45 min. The mixture was then cooled back to -78 °C, for the addition of (£)-l,4-dibromobut-2-ene (12 g, 56.1 1 mmol) in THF (30 mL) and then strired at 0 °C for 45 min and cooled back to -78 °C, for the addition of t-BuOK (6.9 g, 61.72 mmol) in THF (45 mL). The reaction mixture was finally strired for 2 h at 0 °C. The mixture was removed under reduced pressure to give compound 37A (15 g, crude), which was used directly for the next step without further purification. 1H NNMMRR ((440000MMHHzz,, CCDDCC11 33 )) δδ 77..8822 ((dd,, ..// 77..22 HHzz,, 1IHH)),, 77,,6644 -- 77..5588 ((mm,, 1I HH)),, 77.,5500 ((tt,, JJ == 77..00 HHzz,, I 1H1 1)),, 77,,4422 -- 77..2299 ((mm,, 55HH)),, 77..1166 -- 77..11 11 ((mm,, 22HH)),, 55..9933 -- 55..8811 ((mm,, I IHH)),, 55..0066 -- 44,,9966 ((mm,, I I HI S)),, 44,,2299 ((dd.. JJ 77,,33 HHzz,, IIHH)),, 33 ,,7788 -- 33..7733 ((mm,, 22HH)),, 11..4499 -- 11..4400 O (mn.,. I 1H1 1)),,. 11..3377 -- 11..2211 ((mm,, 33HH)),, 11..1133 iitt.. ..// 77..22 HHzz,, 22HH))..

[[0000551199]] SStteepp 22:: SSyynntthheessiiss ooff ccoommppoouunndd 3377BB

CCoommppoouunndd 3377AA ((1155 gg,, 4466..9966 mmmmooll)) wwaass ttaakkeenn uupp iinn M MTTBBEE ((115500 mmLL)) aanndd ttrreeaatteedd wwiitthh HHCCll ((11 MM,, 110000 mmLL)).. TThheenn tthhee mmiixxttuurree wwaass ssttiirrrreedd a att 1155 °°CC ffoorr 33..55 hhrrss.. TThhee llaayyeerrss wweerree sseeppaarraatteedd aanndd tthhee aaqquueeoouuss llaayyeerr wwaass eexxttrraacctteedd wwiitthh MMTTBBEE ((22 xx 110000 mmLL)),, tthhee aaqquueeoouuss llaayyeerr wwaass bbaassiiffiieedd ttoo ppHH ~~ 88--99 wwiitthh ssaatt NNaaHHCC00 33 ssoolluuttiioonn.. TThhee ddeessiirreedd aammiinnee wwaass eexxttrraacctteedd wwiitthh MMTTBBEE ((33 xx 110000 mmLL)) aanndd tthhee oorrggaaiinniicc pphhaassee wwaass wwaasshheedd wwiitthh bbrriinnee (( 0000 mmLL)),, ddrriieedd oovveerr aannhhyyddrroouuss NNaa>>SS(()) ..,, fifilltteerreedd aanndd ccoonncceennttrraatteedd.. TThhee rreessiidduuee wwaass ttrreeaatteedd wwiitthh 44NN HHCCll//ddiiooxxaannee ((4400 mmLL)).. TThheenn tthhee mmiixxttuurree wwaass ccoonncceennttrraatteedd uunnddeerr rreedduucceedd pprreessssuurree ttoo ggiivvee ccoommppoouunndd 3377BB ((33,,66 gg,, ccrruuddee,, HHCCll ssaalltt)),. ΊΊ \\ NNMMRR ((440000MMHHzz,, DDMMSSOO--ddfefe)) δδ 99,.0099 ((bbrr ss,, 33HH)),, 55,,6677 -- 55..5566 ((mm,, IIHH)),, 55.,3355 ((dd,, JJ== 1177..22 HHzz,, IIHH)),, 55..1188 ((dd,, JJ == 1100..11 HHzz,, I IHH)),, 44..2255 -- 44..1111 ((mm,, 22HH)),, 22..4477 -- 22..4422 ((mm,, I IHH)),, 11..8822 ((dddd,, JJ== 66..00,, 99..99 HHzz,, IIHH)),, 11..6633 ((dddd,, JJ == 66,, 11,, 88,,00 HHzz,, IIHH)),, 11..2211 ((tt,, JJ== 77..11 HHzz,, 33HH))..

To a mixture of compound 37B (4.8 g, 25.05 mmol, HCl), Boc 2 0 (5,7 g, 26.30 mmol, 6 mL), DMAP (612 mg, 5.01 mmol) in THF (50 mL) was added DffiA (9.7 g, 75.15 mmol, 13 mL) at 0 °C. The mixture was stirred at 15 °C for 24 hrs. The mixture was concentrated in vacuum. The residue was treated with EtOAc (100 mL), washed with H 2 0 (50 mL). The organic layer was separated and the aqueous layer was extracted with EA (2 x 30 mL). The combined organic layers was washed with 0.5 N HCl (2 x 30 mL), saturated NaHC0 3 (2 x 30 mL), brine (50 mL), dried over anhydrous MgS0 4 , filtered and concentrated. The residue was purified by column chromatography (SiC% Petroleum ether: Ethyl acetate = 5: 1) to give compound 37C (2.3 g, yield: 35%). 1H NMR (400MHz, DMSO-< ) δ 7.63 (s, IH), 5.68 - 5.55 (m, IH), 5.21 (br d, J = 17.2 Hz, IH), 5.04 (dd, J = 1.9, 10.3 Hz, IH), 4.09 - 3.96 (m, 2H), 2.08 (q, J = 8.7 Hz, IH), 1.54 (br dd, J = 5.3, 7.3 Hz, IH), 1.40 - 1.30 (m, 9H), 1.27 - 1.23 (m, IH), 1.19 - 1.1 1 (m, 3H), MS (ESI) m/z (M-100) + 155.8.

[00521] Step 4: Synthesis of compounds 37D and 37E

The compound 37C (1 g) was resolved by SFC to give two isomers. SSFFCC mmeetthhoodd:: CCoolluummnn:: AAYY((225500mmmm**3300mmmm,,1100uumm)) M Moobbiillee pphhaassee:: AA:: CC00 22 Β R:.0O,Γ l%%\Η! F 3 ΗIF 2 ΟO EETTOOHFI;, GGrraaddiieenntt:: 1155%% ooff BB;; FFllooww rraattee:: 6600mmLL//mmiinn..

TThhee ccoommppoouunndd 3377CC wwaass sseeppaarraatteedd bbyy SSFFCC ttoo ggiivvee ccoommppoouunndd 3377DD ((550000 mmgg,, yyiieelldd:: 4499..99%%)) ((RRtt == 11..4499 mmiinn)) aanndd ccoommppoouunndd 3377EE ((445500 mmgg,, yyiieelldd:: 2255,,99%%)) ((RRtt == 22..6688 ramiinn)) bbootthh aass yyeellllooww ooiill.. ll HH N NMMRR ((440000MMHFizz,, DDMMSSOO--ii¾¾)) δδ 77,,6644 ((ss,, IIHH)),, 55,,6677 -- 55,,5544 ((mm,, IIHH)),, 55..2211 ((bbrr dd,, J 1177..22 F 1I1/z,, IIHH)),, 55..0077 -- 55..0011 ((mm,, IHII)),, 44..1100 -- 33,,9944 ((mm,, 22HH)),, 22..0088 ((qq,, JJ== 88..77 H FIzz,, IIHH)),, 11.,5577 -- 11.,4444 ((mm,, IIHFF)),, 11..3388 -~ 11..3311 ((mm,, 99HFI)),, 11,,2277 -- 11..2233 ((mm,, IIHFF)),, 11..1188-- 11..1100 ((mm,, 33HFI)).. MMSS ((EESSII)) mmzz ((MM-- 110000)) ' 115555..88..

1H1H NMMRR ((440000MMHHzz,, DDMMSSGG-- δδ 77..6644 ((ss,, IIHH)),, 55..6677 -- 55..5544 ((mm,, IIHH)),, 55..2211 ((bbrr dd,, JJ == 1177..00 HHzz,, IIHH)),, 55..0044 ((dddd,, JJ== 11..77,, 1100..33 HHzz,, IIHH)),, 44..11 -- 33..9955 ((mm,, 22HH)),, 22..0088 ((qq,, JJ== 88..88 HHzz,, IIHH)),, 11..5544 ((bbrr dddd,, JJ 55..44,, 77..22 HHzz,, I 1H11)),.11..3388 -- 11..3311 ((mm,, 99HH)),, 11..2277 -- 11..2233 ((mm,, I 1H11)),.11..1199 -- 11..1100 ((mm,, 33HH)).. MMSS ((EESSII)) mm//zz ( (MM--110000)) +' 115555,,99....

Compound 37F was prepared from compound 37E following the procedure of compound 12A. Compound 37F (380 mg, crude, HC1 salt) was used directly for the next step. 1H NMR (400MHz, D SO-fifc) '> 9,12 (br s, 2H), 5.62 (qd, ./ 8,7, 10.1 FIz, 111), 5.34 (dd, ./ 1.3, 17,2 FIz, IH), 5,17 (dd, ./ 1,3, 10.4 Hz, IH), 4.25 - 4.10 (m, 2FI), 2.47 - 2.42 (m, IH), 1.83 (dd, ./ 6.0, 9.9 Hz, IH), 1.64 ·· 1.60 (m, IH), 1.23 - 1.13 (m, 31!)

[00523] Step 6: Synthesis of compound 37G

Compound 37G was prepared from compound 37F and (/er/-butoxycarbonyl)-L~ leucine following the procedure of compound 7D. Compound 37G (800 mg, yield 50.5 %) was obtained as an off-white solid and used directly for next step without purification. ! H NMR (400MHz, DMSC g) δ 8.63 (s, IH), 6.74 (br d, 8.4 Hz, H), 5.56 (br d, J= 9.5 Hz, IH), 5.27 (br dd, ,/ 1.4, 17.1 Hz, IH), 5.11 - 5,03 (m, IH), 4.00 (td, ./ 7.0, 13,8 Hz, 21!}. 3.92 - 3.84 (m, IH), 2,13 (br d, 8.4 Hz, IH), 1.67 - 1.56 (m, 2H), 1.45 - 1.28 (m, ill!}. 1.20 (br dd, J = 5,2, 9,4 Hz, IH), 1.14 - 1.09 (m, 3H), 0.85 (t, . 7.3 FIz, 6FI). MS (ESI) m/z (\! Na) ' 391.1.

[00524] Step 7: Synthesis of compound 37H

Compound 3711 was prepared from compound 37G following the procedure of compound 14A. Compound 37H (700 mg, crude, HC1 salt) was obtained as a white solid and used directly for the next step. 1H NMR (400MHz, DMSO-t ) δ 9.41 (s, IH), 8.41 - 8.21 (s, 3H), 5.65 - 5.51 (m, I H), 5,29 (dd, 1 .5, 17.2 Hz, I I I ). 5.1 1 (dd, ./ 1.5, 10.4 Hz, I I I ). 4. 1 1 - 3.94 (m, 2H), 3.65 - 3.61 (rn, I H), 2.24 (q, J = 8.4 Hz, IH), 1.76 - 1 .52 (m, 4H), 1.40 - 1.35 (rn, I H), 1.14 - 1.10 ( rn. 3H), 0.94 - 0,87 (rn, (:»! ! }.

[00525] Step 8: Synthesis of compound 37J

Compound 37,1 was prepared from compound 37H and (5)~2~((tert- butoxycarbonyl)amino)non-8-enoic acid following the procedure of compound 7D. Compound 37J (600 mg, yield 44.8 %) was obtained as an off-white solid. ! H NMR (400MHz, CDCI3) δ 6.95 (br s, I H), 6.45 (d, J = 8.4 Hz, I H), 5.84 - 5.72 (m, 2H), 5.31 (dd, J = 1.1 , 17.2 Hz, I H), 5.13 (dd, ./ 1.4, 10.5 Hz, IH), 5 ,02 - 4.92 (m, 2H), 4.46 - 4,42 (m, I H), 4, 19 - 4.07 (m, 2H), 4,01 - 3.95 (m, IH), 2.16 - 2.09 (m, IH), 2,06 - 2.01 (m, 2H), 1.85 - 1.72 (m, 4H), 1.66 - 1.52 (m, 3H), 1.46 - 1.44 (m, 9H), 1.42 - 1.31 (m, 6H), 1.23 ( 1, ./ 7.1 Hz, 3H), 0.96 - 0.93 (m, 6H). MS (ESI) m/z { W W ) 522.3.

[00526] Step 9: Synthesis of compound 37K

Compound 37K was prepared from compound 37 J following the procedure of compound 14E, Compound 37K (100 mg, yield 47,24 %) was obtained as a white flocculus, Ή NMR (400MHz, CDCI 3 ) δ 7.24 (br s, IH), 6.30 (br d, ../ 7.9 Hz, I H), 5.68 - 5.59 (m, IH), 5.28 (dd, J = 7.6, 10.5 Hz, IH), 5.15 (br d, J = 8.2 Hz, IH), 4.59 - 4.43 (m, IH), 4.16 (q, 7.2 Hz, 2H), 4.09 - 3.95 (m, IH), 2,43 (br d, J ------- 8,6 Hz, I H), 2.03 - 1.76 (m, 6H), 1.71 -

1.65 (m, 2H), 1.52 - 1.42 (m, 1 1H), 1.37 - 1 .21 (m, 8H), 0.91 (dd, ./ 6,4, 18.1 Hz, 6H). MS (ESI) m/z (M+Na) + 516.3 ,

[00527] Step 10: Synthesis of compound 37L

Compound 37L was prepared from compound 37K following the procedure of compound 14F, Compound 37L (40 mg, yield 43 ,7 %) was obtained as an off-white solid, 1H NMR (400MHz, CDCI 3 ) δ 6.83 (br s, IH), 5.82 ·· 5.58 (m, IH), 5.49 ·· 5.46 (m, IH), 5.20 - 5.00 (m, IH), 4.40 (br s, IH), 4.10 (br s, IH), 3.69 - 3.65 (m, IH), 3.54 - 3.50 (m, IH), 2.21 (br s, I H), 2.00 (br s, IH), 1.88 - 1.50 (m, 7H), 1.41 (s, 9H), 1.35 - 1.25 (m, 6H), 0.99 (s, IH), 0.95 - 0.84 (m, 6H). MS (ESI) m/z (M+H) + 452.3.

[00528] Step 11: Synthesis of compound 37

Compound 37 was prepared from compound 37L following the procedure of compound 7. Compound 37 (50 mg, yield 50 %) was obtained as an off-white solid. 1H NMR (400MHz, CDC1 3 ) δ 8.88 (s, I H), 7.48 (br s, I H), 6.46 - 6.36 (m, I H), 5.61 - 5.59 (rn. II I), 5.41 - 5.23 (m, 2H), 4,59 - 4.43 (m, IH), 4.10 - 3 ,98 (m, I H), 2.52 - 2.48 (m, I H), 2.32 - 2.06 (rn, I H), 2.01 - 1.91 (rn, H I ), 1.80 - 1 .58 (m, 8H), 1.43 (s, 9H), 1.29 - 1 .27 (m, 5H), 0,95 - 0.88 (m, 6H). MS (ESI) m/z (M i l ) ' 450, 3.

[00529] Biochemical inhibition of Calpains 1, 2, and 9

[00530] Caipain 1 , 2, and 9 activity and inhibition thereof are assessed by means of a continuous fluorescence assay. The SensoLyte 520 Caipain substrate (Anaspec Inc) is optimized for detecting caipain activity. This substrate contains a novel internally quenched 5-FAM/QXLTM 520 FRET pair. Calpains 1 ,2 and 9 cleave the FRET substrate into two separate fragments resulting in an increase of 5 -F AM fluorescence that is proportional to caipain activity.

[00531] Assays are typically setup in black 384-weli plates using automated liquid handling as follows. Caipain assay base buffer typically contains 50mM: Tri s, pH 7.5, lOOmM NaCl and ImM DTT. Inhibitors are serially diluted in DMSO and used to setup 2x mixtures with calpains in the aforementioned buffer. After incubation at ambient temperature (25C), the reaction is initiated by adding a 2x mix of the fluorescent peptide substrate and CaCl? (required for in-situ caipain activation) in the same buffer. Reaction progress curve data are typically collected for lOmin using excitation/emission wavelengths of 490 nm/520 nm on SpectraMax i3x or the FLIPR-Tetra plate readers (Molecular Devices Inc). Reaction rates were calculated from progress curve slopes typically over l -5min. Dose response curves (rate vs. log inhibitor concentration) were typically fit to a 4-parameter logistic function to extract IC 50 values.

Inhibition of cellular caipain activity

[00532] Caipain activity in SH-SY5Y cells and inhibition thereof are assessed by means of the Caipain-Glo™ platform (Promega, Inc) which is a homogeneous, luminescence assay that uses the cell-permeable and pro-luminescent caipain substrate Suc-LLVY-amino-luciferin. Upon caipain cleavage followed by cell lysis and quenching the luminescence signal developed is proportional to intra-cellular caipain activity.

[00533] Assays are typically setup by seeding SH-SY5Y cells in white 384- well plates at 40k/per well in RPMI-1640 containing 1% serum followed by 37C overnight incubation. Next morning, ceils are pre-incubated for Ihr with serially diluted compounds followed by addition of 20uM each of Suc-LLVY-aminoluciferin substrate and A23187 (ionophore used to induce Ca flux and calpain activity) diluted in Calpain-Glo buffer. After a 4hr incubation at 37C (calpain reaction), cells are lysed at 37C for ihr using 0.9% Triton X-100 containing PBS with lOOuM MDL-28170 (excess calpain inhibitor to quench calpain activity). After centrifugation at 300rpm, the Calpain-Glo™ luciferase detection reagent in Calpain-Glo™ buffer is added followed by lOmin incubation prior to reading luminescence counts using an En Vision plate reader (Perkin Elmer Inc). Dose response curves (luminescence vs. log inhibitor concentration) were typically fit to a 4-parameter logistic function to extract IC 50 values.

[00534] Calpain Inhibition

[00535] Column A: Human Calpain 1/NS l IC50 (nM) MEAN

[00536] Column B: Human Calpain 2/NS1 IC50 (nM)_MEAN

[00537] Column C: Human Calpain 9/N S I IC50 (nM)_MEAN

A: < 3 uM;

B: 3-10 uM;

C: > 10 uM;

IJ99

300

302 Bleomycin-induced pulmonary fibrosis in mice or rats

[00538] The method for inducing pulmonary fibrosis in mice is described in Current Protocols in Pharmacology: 5.46.1, entitled "Mouse Models of Bleomycin-induced

Pulmonary Fibrosis ' ". In order to induce pulmonary fibrosis, 6-8 week old C57B1/6 mice or Wistar rats are instilled once oropharyngeaily with -1.5 U/kg of bleomycin sulfate

(Calbiochem, Billerica, MA). Briefly, for oropharyngeal administration of bleomycin, mice or rats are anesthetized with isofluorane and then suspended on its back at a -60 degree angle on an inclined surface with a rubber band running under the upper incisors. The airway is opened while securing the tongue with one arm of padded forceps and bleomycin is administered into the back of the oral cavity with a syringe. The study is terminated on day 14-28 for orophaiyngeaily administered bleomycin in mice and rats.

[00539] Alternatively, for systemic bleomycin administration by osmotic pumps in mice, the pumps are loaded with bleomycin and implanted subcutaneously under isofluorane anesthesia as described in Lee, Am J Physiol Lung Cell Mol Physiol, 2014. Briefly, mice are systemic-ally administered -50 U/kg bleomycin (Blenoxane; Teva Phanna, North Wales, PA) via osmotic pumps for 7 days. On day 10, the osmotic pumps are removed, and the study is continued until day 35.

[00540] All animals are euthanized at the termination of the studies by cervical dislocation for gross necropsy, and blood collected by cardiac puncture. The lungs from each animal are dissected from the animal and weighed. The BAL cells and fluid are collected by lavaging the lung twice with 0.5 ml Hanks Balanced Salt Solution (HBSS; VWR, Radnor, PA). After collection of BAL cells and fluid, lungs are dissected and removed from each animal. Whole lungs are inflated with 10% NBF and then fixed in 10% NBF for histology. Severity of fibrosis in the lungs is evaluated using a modified Ashcroft score (Hubner, Biotechniques, 2008).

Carbon tetrachlori de-induced liver fibrosis in mice or rats

[00541] Carbon tetrachlori de-induced liver fibrosis is a widely used and acceped model for evaluating novel antifibrotic therapies. The methods for inducing liver fibrosis by carbon tetrachloride administration is described in Lee, J Clin Invest. 1995 and Tsukamoto, Semin Liver Dis, 1990. Briefly, male C57BL/6 mice are challenged with Img/kg carbon

tetrachloride (Sigma Aidrich, diluted 1 :7 in corn or olive oil) administered by intraperitoneal injection twice weekly for a period of 4 weeks. Mice are euthanized on day 28. In an alternative implementation, Wistar rats are administered carbon tetrachloride by intraperitoneal injection three times per week for 8-12 weeks. Rats are euthanized at the termination of the experiment, 8-12 after study initiation.

[00542] Blood is collected by cardiac puncture and processed into serum for evaluation of liver enzymes (including ALT, AST, ALP, etc) at several timepoints throughout the study and at termination of the study. The liver tissues from all animals are collected and fixed by immersion in 10% neutral buffered formalin, processed, paraffin embedded, sectioned, mounted, and stained with Masson's Trichrome (Tri) or Picrosirius Red (PSR) using standard histological methods for evaluation of fibrosis severity.

Mouse Unilateral Ureteral Obstruction Kidney Fibrosis Model

[00543] Female C57BL/6 mice (Harlan, 4-6 weeks of age) will be given free access to food and water and allowed to acclimate for at least 7 days prior to test initiation. After

acclimation, mice are anesthetized and undergo unilateral ureteral obstruction (UUO) surgery or sham to left kidney. Briefly, a longitudinal, upper left incision is performed to expose the left kidney. The renal arten,' is located and 6/0 silk thread is passed between the artery and the ureter. The thread is looped around the ureter and knotted 3 times insuring full ligation of ureter. The kidney is returned to abdomen, the abdominal muscle is sutured and the skin is stapled closed. All animals are euthanized 4, 8, 14, 21 , or 28 days after UUO surgery.

Following sacrifice blood is collected via cardiac puncture, the kidneys are harvested and one half of the kidney is frozen at -80 0 C. and the other half is fixed in 10% neutral buffered formalin for histopathological assessment of kidney fibrosis.

Bleomycin Dermal Fibrosis Model

[00544] Bleomycin (Calbiochem, Billerica MA) is dissolved in phosphate buffered saline (PBS) at 10 ug/ml, and sterilized by filtration. Bleomycin or PBS control (100 ul) is injected subcutaneously into two locations on the shaved back of C57/BL6 or S129 mice (Charles River/Harlan Labs, 20-25 g) once daily for 28 days while under isoflourane anesthesia (5% in 100% 02). After 28 days, mice are euthanized and 6 mm -full thickness punch biopsies are obtained from each injection site. Dermal fibrosis is assessed by standard histopathology and hydroxy-proline biochemical assays.

EXAMPLE 25: Targeting Calpains

Targeting calpains as a novel strategy towards inhibition of EpMT [00545] Calpains are calcium dependent non-lysosomal cysteine proteases that selectively cleavage their target substrates, often leaving behind a functional domain and can be a form of post-translational modification. There are currently 14 known genes encoding for the large calpain subunit in humans: CAPN1 (NCBI Gene ID 823), CAPN2(NCBI Gene ID 824), CAPN3(NCBI Gene ID 825), CAPN5(NCBI Gene ID 726), CAPN6 (NCBI Gene ID 827), CAPN7 (NCBI Gene ID 23473), CAPN8(NCBI Gene ID 388743), CAPN9(NCBI Gene ID 10753), CAPN10(NCBI Gene ID 11 132), CAPN1 1(NCBI Gene ID 11131), CAPN12(NCBI Gene ID 147968), CAPN13(NCBI Gene ID 92251), CAPN14 (NCBI Gene ID 440854), CAPN 15(NCBI Gene ID 6650) and 2 known genes for the small regulator}' subunits

CAPN4/CAPNS1(NCBI Gene ID 826) and CAPNS2 (NCBI Gene ID 84290) (Goll 2003; Schad 2002) . The majority of research has focused on the ubiquitously expressed CAPN1 (aka mu-calpain) and CAPN2 (aka m-cafpain), which require micro or miliimolar levels of calcium for their activation, respectively. These isoforms, along with CAPN9, have been shown to form a heterodimer with the small regulatory subunit (CAPNS1 aka CAPN4) (Ravulapalli 2009). Calpains have been implicated in numerous cellular processes, including cytoskeletal rearrangement (Dourdin 2001), migration (Leloup 2006), signaling (Janossy 2004), and differentiation (Santos 2012).

[00546] For assessment of in vitro EMT, NMuMG cells (ATCC) were grown to confluence in 10% serum (Fetal Bovine Serum) growth media (Dubecco's Modified Eagles Medium supplemented with lOug/mL insulin) and then were followed by 24h starvation in 0.5% serum media +/- drug inhibitors. Cells were then treated with recombinant human TGFb l (R&D Systems 5ng/mL) +/- drug inhibitors in 0.5% serum media. For time points greater than 24h, the aforementioned media was refreshed every 24 hours. Cell lysates were analyzed for aSMA protein expression by western blot.

[00547] Miettinen et al. (1994). "TGF-beta induced transdifferentiation of mammary epithelial cells to mesenchymal cells: involvement of type I receptors." J Cell Biol 127(6 Pt 2):2021-36

[00548] Lamouille et ai. (2014). "Molecular mechanisms of epithelial-mesenchymal transition." Nat Rev Mol Ceil Biol 15(3): 178-96

[00549] For assessment of in vitro FMT, Normal Human Lung Fibroblasts (NHLF) cells (Lonza) were grown in Fibroblast Growth Media-2 (Lonza CC-3131/with CC-4126 bullet kit) and then were followed by 24h starvation in serum/growth factor free Fibroblast Basal Media- 2 (Lonza CC-3131) +/- drug inhibitors. Cells were then treated with TGFbl (5ng raL) Fibroblast Basal Media +/- drug inhibitors. Cell lysates were analyzed for aSMA protein expression by western blot.

[00550] Pegorier et ai. (2010). "Bone Morphogenetic Protein (BM P)- ] and BMP-7 regulate differentially Transforming Growth Factor (TGF)-Bl in normal human lung fibroblasts (NHLF)" Respir Res 1 1 :85

EXAMPLE 26: Human Treatment

[00551] The efficacy of treatment with a compound of a preferred embodiment compared with placebo in patients with idiopathic pulmonary fi brosis (IPF) and the safety of treatment with a compound of a preferred embodiment compared with placebo in patients with IPF is assessed. The primary outcome variable is the absolute change in percent predicted forced vital capacity (FVC) from baseline to Week 52, Other possible end-points would include, but are not limited to: mortality, progression free survival, change in rate of FVC decline, change in Sp02, and change in biomarkers (HRCT image analysis; molecular and cellular markers of disease activity). Secondary outcome measures include: composite outcomes of important !PF-reiated events; progression-free survival, the rate of death from any cause, the rate of death from IPF; categorical assessment of absolute change in percent predicted FVC from baseline to Week 52; change in Shortness-of-Breath from baseline to Week 52, change in percent predicted hemoglobin (Hb)-corrected carbon monoxide diffusing capacity (DLco) of the lungs from baseline to Week 52; change in oxygen saturation during the 6 minute walk test (6MWT) from baseline to Week 52; change in high-resolution computed tomography (HRCT) assessment from baseline to Week 52; change in distance walked in the 6MWT from baseline to Week 52. Patients eligible for this study include, but are not limited to: those patients that satisfy the following inclusion criteria: diagnosis of IPF; 40 to 80 years of age; FVC≥50% predicted value; DLco≥35% predicted value; either FVC or DLco≤ 90% predicted value; no improvement in past year; a ratio of the forced expiratory volume in 1 second (FEVi) to the FVC of 0.80 or more; able to walk 150 meters in 6 minutes and maintain saturation≥83 % while on no more than 6 L/min supplemental oxygen. Patients are excluded from this study if they satisfy any of the following criteria: unable to undergo pulmonary function testing; evidence of significant obstructive lung disease or airway hyper- responsiveness; in the clinical opinion of the investigator, the patient is expected to need and be eligible for a lung transplant within 52 weeks of randomization; active infection; liver disease; cancer or other medical condition likely to result in death within 2 years; diabetes, pregnancy or lactation; substance abuse; personal or family history of long QT syndrome, other IPF treatment; unable to take study medication; withdrawal from other IPF trials.

Patients are orally dosed with either placebo or an amount of a compound of a preferred embodiment (1 mg/day-1000 nig/day). The primary outcome variable will be the absolute change in percent predicted FVC from Baseline to Week 52. Patients will receive blinded study treatment from the time of randomization until the last patient randomized has been treated for 52 weeks. Physical and clinical laboratory assessments will be performed at defined intervals during the treatment duration, for example at weeks 2, 4, 8, 13, 26, 39, and 52. Pulmonary function, exercise tolerance, and shortness-of-breath will be assessed at defined intervals during the treatment duration, for example at weeks 13, 26, 39, and 52. A Data Monitoring Committee (DMC) will periodically review safety and efficacy data to ensure patient safety.

Example Trial in SSc

[00552] The efficacy of treatment with a compound of a preferred embodiment compared with placebo in patients with systemic sclerosis (SSc) and the safety of treatment with a compound of a preferred embodiment compared with placebo in patients with SSc is assessed. The primary outcome variable is the absolute change in Modified Rodnan Skin Score (mRSS) from baseline to Week 48, Other possible end-points would include, but are not limited to: mortality, percentage of patients with treatment-emergent adverse events (AEs) and serious adverse events (SAEs), composite measurement of disease progression, and change in biomarkers (molecular and cellular markers of disease activity, such as C-reactive protein). Secondary outcome measures include, but are not limited to: Scleroderma Health Assessment Questionnaire (SHAQ) score; the Health Assessment Questionnaire Disability Index (HAQ-DI); Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT) score, severity of pruritus as measured by a standardized scale, such as the 5-D Itch Scale; St.

George's Respiratory Questionnaire (SGRQ) score; Tender Joint Count 28 (TCJ28); lung function parameters; standard vital signs (including blood pressure, heart rate, and

temperature); electrocardiogram measurements (ECGs); laborator' tests (clinical chemistry, hematology, and urinalysis); pharmacokinetics (PK) measurements, included in these measurements and in addition, clinical and biomarker samples, such as skin biopsies and blood (or serum and/or plasma), will also be collected prior to initiation of treatment. Additionally, patients eligible for this study include, but are not limited to, those patients that satisfy the following criteria: Patients at least 18 years of age; diagnosis of SSc according to the American College of Rheumatol ogy (ACR) and European League Against Rheumati sm (EULAR) Criteria, meeting criteria for active disease and with a total disease duration of less than or equal to 60 months; 10≤ mRSS≤ 35. Patients are excluded from this study if they satisfy any of the following criteria: major surgery within 8 weeks prior to screening;

scleroderma limited to area distal to the elbows or knees; rheumatic autoimmune disease other than SSc; use of any investigational, biologic, or immunosuppressive therapies, including intra-articular or parenteral corticosteroids within 4 weeks of screening. Patients are orally dosed with either placebo or an amount of a compound of a preferred embodiment (1 mg/day-1000 mg/day). The primary outcome variable will be the absolute change in mRSS from Baseline to Week 48. Patients will receive blinded study treatment from the time of randomization until the last patient randomized has been treated for 48 weeks. Physical and clinical laboratory assessments will be performed at defined intervals during the treatment duration, such as Weeks 2, 4, 8, 12, 24, 36, and 48. Clinical and biomarker samples will also be collected at Week 48. A Data Monitoring Committee (DMC) will periodically review safety and efficacy data to ensure patient safety.

[00553] In some embodiments, patients may be treated with a calpain inhibitor in combination with additional therapies such as anti -inflammatories including glucocorticoids, analgesics (e.g. ibuprofen), aspirin, and agents that modulate a Th2 -immune response, immunosuppressants including methotrexate, mycophenolate, cyclophosphamide, cyclosporine, thalidomide, pomalidomide, leflunomide, hydroxychloroquine, azathioprine, soluble bovine cartilage, vasodilators including endothelin receptor antagonists, prostacyclin analogues, nifedipine, and sildenafil, IL-6 receptor antagonists, selective and non-selective tyrosine kinase inhibitors, Wnt-pathway modulators, PPAR activators, caspase-3 inhibitors, LPA receptor antagonists, B cell depleting agents, CCR2 antagonists, pirfenidone, cannabinoid receptor agonists, ROCK inhibitors, miRNA-targeting agents, toil-like receptor antagonists, CTGF -targeting agents, NADPH oxidase inhibitors, tryptase inhibitors, TGF □ inhibitors, relaxin receptor agonists, and autologous adipose derived regenerative cells.

[00554] While some embodiments have been illustrated and described, a person with ordinary skill in the art, after reading the foregoing specification, can effect changes, substitutions of equivalents and other types of alterations to the compounds of the present technology or salts, pharmaceutical compositions, derivatives, prodrugs, metabolites, tautomers or racemic mixtures thereof as set forth herein. Each aspect and embodiment described above can also have included or incorporated therewith such variations or aspects as disclosed in regard to any or all of the other aspects and embodiments.

[00555] The present technology is also not to be limited in terms of the particular aspects described herein, which are intended as single illustrations of individual aspects of the present technology. Many modifications and variations of this present technology can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. Functionally equivalent methods within the scope of the present technology, in addition to those enumerated herein, will be apparent to those skilled in the art from the foregoing descriptions. Such modifications and variations are intended to fail within the scope of the appended claims. It is to be understood that this present technology is not limited to particular methods, reagents, compounds, compositions, labeled compounds or biological systems, which can, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular aspects only, and is not intended to be limiting. Thus, it is intended that the specification be considered as exemplar}' only with the breadth, scope and spirit of the present technology indicated only by the appended claims, definitions therein and any equivalents thereof.

[00556] The embodiments, illustratively described herein may suitably be practiced in the absence of any element or elements, limitation or limitations, not specifically disclosed herein. Thus, for example, the terms "comprising," "including," "containing," etc, shall be read expansively and without limitation. Additionally, the terms and expressions employed herein have been used as terms of description and not of limitation, and there is no intention in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the claimed technology. Additionally, the phrase "consisting essentially of will be understood to include those elements specifically recited and those additional elements that do not materially affect the basic and novel characteristics of the claimed technology. The phrase "consisting of excludes any element not specified.

[00557] In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that the disclosure is also thereby- described in terms of any individual member or subgroup of members of the Markush group. Each of the narrower species and subgeneric groupings falling within the generic disclosure also form part of the present technology. This includes the generic description of the present technology with a proviso or negative limitation removing any subject matter from the genus, regardless of whether or not the excised material is specifically recited herein.

[00558] All publications, patent applications, issued patents, and other documents (for example, journals, articles and/or textbooks) referred to in this specification are herein incorporated by reference as if each individual publication, patent application, issued patent, or other document was specifically and individually indicated to be incorporated by reference in its entirety. Definitions that are contained in text incorporated by reference are excluded to the extent that they contradict definitions in this disclosure.

[00559] Other embodiments are set forth in the following claims, along with the full scope of equivalents to which such claims are entitled.

[00560] While the invention has been particularly shown and described with reference to a preferred embodiment and various alternate embodiments, it will be understood by persons skilled in the relevant art that various changes in form and details can be made therein without departing from the spirit and scope of the invention.

[00561] All references, issued patents and patent applications cited within the body of the instant specification are hereby incorporated by reference in their entirety, for all purposes.

REFERENCES CITED

1. U.S. Patent No. 5, 145,684

2. Goll et al. (2003). "The calpain system." Physiol Rev 83(3):731-801.

3. Schad et al . (2002), "A novel human small subunit of calpains." Biochem J 362(Pt 2):383-8.

4. Ravulapalli et al. (2009). "Distinguishing between calpain heterodimerization and homodimerization." FEBS J 276(4):973-82.

5. Dourdiii et al . (2001), "Reduced cell migration and disruption of the actin cytoskeleton in calpain-deficient embryonic fibroblasts." J Biol Chem 276(51):48382-8.

6. Leloup et al. (2006). "Involvement of calpains in growth factor-mediated migration." Int J Biochem Cell Biol 38(12):2049-63.

7. Janossy et al . (2004), "Calpain as a multi-site regulator of cell cycle." Biochem Pharmacol 67(8): 1513-21.

8. Santos et al. (2012). "Distinct regulatory functions of calpain 1 and 2 during neural stem cell self-renewal and differentiation." PLoS One 7(3):e33468.

9. Miettinen et al. (1994). "TGF-beta induced transdifferentiation of mammary epithelial cells to mesenchymal cells: involvement of type I receptors." J Cell Biol 127(6 Pt 2):2021 -36,

10. Lamouille et al. (2014). "Molecular mechanisms of epithelial -mesenchymal transition " Nat Rev Mol Cell Biol 15(3): 178-96.

11. Pegorier et al. (2010). "Bone Morphogenetic Protein (BMP)-4 and BMP-7 regulate differentially Transforming Growth Factor (TGF)-Bl in normal human lung fibroblasts (NHLF)" Respir Res 11 :85.